1
|
Su J, Zhang J, Wu Y, Ni C, Ding Y, Cai Z, Xu M, Lai M, Wang J, Lin S, Lu J. Cabozantinib in combination with immune checkpoint inhibitors for renal cell carcinoma: a systematic review and meta-analysis. Front Pharmacol 2024; 15:1322473. [PMID: 38694912 PMCID: PMC11061414 DOI: 10.3389/fphar.2024.1322473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 04/04/2024] [Indexed: 05/04/2024] Open
Abstract
Context Cabozantinib combined with immune checkpoint inhibitors (ICIs) has brought a new therapeutic effect for the medical treatment of renal cell carcinoma (RCC). Objectives We performed a meta-analysis of randomized controlled trials and single-arm trials to evaluate the efficacy and safety of cabozantinib plus ICIs in RCC. Methods We extracted data from PubMed, Cochrane, Medline and Embase databases, and rated literature quality through Cochrane risk of bias tool and MINORS. RevMan5.3 software was used to analyze the results of randomized controlled trials and single-arm trials. Results A total of 7 studies were included. Treatment with cabozantinib plus ICIs improved PFS [HR 0.75, (95%CI: 0.52, 1.08), p = 0.12] and the OS [HR 0.80, (95%CI: 0.60, 1.07), p = 0.13] in randomized controlled trials. Meanwhile, the result of the ORR in randomized controlled trials was [risk ratio (RR) 1.37, (95%CI: 1.21, 1.54), p < 0.00001] and in single-arm trials was [risk difference (RD) 0.49, (95%CI: 0.26, 0.71), p < 0.0001]. Conclusion Cabozantinib plus ICIs prolonged the PFS and OS, and improved ORR in patients with RCC. Our recommendation is to use cabozantinib plus ICIs to treat advanced RCC, and to continuous monitor and manage the drug-related adverse events. Systematic Review Registration identifier CRD42023455878.
Collapse
Affiliation(s)
- Jingyang Su
- Department of Oncology, Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
- Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Jialin Zhang
- Department of Oncology, Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuqian Wu
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Cui Ni
- Department of Oncology, Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Yueyue Ding
- Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Zelin Cai
- Department of Oncology, Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Ming Xu
- Department of Traditional Chinese Medicine, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Mingyang Lai
- Department of Oncology, Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Jue Wang
- Department of Oncology, Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Shengyou Lin
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jinhua Lu
- Department of Oncology, Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
2
|
Kim GR, Kang JH, Kim HJ, Im E, Bae J, Kwon WS, Rha SY, Chung HC, Cho EY, Kim SY, Kim YC. Discovery of novel 1H-benzo[d]imidazole-4,7-dione based transglutaminase 2 inhibitors as p53 stabilizing anticancer agents in renal cell carcinoma. Bioorg Chem 2024; 143:107061. [PMID: 38154386 DOI: 10.1016/j.bioorg.2023.107061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/21/2023] [Accepted: 12/23/2023] [Indexed: 12/30/2023]
Abstract
Overexpression of transglutaminase 2 (TGase 2; TG2) has been implicated in the progression of renal cell carcinoma (RCC) through the inactivation of p53 by forming a protein complex. Because most p53 in RCC has no mutations, apoptosis can be increased by inhibiting the binding between TG2 and p53 to increase the stability of p53. In the present study, a novel TG2 inhibitor was discovered by investigating the structure of 1H-benzo[d]imidazole-4,7-dione as a simpler chemotype based on the amino-1,4-benzoquinone moiety of streptonigrin, a previously reported inhibitor. Through structure-activity relationship (SAR) studies, compound 8j (MD102) was discovered as a potent TG2 inhibitor with an IC50 value of 0.35 µM, p53 stabilization effect and anticancer effects in the ACHN and Caki-1 RCC cell lines with sulforhodamine B (SRB) GI50 values of 2.15 µM and 1.98 µM, respectively. The binding property of compound 8j (MD102) with TG2 was confirmed to be reversible in a competitive enzyme assay, and the binding interaction was expected to be formed at the β-sandwich domain, a p53 binding site, in the SPR binding assay with mutant proteins. The mode of binding of compound 8j (MD102) to the β-sandwich domain of TG2 was analyzed by molecular docking using the crystal structure of the active conformation of human TG2. Compound 8j (MD102) induced a decrease in the downstream signaling of p-AKT and p-mTOR through the stabilization of p53 by TG2 inhibition, resulting in tumor cell apoptosis. In a xenograft animal model using ACHN cancer cells, oral administration and intraperitoneal injection of compound 8j (MD102) showed an inhibitory effect on tumor growth, confirming increased levels of p53 and decreased levels of Ki-67 in tumor tissues through immunohistochemical (IHC) tissue staining. These results indicated that the inhibition of TG2 by compound 8j (MD102) could enhance p53 stabilization, thereby ultimately showing anticancer effects in RCC. Compound 8j (MD102), a novel TG2 inhibitor, can be further applied for the development of an anticancer candidate drug targeting RCC.
Collapse
Affiliation(s)
- Ga-Ram Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Joon Hee Kang
- Graduate School of Cancer Science and Policy, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang 10408, Republic of Korea
| | - Hyeon Joo Kim
- MDbiopharm Corp., 114 Beobwon-ro, Songpa-gu, Seoul 05854, Republic of Korea
| | - Eunji Im
- MDbiopharm Corp., 114 Beobwon-ro, Songpa-gu, Seoul 05854, Republic of Korea
| | - Jinsu Bae
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Woo Sun Kwon
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Sun Young Rha
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea; Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Hyun Cheol Chung
- MDbiopharm Corp., 114 Beobwon-ro, Songpa-gu, Seoul 05854, Republic of Korea; Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Eun Yi Cho
- MDbiopharm Corp., 114 Beobwon-ro, Songpa-gu, Seoul 05854, Republic of Korea.
| | - Soo-Youl Kim
- Cancer Molecular Biology Branch, Division of Cancer Biology, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang 10408, Republic of Korea.
| | - Yong-Chul Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Republic of Korea.
| |
Collapse
|
3
|
Santorsola M, Capuozzo M, Nasti G, Sabbatino F, Di Mauro A, Di Mauro G, Vanni G, Maiolino P, Correra M, Granata V, Gualillo O, Berretta M, Ottaiano A. Exploring the Spectrum of VEGF Inhibitors' Toxicities from Systemic to Intra-Vitreal Usage in Medical Practice. Cancers (Basel) 2024; 16:350. [PMID: 38254839 PMCID: PMC10813960 DOI: 10.3390/cancers16020350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
The use of Vascular Endothelial Growth Factor inhibitors (VEGFi) has become prevalent in the field of medicine, given the high incidence of various pathological conditions necessitating VEGF inhibition within the general population. These conditions encompass a range of advanced neoplasms, such as colorectal cancer, non-small cell lung cancer, renal cancer, ovarian cancer, and others, along with ocular diseases. The utilization of VEGFi is not without potential risks and adverse effects, requiring healthcare providers to be well-prepared for identification and management. VEGFi can be broadly categorized into two groups: antibodies or chimeric proteins that specifically target VEGF (bevacizumab, ramucirumab, aflibercept, ranibizumab, and brolucizumab) and non-selective and selective small molecules (sunitinib, sorafenib, cabozantinib, lenvatinib, regorafenib, etc.) designed to impede intracellular signaling of the VEGF receptor (RTKi, receptor tyrosine kinase inhibitors). The presentation and mechanisms of adverse effects resulting from VEGFi depend primarily on this distinction and the route of drug administration (systemic or intra-vitreal). This review provides a thorough examination of the causes, recognition, management, and preventive strategies for VEGFi toxicities with the goal of offering support to oncologists in both clinical practice and the design of clinical trials.
Collapse
Affiliation(s)
- Mariachiara Santorsola
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, via M. Semmola, 80131 Naples, Italy; (M.S.); (G.N.); (A.D.M.); (P.M.); (M.C.); (V.G.)
| | | | - Guglielmo Nasti
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, via M. Semmola, 80131 Naples, Italy; (M.S.); (G.N.); (A.D.M.); (P.M.); (M.C.); (V.G.)
| | - Francesco Sabbatino
- Oncology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Salerno, Italy;
| | - Annabella Di Mauro
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, via M. Semmola, 80131 Naples, Italy; (M.S.); (G.N.); (A.D.M.); (P.M.); (M.C.); (V.G.)
| | - Giordana Di Mauro
- Department of Human Pathology “G. Barresi”, University of Messina, 98125 Messina, Italy;
| | - Gianluca Vanni
- Breast Unit, Department of Surgical Science, PTV Policlinico Tor Vergata University, 00133 Rome, Italy;
| | - Piera Maiolino
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, via M. Semmola, 80131 Naples, Italy; (M.S.); (G.N.); (A.D.M.); (P.M.); (M.C.); (V.G.)
| | - Marco Correra
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, via M. Semmola, 80131 Naples, Italy; (M.S.); (G.N.); (A.D.M.); (P.M.); (M.C.); (V.G.)
| | - Vincenza Granata
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, via M. Semmola, 80131 Naples, Italy; (M.S.); (G.N.); (A.D.M.); (P.M.); (M.C.); (V.G.)
| | - Oreste Gualillo
- SERGAS (Servizo Galego de Saude), NEIRID Laboratory (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), IDIS (Instituto de Investigación Sanitaria de Santiago), Research Laboratory 9, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain;
| | - Massimiliano Berretta
- Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Alessandro Ottaiano
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, via M. Semmola, 80131 Naples, Italy; (M.S.); (G.N.); (A.D.M.); (P.M.); (M.C.); (V.G.)
| |
Collapse
|
4
|
Silvestris N, Franchina T, Gallo M, Argentiero A, Avogaro A, Cirino G, Colao A, Danesi R, Di Cianni G, D'Oronzo S, Faggiano A, Fogli S, Giuffrida D, Gori S, Marrano N, Mazzilli R, Monami M, Montagnani M, Morviducci L, Natalicchio A, Ragni A, Renzelli V, Russo A, Sciacca L, Tuveri E, Zatelli MC, Giorgino F, Cinieri S. Diabetes management in cancer patients. An Italian Association of Medical Oncology, Italian Association of Medical Diabetologists, Italian Society of Diabetology, Italian Society of Endocrinology and Italian Society of Pharmacology multidisciplinary consensus position paper. ESMO Open 2023; 8:102062. [PMID: 38070434 PMCID: PMC10714217 DOI: 10.1016/j.esmoop.2023.102062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/07/2023] [Accepted: 10/11/2023] [Indexed: 12/31/2023] Open
Abstract
Cancer management has significantly evolved in recent years, focusing on a multidisciplinary team approach to provide the best possible patient care and address the various comorbidities, toxicities, and complications that may arise during the patient's treatment journey. The co-occurrence of diabetes and cancer presents a significant challenge for health care professionals worldwide. Management of these conditions requires a holistic approach to improve patients' overall health, treatment outcomes, and quality of life, preventing diabetes complications and cancer treatment side-effects. In this article, a multidisciplinary panel of experts from different Italian scientific societies provide a critical overview of the co-management of cancer and diabetes, with an increasing focus on identifying a novel specialty field, 'diabeto-oncology', and suggest new co-management models of cancer patients with diabetes to improve their care. To better support cancer patients with diabetes and ensure high levels of coordinated care between oncologists and diabetologists, 'diabeto-oncology' could represent a new specialized field that combines specific expertise, skills, and training.
Collapse
Affiliation(s)
- N Silvestris
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina.
| | - T Franchina
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina
| | - M Gallo
- Endocrinology and Metabolic Diseases Unit, AO SS Antonio e Biagio e Cesare Arrigo of Alessandria, Alessandria
| | - A Argentiero
- Medical Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari
| | - A Avogaro
- Department of Medicine, University of Padova, Padua
| | - G Cirino
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples
| | - A Colao
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples; UNESCO Chair, Education for Health and Sustainable Development, Federico II University, Naples
| | - R Danesi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa
| | | | - S D'Oronzo
- Interdisciplinary Department of Medicine, University of Bari Aldo Moro, Bari
| | - A Faggiano
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, ENETS Center of Excellence, Sapienza University of Rome, Rome
| | - S Fogli
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa
| | - D Giuffrida
- Department of Oncology, Istituto Oncologico del Mediterraneo, Viagrande, Catania
| | - S Gori
- Oncologia Medica, IRCCS Ospedale Don Calabria-Sacro Cuore di Negrar, Verona
| | - N Marrano
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari
| | - R Mazzilli
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, ENETS Center of Excellence, Sapienza University of Rome, Rome
| | - M Monami
- Diabetology, Careggi Hospital and University of Florence, Firenze
| | - M Montagnani
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Pharmacology, Medical School, University of Bari Aldo Moro, Bari
| | - L Morviducci
- Diabetology and Nutrition Unit, Department of Medical Specialties, ASL Roma 1 - S, Spirito Hospital, Rome
| | - A Natalicchio
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari
| | - A Ragni
- Endocrinology and Metabolic Diseases Unit, AO SS Antonio e Biagio e Cesare Arrigo of Alessandria, Alessandria
| | - V Renzelli
- Diabetologist and Endocrinologist, Italian Association of Medical Diabetologists, Rome
| | - A Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo
| | - L Sciacca
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania, Catania
| | - E Tuveri
- Diabetology, Endocrinology and Metabolic Diseases Service, ASL-Sulcis, Carbonia
| | - M C Zatelli
- Section of Endocrinology, Geriatrics, and Internal Medicine, Department of Medical Sciences, University of Ferrara, Ferrara
| | - F Giorgino
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari
| | - S Cinieri
- Medical Oncology Division and Breast Unit, Senatore Antonio Perrino Hospital, ASL Brindisi, Brindisi, Italy
| |
Collapse
|
5
|
Du X, Zhao Z, Zhao X, Wang H, Jiang L, Tang W. Risk signature identification and NPRL2 affects sunitinib sensitivity in clear cell renal cell carcinoma. Biochem Biophys Res Commun 2023; 663:122-131. [PMID: 37121122 DOI: 10.1016/j.bbrc.2023.04.063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/09/2023] [Accepted: 04/19/2023] [Indexed: 05/02/2023]
Abstract
Tumor suppressor genes (TSGs) play a crucial role in tumorigenesis and drug resistance. We analyzed the subtypes of clear cell renal cell carcinoma (ccRCC) mediated by 8 genes contained in the 3p21.3 tumor suppressor gene cluster and their effects on TME cell infiltration based on the TCGA database. The risk score model was established by principal component analysis. The hub gene NPRL2 was selected by protein-protein interactions (PPI) analysis. The effect of NPRL2 on sunitinib sensitivity of ccRCC was verified by using CCK-8, colony formation assay, wound healing assay, transwell assay and xenograft tumor model. Changes in protein expression were detected by Western blotting. We found that 8 TSGs were all differentially expressed in ccRCC samples, which could divide ccRCC into two subtypes. The constructed risk score model could predict the prognosis and drug sensitivity of ccRCC patients, and was an independent prognostic factor for ccRCC. Over-expression of NPRL2 promoted apoptosis, inhibited EMT, decreased the phosphorylation of the PI3K/AKT/mTOR signaling pathway to inhibit its activity, and promoted the sensitivity of sunitinib to ccRCC cells. Collectively, our findings increased the understanding of TSGs in ccRCC, suggesting that NPRL2 as a TSG could enhance sunitinib sensitivity to ccRCC cells.
Collapse
Affiliation(s)
- Xiaoyi Du
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhipeng Zhao
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xin Zhao
- Department of Urology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Hexi Wang
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Jiang
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Wei Tang
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
6
|
Liu Q, Li S, Qiu Y, Zhang J, Rios FJ, Zou Z, Touyz RM. Cardiovascular toxicity of tyrosine kinase inhibitors during cancer treatment: Potential involvement of TRPM7. Front Cardiovasc Med 2023; 10:1002438. [PMID: 36818331 PMCID: PMC9936099 DOI: 10.3389/fcvm.2023.1002438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 01/18/2023] [Indexed: 02/05/2023] Open
Abstract
Receptor tyrosine kinases (RTKs) are a class of membrane spanning cell-surface receptors that transmit extracellular signals through the membrane to trigger diverse intracellular signaling through tyrosine kinases (TKs), and play important role in cancer development. Therapeutic approaches targeting RTKs such as vascular endothelial growth factor receptor (VEGFR), epidermal growth factor receptor (EGFR), and platelet-derived growth factor receptor (PDGFR), and TKs, such as c-Src, ABL, JAK, are widely used to treat human cancers. Despite favorable benefits in cancer treatment that prolong survival, these tyrosine kinase inhibitors (TKIs) and monoclonal antibodies targeting RTKs are also accompanied by adverse effects, including cardiovascular toxicity. Mechanisms underlying TKI-induced cardiovascular toxicity remain unclear. The transient receptor potential melastatin-subfamily member 7 (TRPM7) is a ubiquitously expressed chanzyme consisting of a membrane-based ion channel and intracellular α-kinase. TRPM7 is a cation channel that regulates transmembrane Mg2+ and Ca2+ and is involved in a variety of (patho)physiological processes in the cardiovascular system, contributing to hypertension, cardiac fibrosis, inflammation, and atrial arrhythmias. Of importance, we and others demonstrated significant cross-talk between TRPM7, RTKs, and TK signaling in different cell types including vascular smooth muscle cells (VSMCs), which might be a link between TKIs and their cardiovascular effects. In this review, we summarize the implications of RTK inhibitors (RTKIs) and TKIs in cardiovascular toxicities during anti-cancer treatment, with a focus on the potential role of TRPM7/Mg2+ as a mediator of RTKI/TKI-induced cardiovascular toxicity. We also describe the important role of TRPM7 in cancer development and cardiovascular diseases, and the interaction between TRPM7 and RTKs, providing insights for possible mechanisms underlying cardiovascular disease in cancer patients treated with RTKI/TKIs.
Collapse
Affiliation(s)
- Qing Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Suyao Li
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuran Qiu
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiayu Zhang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Francisco J. Rios
- Research Institute of McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Zhiguo Zou
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,Zhiguo Zou ✉
| | - Rhian M. Touyz
- Research Institute of McGill University Health Centre, McGill University, Montreal, QC, Canada,*Correspondence: Rhian M. Touyz ✉
| |
Collapse
|
7
|
Hong SH, Chung HS, Seo IY, Kwon TG, Jeong H, Chung JI, Jeon SH, Park JY, Ha HK, Chung BH, Song W, Kim YJ, Kim SH, Lee JS, Lee J, Chung J. Patients' self-management of adverse events and patient-reported outcomes in advanced renal cell carcinoma treated with targeted therapies: A prospective, longitudinal, observational study. J Patient Rep Outcomes 2022; 6:125. [PMID: 36525150 PMCID: PMC9758263 DOI: 10.1186/s41687-022-00532-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Early intervention to reduce the impact of adverse events (AEs) may improve patients' quality of life and enable optimal treatment duration. METHODS This nationwide, multicenter, prospective, longitudinal, 1-year observational study investigated patients' self-management of AEs associated with targeted therapy for advanced renal cell carcinoma (RCC) and explored corresponding outcomes, including treatment duration and patient-reported outcomes (PROs). RESULTS We enrolled 77 advanced RCC patients (mean age 62 years) treated with a first targeted therapy. 210 cases of seven AEs of interest (fatigue, hand-foot syndrome, oral mucosal inflammation, diarrhea, gastrointestinal symptoms, hypertension, and anorexia) were observed. Most AEs were mild to moderate. Overall, 63.4% of patients were identified as managing their AEs well, reporting numerically longer treatment duration and significantly higher PRO scores than patients identified as poor managers. CONCLUSIONS Longer treatment duration and improved PROs were observed when advanced RCC patients managed targeted therapy-associated AEs well. Repeated education for consolidating AE self-management could be considered to enhance overall treatment outcomes.
Collapse
Affiliation(s)
- Sung-Hoo Hong
- Department of Urology, The Catholic University of Korea Seoul St. Mary's Hospital at Seocho-gu, Seoul, Republic of Korea
| | - Ho Seok Chung
- Department of Urology, Chonnam National University Hwasun Hospital at Hwasun-gun, Jeollanam-do, Republic of Korea
| | - Ill-Young Seo
- Department of Urology, Wonkwang University Hospital at Iksan, Jeonlabuk-do, Republic of Korea
| | - Tae Gyun Kwon
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Hyeon Jeong
- Department of Urology, SMG-SNU Boramae Medical Center at Dongjak-gu, Seoul, Republic of Korea
| | - Jae-Il Chung
- Department of Urology, Inje University Busan Paik Hospital, Busanjin-gu, Busan, Republic of Korea
| | - Seung Hyun Jeon
- Department of Urology, Kyung Hee University School of Medicine at Dongdaemun-gu, Seoul, Republic of Korea
| | - Jae Young Park
- Department of Urology, Korea University Ansan Hospital at Ansan-si, Danwon-gu, Gyeonggi-do, Republic of Korea
| | - Hong Koo Ha
- Department of Urology, Pusan National University Hospital at Seo-gu, Busan, Republic of Korea
| | - Byung-Ha Chung
- Department of Urology, Gangnam Severance Hospital, Yonsei University Health System at Gangnam-gu, Seoul, Republic of Korea
| | - Wan Song
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Young-Joo Kim
- Medical Division, Pfizer Biopharmaceuticals Group, Pfizer Pharmaceuticals Korea Limited, Seoul, Republic of Korea
| | - Sang-Hee Kim
- Medical Division, Pfizer Biopharmaceuticals Group, Pfizer Pharmaceuticals Korea Limited, Seoul, Republic of Korea
| | - Jee-Sun Lee
- Medical Division, Pfizer Biopharmaceuticals Group, Pfizer Pharmaceuticals Korea Limited, Seoul, Republic of Korea
| | - Juneyoung Lee
- Department of Biostatistics, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Jinsoo Chung
- Center for Urologic Cancer, National Cancer Center, Goyang, Republic of Korea.
| |
Collapse
|
8
|
Kastrati K, Mathies V, Kipp AP, Huebner J. Patient-reported experiences with side effects of kidney cancer therapies and corresponding information flow. J Patient Rep Outcomes 2022; 6:126. [PMID: 36525162 PMCID: PMC9758261 DOI: 10.1186/s41687-022-00533-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Treatment options for metastatic renal cell carcinoma (mRCC) have improved over recent years. Various therapies for metastatic renal cell carcinoma are currently approved for first and successive lines. Having various treatment options makes it important to reflect how patients experience side effects in the real-world setting. So far, data on the side effects of these treatments have only been collected within clinical trials, and have been mostly assessed by the investigator and not as patient-reported outcomes. Our aim was to determine patient-reported experiences of side effects in the real-world setting and to evaluate the doctor-patient communication regarding side effects. Data were collected via an anonymous, voluntary online survey given to members of a support group for RCC; the questionnaire was completed by 104 mRCC patients. RESULTS 89.1% of participants were suffering from side effects of any grade. These appeared to be higher for patients treated with tyrosine kinase inhibitors compared to those treated with immune-checkpoint inhibitors (98.4% vs. 68.4%). However, information on side effects is scarce: 4.0% had never heard anything about them while only 18.8% of participants received detailed information on possible side effects. Although 85.6% of participants reported side effects to their physician, 34.6% did not encounter an improvement. Limitations of the study include the design as an online questionnaire and the small sample, consisting only of members of a support group. CONCLUSIONS Differences can be seen between patient-reported side effects within our survey and those based on clinical trials. A shift towards more patient-reported outcomes is needed. In addition, patients seeking the advice of their physician on side effects are in need of more-or better-information and support.
Collapse
Affiliation(s)
- Karin Kastrati
- grid.275559.90000 0000 8517 6224Klinik Für Innere Medizin II, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Viktoria Mathies
- grid.275559.90000 0000 8517 6224UniversitätsTumorCentrum Jena, University Hospital Jena, Jena, Germany
| | - Anna P. Kipp
- grid.9613.d0000 0001 1939 2794Department of Nutritional Physiology, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena, Germany
| | - Jutta Huebner
- grid.275559.90000 0000 8517 6224Klinik Für Innere Medizin II, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany
| |
Collapse
|
9
|
Mehta A. Tracking the Development of Cancer Care After 75 Years of Independence: India's Fight Against Cancer Since 1947. Indian J Surg Oncol 2022; 13:12-26. [PMID: 36691502 PMCID: PMC9859970 DOI: 10.1007/s13193-022-01689-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 12/12/2022] [Indexed: 01/13/2023] Open
Abstract
India is one of the fastest developing countries with tremendous growth in industrialization and healthcare facilities. Research and development in the field of healthcare improved the quality of life and well-being of our population. Despite the availability of healthcare facilities and infrastructure, we are still facing considerable challenges in the prevention, diagnosis, and treatment of cancer. The present review focuses on the history and development of cancer care facilities since independence. The advances in cancer diagnostics for early detection of cancer and developments in the field of conventional surgery, including laparoscopic and robotic surgeries, chemotherapy, and radiation therapy, are reviewed. Immunotherapy, vaccines, and selective targeting of tumor cells using nanotechnology are emerging areas in the field of cancer research.
Collapse
Affiliation(s)
- Ashok Mehta
- Nanavati Max Super Speciality Hospital, Mumbai, India
- L S Raheja Hospital, Mumbai, India
- HCG Cancer Centre Colaba, Mumbai, India
| |
Collapse
|
10
|
Paillasse MR, Esquerré M, Bertrand FA, Poussereau-Pomié C, Pichery M, Visentin V, Gueguen-Dorbes G, Gaujarengues F, Barron P, Badet G, Briaux A, Ancey PB, Sibrac D, Erdociain E, Özcelik D, Meneyrol J, Martin V, Gomez-Brouchet A, Selves J, Rochaix P, Battistella M, Lebbé C, Delord JP, Dol-Gleizes F, Bono F, Blanc I, Alam A, Hunneyball I, Whittaker M, Fons P. Targeting Tumor Angiogenesis with the Selective VEGFR-3 Inhibitor EVT801 in Combination with Cancer Immunotherapy. CANCER RESEARCH COMMUNICATIONS 2022; 2:1504-1519. [PMID: 36970050 PMCID: PMC10035370 DOI: 10.1158/2767-9764.crc-22-0151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/13/2022] [Accepted: 11/02/2022] [Indexed: 11/30/2022]
Abstract
The receptor tyrosine kinase VEGFR-3 plays a crucial role in cancer-induced angiogenesis and lymphangiogenesis, promoting tumor development and metastasis. Here, we report the novel VEGFR-3 inhibitor EVT801 that presents a more selective and less toxic profile than two major inhibitors of VEGFRs (i.e., sorafenib and pazopanib). As monotherapy, EVT801 showed a potent antitumor effect in VEGFR-3–positive tumors, and in tumors with VEGFR-3–positive microenvironments. EVT801 suppressed VEGF-C–induced human endothelial cell proliferation in vitro and tumor (lymph)angiogenesis in different tumor mouse models. In addition to reduced tumor growth, EVT801 decreased tumor hypoxia, favored sustained tumor blood vessel homogenization (i.e., leaving fewer and overall larger vessels), and reduced important immunosuppressive cytokines (CCL4, CCL5) and myeloid-derived suppressor cells (MDSC) in circulation. Furthermore, in carcinoma mouse models, the combination of EVT801 with immune checkpoint therapy (ICT) yielded superior outcomes to either single treatment. Moreover, tumor growth inhibition was inversely correlated with levels of CCL4, CCL5, and MDSCs after treatment with EVT801, either alone or combined with ICT. Taken together, EVT801 represents a promising anti(lymph)angiogenic drug for improving ICT response rates in patients with VEGFR-3 positive tumors.
Significance:
The VEGFR-3 inhibitor EVT801 demonstrates superior selectivity and toxicity profile than other VEGFR-3 tyrosine kinase inhibitors. EVT801 showed potent antitumor effects in VEGFR-3–positive tumors, and tumors with VEGFR-3–positive microenvironments through blood vessel homogenization, and reduction of tumor hypoxia and limited immunosuppression. EVT801 increases immune checkpoint inhibitors’ antitumor effects.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Gaelle Badet
- 1Evotec France, Campus Curie, Toulouse CEDEX, France
| | - Anne Briaux
- 1Evotec France, Campus Curie, Toulouse CEDEX, France
| | | | - David Sibrac
- 1Evotec France, Campus Curie, Toulouse CEDEX, France
| | | | | | | | | | - Anne Gomez-Brouchet
- 4Institut Universitaire du Cancer Toulouse Oncopole (IUCT-O), Toulouse, Occitanie, France
| | - Janik Selves
- 4Institut Universitaire du Cancer Toulouse Oncopole (IUCT-O), Toulouse, Occitanie, France
| | - Philippe Rochaix
- 4Institut Universitaire du Cancer Toulouse Oncopole (IUCT-O), Toulouse, Occitanie, France
| | - Maxime Battistella
- 5Université de Paris, Department of Pathology, AP-HP Hôpital Saint Louis, INSERM U976, Paris, France
| | - Céleste Lebbé
- 6Université de Paris, Department of Dermatology, AP-HP Hôpital Saint Louis, INSERM U976, Paris, France
| | - Jean-Pierre Delord
- 4Institut Universitaire du Cancer Toulouse Oncopole (IUCT-O), Toulouse, Occitanie, France
| | | | | | | | | | | | | | - Pierre Fons
- 1Evotec France, Campus Curie, Toulouse CEDEX, France
| |
Collapse
|
11
|
Assaf L, Eid AA, Nassif J. Role of AMPK/mTOR, mitochondria, and ROS in the pathogenesis of endometriosis. Life Sci 2022; 306:120805. [PMID: 35850246 DOI: 10.1016/j.lfs.2022.120805] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 06/27/2022] [Accepted: 07/10/2022] [Indexed: 12/19/2022]
Abstract
Endometriosis is the presence of endometrial tissue outside the uterine cavity usually in the ovaries, fallopian tube, and pelvic cavity. It's a chronic enigmatic gynecological condition associated with dysmenorrhea, dyspareunia, pelvic pain, and infertility. Endometriosis lesions exist in a unique microenvironment characterized by increased concentrations of hormones, inflammation, and oxidative stress. This environment promotes cell survival through the binding of membrane receptors and subsequent cascading activation of intracellular kinases that stimulate a cellular response. In endometriosis, well-established signaling pathways, mTOR and AMPK, are altered via steroid hormones and other factors to promote cell growth, migration, and proliferation. This is accompanied by dysfunction in the mitochondria that increase energy production to sustain proliferation demands consequently leading to reactive oxygen species overproduction. This review aims to summarize the role of altered mTOR/AMPK signaling pathway, mitochondrial dysfunction, and reactive oxygen species overproduction along with providing therapeutic and diagnostic approaches. Highlighting these factors would provide a better understanding to reach a coherent theory for the pathogenesis of endometriosis.
Collapse
Affiliation(s)
- Lama Assaf
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon; AUB Diabetes, American University of Beirut, Beirut, Lebanon
| | - Assaad A Eid
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon; AUB Diabetes, American University of Beirut, Beirut, Lebanon.
| | - Joseph Nassif
- Division of Minimally Invasive Gynecology, Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
12
|
Lifton J, Chow A, Moshfeghi AA. FAILED LASER RETINOPEXY IN A PATIENT UNDERGOING PAZOPANIB THERAPY. Retin Cases Brief Rep 2021; 15:752-755. [PMID: 31205141 DOI: 10.1097/icb.0000000000000877] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE To report a case of failed endolaser retinopexy in patient taking pazopanib and possible association with impaired retinal remodeling. METHODS Case report. RESULTS A 68-year-old man with a history of radiation-induced high-grade metastatic mediastinal leiomyosarcoma, on pazopanib 600 mg daily, presented with a superior retinal tear 2 months after initiating pazopanib. The patient was treated with pars plana vitrectomy and endolaser retinopexy without any immediate complications. One month postoperatively, the patient was found to have a new superior macula-sparing rhegmatogenous retinal detachment extending through the laser barricade nasally. A large proportion of the laser lesions in this area had notably remained chalk-white. The patient underwent pars plana vitrectomy with scleral buckling, and repeat endolaser retinopexy. Pazopanib therapy was stopped 1 week later in consultation with the oncology team and was reinitiated after the retina seemed stable under silicone oil for several weeks. The scars from the patient's repeat endolaser retinopexy remained pale white at his clinic visit 2 months after the procedure, and at that time, his initial laser scars had only just begun to darken with pigment. Four and a half months after the initial vitrectomy and endolaser retinopexy, and three and a half months after his repeat vitrectomy and endolaser retinopexy, all the patient's laser scars have developed increased pigment, and his retina remains completely flat under silicone oil tamponade. CONCLUSION Pazopanib may have had an adverse effect on retinal wound healing after vitrectomy with endolaser retinopexy, leading to persistence of pale laser lesions many weeks after laser application. It is possible that this may have impaired the typical development of chorioretinal laser scar formation and led to subsequent retinal detachment.
Collapse
Affiliation(s)
- Jacob Lifton
- Department of Ophthalmology, USC Roski Eye Institute, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | | | | |
Collapse
|
13
|
Continued versus Interrupted Targeted Therapy during Metastasis-Directed Stereotactic Radiotherapy: A Retrospective Multi-Center Safety and Efficacy Analysis. Cancers (Basel) 2021; 13:cancers13194780. [PMID: 34638265 PMCID: PMC8507709 DOI: 10.3390/cancers13194780] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/20/2021] [Accepted: 08/30/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary The increasing use of targeted therapy (TT) has resulted in prolonged disease control and survival in many metastatic cancers. In parallel, stereotactic radiotherapy (SRT) is increasingly performed in patients receiving TT to obtain a durable control of resistant metastases, and thereby to prolong the time to disseminated disease progression and switch of systemic therapy. The aim of this study was to analyze the safety and efficacy of SRT combined with TT in metastatic cancer patients and to assess the influence of continuous vs. interrupted TT during metastasis-directed SRT. The data of 454 SRTs in 158 patients from the international multicenter database (TOaSTT) on metastatic cancer patients treated with SRT and concurrent TT (within 30 days) were analyzed. We found that there was no significant difference in survival, progression, or severe toxicity, whether TT was interrupted during SRT or not. Although any-grade acute and late SRT-related toxicity occurred in 63 (40%) and 52 (33%) patients, severe SRT-related toxicity rates were low (3% and 4%, respectively). The highest toxicity rates were observed for the combination of SRT and EGFRi or BRAF/MEKi, and any grade of toxicity was significantly increased when EGFRi or BRAF/MEKi were continued during SRT. However, this did not account for severe toxicity. Abstract The increasing use of targeted therapy (TT) has resulted in prolonged disease control and survival in many metastatic cancers. In parallel, stereotactic radiotherapy (SRT) is increasingly performed in patients receiving TT to obtain a durable control of resistant metastases, and thereby to prolong the time to disseminated disease progression and switch of systemic therapy. The aims of this study were to analyze the safety and efficacy of SRT combined with TT in metastatic cancer patients and to assess the influence of continuous vs. interrupted TT during metastasis-directed SRT. The data of 454 SRTs in 158 patients from the international multicenter database (TOaSTT) on metastatic cancer patients treated with SRT and concurrent TT (within 30 days) were analyzed using Kaplan–Meier and log rank testing. Toxicity was defined by the CTCAE v4.03 criteria. The median FU was 19.9 mo (range 1–102 mo); 1y OS, PFS and LC were 59%, 24% and 84%, respectively. Median TTS was 25.5 mo (95% CI 11–40). TT was started before SRT in 77% of patients. TT was interrupted during SRT in 44% of patients, with a median interruption of 7 (range 1–42) days. There was no significant difference in OS or PFS whether TT was temporarily interrupted during SRT or not. Any-grade acute and late SRT-related toxicity occurred in 63 (40%) and 52 (33%) patients, respectively. The highest toxicity rates were observed for the combination of SRT and EGFRi or BRAF/MEKi, and any-grade toxicity was significantly increased when EGFRi (p = 0.016) or BRAF/MEKi (p = 0.009) were continued during SRT. Severe (≥grade 3) acute and late SRT-related toxicity were observed in 5 (3%) and 7 (4%) patients, respectively, most frequently in patients treated with EGFRi or BRAF/MEKi and in the intracranial cohort. There was no significant difference in severe toxicity whether TT was interrupted before and after SRT or not. In conclusion, SRT and continuous vs. interrupted TT in metastatic cancer patients did not influence OS or PFS. Overall, severe toxicity of combined treatment was rare; a potentially increased toxicity after SRT and continuous treatment with EGFR inhibitors or BRAF(±MEK) inhibitors requires further evaluation.
Collapse
|
14
|
Demlová R, Turjap M, Peš O, Kostolanská K, Juřica J. Therapeutic Drug Monitoring of Sunitinib in Gastrointestinal Stromal Tumors and Metastatic Renal Cell Carcinoma in Adults-A Review. Ther Drug Monit 2021; 42:20-32. [PMID: 31259881 DOI: 10.1097/ftd.0000000000000663] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Sunitinib is an inhibitor of multiple receptor tyrosine kinases and is a standard-of-care treatment for advanced and metastatic renal cell carcinoma and a second-line treatment in locally advanced inoperable and metastatic gastrointestinal stromal tumors. A fixed dose of the drug, however, does not produce a uniform therapeutic outcome in all patients, and many face adverse effects and/or toxicity. One of the possible causes of the interindividual variability in the efficacy and toxicity response is the highly variable systemic exposure to sunitinib and its active metabolite. This review aims to summarize all available clinical evidence of the treatment of adult patients using sunitinib in approved indications, addressing the necessity to introduce proper and robust therapeutic drug monitoring (TDM) of sunitinib and its major metabolite, N-desethylsunitinib. METHODS The authors performed a systematic search of the available scientific literature using the PubMed online database. The search terms were "sunitinib" AND "therapeutic drug monitoring" OR "TDM" OR "plasma levels" OR "concentration" OR "exposure." The search yielded 520 journal articles. In total, 447 publications were excluded because they lacked sufficient relevance to the reviewed topic. The remaining 73 articles were, together with currently valid guidelines, thoroughly reviewed. RESULTS There is sufficient evidence confirming the concentration-efficacy and concentration-toxicity relationship in the indications of gastrointestinal stromal tumors and metastatic renal clear-cell carcinoma. For optimal therapeutic response, total (sunitinib + N-desethylsunitinib) trough levels of 50-100 ng/mL serve as a reasonable target therapeutic range. To avoid toxicity, the total trough levels should not exceed 100 ng/mL. CONCLUSIONS According to the current evidence presented in this review, a TDM-guided dose modification of sunitinib in selected groups of patients could provide a better treatment outcome while simultaneously preventing sunitinib toxicity.
Collapse
Affiliation(s)
- Regina Demlová
- Department of Pharmacology, Medical Faculty, Masaryk University, Brno
| | - Miroslav Turjap
- Department of Clinical Pharmacy, University Hospital Ostrava, Ostrava
| | - Ondřej Peš
- Department of Biochemistry, Medical Faculty, Masaryk University
| | | | - Jan Juřica
- Department of Pharmacology, Medical Faculty, Masaryk University, Masaryk Memorial Cancer Institute; and.,Department of Human Pharmacology and Toxicology, University of Veterinary and Pharmaceutical Sciences, Brno, Czech Republic
| |
Collapse
|
15
|
Dufies M, Verbiest A, Cooley LS, Ndiaye PD, He X, Nottet N, Souleyreau W, Hagege A, Torrino S, Parola J, Giuliano S, Borchiellini D, Schiappa R, Mograbi B, Zucman-Rossi J, Bensalah K, Ravaud A, Auberger P, Bikfalvi A, Chamorey E, Rioux-Leclercq N, Mazure NM, Beuselinck B, Cao Y, Bernhard JC, Ambrosetti D, Pagès G. Plk1, upregulated by HIF-2, mediates metastasis and drug resistance of clear cell renal cell carcinoma. Commun Biol 2021; 4:166. [PMID: 33547392 PMCID: PMC7865059 DOI: 10.1038/s42003-021-01653-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 11/20/2020] [Indexed: 11/13/2022] Open
Abstract
Polo-like kinase 1 (Plk1) expression is inversely correlated with survival advantages in many cancers. However, molecular mechanisms that underlie Plk1 expression are poorly understood. Here, we uncover a hypoxia-regulated mechanism of Plk1-mediated cancer metastasis and drug resistance. We demonstrated that a HIF-2-dependent regulatory pathway drives Plk1 expression in clear cell renal cell carcinoma (ccRCC). Mechanistically, HIF-2 transcriptionally targets the hypoxia response element of the Plk1 promoter. In ccRCC patients, high expression of Plk1 was correlated to poor disease-free survival and overall survival. Loss-of-function of Plk1 in vivo markedly attenuated ccRCC growth and metastasis. High Plk1 expression conferred a resistant phenotype of ccRCC to targeted therapeutics such as sunitinib, in vitro, in vivo, and in metastatic ccRCC patients. Importantly, high Plk1 expression was defined in a subpopulation of ccRCC patients that are refractory to current therapies. Hence, we propose a therapeutic paradigm for improving outcomes of ccRCC patients.
Collapse
Affiliation(s)
- Maeva Dufies
- Centre Scientifique de Monaco, Biomedical Department, 8 quai Antoine Premier, 98 000, Monaco, Monaco.
- LIA ROPSE, Laboratoire International Associé Université Côte d'Azur - Centre Scientifique de Monaco, Nice, France.
| | - Annelies Verbiest
- Department of General Medical Oncology, University Hospitals Leuven, 3000, Leuven, Belgium
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, 3000, Leuven, Belgium
| | | | - Papa Diogop Ndiaye
- LIA ROPSE, Laboratoire International Associé Université Côte d'Azur - Centre Scientifique de Monaco, Nice, France
- University Côte d'Azur, Institute for Research on Cancer and Aging of Nice (IRCAN), CNRS UMR 7284; INSERM U1081, Centre Antoine Lacassagne, 06189, Nice, France
| | - Xingkang He
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | - Nicolas Nottet
- University Côte d'Azur, C3M, Inserm U1065, 06204, Nice, France
| | | | - Anais Hagege
- LIA ROPSE, Laboratoire International Associé Université Côte d'Azur - Centre Scientifique de Monaco, Nice, France
- University Côte d'Azur, Institute for Research on Cancer and Aging of Nice (IRCAN), CNRS UMR 7284; INSERM U1081, Centre Antoine Lacassagne, 06189, Nice, France
| | - Stephanie Torrino
- University Côte d'Azur, Institut de Pharmacologie Cellulaire et Moléculaire, CNRS UMR7275, 06560, Valbonne, France
| | - Julien Parola
- LIA ROPSE, Laboratoire International Associé Université Côte d'Azur - Centre Scientifique de Monaco, Nice, France
- University Côte d'Azur, Institute for Research on Cancer and Aging of Nice (IRCAN), CNRS UMR 7284; INSERM U1081, Centre Antoine Lacassagne, 06189, Nice, France
- Centre Antoine Lacassagne, 06189, Nice, France
| | - Sandy Giuliano
- LIA ROPSE, Laboratoire International Associé Université Côte d'Azur - Centre Scientifique de Monaco, Nice, France
- University Côte d'Azur, Institute for Research on Cancer and Aging of Nice (IRCAN), CNRS UMR 7284; INSERM U1081, Centre Antoine Lacassagne, 06189, Nice, France
| | | | | | - Baharia Mograbi
- University Côte d'Azur, Institute for Research on Cancer and Aging of Nice (IRCAN), CNRS UMR 7284; INSERM U1081, Centre Antoine Lacassagne, 06189, Nice, France
| | - Jessica Zucman-Rossi
- Inserm, UMR-1138, Génomique fonctionnelle des tumeurs solides, IUH, 75010, Paris, France
| | - Karim Bensalah
- Centre Hospitalier Universitaire (CHU) de Pontchaillou Rennes, service d'urologie, 35000, Rennes, France
| | - Alain Ravaud
- Centre Hospitalier Universitaire (CHU) de Bordeaux, service d'oncologie médicale, 33000, Bordeaux, France
| | | | | | | | | | | | - Benoit Beuselinck
- Department of General Medical Oncology, University Hospitals Leuven, 3000, Leuven, Belgium
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, 3000, Leuven, Belgium
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | | | - Damien Ambrosetti
- University Côte d'Azur, Centre Hospitalier Universitaire (CHU) de Nice, Hôpital Pasteur, Central laboratory of Pathology, 06000, Nice, France
| | - Gilles Pagès
- Centre Scientifique de Monaco, Biomedical Department, 8 quai Antoine Premier, 98 000, Monaco, Monaco.
- LIA ROPSE, Laboratoire International Associé Université Côte d'Azur - Centre Scientifique de Monaco, Nice, France.
- University Côte d'Azur, Institute for Research on Cancer and Aging of Nice (IRCAN), CNRS UMR 7284; INSERM U1081, Centre Antoine Lacassagne, 06189, Nice, France.
| |
Collapse
|
16
|
Zhu X, Xu A, Zhang Y, Huo N, Cong R, Ma L, Chu Z, Tang Z, Kang X, Xian S, Xu X. ITPKA1 Promotes Growth, Migration and Invasion of Renal Cell Carcinoma via Activation of mTOR Signaling Pathway. Onco Targets Ther 2020; 13:10515-10523. [PMID: 33116630 PMCID: PMC7573328 DOI: 10.2147/ott.s266095] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/04/2020] [Indexed: 12/12/2022] Open
Abstract
Background Renal cell cancer (RCC) is one of the most lethal malignancies of the kidney in adults. mTOR (mammalian target of rapamycin) signaling pathway plays a pivotal role in RCC tumorigenesis and progression and inhibitors targeting the mTOR pathway have been widely used in advanced RCC treatment. Therefore, it is of great significance to explore the potential regulators of the mTOR pathway as RCC therapeutic targets. Materials and Methods Bioinformatics analysis was used to screen out the most significant differentially expressed genes in the RCC dataset of The Cancer Genome Atlas (TCGA). Real-time PCR and Western-blot analysis were utilized to examine the expression of inositol-1,4,5-trisphosphate-3-kinase-A (ITPKA) in four RCC cell lines and one human embryonic kidney cell line. Cell counting Kit-8 and colony formation assay were performed to estimate the effect of ITPKA on the proliferation ability of RCC cells. Wound healing and Transwell assays were used to test the effect of ITPKA on RCC cell migration and invasion. Xenograft formation assay was performed in nude mice to investigate the effect of ITPKA in vivo. mTORC1 pathway inhibitor was added to explore the mechanisms by which ITPKA regulates RCC cell growth and progression. Results Based on bioinformatics analysis, ITPKA is screened out as one of the most significant differentially expressed genes in RCC. ITPKA is upregulated and positively correlated with RCC malignancy and poorer prognosis. ITPKA promotes RCC growth, migration and invasion in cultured cells, and accelerates tumor growth in nude mice. Mechanistically, ITPKA stimulates the mTORC1 signaling pathway which is a requirement for ITPKA modulation of RCC cell proliferation, migration and invasion. Conclusion Our data demonstrate a critical regulatory role of the ITPKA in RCC and suggest that ITPKA/mTORC1 axis may be a promising target for diagnosis and treatment of RCC.
Collapse
Affiliation(s)
- Xiang Zhu
- Department of Infectious Disease, Army No.82 Group Military Hospital, Baoding, People's Republic of China.,Department of Cellular Engineering Lab, Beijing Institute of Biotechnology, Beijing, People's Republic of China
| | - An Xu
- Department of Oncology, Second Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Yang Zhang
- Department of Radiation Oncology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Nan Huo
- Department of Cellular Engineering Lab, Beijing Institute of Biotechnology, Beijing, People's Republic of China
| | - Rui Cong
- Department of Cellular Engineering Lab, Beijing Institute of Biotechnology, Beijing, People's Republic of China
| | - Luyuan Ma
- Department of Cellular Engineering Lab, Beijing Institute of Biotechnology, Beijing, People's Republic of China
| | - Zhong Chu
- Department of Cellular Engineering Lab, Beijing Institute of Biotechnology, Beijing, People's Republic of China
| | - Zhi Tang
- Department of Cellular Engineering Lab, Beijing Institute of Biotechnology, Beijing, People's Republic of China
| | - Xiaofeng Kang
- Department of Cellular Engineering Lab, Beijing Institute of Biotechnology, Beijing, People's Republic of China
| | - Shaozhong Xian
- Department of Urology, Beijing Luhe Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Xiaojie Xu
- Department of Cellular Engineering Lab, Beijing Institute of Biotechnology, Beijing, People's Republic of China
| |
Collapse
|
17
|
Wu J, Huang H. Acquired Hypothyroidism in Patients with Metastatic Renal Cell Carcinoma Treated with Tyrosine Kinase Inhibitors. Drug Des Devel Ther 2020; 14:3977-3982. [PMID: 33061302 PMCID: PMC7532040 DOI: 10.2147/dddt.s270210] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/02/2020] [Indexed: 02/05/2023] Open
Abstract
PURPOSE We analyze a number of studies that describe the relationship between the onset of hypothyroidism and the prognosis of patients with metastatic renal cell carcinoma (mRCC) treated with TKIs. PATIENTS AND METHODS Targeted therapies are currently considered as the first-line treatment for patients with mRCC. The occurrence of hypothyroidism in the treatment of mRCC with TKIs is a major side effect. A comprehensive search was performed in Pubmed, Cochrane Library, Institute for Scientific Information, and CKNI. The following keywords and descriptors were used during the search and were combined in a number of sequences in English and Chinese languages: protein kinase inhibitors (including sunitinib, pazopanib, tivozanib, cabozantinib, axitinib, sorafenib), hypothyroidism and renal cell carcinoma. RESULTS Scholars have studied the clinical relationship between hypothyroidism and TKI treatment and its molecular biology mechanism. Most studies hold that hypothyroidism occurring during TKI therapy may serve as potential biomarkers of response and efficacy of treatment, but there is no consensus on this point. CONCLUSION The mechanism of hypothyroidism occurring is still unclear. Therefore, more studies are needed to clarify whether better outcomes are associated with TKI-induced hypothyroidism in mRCC patients, helping to maximize the treatment outcome of mRCC patients.
Collapse
Affiliation(s)
- Jialu Wu
- Sichuan University, Department of Endocrinology and Metabolism, West China Hospital, Chengdu610064, Sichuan Province, People’s Republic of China
| | - Hui Huang
- Sichuan University, Department of Endocrinology and Metabolism, West China Hospital, Chengdu610064, Sichuan Province, People’s Republic of China
| |
Collapse
|
18
|
Milluzzo A, Vigneri P, Martorana F, Vigneri R, Sciacca L. Type 2 diabetes and cancer: problems and suggestions for best patient management. EXPLORATION OF MEDICINE 2020. [DOI: 10.37349/emed.2020.00013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Diabetes and cancer are widespread worldwide and the number of subjects presenting both diseases increased over the years. The management of cancer patients having diabetes represents a challenge not only because of the complexity and heterogeneity of these pathologies but also for the lack of standardised clinical guidelines. The diagnosis of cancer is traumatizing and monopolizes the attention of both patients and caregivers. Thus, pre-existent or new-onset diabetes can be overshadowed thus increasing the risk for short- and long-term adverse events. Moreover, drugs used for each disease can interfere with the clinical course of the concomitant disease, making challenging the management of these patients. Over the years, this issue has become more relevant because of the increased patients’ life expectancy due to the improved efficacy of diabetes and cancer therapies.
The purpose of this review is to highlight what is known and what should be taken into consideration to optimise the clinical management of patients with diabetes and cancer. Due to the complexity of these diseases, a multidisciplinary, shared approach, including all the protagonists involved, is necessary to improve patients’ quality of life and lifespan.
Collapse
Affiliation(s)
- Agostino Milluzzo
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania Medical School, 95122 Catania, Italy
| | - Paolo Vigneri
- Center of Experimental Oncology and Hematology, Department of Clinical and Experimental Medicine, University of Catania, A.O.U. Policlinico-Vittorio Emanuele, 95124 Catania, Italy
| | - Federica Martorana
- Center of Experimental Oncology and Hematology, Department of Clinical and Experimental Medicine, University of Catania, A.O.U. Policlinico-Vittorio Emanuele, 95124 Catania, Italy
| | - Riccardo Vigneri
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania Medical School, 95122 Catania, Italy; Institute of Crystallography, Catania Section, National Research Council, CNR, 95126 Catania, Italy
| | - Laura Sciacca
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania Medical School, 95122 Catania, Italy
| |
Collapse
|
19
|
Outeiro-Pinho G, Barros-Silva D, Correia MP, Henrique R, Jerónimo C. Renal Cell Tumors: Uncovering the Biomarker Potential of ncRNAs. Cancers (Basel) 2020; 12:cancers12082214. [PMID: 32784737 PMCID: PMC7465320 DOI: 10.3390/cancers12082214] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/30/2020] [Accepted: 08/05/2020] [Indexed: 12/24/2022] Open
Abstract
Renal cell tumors (RCT) remain as one of the most common and lethal urological tumors worldwide. Discrimination between (1) benign and malignant disease, (2) indolent and aggressive tumors, and (3) patient responsiveness to a specific therapy is of major clinical importance, allowing for a more efficient patient management. Nonetheless, currently available tools provide limited information and novel strategies are needed. Over the years, a putative role of non-coding RNAs (ncRNAs) as disease biomarkers has gained relevance and is now one of the most prolific fields in biological sciences. Herein, we extensively sought the most significant reports on ncRNAs as potential RCTs' diagnostic, prognostic, predictive, and monitoring biomarkers. We could conclude that ncRNAs, either alone or in combination with currently used clinical and pathological parameters, might represent key elements to improve patient management, potentiating the implementation of precision medicine. Nevertheless, most ncRNA biomarkers require large-scale validation studies, prior to clinical implementation.
Collapse
Affiliation(s)
- Gonçalo Outeiro-Pinho
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (G.O.-P.); (D.B.-S.); (M.P.C.); (R.H.)
| | - Daniela Barros-Silva
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (G.O.-P.); (D.B.-S.); (M.P.C.); (R.H.)
| | - Margareta P. Correia
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (G.O.-P.); (D.B.-S.); (M.P.C.); (R.H.)
| | - Rui Henrique
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (G.O.-P.); (D.B.-S.); (M.P.C.); (R.H.)
- Department of Pathology, Portuguese Oncology Institute of Porto, 4200-072 Porto, Portugal
- Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar-University of Porto (ICBAS-UP), Rua de Jorge Viterbo Ferreira n. 228, 4050-313 Porto, Portugal
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (G.O.-P.); (D.B.-S.); (M.P.C.); (R.H.)
- Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar-University of Porto (ICBAS-UP), Rua de Jorge Viterbo Ferreira n. 228, 4050-313 Porto, Portugal
- Correspondence: ; Tel.: +351-225084000; Fax: +351-225084199
| |
Collapse
|
20
|
Keefe DMK, Bateman EH. Potential Successes and Challenges of Targeted Cancer Therapies. J Natl Cancer Inst Monogr 2020; 2019:5551349. [PMID: 31425592 DOI: 10.1093/jncimonographs/lgz008] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/29/2019] [Accepted: 04/19/2019] [Indexed: 01/15/2023] Open
Abstract
The concept and realization of targeted anticancer therapy (TAT) have existed for at least two decades and continue to expand rapidly. It has become clear that there is no "magic bullet" to cure cancer and that even TATs are unlikely to be successful as single agents, necessitating combination with chemotherapy, radiotherapy, or even other targeting agents. The other promise that has not been fulfilled by TAT is that of reduced toxicity. It was thought that by targeting receptors on or within cells, rather than particular phases of the cell cycle, TATs would not be toxic. However, it turns out that the targets also exist on or within normal cells and that there is even cross-reactivity between receptors on nontarget tissues. All of this results in toxicity, the mechanism of which are the same as the mechanism of action of the drugs, making toxicity reduction or prevention very difficult. This leads to new toxicities with new targeted treatments. Nevertheless, all of the above should not detract from the obvious successes of targeted agents, which have turned several acutely fatal cancers into chronic diseases and rendered some hitherto untreatable cancers into treatable diseases.
Collapse
Affiliation(s)
- Dorothy M K Keefe
- Mucositis Research Group, Discipline of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia
| | - Emma H Bateman
- Mucositis Research Group, Discipline of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia
| |
Collapse
|
21
|
Raoul JL, Faivre S, Frenel JS, Rimassa L. Medical oncologists must get more involved in systemic treatment. Ann Oncol 2020; 30:6-8. [PMID: 32081277 DOI: 10.1093/annonc/mdy471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- J L Raoul
- Department of Medical Oncology, Institut de Cancérologie de l'Ouest, Nantes.
| | - S Faivre
- Department of Medical Oncology, Hôpitaux Universitaires Paris Nord Val de Seine, Paris, France
| | - J S Frenel
- Department of Medical Oncology, Institut de Cancérologie de l'Ouest, Nantes
| | - L Rimassa
- Medical Oncology Unit, Humanitas Cancer Center, IRCCS, Milan, Italy
| |
Collapse
|
22
|
Paluri RK, Sonpavde G, Morgan C, Rojymon J, Mar AH, Gangaraju R. Renal toxicity with mammalian target of rapamycin inhibitors: A meta-analysis of randomized clinical trials. Oncol Rev 2019; 13:455. [PMID: 31857859 PMCID: PMC6886005 DOI: 10.4081/oncol.2019.455] [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] [Received: 09/11/2019] [Accepted: 11/12/2019] [Indexed: 12/30/2022] Open
Abstract
A meta-analysis of randomized clinical trials (RCT) was done to determine the relative risk (RR) of acute kidney injury (AKI) with the use of mammalian target of rapamycin (mTOR) inhibitors. Citations from PubMed/Medline, clinical trials.gov, package inserts and abstracts from major conferences were reviewed to include RCTs comparing arms with or without mTOR inhibitors. The RR of all grade AKI in patients taking mTOR inhibitors compared to patients not on mTOR inhibitors was 1.55 (95% CI: 1.11 to 2.16, P=0.010). There was no significant difference in the risk of high-grade AKI for the two groups (RR=1.29, P=0.118, 95% CI: 0.94 to 1.77). There was no significant difference in the incidence rates for either all grade or high-grade AKI between the two groups. There was no publication bias and the trials were of high quality per Jadad scoring.
Collapse
Affiliation(s)
- Ravi K Paluri
- Division of Hematology-Oncology, Department of Medicine, University of Alabama at Birmingham, AL
| | - Guru Sonpavde
- Section of Medical Oncology, Department of Medicine, Dana-Farber Cancer Institute, Harvard Medical School, MA
| | - Charity Morgan
- Department of Biostatistics, University of Alabama at Birmingham, AL
| | - Jacob Rojymon
- Department of Radiation Oncology, University of Alabama at Birmingham, AL, USA
| | | | - Radhika Gangaraju
- Division of Hematology-Oncology, Department of Medicine, University of Alabama at Birmingham, AL
| |
Collapse
|
23
|
Gabora K, Piciu A, Bădulescu IC, Larg MI, Stoian IA, Piciu D. Current evidence on thyroid related adverse events in patients treated with protein tyrosine kinase inhibitors. Drug Metab Rev 2019; 51:562-569. [PMID: 31718371 DOI: 10.1080/03602532.2019.1687512] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Tyrosine kinase inhibitors (TKI) are gaining more ground in oncology, they are widely used in the treatment of multiple types of cancers; still important side effects limit their efficacy. The aim of this study is to evaluate the existing medical literature on TKI induced thyroid dysfunction, to assess the adverse effects of targeted therapy on thyroid function in oncological patients and to evaluate the effects of thyroid dysfunction on disease prognosis. We included in this review 22 original studies published between 2010 and 2019. We used the PubMed database to search for articles upon the development of hypothyroidism and hyperthyroidism in TKI treated patients. After a careful review of the existing literature, we selected the relevant studies and cross-referenced the bibliography of each paper. A number of 1641 patients were included in our review. We found that thyroid dysfunction is not a rare side effect of TKI treatment, approximately 33% of the total number of patients presented clinical hypothyroidism. We also studied the necessity of thyroid hormone substitution treatment, a quarter of evaluated patients needed substitution therapy. Multiple studies showed that there is a link between a patient developing hypothyroidism and progression free survival. Hypothyroidism is a frequent side effect of TKI treatment, which affects the quality of life, sometimes even determines physicians to stop TKI treatment altogether. Our study underlines the necessity of TSH baseline testing and monitoring in patients treated with TKI agents.
Collapse
Affiliation(s)
- Katalin Gabora
- Department of Nuclear Medicine, Prof. Dr. Ion Chiricuță Institute of Oncology, Cluj Napoca, Romania.,Iuliu Hațieganu Univerisity of Pharmacy and Medicine, Cluj Napoca, Romania
| | - Andra Piciu
- Iuliu Hațieganu Univerisity of Pharmacy and Medicine, Cluj Napoca, Romania
| | | | - Maria Iulia Larg
- Department of Nuclear Medicine, Prof. Dr. Ion Chiricuță Institute of Oncology, Cluj Napoca, Romania.,Iuliu Hațieganu Univerisity of Pharmacy and Medicine, Cluj Napoca, Romania
| | - Ioan-Adrian Stoian
- Iuliu Hațieganu Univerisity of Pharmacy and Medicine, Cluj Napoca, Romania
| | - Doina Piciu
- Department of Nuclear Medicine, Prof. Dr. Ion Chiricuță Institute of Oncology, Cluj Napoca, Romania.,Iuliu Hațieganu Univerisity of Pharmacy and Medicine, Cluj Napoca, Romania
| |
Collapse
|
24
|
Kim JK, Lee JH. Mechanistic Target of Rapamycin Pathway in Epileptic Disorders. J Korean Neurosurg Soc 2019; 62:272-287. [PMID: 31085953 PMCID: PMC6514310 DOI: 10.3340/jkns.2019.0027] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 03/12/2019] [Indexed: 12/19/2022] Open
Abstract
The mechanistic target of rapamycin (mTOR) pathway coordinates the metabolic activity of eukaryotic cells through environmental signals, including nutrients, energy, growth factors, and oxygen. In the nervous system, the mTOR pathway regulates fundamental biological processes associated with neural development and neurodegeneration. Intriguingly, genes that constitute the mTOR pathway have been found to be germline and somatic mutation from patients with various epileptic disorders. Hyperactivation of the mTOR pathway due to said mutations has garnered increasing attention as culprits of these conditions : somatic mutations, in particular, in epileptic foci have recently been identified as a major genetic cause of intractable focal epilepsy, such as focal cortical dysplasia. Meanwhile, epilepsy models with aberrant activation of the mTOR pathway have helped elucidate the role of the mTOR pathway in epileptogenesis, and evidence from epilepsy models of human mutations recapitulating the features of epileptic patients has indicated that mTOR inhibitors may be of use in treating epilepsy associated with mutations in mTOR pathway genes. Here, we review recent advances in the molecular and genetic understanding of mTOR signaling in epileptic disorders. In particular, we focus on the development of and limitations to therapies targeting the mTOR pathway to treat epileptic seizures. We also discuss future perspectives on mTOR inhibition therapies and special diagnostic methods for intractable epilepsies caused by brain somatic mutations.
Collapse
Affiliation(s)
- Jang Keun Kim
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Jeong Ho Lee
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| |
Collapse
|
25
|
Levin AS, Alcorn SR, Neuman BJ, Meyer CF. Team Approach: Emergencies in Patients with Skeletal Metastases. JBJS Rev 2019; 7:e8. [PMID: 30920480 DOI: 10.2106/jbjs.rvw.18.00036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Adam S Levin
- Department of Orthopaedic Surgery (A.S.L. and B.J.N.), Department of Radiation and Molecular Radiation Sciences (S.R.A.), Sidney Kimmel Comprehensive Cancer Center (C.F.M.), The Johns Hopkins School of Medicine, Baltimore, Maryland
| | | | | | | |
Collapse
|
26
|
Milluzzo A, Tumminia A, Vella V, Gianì F, Manzella L, Frittitta L, Belfiore A, Vigneri R, Sciacca L. Short-term adverse effects of anticancer drugs in patients with type 2 diabetes. J Chemother 2019; 31:150-159. [DOI: 10.1080/1120009x.2019.1572297] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Agostino Milluzzo
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania Medical School, Garibaldi-Nesima Hospital, Catania, Italy
| | - Andrea Tumminia
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania Medical School, Garibaldi-Nesima Hospital, Catania, Italy
| | - Veronica Vella
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania Medical School, Garibaldi-Nesima Hospital, Catania, Italy
- School of Human and Social Science, University “Kore” of Enna, Enna, Italy
| | - Fiorenza Gianì
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania Medical School, Garibaldi-Nesima Hospital, Catania, Italy
| | - Livia Manzella
- Department of Clinical and Experimental Medicine, University of Catania Medical School, Center of Experimental Oncology and Hematology, Catania, Italy
| | - Lucia Frittitta
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania Medical School, Garibaldi-Nesima Hospital, Catania, Italy
| | - Antonino Belfiore
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania Medical School, Garibaldi-Nesima Hospital, Catania, Italy
| | - Riccardo Vigneri
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania Medical School, Garibaldi-Nesima Hospital, Catania, Italy
- Institute of Bioimages and Biostructures, CNR, Catania, Italy
| | - Laura Sciacca
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania Medical School, Garibaldi-Nesima Hospital, Catania, Italy
| |
Collapse
|
27
|
Ndiaye PD, Dufies M, Giuliano S, Douguet L, Grépin R, Durivault J, Lenormand P, Glisse N, Mintcheva J, Vouret-Craviari V, Mograbi B, Wurmser M, Ambrosetti D, Rioux-Leclercq N, Maire P, Pagès G. VEGFC acts as a double-edged sword in renal cell carcinoma aggressiveness. Am J Cancer Res 2019; 9:661-675. [PMID: 30809300 PMCID: PMC6376471 DOI: 10.7150/thno.27794] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 07/30/2018] [Indexed: 12/17/2022] Open
Abstract
Hypoxic zones are common features of metastatic tumors. Due to inactivation of the von Hippel-Lindau gene (VHL), renal cell carcinomas (RCC) show constitutive stabilization of the alpha subunit of the hypoxia-inducible factor (HIF). Thus, RCC represents a model of chronic hypoxia. Development of the lymphatic network is dependent on vascular endothelial growth factor C (VEGFC) and lies at the front line of metastatic spreading. Here, we addressed the role of VEGFC in RCC aggressiveness and the regulation of its expression in hypoxia. Methods: Transcriptional and post transcriptional regulation of VEGFC expression was evaluated by qPCR and with reporter genes. The involvement of HIF was evaluated using a siRNA approach. Experimental RCC were performed with immuno-competent/deficient mice using human and mouse cells knocked-out for the VEGFC gene by a CRISPR/Cas9 method. The VEGFC axis was analyzed with an online available data base (TCGA) and using an independent cohort of patients. Results: Hypoxia induced VEGFC protein expression but down-regulated VEGFC gene transcription and mRNA stability. Increased proliferation, migration, over-activation of the AKT signaling pathway and enhanced expression of mesenchymal markers characterized VEGFC-/- cells. VEGFC-/- cells did not form tumors in immuno-deficient mice but developed aggressive tumors in immuno-competent mice. These tumors showed down-regulation of markers of activated lymphocytes and M1 macrophages, and up-regulation of M2 macrophages markers and programmed death ligand 1 (PDL1). Over-expression of lymphangiogenic genes including VEGFC was linked to increased disease-free and overall survival in patients with non-metastatic tumors, whereas its over-expression correlated with decreased progression-free and overall survival of metastatic patients. Conclusion: Our study revisited the admitted dogma linking VEGFC to tumor aggressiveness. We conclude that targeting VEGFC for therapy must be considered with caution.
Collapse
|
28
|
Cabanillas ME, Takahashi S. Managing the adverse events associated with lenvatinib therapy in radioiodine-refractory differentiated thyroid cancer. Semin Oncol 2018; 46:57-64. [PMID: 30685073 DOI: 10.1053/j.seminoncol.2018.11.004] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 11/21/2018] [Indexed: 02/06/2023]
Abstract
Lenvatinib is a multikinase inhibitor of vascular endothelial growth factor (VEGF) receptors 1-3, fibroblast growth factor receptors 1-4, RET, KIT, and platelet-derived growth factor receptor-α. Lenvatinib is approved as a monotherapy for the treatment of radioiodine-refractory differentiated thyroid cancer and in combination with everolimus for the second-line treatment of advanced renal cell carcinoma. Lenvatinib is also under investigation for the treatment of several malignancies including unresectable hepatocellular carcinoma. Although lenvatinib is associated with favorable efficacy, it is associated with adverse events (AEs) that the clinician will have to closely monitor for and proactively manage. Most of these AEs are known class effects of VEGF-targeted therapies, including hypertension, diarrhea, fatigue or asthenia, decreased appetite, and weight loss. This review summarizes the safety profile of lenvatinib and offers guidance for the management of both frequent and rare AEs. We discuss the potential mechanisms underlying these AEs and present practical recommendations for managing toxicities. The development of treatment plans that include prophylactic and therapeutic strategies for the management of lenvatinib-associated AEs has the potential to improve patient quality of life, optimize adherence, minimize the need for dose reductions, treatment interruptions, or discontinuations, and maximize patient outcomes.
Collapse
Affiliation(s)
- Maria E Cabanillas
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Shunji Takahashi
- Department of Medical Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
29
|
Woldu SL, Matulay JT, Clinton TN, Singla N, Freifeld Y, Sanli O, Krabbe LM, Hutchinson RC, Lotan Y, Hammers H, Hannan R, Brugarolas J, Bagrodia A, Margulis V. Incidence and Outcomes of Delayed Targeted Therapy After Cytoreductive Nephrectomy for Metastatic Renal-Cell Carcinoma: A Nationwide Cancer Registry Study. Clin Genitourin Cancer 2018; 16:e1221-e1235. [DOI: 10.1016/j.clgc.2018.08.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/26/2018] [Accepted: 08/05/2018] [Indexed: 12/24/2022]
|
30
|
Tsironis G, Koutsoukos K, Athanasakis K, Tsiara A, Tzannis K, Gerolympou M, Visvikis A, Oikonomopoulos G, Kollia A, Giannopoulou E, Dimitra M, Kostouros E, Papatsoris A, Dellis A, Stravodimos K, Varkarakis I, Samantas E, Aravantinos G, Kentepozidis N, Christodoulou C, Bozionelou V, Dimopoulos MA, Bamias A. Patterns of practice and pharmacoeconomic analysis of the management of patients with metastatic renal cell carcinoma (mRCC) in Greece--the CRISIS study. A retrospective analysis by the Hellenic Genitourinary Cancer Group (HGUCG). Expert Rev Pharmacoecon Outcomes Res 2018; 19:491-501. [PMID: 30417707 DOI: 10.1080/14737167.2019.1546121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Background:Metastatic RCC (mRCC) treatment has been revolutionized with 11 approved targeted agents. We report patterns of practice, outcomes and pharmacoeconomic analyses after the introduction of targeted therapy. Patients and methods: CRISIS was a retrospective multicenter study of mRCCpatients who received targeted therapy . Results were related to the start of 1st-line therapy, with a cut off at 1 January 2011 in order to depict the impact of increased availability of effective options. Results: 164 patients, were included. 70.1% and 44.5% received 2nd and 3rd-line therapy, respectively. More patients were treated in 2nd-line after 1 January 2011. After a median follow-up of 55.1 months, median progression-free (PFS) and overall survival (OS) were 10.7 (95% confidence intervals [CI]: 8.3-13.7), 7.3 (95% CI: 5.1-8.6), 5.8 (95% CI: 3.8-7.8) and 34 (95% CI: 28.5-39.8), 22.4 (95% CI: 16-32.1), 18.3 (95% CI: 12.4-26.4) months for first, second and third line, respectively. Efficacy of sunitinib and pazopanib in 1st-line were similar. The mean total cost/patient was 35,012.2 Euros (standard deviation [SD]: 28,971.5). Conclusions: Our study confirms previous real-world data suggesting that continuing advances in the treatment of mRCC produce favorable outcomes in everyday practice. Pharmacoeconomic analyses are important for cost-effective utilization of emerging novel therapies.
Collapse
Affiliation(s)
- Georgiops Tsironis
- a Oncology Unit, Department of Clinical Therapeutics, Alexandra Hospital , University of Athens , Athens , Greece.,b Hellenic Genito-Urinary Cancer Group , Athens , Greece
| | - Konstantinos Koutsoukos
- a Oncology Unit, Department of Clinical Therapeutics, Alexandra Hospital , University of Athens , Athens , Greece.,b Hellenic Genito-Urinary Cancer Group , Athens , Greece
| | | | - Anna Tsiara
- a Oncology Unit, Department of Clinical Therapeutics, Alexandra Hospital , University of Athens , Athens , Greece
| | - Kimon Tzannis
- b Hellenic Genito-Urinary Cancer Group , Athens , Greece
| | - Margarita Gerolympou
- d 3rd Oncology Clinic , General Oncology Hospital of Kifisias "Ag. Anargyroi" , Athens , Greece
| | - Anastasios Visvikis
- d 3rd Oncology Clinic , General Oncology Hospital of Kifisias "Ag. Anargyroi" , Athens , Greece
| | | | | | | | | | - Efthymios Kostouros
- a Oncology Unit, Department of Clinical Therapeutics, Alexandra Hospital , University of Athens , Athens , Greece
| | - Athanasios Papatsoris
- b Hellenic Genito-Urinary Cancer Group , Athens , Greece.,f 2nd Department of Urology, Sismanoglio General Hospital , University of Athens , Athens , Greece
| | - Athanasios Dellis
- b Hellenic Genito-Urinary Cancer Group , Athens , Greece.,g 2nd Department of Surgery, Aretaieion Academic Hospital , University of Athens , Athens , Greece
| | - Konstantinos Stravodimos
- b Hellenic Genito-Urinary Cancer Group , Athens , Greece.,h 1st University Urology Clinic, Laiko Hospital , University of Athens , Athens , Greece
| | - Ioannis Varkarakis
- b Hellenic Genito-Urinary Cancer Group , Athens , Greece.,f 2nd Department of Urology, Sismanoglio General Hospital , University of Athens , Athens , Greece
| | - Epaminontas Samantas
- d 3rd Oncology Clinic , General Oncology Hospital of Kifisias "Ag. Anargyroi" , Athens , Greece
| | - Gerasimos Aravantinos
- i 2nd Oncology Clinic , General Oncology Hospital of Kifisias "Ag. Anargyroi" , Athens , Greece
| | | | | | - Vasiliki Bozionelou
- k Department of Medical Oncology , University Hospital of Heraklion , Heraklion , Greece
| | - Meletios Athanasios Dimopoulos
- a Oncology Unit, Department of Clinical Therapeutics, Alexandra Hospital , University of Athens , Athens , Greece.,b Hellenic Genito-Urinary Cancer Group , Athens , Greece
| | - Aristotle Bamias
- a Oncology Unit, Department of Clinical Therapeutics, Alexandra Hospital , University of Athens , Athens , Greece.,b Hellenic Genito-Urinary Cancer Group , Athens , Greece
| |
Collapse
|
31
|
Percutaneous microwave ablation for local control of metastatic renal cell carcinoma. Abdom Radiol (NY) 2018; 43:2446-2454. [PMID: 29464274 DOI: 10.1007/s00261-018-1498-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE The purpose of the article is to evaluate the safety and oncologic efficacy of microwave ablation for metastatic renal cell carcinoma (mRCC). MATERIALS AND METHODS From September 2011 to December 2016, 33 mRCC were ablated in 18 patients using percutaneous microwave ablation. Sites of mRCC include retroperitoneum (n = 12), contralateral kidney (n = 6), liver (n = 6), lung (n = 5), adrenal gland (n = 5). Technical success, local, and distant tumor progression, and complications were assessed at immediate and follow-up imaging. The Kaplan-Meier method was used for survival analysis. RESULTS Technical success was achieved for 33/33 (100%) mRCC tumors. Ablation provided durable local control for 28/30 (93%) mRCC tumors in 17 patients at a median duration of clinical and imaging follow-up of 1.6 years (IQR 0.7-3.6) and 0.8 years (IQR 0.5-2.7), respectively. In-hospital and perioperative mortality was 0%. There were 5 (15%) procedure-related complications including one high-grade event (Clavien-Dindo III). Four patients have died from mRCC at a median of 1.3 years (range 0.7-5.1) following ablation. Estimated OS (95% CI number still at risk) at 1, 2, and 5 years were 86% (53-96%, 11), 75% (39-92%, 8), and 75% (39-92%, 3), respectively. CONCLUSIONS Microwave ablation of oligometastatic renal cell carcinoma is safe and provides durable local control in appropriately selected patients.
Collapse
|
32
|
Treatment Facility Volume and Survival in Patients with Metastatic Renal Cell Carcinoma: A Registry-based Analysis. Eur Urol 2018; 74:387-393. [PMID: 29880274 DOI: 10.1016/j.eururo.2018.05.025] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 05/18/2018] [Indexed: 11/24/2022]
Abstract
BACKGROUND Higher treatment facility (TF) volume has been linked with improved oncologic treatment outcomes. OBJECTIVE To determine the association between TF volume and overall survival in patients with metastatic renal cell carcinoma (mRCC). DESIGN, SETTING, AND PARTICIPANTS The National Cancer Database (NCDB) was queried for all patients with mRCC with survival data available (2004-2013, cohort A). Overall survival was assessed based on TF volumes, and increasingly narrow inclusion criteria were used to confirm the cohort A association: cohort B=mRCC patients with active treatment; cohort C=mRCC patients with systemic therapy; cohort D=mRCC patients with systemic therapy at the reporting institution; and cohort E=mRCC patients with systemic therapy at the reporting institution with known liver and lung metastatic status. Sensitivity analyses were also performed on subcohorts of mRCC who never underwent a nephrectomy (C1, D1, and E1). OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The effect of volume on time to death (from any cause) was determined using Cox regression models, adjusting for multiple clinical pathologic factors. Volume effects (assessed continuously) were modeled using flexible cubic splines, and adjusted 1-yr survivals were obtained from the model. RESULTS AND LIMITATIONS A total of 41 836 mRCC patients were treated at 1222 TFs. The median age was 65 yr. Of the patients, 66% were men and 79% had clear cell mRCC. Median TF volume was 2.2 patients per year (pts/yr). Across all cohorts, higher TF volume was associated with improved outcomes. Adjusted 1-yr survival in cohort A was 0.36 at 2 pts/yr, 0.39 at 5 pts/yr, 0.42 at 10 pts/yr, and 0.46 at 20 pts/yr, with similar magnitudes of effect in cohorts B-E. Limitations include the retrospective nature of NCDB analysis and the lack of information on treatment regimens used at specific facilities, which may explain mechanisms of effects. CONCLUSIONS Higher facility volume is associated with improvements in survival for patients being treated for mRCC. Steps should be taken to standardize management of mRCC patients, such as evidence-based pathway development, clinical trial access, and multidisciplinary resource availability at lower-volume TFs. PATIENT SUMMARY In this report, we analyzed a large cancer database and found that patients with metastatic kidney cancer survived longer if they were managed at facilities that treated a higher volume of such patients. This information can help find the best treatment environment for patients with metastatic kidney cancer.
Collapse
|
33
|
The microRNA signature of patients with sunitinib failure: regulation of UHRF1 pathways by microRNA-101 in renal cell carcinoma. Oncotarget 2018; 7:59070-59086. [PMID: 27487138 PMCID: PMC5312296 DOI: 10.18632/oncotarget.10887] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 07/19/2016] [Indexed: 12/21/2022] Open
Abstract
Molecular targeted therapy is a standard treatment for patients with advanced renal cell carcinoma (RCC). Sunitinib is one of the most common molecular-targeted drugs for metastatic RCC. Molecular mechanisms of sunitinib resistance in RCC cells is still ambiguous. The microRNA (miRNA) expression signature of patients with sunitinib failure in RCC was constructed using a polymerase chain reaction (PCR)-based array. Several miRNAs that were aberrantly expressed in RCC tissues from patients treated with sunitinib were identified in this analysis. MicroRNA-101 (miR- 101) was markedly suppressed in sunitinib treated RCC tissues. Restoration of miR-101 significantly inhibited cell migration and invasion in Caki-1 and 786-O cells. Ubiquitin-like with PHD and ring finger domains 1 (UHRF1) was directly suppressed by miR-101 in RCC cells, and overexpression of UHRF1 was confirmed in sunitinib-treated RCC tissues. The pathways of nucleotide excision repair and mismatch repair were significantly suppressed by knockdown of UHRF1. Our findings showed that antitumor miR-101- mediated UHRF1 pathways may be suppressed by sunitinib treatment.
Collapse
|
34
|
Gerendash BS, Creel PA. Practical management of adverse events associated with cabozantinib treatment in patients with renal-cell carcinoma. Onco Targets Ther 2017; 10:5053-5064. [PMID: 29089775 PMCID: PMC5656352 DOI: 10.2147/ott.s145295] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cabozantinib is an oral tyrosine-kinase inhibitor whose targets include VEGFR, MET, and AXL. Cabozantinib is approved for the treatment of patients with advanced clear-cell renal-cell carcinoma (RCC) who have received prior antiangiogenic therapy. In the pivotal Phase III trial of second-line RCC, cabozantinib was associated with a significant improvement in overall survival, progression-free survival, and antitumor response compared with everolimus. Adverse events (AEs) were common for patients receiving cabozantinib, but were effectively managed with supportive care and dose modifications, as discontinuations of cabozantinib due to an AE were infrequent. This article reviews the management of the more common AEs associated with cabozantinib based on findings from the pivotal study, clinical practice guidelines, and the authors' real-world clinical experience, with support from published literature. We focus on hypertension, palmar-plantar erythrodysesthesia, diarrhea, nausea, vomiting, decreased appetite, fatigue, and stomatitis. Effective management of these AEs involves a multimodal strategy that includes patient education, prophylactic and supportive care, and dose modifications. Effective AE management can allow patients to maintain antitumor activity with cabozantinib while mitigating the impact on quality of life.
Collapse
Affiliation(s)
- Benjamin S Gerendash
- Department of Medical Oncology and Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Patricia A Creel
- Oncology, Clinical Operations, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
35
|
Wen T, Xiao H, Luo C, Huang L, Xiong M. Efficacy of sequential therapies with sorafenib-sunitinib versus sunitinib-sorafenib in metastatic renal cell carcinoma: A systematic review and meta-analysis. Oncotarget 2017; 8:20441-20451. [PMID: 28099901 PMCID: PMC5386775 DOI: 10.18632/oncotarget.14671] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 01/09/2017] [Indexed: 01/11/2023] Open
Abstract
The most efficient sequence of targeted agents for metastatic renal cell carcinoma patients has yet to be identified. Whether the sequence of sorafenib and sunitinib really matters is controversial and not answered clearly until now. This meta-analysis aims to estimate the efficacy of receptor tyrosine kinase inhibitors sorafenib-sunitinib and sunitinib-sorafenib for metastatic renal cell carcinoma, on the outcome of first-line progression-free survival, second-line progression-free survival, total progression-free survival and overall survival. We searched PubMed, Embase, Cochrane Library and ClinicalTrails.gov for eligible studies. Data were analyzed using random or fixed effects model depending on the heterogeneity of the eligible studies. Heterogeneity across studies were analyzed using Q and I2 statistics. Of 902 identified studies, ten were qualified in our analysis (N = 1732 patients). Sorafenib-sunitinib yielded no statistically significant benefit in first-line progression-free survival (fixed effects; HR = 0.95; 95%CI 0.75-1.21; p = 0.702), total progression-free survival (random effects; HR = 0.92; 95%CI 0.71-1.19; p = 0.531) and overall survival (fixed effects; HR = 0.89; 95%CI 0.72-1.09; p = 0.257), compared with sunitinib-sorafenib. Second-line progression-free survival was longer for sorafenib-sunitinib than sunitinib-sorafenib (fixed effects; HR = 0.55; 95%CI 0.44-0.68; p = 0.000). Sequential therapies with sorafenib and sunitinib is well tolerated and efficient in mRCC. However, there are no evidence supported that sorafenib–sunitinib has the superiority to sunitinib-sorafenib in sequence. The ideal sequence of targeted agents requires further elucidation.
Collapse
Affiliation(s)
- Tingyu Wen
- College of Basic Medical Sciences, Gannan Medical University, Ganzhou, China
| | - Hai Xiao
- Department of Pathology, Gannan Medical University, Ganzhou, China
| | - Chao Luo
- Department of Urology, People's Hosptial of Pingxiang, Pingxiang, China
| | - Li Huang
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Meimei Xiong
- Department of Nephrology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
36
|
Srinivas S, Stein D, Teltsch DY, Tao S, Cisar L, Ramaswamy K. Real-world chart review study of adverse events management in patients taking tyrosine kinase inhibitors to treat metastatic renal cell carcinoma. J Oncol Pharm Pract 2017; 24:574-583. [PMID: 28732453 DOI: 10.1177/1078155217719583] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The purpose is to describe management of adverse events of special interest across tyrosine kinase inhibitors approved for metastatic renal cell carcinoma. METHODS We conducted a retrospective chart review in metastatic renal cell carcinoma patients initiating tyrosine kinase inhibitor monotherapy between 15 November 2010 and 15 November 2013, and experiencing ≥ 1 adverse events of special interest (diarrhea, fatigue, hand-foot syndrome, hypertension, or stomatitis/mucositis) within 3 months of initiation. Demographics, medical history, treatment regimens, and adverse events of special interest management data for 3.5 months postonset were collected. RESULTS In 220 charts from 27 centers, tyrosine kinase inhibitors prescribed included sunitinib (55%), pazopanib (27%), axitinib (9%), and sorafenib (8%). During the study period, patients experienced 376 adverse events of special interest (13% serious). Fatigue was most common (62% of patients), followed by hypertension (37%), diarrhea (30%), stomatitis/mucositis (29%), and hand-foot syndrome (12%). Over half (56%) the adverse events of special interest were resolved or resolving. Treatment discontinuation due to adverse events of special interest occurred in 15% of patients. Prophylaxis was rarely provided (8%), whereas 59% of patients received adverse events of special interest treatment (pharmacologic (55%) and/or nonpharmacologic (7%)), 27% received tyrosine kinase inhibitor dose management, 23% received both adverse events of special interest treatment and dose management, and 31% received neither. Hypertension was the most treated (72% of all events), and fatigue was the least treated (9%); only 4% of patients received pharmacologic treatment for fatigue. CONCLUSIONS Most adverse events of special interest were nonserious and more than half of the patients received pharmacologic and/or nonpharmacologic treatment. Fatigue was the most common yet least frequently treated adverse event of special interest, and few patients received prophylaxis or nonpharmacologic treatment. More emphasis on treatment and prophylaxis options for bothersome adverse events is warranted.
Collapse
Affiliation(s)
- Sandy Srinivas
- 1 Division of Medical Oncology, Stanford University Medical Center, USA
| | - Dara Stein
- 2 Safety, Epidemiology, Registries, and Risk Management (SERRM), United BioSource Corporation, Canada
| | | | - Sunning Tao
- 2 Safety, Epidemiology, Registries, and Risk Management (SERRM), United BioSource Corporation, Canada
| | | | | |
Collapse
|
37
|
Rini BI, Gruenwald V, Jonasch E, Fishman MN, Tomita Y, Michaelson MD, Tarazi J, Cisar L, Hariharan S, Bair AH, Rosbrook B, Hutson TE. Long-term Duration of First-Line Axitinib Treatment in Advanced Renal Cell Carcinoma. Target Oncol 2017; 12:333-340. [DOI: 10.1007/s11523-017-0487-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
38
|
Zhang HL, Qin XJ, Wang HK, Gu WJ, Ma CG, Shi GH, Zhou LP, Ye DW. Clinicopathological and prognostic factors for long-term survival in Chinese patients with metastatic renal cell carcinoma treated with sorafenib: a single-center retrospective study. Oncotarget 2017; 6:36870-83. [PMID: 26472104 PMCID: PMC4742216 DOI: 10.18632/oncotarget.4874] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 10/01/2015] [Indexed: 01/13/2023] Open
Abstract
Data on long-term survival and prognostic significance of demographic factors and adverse events (AEs) associated with sorafenib, an orally administered multikinase inhibitor in Chinese population with advanced renal cell carcinoma (RCC) are limited. Outcome data from adult patients (n = 256) with advanced RCC who received sorafenib (400 mg twice daily) either as first-line or second-line therapy between April 2006 and May 2013 were analyzed retrospectively. The primary endpoint was median overall survival (OS), determined to be 22.2 (95% CI: 17.1–27.4) months, and the secondary endpoint was overall median progression-free survival (PFS), determined to be 13.6 (95% CI: 10.7–16.4) months at a median follow-up time of 61.8 (95% CI: 16.2–97.4) months. Analysis of the incidence of AEs revealed the most common side effect as hand-foot skin reactions (60.5%) followed by diarrhea (38.7%), fatigue (35.5%), alopecia (34.0%), rash (24.6%), hypertension (21.5%) and gingival hemorrhage (21.1%). Multivariate regression analysis revealed older age (≥ 58 years), lower Memorial Sloan-Kettering Cancer Center score, time from nephrectomy to sorafenib treatment, number of metastatic tumors and best response as significant and independent demographic predictors for improved PFS and/or OS (p ≤ 0.05). Alopecia was identified as a significant and independent predictor of increased OS, whereas vomiting and weight loss were identified as significant predictors of decreased OS (p ≤ 0.05). Sorafenib significantly improved OS and PFS in Chinese patients with advanced RCC. Considering the identified significant prognostic demographic factors along with the advocated prognostic manageable AEs while identifying treatment strategy may help clinicians select the best treatment modality and better predict survival in these patients.
Collapse
Affiliation(s)
- Hai-Liang Zhang
- Department of Urology, Fudan University, Shanghai Cancer Center, Shanghai 200032, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Xiao-Jian Qin
- Department of Urology, Fudan University, Shanghai Cancer Center, Shanghai 200032, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Hong-Kai Wang
- Department of Urology, Fudan University, Shanghai Cancer Center, Shanghai 200032, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Wei-Jie Gu
- Department of Urology, Fudan University, Shanghai Cancer Center, Shanghai 200032, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Chun-Guang Ma
- Department of Urology, Fudan University, Shanghai Cancer Center, Shanghai 200032, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Guo-Hai Shi
- Department of Urology, Fudan University, Shanghai Cancer Center, Shanghai 200032, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Liang-Ping Zhou
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China.,Department of Radiology, Fudan University, Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - Ding-Wei Ye
- Department of Urology, Fudan University, Shanghai Cancer Center, Shanghai 200032, P.R. China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
39
|
Hwangbo Y, Lee EK. Acute Hyperglycemia Associated with Anti-Cancer Medication. Endocrinol Metab (Seoul) 2017; 32:23-29. [PMID: 28345313 PMCID: PMC5368117 DOI: 10.3803/enm.2017.32.1.23] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 02/01/2017] [Accepted: 02/16/2017] [Indexed: 12/16/2022] Open
Abstract
Hyperglycemia during chemotherapy occurs in approximately 10% to 30% of patients. Glucocorticoids and L-asparaginase are well known to cause acute hyperglycemia during chemotherapy. Long-term hyperglycemia is also frequently observed, especially in patients with hematologic malignancies treated with L-asparaginase-based regimens and total body irradiation. Glucocorticoid-induced hyperglycemia often develops because of increased insulin resistance, diminished insulin secretion, and exaggerated hepatic glucose output. Screening strategies for this condition include random glucose testing, hemoglobin A1c testing, oral glucose loading, and fasting plasma glucose screens. The management of hyperglycemia starts with insulin or sulfonylurea, depending on the type, dose, and delivery of the glucocorticoid formulation. Mammalian target of rapamycin (mTOR) inhibitors are associated with a high incidence of hyperglycemia, ranging from 13% to 50%. Immunotherapy, such as anti-programmed death 1 (PD-1) antibody treatment, induces hyperglycemia with a prevalence of 0.1%. The proposed mechanism of immunotherapy-induced hyperglycemia is an autoimmune process (insulitis). Withdrawal of the PD-1 inhibitor is the primary treatment for severe hyperglycemia. The efficacy of glucocorticoid therapy is not fully established and the decision to resume PD-1 inhibitor therapy depends on the severity of the hyperglycemia. Diabetic patients should achieve optimized glycemic control before initiating treatment, and glucose levels should be monitored periodically in patients initiating mTOR inhibitor or PD-1 inhibitor therapy. With regard to hyperglycemia caused by anti-cancer therapy, frequent monitoring and proper management are important for promoting the efficacy of anti-cancer therapy and improving patients' quality of life.
Collapse
Affiliation(s)
- Yul Hwangbo
- Department of Internal Medicine, Center for Thyroid Cancer, National Cancer Center, Goyang, Korea
| | - Eun Kyung Lee
- Department of Internal Medicine, Center for Thyroid Cancer, National Cancer Center, Goyang, Korea.
| |
Collapse
|
40
|
Dufies M, Giuliano S, Ambrosetti D, Claren A, Ndiaye PD, Mastri M, Moghrabi W, Cooley LS, Ettaiche M, Chamorey E, Parola J, Vial V, Lupu-Plesu M, Bernhard JC, Ravaud A, Borchiellini D, Ferrero JM, Bikfalvi A, Ebos JM, Khabar KS, Grépin R, Pagès G. Sunitinib Stimulates Expression of VEGFC by Tumor Cells and Promotes Lymphangiogenesis in Clear Cell Renal Cell Carcinomas. Cancer Res 2017; 77:1212-1226. [PMID: 28087600 DOI: 10.1158/0008-5472.can-16-3088] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/07/2016] [Accepted: 12/08/2016] [Indexed: 11/16/2022]
Abstract
Sunitinib is an antiangiogenic therapy given as a first-line treatment for renal cell carcinoma (RCC). While treatment improves progression-free survival, most patients relapse. We hypothesized that patient relapse can stem from the development of a lymphatic network driven by the production of the main growth factor for lymphatic endothelial cells, VEGFC. In this study, we found that sunitinib can stimulate vegfc gene transcription and increase VEGFC mRNA half-life. In addition, sunitinib activated p38 MAPK, which resulted in the upregulation/activity of HuR and inactivation of tristetraprolin, two AU-rich element-binding proteins. Sunitinib stimulated a VEGFC-dependent development of lymphatic vessels in experimental tumors. This may explain our findings of increased lymph node invasion and new metastatic sites in 30% of sunitinib-treated patients and increased lymphatic vessels found in 70% of neoadjuvant treated patients. In summary, a therapy dedicated to destroying tumor blood vessels induced the development of lymphatic vessels, which may have contributed to the treatment failure. Cancer Res; 77(5); 1212-26. ©2017 AACR.
Collapse
Affiliation(s)
- Maeva Dufies
- University of Nice Sophia Antipolis, Institute for Research on Cancer and Aging of Nice, CNRS UMR 7284, INSERM U1081, Centre Antoine Lacassagne, Nice, France
| | - Sandy Giuliano
- University of Nice Sophia Antipolis, Institute for Research on Cancer and Aging of Nice, CNRS UMR 7284, INSERM U1081, Centre Antoine Lacassagne, Nice, France
- Biomedical Department, Centre Scientifique de Monaco, Monaco, Principality of Monaco
| | - Damien Ambrosetti
- Central Laboratory of Pathology, Centre Hospitalier Universitaire (CHU) de Nice, Hôpital Pasteur, Nice, France
| | - Audrey Claren
- University of Nice Sophia Antipolis, Institute for Research on Cancer and Aging of Nice, CNRS UMR 7284, INSERM U1081, Centre Antoine Lacassagne, Nice, France
- Radiotherapy Department, Centre Antoine Lacassagne, Nice, France
| | - Papa Diogop Ndiaye
- University of Nice Sophia Antipolis, Institute for Research on Cancer and Aging of Nice, CNRS UMR 7284, INSERM U1081, Centre Antoine Lacassagne, Nice, France
| | - Michalis Mastri
- Center for Genetics and Pharmacology, Roswell Park Cancer Institute, Buffalo, New York
| | - Walid Moghrabi
- King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | | | - Marc Ettaiche
- Statistics Department, Centre Antoine Lacassagne, Nice, France
| | | | - Julien Parola
- University of Nice Sophia Antipolis, Institute for Research on Cancer and Aging of Nice, CNRS UMR 7284, INSERM U1081, Centre Antoine Lacassagne, Nice, France
| | - Valerie Vial
- Biomedical Department, Centre Scientifique de Monaco, Monaco, Principality of Monaco
| | - Marilena Lupu-Plesu
- University of Nice Sophia Antipolis, Institute for Research on Cancer and Aging of Nice, CNRS UMR 7284, INSERM U1081, Centre Antoine Lacassagne, Nice, France
| | | | - Alain Ravaud
- Service d'Oncologie Médicale, Centre Hospitalier Universitaire (CHU) de Bordeaux, Bordeaux, France
| | | | | | | | - John M Ebos
- Center for Genetics and Pharmacology, Roswell Park Cancer Institute, Buffalo, New York
| | - Khalid Saad Khabar
- King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Renaud Grépin
- Biomedical Department, Centre Scientifique de Monaco, Monaco, Principality of Monaco
| | - Gilles Pagès
- University of Nice Sophia Antipolis, Institute for Research on Cancer and Aging of Nice, CNRS UMR 7284, INSERM U1081, Centre Antoine Lacassagne, Nice, France.
| |
Collapse
|
41
|
Divers J, O'Shaughnessy J. Stomatitis Associated With Use of mTOR Inhibitors: Implications for Patients With Invasive Breast Cancer. Clin J Oncol Nurs 2017. [PMID: 26207713 DOI: 10.1188/15.cjon.468-474] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND The mammalian target of rapamycin (mTOR) inhibitor everolimus is approved (in combination with exemestane) for the treatment of postmenopausal women with advanced hormone receptor-positive, human epidermal growth factor receptor 2-negative breast cancer resistant to endocrine therapy. Stomatitis is among the most frequently reported dose-limiting adverse events associated with everolimus use, often requiring treatment interruption or dose reduction. OBJECTIVES This article aims to educate nurses on the identification and management of stomatitis associated with mTOR inhibitors in hormone receptor-positive advanced breast cancer and to assist nurses with additional management techniques to improve patient outcomes. METHODS An evaluation of the literature highlighting the incidence, identification, and management of stomatitis in cancer was performed with a particular focus on breast cancer. In addition, the experiences of the authors' cancer center on managing stomatitis are described. FINDINGS A growing body of clinical evidence shows the benefits of adding steroid-based mouth rinses to the treatment plan. Clinical experience provides additional insight into stomatitis preventive and management strategies for patients with breast cancer receiving treatment with everolimus.
Collapse
Affiliation(s)
- Josephine Divers
- Texas Oncology-Baylor Charles A. Sammons Cancer Center in Dallas
| | | |
Collapse
|
42
|
de Velasco G, Gray KP, Hamieh L, Urun Y, Carol HA, Fay AP, Signoretti S, Kwiatkowski DJ, McDermott DF, Freedman M, Pomerantz MM, Choueiri TK. Pharmacogenomic Markers of Targeted Therapy Toxicity in Patients with Metastatic Renal Cell Carcinoma. Eur Urol Focus 2016; 2:633-639. [PMID: 28723497 PMCID: PMC5520643 DOI: 10.1016/j.euf.2016.03.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 03/04/2016] [Accepted: 03/26/2016] [Indexed: 01/05/2023]
Abstract
BACKGROUND Targeted therapy (TT) in metastatic renal cell carcinoma (mRCC) may be associated with a high rate of toxicity that undermines treatment efficacy and patient quality of life. Polymorphisms in genes involved in the pharmacokinetic pathways of TTs may predict toxicity. OBJECTIVE To investigate whether selected single-nucleotide polymorphisms (SNPs) in three core genes involved in the metabolism and transport of sunitinib and the mTOR inhibitors everolimus and temsirolimus are associated with adverse events (AEs). DESIGN, SETTING, AND PARTICIPANTS Germline DNA was extracted from blood or normal kidney tissue from mRCC patients of Caucasian ethnicity in two cohorts treated with either sunitinib (n=159) or mTOR inhibitors (n=62). Six SNPs in three candidate genes (CYP3A4: rs2242480, rs4646437, and rs2246709; CYP3A5: rs15524; and ABCB1: rs2032582 and rs1045642) were analyzed. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Primary endpoints were grade ≥3 AEs for all patients; grade ≥3 hypertension in the sunitinib cohort, and any grade pneumonitis in the mTOR inhibitors cohort. A logistic regression model was used to assess the association between SNPs and AEs, with adjustment for relevant clinical factors. RESULTS AND LIMITATIONS In total, 221 samples were successfully genotyped for the selected SNPs. In the sunitinib cohort, the CYP3A4 rs464637 AG variant was associated with a lower risk of high-grade AEs (odds ratio 0.27, 95% confidence interval 0.08-0.88; p=0.03), but no SNPs were associated with hypertension. In the mTOR inhibitor cohort, none of the selected SNPs was associated with analyzed toxicities. CONCLUSIONS We observed an association between CYP3A4 polymorphisms and toxicity outcomes in mRCC patients treated with sunitinib, but not with everolimus or temsirolimus. Our findings are exploratory in nature, and further validation in independent and larger cohorts is needed. PATIENT SUMMARY We found that variants of CYP3A4, a gene involved in drug metabolism, are associated with sunitinib toxicity. This information may help in better selection of patients for targeted therapies in metastatic renal cell carcinoma.
Collapse
Affiliation(s)
| | - Kathryn P Gray
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Biostatistics and Computational Biology, Harvard School of Public Health, Boston, MA, USA
| | - Lana Hamieh
- Division of Pulmonary Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Yuksel Urun
- Department of Medical Oncology, Ankara University School of Medicine, Turkey
| | - Hallie A Carol
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Andre P Fay
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; PUCRS School of Medicine, Porto Alegre, Brazil
| | - Sabina Signoretti
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - David J Kwiatkowski
- Division of Pulmonary Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - David F McDermott
- Department of Medical Oncology, Beth-Israel Deaconess Medical Center, Boston, MA, USA
| | - Matthew Freedman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Mark M Pomerantz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Toni K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
43
|
Ravaud A, Motzer RJ, Pandha HS, George DJ, Pantuck AJ, Patel A, Chang YH, Escudier B, Donskov F, Magheli A, Carteni G, Laguerre B, Tomczak P, Breza J, Gerletti P, Lechuga M, Lin X, Martini JF, Ramaswamy K, Casey M, Staehler M, Patard JJ. Adjuvant Sunitinib in High-Risk Renal-Cell Carcinoma after Nephrectomy. N Engl J Med 2016; 375:2246-2254. [PMID: 27718781 DOI: 10.1056/nejmoa1611406] [Citation(s) in RCA: 549] [Impact Index Per Article: 68.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Sunitinib, a vascular endothelial growth factor pathway inhibitor, is an effective treatment for metastatic renal-cell carcinoma. We sought to determine the efficacy and safety of sunitinib in patients with locoregional renal-cell carcinoma at high risk for tumor recurrence after nephrectomy. METHODS In this randomized, double-blind, phase 3 trial, we assigned 615 patients with locoregional, high-risk clear-cell renal-cell carcinoma to receive either sunitinib (50 mg per day) or placebo on a 4-weeks-on, 2-weeks-off schedule for 1 year or until disease recurrence, unacceptable toxicity, or consent withdrawal. The primary end point was disease-free survival, according to blinded independent central review. Secondary end points included investigator-assessed disease-free survival, overall survival, and safety. RESULTS The median duration of disease-free survival was 6.8 years (95% confidence interval [CI], 5.8 to not reached) in the sunitinib group and 5.6 years (95% CI, 3.8 to 6.6) in the placebo group (hazard ratio, 0.76; 95% CI, 0.59 to 0.98; P=0.03). Overall survival data were not mature at the time of data cutoff. Dose reductions because of adverse events were more frequent in the sunitinib group than in the placebo group (34.3% vs. 2%), as were dose interruptions (46.4% vs. 13.2%) and discontinuations (28.1% vs. 5.6%). Grade 3 or 4 adverse events were more frequent in the sunitinib group (48.4% for grade 3 events and 12.1% for grade 4 events) than in the placebo group (15.8% and 3.6%, respectively). There was a similar incidence of serious adverse events in the two groups (21.9% for sunitinib vs. 17.1% for placebo); no deaths were attributed to toxic effects. CONCLUSIONS Among patients with locoregional clear-cell renal-cell carcinoma at high risk for tumor recurrence after nephrectomy, the median duration of disease-free survival was significantly longer in the sunitinib group than in the placebo group, at a cost of a higher rate of toxic events. (Funded by Pfizer; S-TRAC ClinicalTrials.gov number, NCT00375674 .).
Collapse
Affiliation(s)
- Alain Ravaud
- From the Department of Medical Oncology, Bordeaux University Hospital, Bordeaux (A.R.), Department of Medical Oncology, Institut Gustave Roussy, Villejuif (B.E.), Medical Oncology, Centre Eugene Marquis, Rennes (B.L.), and Department of Urology, Bicêtre Hospital, Paris-Saclay University, Le Kremlin Bicêtre (J.-J.P.) - all in France; Department of Medicine, Memorial Sloan Kettering Cancer Center (R.J.M.), and Pfizer (K.R.) - both in New York; Department of Clinical and Experimental Medicine, University of Surrey, Surrey, United Kingdom (H.S.P.); Division of Medical Oncology, Duke Cancer Institute, Durham, NC (D.J.G.); Department of Urology, Ronald Reagan UCLA Medical Center, Los Angeles (A.J.P.), and Pfizer, La Jolla (X.L., J.-F.M.) - both in California; Spire Roding Hospital, London (A.P.); Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan (Y.-H.C.); Department of Oncology, Aarhus University Hospital, Aarhus, Denmark (F.D.); Department of Urology, Charité Universitätsmedizin Berlin, Berlin (A.M.), and Department of Urology, University Hospital of Munich, Munich (M.S.) - both in Germany; Divisions of Oncology and Urology, Azienda Ospedaliera di Rilievo Nazionale A. Cardarelli, Naples, Italy (G.C.); Klinika Onkologii Oddzial Chemioterapii, Poznan, Poland (P.T.); Department of Urology, Slovak Medical University, Bratislava, Slovakia (J.B.); and Pfizer, Milan (P.G., M.L.), and Collegeville, PA (M.C.)
| | - Robert J Motzer
- From the Department of Medical Oncology, Bordeaux University Hospital, Bordeaux (A.R.), Department of Medical Oncology, Institut Gustave Roussy, Villejuif (B.E.), Medical Oncology, Centre Eugene Marquis, Rennes (B.L.), and Department of Urology, Bicêtre Hospital, Paris-Saclay University, Le Kremlin Bicêtre (J.-J.P.) - all in France; Department of Medicine, Memorial Sloan Kettering Cancer Center (R.J.M.), and Pfizer (K.R.) - both in New York; Department of Clinical and Experimental Medicine, University of Surrey, Surrey, United Kingdom (H.S.P.); Division of Medical Oncology, Duke Cancer Institute, Durham, NC (D.J.G.); Department of Urology, Ronald Reagan UCLA Medical Center, Los Angeles (A.J.P.), and Pfizer, La Jolla (X.L., J.-F.M.) - both in California; Spire Roding Hospital, London (A.P.); Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan (Y.-H.C.); Department of Oncology, Aarhus University Hospital, Aarhus, Denmark (F.D.); Department of Urology, Charité Universitätsmedizin Berlin, Berlin (A.M.), and Department of Urology, University Hospital of Munich, Munich (M.S.) - both in Germany; Divisions of Oncology and Urology, Azienda Ospedaliera di Rilievo Nazionale A. Cardarelli, Naples, Italy (G.C.); Klinika Onkologii Oddzial Chemioterapii, Poznan, Poland (P.T.); Department of Urology, Slovak Medical University, Bratislava, Slovakia (J.B.); and Pfizer, Milan (P.G., M.L.), and Collegeville, PA (M.C.)
| | - Hardev S Pandha
- From the Department of Medical Oncology, Bordeaux University Hospital, Bordeaux (A.R.), Department of Medical Oncology, Institut Gustave Roussy, Villejuif (B.E.), Medical Oncology, Centre Eugene Marquis, Rennes (B.L.), and Department of Urology, Bicêtre Hospital, Paris-Saclay University, Le Kremlin Bicêtre (J.-J.P.) - all in France; Department of Medicine, Memorial Sloan Kettering Cancer Center (R.J.M.), and Pfizer (K.R.) - both in New York; Department of Clinical and Experimental Medicine, University of Surrey, Surrey, United Kingdom (H.S.P.); Division of Medical Oncology, Duke Cancer Institute, Durham, NC (D.J.G.); Department of Urology, Ronald Reagan UCLA Medical Center, Los Angeles (A.J.P.), and Pfizer, La Jolla (X.L., J.-F.M.) - both in California; Spire Roding Hospital, London (A.P.); Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan (Y.-H.C.); Department of Oncology, Aarhus University Hospital, Aarhus, Denmark (F.D.); Department of Urology, Charité Universitätsmedizin Berlin, Berlin (A.M.), and Department of Urology, University Hospital of Munich, Munich (M.S.) - both in Germany; Divisions of Oncology and Urology, Azienda Ospedaliera di Rilievo Nazionale A. Cardarelli, Naples, Italy (G.C.); Klinika Onkologii Oddzial Chemioterapii, Poznan, Poland (P.T.); Department of Urology, Slovak Medical University, Bratislava, Slovakia (J.B.); and Pfizer, Milan (P.G., M.L.), and Collegeville, PA (M.C.)
| | - Daniel J George
- From the Department of Medical Oncology, Bordeaux University Hospital, Bordeaux (A.R.), Department of Medical Oncology, Institut Gustave Roussy, Villejuif (B.E.), Medical Oncology, Centre Eugene Marquis, Rennes (B.L.), and Department of Urology, Bicêtre Hospital, Paris-Saclay University, Le Kremlin Bicêtre (J.-J.P.) - all in France; Department of Medicine, Memorial Sloan Kettering Cancer Center (R.J.M.), and Pfizer (K.R.) - both in New York; Department of Clinical and Experimental Medicine, University of Surrey, Surrey, United Kingdom (H.S.P.); Division of Medical Oncology, Duke Cancer Institute, Durham, NC (D.J.G.); Department of Urology, Ronald Reagan UCLA Medical Center, Los Angeles (A.J.P.), and Pfizer, La Jolla (X.L., J.-F.M.) - both in California; Spire Roding Hospital, London (A.P.); Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan (Y.-H.C.); Department of Oncology, Aarhus University Hospital, Aarhus, Denmark (F.D.); Department of Urology, Charité Universitätsmedizin Berlin, Berlin (A.M.), and Department of Urology, University Hospital of Munich, Munich (M.S.) - both in Germany; Divisions of Oncology and Urology, Azienda Ospedaliera di Rilievo Nazionale A. Cardarelli, Naples, Italy (G.C.); Klinika Onkologii Oddzial Chemioterapii, Poznan, Poland (P.T.); Department of Urology, Slovak Medical University, Bratislava, Slovakia (J.B.); and Pfizer, Milan (P.G., M.L.), and Collegeville, PA (M.C.)
| | - Allan J Pantuck
- From the Department of Medical Oncology, Bordeaux University Hospital, Bordeaux (A.R.), Department of Medical Oncology, Institut Gustave Roussy, Villejuif (B.E.), Medical Oncology, Centre Eugene Marquis, Rennes (B.L.), and Department of Urology, Bicêtre Hospital, Paris-Saclay University, Le Kremlin Bicêtre (J.-J.P.) - all in France; Department of Medicine, Memorial Sloan Kettering Cancer Center (R.J.M.), and Pfizer (K.R.) - both in New York; Department of Clinical and Experimental Medicine, University of Surrey, Surrey, United Kingdom (H.S.P.); Division of Medical Oncology, Duke Cancer Institute, Durham, NC (D.J.G.); Department of Urology, Ronald Reagan UCLA Medical Center, Los Angeles (A.J.P.), and Pfizer, La Jolla (X.L., J.-F.M.) - both in California; Spire Roding Hospital, London (A.P.); Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan (Y.-H.C.); Department of Oncology, Aarhus University Hospital, Aarhus, Denmark (F.D.); Department of Urology, Charité Universitätsmedizin Berlin, Berlin (A.M.), and Department of Urology, University Hospital of Munich, Munich (M.S.) - both in Germany; Divisions of Oncology and Urology, Azienda Ospedaliera di Rilievo Nazionale A. Cardarelli, Naples, Italy (G.C.); Klinika Onkologii Oddzial Chemioterapii, Poznan, Poland (P.T.); Department of Urology, Slovak Medical University, Bratislava, Slovakia (J.B.); and Pfizer, Milan (P.G., M.L.), and Collegeville, PA (M.C.)
| | - Anup Patel
- From the Department of Medical Oncology, Bordeaux University Hospital, Bordeaux (A.R.), Department of Medical Oncology, Institut Gustave Roussy, Villejuif (B.E.), Medical Oncology, Centre Eugene Marquis, Rennes (B.L.), and Department of Urology, Bicêtre Hospital, Paris-Saclay University, Le Kremlin Bicêtre (J.-J.P.) - all in France; Department of Medicine, Memorial Sloan Kettering Cancer Center (R.J.M.), and Pfizer (K.R.) - both in New York; Department of Clinical and Experimental Medicine, University of Surrey, Surrey, United Kingdom (H.S.P.); Division of Medical Oncology, Duke Cancer Institute, Durham, NC (D.J.G.); Department of Urology, Ronald Reagan UCLA Medical Center, Los Angeles (A.J.P.), and Pfizer, La Jolla (X.L., J.-F.M.) - both in California; Spire Roding Hospital, London (A.P.); Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan (Y.-H.C.); Department of Oncology, Aarhus University Hospital, Aarhus, Denmark (F.D.); Department of Urology, Charité Universitätsmedizin Berlin, Berlin (A.M.), and Department of Urology, University Hospital of Munich, Munich (M.S.) - both in Germany; Divisions of Oncology and Urology, Azienda Ospedaliera di Rilievo Nazionale A. Cardarelli, Naples, Italy (G.C.); Klinika Onkologii Oddzial Chemioterapii, Poznan, Poland (P.T.); Department of Urology, Slovak Medical University, Bratislava, Slovakia (J.B.); and Pfizer, Milan (P.G., M.L.), and Collegeville, PA (M.C.)
| | - Yen-Hwa Chang
- From the Department of Medical Oncology, Bordeaux University Hospital, Bordeaux (A.R.), Department of Medical Oncology, Institut Gustave Roussy, Villejuif (B.E.), Medical Oncology, Centre Eugene Marquis, Rennes (B.L.), and Department of Urology, Bicêtre Hospital, Paris-Saclay University, Le Kremlin Bicêtre (J.-J.P.) - all in France; Department of Medicine, Memorial Sloan Kettering Cancer Center (R.J.M.), and Pfizer (K.R.) - both in New York; Department of Clinical and Experimental Medicine, University of Surrey, Surrey, United Kingdom (H.S.P.); Division of Medical Oncology, Duke Cancer Institute, Durham, NC (D.J.G.); Department of Urology, Ronald Reagan UCLA Medical Center, Los Angeles (A.J.P.), and Pfizer, La Jolla (X.L., J.-F.M.) - both in California; Spire Roding Hospital, London (A.P.); Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan (Y.-H.C.); Department of Oncology, Aarhus University Hospital, Aarhus, Denmark (F.D.); Department of Urology, Charité Universitätsmedizin Berlin, Berlin (A.M.), and Department of Urology, University Hospital of Munich, Munich (M.S.) - both in Germany; Divisions of Oncology and Urology, Azienda Ospedaliera di Rilievo Nazionale A. Cardarelli, Naples, Italy (G.C.); Klinika Onkologii Oddzial Chemioterapii, Poznan, Poland (P.T.); Department of Urology, Slovak Medical University, Bratislava, Slovakia (J.B.); and Pfizer, Milan (P.G., M.L.), and Collegeville, PA (M.C.)
| | - Bernard Escudier
- From the Department of Medical Oncology, Bordeaux University Hospital, Bordeaux (A.R.), Department of Medical Oncology, Institut Gustave Roussy, Villejuif (B.E.), Medical Oncology, Centre Eugene Marquis, Rennes (B.L.), and Department of Urology, Bicêtre Hospital, Paris-Saclay University, Le Kremlin Bicêtre (J.-J.P.) - all in France; Department of Medicine, Memorial Sloan Kettering Cancer Center (R.J.M.), and Pfizer (K.R.) - both in New York; Department of Clinical and Experimental Medicine, University of Surrey, Surrey, United Kingdom (H.S.P.); Division of Medical Oncology, Duke Cancer Institute, Durham, NC (D.J.G.); Department of Urology, Ronald Reagan UCLA Medical Center, Los Angeles (A.J.P.), and Pfizer, La Jolla (X.L., J.-F.M.) - both in California; Spire Roding Hospital, London (A.P.); Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan (Y.-H.C.); Department of Oncology, Aarhus University Hospital, Aarhus, Denmark (F.D.); Department of Urology, Charité Universitätsmedizin Berlin, Berlin (A.M.), and Department of Urology, University Hospital of Munich, Munich (M.S.) - both in Germany; Divisions of Oncology and Urology, Azienda Ospedaliera di Rilievo Nazionale A. Cardarelli, Naples, Italy (G.C.); Klinika Onkologii Oddzial Chemioterapii, Poznan, Poland (P.T.); Department of Urology, Slovak Medical University, Bratislava, Slovakia (J.B.); and Pfizer, Milan (P.G., M.L.), and Collegeville, PA (M.C.)
| | - Frede Donskov
- From the Department of Medical Oncology, Bordeaux University Hospital, Bordeaux (A.R.), Department of Medical Oncology, Institut Gustave Roussy, Villejuif (B.E.), Medical Oncology, Centre Eugene Marquis, Rennes (B.L.), and Department of Urology, Bicêtre Hospital, Paris-Saclay University, Le Kremlin Bicêtre (J.-J.P.) - all in France; Department of Medicine, Memorial Sloan Kettering Cancer Center (R.J.M.), and Pfizer (K.R.) - both in New York; Department of Clinical and Experimental Medicine, University of Surrey, Surrey, United Kingdom (H.S.P.); Division of Medical Oncology, Duke Cancer Institute, Durham, NC (D.J.G.); Department of Urology, Ronald Reagan UCLA Medical Center, Los Angeles (A.J.P.), and Pfizer, La Jolla (X.L., J.-F.M.) - both in California; Spire Roding Hospital, London (A.P.); Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan (Y.-H.C.); Department of Oncology, Aarhus University Hospital, Aarhus, Denmark (F.D.); Department of Urology, Charité Universitätsmedizin Berlin, Berlin (A.M.), and Department of Urology, University Hospital of Munich, Munich (M.S.) - both in Germany; Divisions of Oncology and Urology, Azienda Ospedaliera di Rilievo Nazionale A. Cardarelli, Naples, Italy (G.C.); Klinika Onkologii Oddzial Chemioterapii, Poznan, Poland (P.T.); Department of Urology, Slovak Medical University, Bratislava, Slovakia (J.B.); and Pfizer, Milan (P.G., M.L.), and Collegeville, PA (M.C.)
| | - Ahmed Magheli
- From the Department of Medical Oncology, Bordeaux University Hospital, Bordeaux (A.R.), Department of Medical Oncology, Institut Gustave Roussy, Villejuif (B.E.), Medical Oncology, Centre Eugene Marquis, Rennes (B.L.), and Department of Urology, Bicêtre Hospital, Paris-Saclay University, Le Kremlin Bicêtre (J.-J.P.) - all in France; Department of Medicine, Memorial Sloan Kettering Cancer Center (R.J.M.), and Pfizer (K.R.) - both in New York; Department of Clinical and Experimental Medicine, University of Surrey, Surrey, United Kingdom (H.S.P.); Division of Medical Oncology, Duke Cancer Institute, Durham, NC (D.J.G.); Department of Urology, Ronald Reagan UCLA Medical Center, Los Angeles (A.J.P.), and Pfizer, La Jolla (X.L., J.-F.M.) - both in California; Spire Roding Hospital, London (A.P.); Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan (Y.-H.C.); Department of Oncology, Aarhus University Hospital, Aarhus, Denmark (F.D.); Department of Urology, Charité Universitätsmedizin Berlin, Berlin (A.M.), and Department of Urology, University Hospital of Munich, Munich (M.S.) - both in Germany; Divisions of Oncology and Urology, Azienda Ospedaliera di Rilievo Nazionale A. Cardarelli, Naples, Italy (G.C.); Klinika Onkologii Oddzial Chemioterapii, Poznan, Poland (P.T.); Department of Urology, Slovak Medical University, Bratislava, Slovakia (J.B.); and Pfizer, Milan (P.G., M.L.), and Collegeville, PA (M.C.)
| | - Giacomo Carteni
- From the Department of Medical Oncology, Bordeaux University Hospital, Bordeaux (A.R.), Department of Medical Oncology, Institut Gustave Roussy, Villejuif (B.E.), Medical Oncology, Centre Eugene Marquis, Rennes (B.L.), and Department of Urology, Bicêtre Hospital, Paris-Saclay University, Le Kremlin Bicêtre (J.-J.P.) - all in France; Department of Medicine, Memorial Sloan Kettering Cancer Center (R.J.M.), and Pfizer (K.R.) - both in New York; Department of Clinical and Experimental Medicine, University of Surrey, Surrey, United Kingdom (H.S.P.); Division of Medical Oncology, Duke Cancer Institute, Durham, NC (D.J.G.); Department of Urology, Ronald Reagan UCLA Medical Center, Los Angeles (A.J.P.), and Pfizer, La Jolla (X.L., J.-F.M.) - both in California; Spire Roding Hospital, London (A.P.); Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan (Y.-H.C.); Department of Oncology, Aarhus University Hospital, Aarhus, Denmark (F.D.); Department of Urology, Charité Universitätsmedizin Berlin, Berlin (A.M.), and Department of Urology, University Hospital of Munich, Munich (M.S.) - both in Germany; Divisions of Oncology and Urology, Azienda Ospedaliera di Rilievo Nazionale A. Cardarelli, Naples, Italy (G.C.); Klinika Onkologii Oddzial Chemioterapii, Poznan, Poland (P.T.); Department of Urology, Slovak Medical University, Bratislava, Slovakia (J.B.); and Pfizer, Milan (P.G., M.L.), and Collegeville, PA (M.C.)
| | - Brigitte Laguerre
- From the Department of Medical Oncology, Bordeaux University Hospital, Bordeaux (A.R.), Department of Medical Oncology, Institut Gustave Roussy, Villejuif (B.E.), Medical Oncology, Centre Eugene Marquis, Rennes (B.L.), and Department of Urology, Bicêtre Hospital, Paris-Saclay University, Le Kremlin Bicêtre (J.-J.P.) - all in France; Department of Medicine, Memorial Sloan Kettering Cancer Center (R.J.M.), and Pfizer (K.R.) - both in New York; Department of Clinical and Experimental Medicine, University of Surrey, Surrey, United Kingdom (H.S.P.); Division of Medical Oncology, Duke Cancer Institute, Durham, NC (D.J.G.); Department of Urology, Ronald Reagan UCLA Medical Center, Los Angeles (A.J.P.), and Pfizer, La Jolla (X.L., J.-F.M.) - both in California; Spire Roding Hospital, London (A.P.); Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan (Y.-H.C.); Department of Oncology, Aarhus University Hospital, Aarhus, Denmark (F.D.); Department of Urology, Charité Universitätsmedizin Berlin, Berlin (A.M.), and Department of Urology, University Hospital of Munich, Munich (M.S.) - both in Germany; Divisions of Oncology and Urology, Azienda Ospedaliera di Rilievo Nazionale A. Cardarelli, Naples, Italy (G.C.); Klinika Onkologii Oddzial Chemioterapii, Poznan, Poland (P.T.); Department of Urology, Slovak Medical University, Bratislava, Slovakia (J.B.); and Pfizer, Milan (P.G., M.L.), and Collegeville, PA (M.C.)
| | - Piotr Tomczak
- From the Department of Medical Oncology, Bordeaux University Hospital, Bordeaux (A.R.), Department of Medical Oncology, Institut Gustave Roussy, Villejuif (B.E.), Medical Oncology, Centre Eugene Marquis, Rennes (B.L.), and Department of Urology, Bicêtre Hospital, Paris-Saclay University, Le Kremlin Bicêtre (J.-J.P.) - all in France; Department of Medicine, Memorial Sloan Kettering Cancer Center (R.J.M.), and Pfizer (K.R.) - both in New York; Department of Clinical and Experimental Medicine, University of Surrey, Surrey, United Kingdom (H.S.P.); Division of Medical Oncology, Duke Cancer Institute, Durham, NC (D.J.G.); Department of Urology, Ronald Reagan UCLA Medical Center, Los Angeles (A.J.P.), and Pfizer, La Jolla (X.L., J.-F.M.) - both in California; Spire Roding Hospital, London (A.P.); Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan (Y.-H.C.); Department of Oncology, Aarhus University Hospital, Aarhus, Denmark (F.D.); Department of Urology, Charité Universitätsmedizin Berlin, Berlin (A.M.), and Department of Urology, University Hospital of Munich, Munich (M.S.) - both in Germany; Divisions of Oncology and Urology, Azienda Ospedaliera di Rilievo Nazionale A. Cardarelli, Naples, Italy (G.C.); Klinika Onkologii Oddzial Chemioterapii, Poznan, Poland (P.T.); Department of Urology, Slovak Medical University, Bratislava, Slovakia (J.B.); and Pfizer, Milan (P.G., M.L.), and Collegeville, PA (M.C.)
| | - Jan Breza
- From the Department of Medical Oncology, Bordeaux University Hospital, Bordeaux (A.R.), Department of Medical Oncology, Institut Gustave Roussy, Villejuif (B.E.), Medical Oncology, Centre Eugene Marquis, Rennes (B.L.), and Department of Urology, Bicêtre Hospital, Paris-Saclay University, Le Kremlin Bicêtre (J.-J.P.) - all in France; Department of Medicine, Memorial Sloan Kettering Cancer Center (R.J.M.), and Pfizer (K.R.) - both in New York; Department of Clinical and Experimental Medicine, University of Surrey, Surrey, United Kingdom (H.S.P.); Division of Medical Oncology, Duke Cancer Institute, Durham, NC (D.J.G.); Department of Urology, Ronald Reagan UCLA Medical Center, Los Angeles (A.J.P.), and Pfizer, La Jolla (X.L., J.-F.M.) - both in California; Spire Roding Hospital, London (A.P.); Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan (Y.-H.C.); Department of Oncology, Aarhus University Hospital, Aarhus, Denmark (F.D.); Department of Urology, Charité Universitätsmedizin Berlin, Berlin (A.M.), and Department of Urology, University Hospital of Munich, Munich (M.S.) - both in Germany; Divisions of Oncology and Urology, Azienda Ospedaliera di Rilievo Nazionale A. Cardarelli, Naples, Italy (G.C.); Klinika Onkologii Oddzial Chemioterapii, Poznan, Poland (P.T.); Department of Urology, Slovak Medical University, Bratislava, Slovakia (J.B.); and Pfizer, Milan (P.G., M.L.), and Collegeville, PA (M.C.)
| | - Paola Gerletti
- From the Department of Medical Oncology, Bordeaux University Hospital, Bordeaux (A.R.), Department of Medical Oncology, Institut Gustave Roussy, Villejuif (B.E.), Medical Oncology, Centre Eugene Marquis, Rennes (B.L.), and Department of Urology, Bicêtre Hospital, Paris-Saclay University, Le Kremlin Bicêtre (J.-J.P.) - all in France; Department of Medicine, Memorial Sloan Kettering Cancer Center (R.J.M.), and Pfizer (K.R.) - both in New York; Department of Clinical and Experimental Medicine, University of Surrey, Surrey, United Kingdom (H.S.P.); Division of Medical Oncology, Duke Cancer Institute, Durham, NC (D.J.G.); Department of Urology, Ronald Reagan UCLA Medical Center, Los Angeles (A.J.P.), and Pfizer, La Jolla (X.L., J.-F.M.) - both in California; Spire Roding Hospital, London (A.P.); Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan (Y.-H.C.); Department of Oncology, Aarhus University Hospital, Aarhus, Denmark (F.D.); Department of Urology, Charité Universitätsmedizin Berlin, Berlin (A.M.), and Department of Urology, University Hospital of Munich, Munich (M.S.) - both in Germany; Divisions of Oncology and Urology, Azienda Ospedaliera di Rilievo Nazionale A. Cardarelli, Naples, Italy (G.C.); Klinika Onkologii Oddzial Chemioterapii, Poznan, Poland (P.T.); Department of Urology, Slovak Medical University, Bratislava, Slovakia (J.B.); and Pfizer, Milan (P.G., M.L.), and Collegeville, PA (M.C.)
| | - Mariajose Lechuga
- From the Department of Medical Oncology, Bordeaux University Hospital, Bordeaux (A.R.), Department of Medical Oncology, Institut Gustave Roussy, Villejuif (B.E.), Medical Oncology, Centre Eugene Marquis, Rennes (B.L.), and Department of Urology, Bicêtre Hospital, Paris-Saclay University, Le Kremlin Bicêtre (J.-J.P.) - all in France; Department of Medicine, Memorial Sloan Kettering Cancer Center (R.J.M.), and Pfizer (K.R.) - both in New York; Department of Clinical and Experimental Medicine, University of Surrey, Surrey, United Kingdom (H.S.P.); Division of Medical Oncology, Duke Cancer Institute, Durham, NC (D.J.G.); Department of Urology, Ronald Reagan UCLA Medical Center, Los Angeles (A.J.P.), and Pfizer, La Jolla (X.L., J.-F.M.) - both in California; Spire Roding Hospital, London (A.P.); Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan (Y.-H.C.); Department of Oncology, Aarhus University Hospital, Aarhus, Denmark (F.D.); Department of Urology, Charité Universitätsmedizin Berlin, Berlin (A.M.), and Department of Urology, University Hospital of Munich, Munich (M.S.) - both in Germany; Divisions of Oncology and Urology, Azienda Ospedaliera di Rilievo Nazionale A. Cardarelli, Naples, Italy (G.C.); Klinika Onkologii Oddzial Chemioterapii, Poznan, Poland (P.T.); Department of Urology, Slovak Medical University, Bratislava, Slovakia (J.B.); and Pfizer, Milan (P.G., M.L.), and Collegeville, PA (M.C.)
| | - Xun Lin
- From the Department of Medical Oncology, Bordeaux University Hospital, Bordeaux (A.R.), Department of Medical Oncology, Institut Gustave Roussy, Villejuif (B.E.), Medical Oncology, Centre Eugene Marquis, Rennes (B.L.), and Department of Urology, Bicêtre Hospital, Paris-Saclay University, Le Kremlin Bicêtre (J.-J.P.) - all in France; Department of Medicine, Memorial Sloan Kettering Cancer Center (R.J.M.), and Pfizer (K.R.) - both in New York; Department of Clinical and Experimental Medicine, University of Surrey, Surrey, United Kingdom (H.S.P.); Division of Medical Oncology, Duke Cancer Institute, Durham, NC (D.J.G.); Department of Urology, Ronald Reagan UCLA Medical Center, Los Angeles (A.J.P.), and Pfizer, La Jolla (X.L., J.-F.M.) - both in California; Spire Roding Hospital, London (A.P.); Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan (Y.-H.C.); Department of Oncology, Aarhus University Hospital, Aarhus, Denmark (F.D.); Department of Urology, Charité Universitätsmedizin Berlin, Berlin (A.M.), and Department of Urology, University Hospital of Munich, Munich (M.S.) - both in Germany; Divisions of Oncology and Urology, Azienda Ospedaliera di Rilievo Nazionale A. Cardarelli, Naples, Italy (G.C.); Klinika Onkologii Oddzial Chemioterapii, Poznan, Poland (P.T.); Department of Urology, Slovak Medical University, Bratislava, Slovakia (J.B.); and Pfizer, Milan (P.G., M.L.), and Collegeville, PA (M.C.)
| | - Jean-Francois Martini
- From the Department of Medical Oncology, Bordeaux University Hospital, Bordeaux (A.R.), Department of Medical Oncology, Institut Gustave Roussy, Villejuif (B.E.), Medical Oncology, Centre Eugene Marquis, Rennes (B.L.), and Department of Urology, Bicêtre Hospital, Paris-Saclay University, Le Kremlin Bicêtre (J.-J.P.) - all in France; Department of Medicine, Memorial Sloan Kettering Cancer Center (R.J.M.), and Pfizer (K.R.) - both in New York; Department of Clinical and Experimental Medicine, University of Surrey, Surrey, United Kingdom (H.S.P.); Division of Medical Oncology, Duke Cancer Institute, Durham, NC (D.J.G.); Department of Urology, Ronald Reagan UCLA Medical Center, Los Angeles (A.J.P.), and Pfizer, La Jolla (X.L., J.-F.M.) - both in California; Spire Roding Hospital, London (A.P.); Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan (Y.-H.C.); Department of Oncology, Aarhus University Hospital, Aarhus, Denmark (F.D.); Department of Urology, Charité Universitätsmedizin Berlin, Berlin (A.M.), and Department of Urology, University Hospital of Munich, Munich (M.S.) - both in Germany; Divisions of Oncology and Urology, Azienda Ospedaliera di Rilievo Nazionale A. Cardarelli, Naples, Italy (G.C.); Klinika Onkologii Oddzial Chemioterapii, Poznan, Poland (P.T.); Department of Urology, Slovak Medical University, Bratislava, Slovakia (J.B.); and Pfizer, Milan (P.G., M.L.), and Collegeville, PA (M.C.)
| | - Krishnan Ramaswamy
- From the Department of Medical Oncology, Bordeaux University Hospital, Bordeaux (A.R.), Department of Medical Oncology, Institut Gustave Roussy, Villejuif (B.E.), Medical Oncology, Centre Eugene Marquis, Rennes (B.L.), and Department of Urology, Bicêtre Hospital, Paris-Saclay University, Le Kremlin Bicêtre (J.-J.P.) - all in France; Department of Medicine, Memorial Sloan Kettering Cancer Center (R.J.M.), and Pfizer (K.R.) - both in New York; Department of Clinical and Experimental Medicine, University of Surrey, Surrey, United Kingdom (H.S.P.); Division of Medical Oncology, Duke Cancer Institute, Durham, NC (D.J.G.); Department of Urology, Ronald Reagan UCLA Medical Center, Los Angeles (A.J.P.), and Pfizer, La Jolla (X.L., J.-F.M.) - both in California; Spire Roding Hospital, London (A.P.); Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan (Y.-H.C.); Department of Oncology, Aarhus University Hospital, Aarhus, Denmark (F.D.); Department of Urology, Charité Universitätsmedizin Berlin, Berlin (A.M.), and Department of Urology, University Hospital of Munich, Munich (M.S.) - both in Germany; Divisions of Oncology and Urology, Azienda Ospedaliera di Rilievo Nazionale A. Cardarelli, Naples, Italy (G.C.); Klinika Onkologii Oddzial Chemioterapii, Poznan, Poland (P.T.); Department of Urology, Slovak Medical University, Bratislava, Slovakia (J.B.); and Pfizer, Milan (P.G., M.L.), and Collegeville, PA (M.C.)
| | - Michelle Casey
- From the Department of Medical Oncology, Bordeaux University Hospital, Bordeaux (A.R.), Department of Medical Oncology, Institut Gustave Roussy, Villejuif (B.E.), Medical Oncology, Centre Eugene Marquis, Rennes (B.L.), and Department of Urology, Bicêtre Hospital, Paris-Saclay University, Le Kremlin Bicêtre (J.-J.P.) - all in France; Department of Medicine, Memorial Sloan Kettering Cancer Center (R.J.M.), and Pfizer (K.R.) - both in New York; Department of Clinical and Experimental Medicine, University of Surrey, Surrey, United Kingdom (H.S.P.); Division of Medical Oncology, Duke Cancer Institute, Durham, NC (D.J.G.); Department of Urology, Ronald Reagan UCLA Medical Center, Los Angeles (A.J.P.), and Pfizer, La Jolla (X.L., J.-F.M.) - both in California; Spire Roding Hospital, London (A.P.); Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan (Y.-H.C.); Department of Oncology, Aarhus University Hospital, Aarhus, Denmark (F.D.); Department of Urology, Charité Universitätsmedizin Berlin, Berlin (A.M.), and Department of Urology, University Hospital of Munich, Munich (M.S.) - both in Germany; Divisions of Oncology and Urology, Azienda Ospedaliera di Rilievo Nazionale A. Cardarelli, Naples, Italy (G.C.); Klinika Onkologii Oddzial Chemioterapii, Poznan, Poland (P.T.); Department of Urology, Slovak Medical University, Bratislava, Slovakia (J.B.); and Pfizer, Milan (P.G., M.L.), and Collegeville, PA (M.C.)
| | - Michael Staehler
- From the Department of Medical Oncology, Bordeaux University Hospital, Bordeaux (A.R.), Department of Medical Oncology, Institut Gustave Roussy, Villejuif (B.E.), Medical Oncology, Centre Eugene Marquis, Rennes (B.L.), and Department of Urology, Bicêtre Hospital, Paris-Saclay University, Le Kremlin Bicêtre (J.-J.P.) - all in France; Department of Medicine, Memorial Sloan Kettering Cancer Center (R.J.M.), and Pfizer (K.R.) - both in New York; Department of Clinical and Experimental Medicine, University of Surrey, Surrey, United Kingdom (H.S.P.); Division of Medical Oncology, Duke Cancer Institute, Durham, NC (D.J.G.); Department of Urology, Ronald Reagan UCLA Medical Center, Los Angeles (A.J.P.), and Pfizer, La Jolla (X.L., J.-F.M.) - both in California; Spire Roding Hospital, London (A.P.); Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan (Y.-H.C.); Department of Oncology, Aarhus University Hospital, Aarhus, Denmark (F.D.); Department of Urology, Charité Universitätsmedizin Berlin, Berlin (A.M.), and Department of Urology, University Hospital of Munich, Munich (M.S.) - both in Germany; Divisions of Oncology and Urology, Azienda Ospedaliera di Rilievo Nazionale A. Cardarelli, Naples, Italy (G.C.); Klinika Onkologii Oddzial Chemioterapii, Poznan, Poland (P.T.); Department of Urology, Slovak Medical University, Bratislava, Slovakia (J.B.); and Pfizer, Milan (P.G., M.L.), and Collegeville, PA (M.C.)
| | - Jean-Jacques Patard
- From the Department of Medical Oncology, Bordeaux University Hospital, Bordeaux (A.R.), Department of Medical Oncology, Institut Gustave Roussy, Villejuif (B.E.), Medical Oncology, Centre Eugene Marquis, Rennes (B.L.), and Department of Urology, Bicêtre Hospital, Paris-Saclay University, Le Kremlin Bicêtre (J.-J.P.) - all in France; Department of Medicine, Memorial Sloan Kettering Cancer Center (R.J.M.), and Pfizer (K.R.) - both in New York; Department of Clinical and Experimental Medicine, University of Surrey, Surrey, United Kingdom (H.S.P.); Division of Medical Oncology, Duke Cancer Institute, Durham, NC (D.J.G.); Department of Urology, Ronald Reagan UCLA Medical Center, Los Angeles (A.J.P.), and Pfizer, La Jolla (X.L., J.-F.M.) - both in California; Spire Roding Hospital, London (A.P.); Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan (Y.-H.C.); Department of Oncology, Aarhus University Hospital, Aarhus, Denmark (F.D.); Department of Urology, Charité Universitätsmedizin Berlin, Berlin (A.M.), and Department of Urology, University Hospital of Munich, Munich (M.S.) - both in Germany; Divisions of Oncology and Urology, Azienda Ospedaliera di Rilievo Nazionale A. Cardarelli, Naples, Italy (G.C.); Klinika Onkologii Oddzial Chemioterapii, Poznan, Poland (P.T.); Department of Urology, Slovak Medical University, Bratislava, Slovakia (J.B.); and Pfizer, Milan (P.G., M.L.), and Collegeville, PA (M.C.)
| |
Collapse
|
44
|
Prospective Evaluation of the Impact of Antiangiogenic Treatment on Cognitive Functions in Metastatic Renal Cancer. Eur Urol Focus 2016; 2:642-649. [DOI: 10.1016/j.euf.2016.04.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 03/31/2016] [Accepted: 04/19/2016] [Indexed: 12/21/2022]
|
45
|
|
46
|
Escudier B, Lougheed JC, Albiges L. Cabozantinib for the treatment of renal cell carcinoma. Expert Opin Pharmacother 2016; 17:2499-2504. [PMID: 27835047 DOI: 10.1080/14656566.2016.1258059] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Agents that target the vascular endothelial growth factor (VEGF) or mammalian target of rapamycin (mTOR) pathway as well as the PD-1 checkpoint inhibitor nivolumab are standard therapies for advanced renal cell carcinoma (RCC). Recently, cabozantinib, an inhibitor of MET, VEGF receptors, and AXL, was approved by the FDA and European Commission based on improved progression-free survival (PFS), overall survival (OS), and objective response rate (ORR) compared to standard of care treatment with everolimus in a randomized phase 3 trial in advanced RCC after prior VEGFR-tyrosine kinase inhibitor (TKI) therapy. Areas covered:The preclinical development and scientific rationale, pharmacokinetics, and clinical efficacy and safety of cabozantinib for the treatment of advanced RCC are reviewed. The use of cabozantinib in clinical practice with the growing number of available treatments for advanced RCC is discussed. Expert opinion: Cabozantinib is the only therapy for advanced RCC that has improved PFS, ORR, and OS in a pivotal phase 3 trial after prior antiangiogenic therapy. While no clinical trials have been published comparing cabozantinib with another TKI, available clinical data suggest it could be the most efficacious TKI for second-line therapy. Preliminary encouraging results have also been reported in a phase 2 trial in untreated poor- and intermediate- risk patients with RCC, indicating that treatment with cabozantinib may also be beneficial in the first-line setting.
Collapse
Affiliation(s)
- Bernard Escudier
- a Department of Medical Oncology , Gustave Roussy , Villejuif , France
| | - Julie C Lougheed
- b Department of Medical Oncology , Exelixis, Inc. , South San Francisco , California
| | - Laurence Albiges
- a Department of Medical Oncology , Gustave Roussy , Villejuif , France
| |
Collapse
|
47
|
Wang J, Xu Y, Zhu L, Zou Y, Kong W, Dong B, Huang J, Chen Y, Xue W, Huang Y, Zhang J. High Expression of Stearoyl-CoA Desaturase 1 Predicts Poor Prognosis in Patients with Clear-Cell Renal Cell Carcinoma. PLoS One 2016; 11:e0166231. [PMID: 27861513 PMCID: PMC5115711 DOI: 10.1371/journal.pone.0166231] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 10/04/2016] [Indexed: 01/20/2023] Open
Abstract
Stearoyl-CoA desaturase 1 (SCD1), the rate-limiting enzymes in the biosynthesis of monounsaturated fatty acids from saturated fatty acids, have been gradually recognized as a potential therapeutic target for various malignancies, particularly in clear-cell renal cell carcinoma (ccRCC). However, the prognostic value of SCD1 in ccRCC is still unknown. The aim of this study is to evaluate the clinical significance of SCD1 expression in patients with ccRCC. SCD1 expression in tumor tissues obtained from 359 patients who underwent nephrectomy for ccRCC are retrospectively assessed. During a median follow-up of 63 months (range: 1–144month), 56 patients in total died before the last follow-up in this study. Survival curves were plotted with the Kaplan–Meier method and compared with the log-rank test. Meanwhile, univariate and multivariate Cox regression models were applied to evaluate the prognostic value of SCD1 expression in overall survival (OS) for ccRCC patients. Moreover, SCD1 was enrolled into a newly built nomogram with factors selected by multivariate analysis, and the calibration was built to evaluate the predictive accuracy of nomogram. High SCD1 expression occurred in 61.6% (221/359) of ccRCC patients, which was significantly associated with age (p = 0.030), TNM stage (p = 0.021), pN stage (p = 0.014), Fuhrman grade (p = 0.014) and tumor sizes (p = 0.040). In multivariate analysis, SCD1 expression was confirmed as an adverse independent prognostic factor for OS. The prognostic accuracy of TNM stage, Fuhrman grade and tumor sizes was significantly increased when SCD1 expression was added. The independent prognostic factors, pT stage, pN stage, Fuhrman grade and tumor sizes, as well as SCD1 expression were integrated to establish a predictive nomogram with high predictive accuracy. Calibration curves revealed optimal consistency between observations and prognosis. In conclusion, high SCD1 expression is an independent prognostic factor for OS in patients with ccRCC. Our data suggest that the expression of SCD1 might guide the clinical decisions for patients with ccRCC.
Collapse
Affiliation(s)
- Jianfeng Wang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yunze Xu
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Liangsong Zhu
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yun Zou
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wen Kong
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Baijun Dong
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jiwei Huang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yonghui Chen
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wei Xue
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yiran Huang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
- * E-mail: (JZ); (YH)
| | - Jin Zhang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
- * E-mail: (JZ); (YH)
| |
Collapse
|
48
|
Choueiri TK, Halabi S, Sanford BL, Hahn O, Michaelson MD, Walsh MK, Feldman DR, Olencki T, Picus J, Small EJ, Dakhil S, George DJ, Morris MJ. Cabozantinib Versus Sunitinib As Initial Targeted Therapy for Patients With Metastatic Renal Cell Carcinoma of Poor or Intermediate Risk: The Alliance A031203 CABOSUN Trial. J Clin Oncol 2016; 35:591-597. [PMID: 28199818 DOI: 10.1200/jco.2016.70.7398] [Citation(s) in RCA: 529] [Impact Index Per Article: 66.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Purpose Cabozantinib is an oral potent inhibitor of vascular endothelial growth factor receptor 2, MET, and AXL and is a standard second-line therapy for metastatic renal cell carcinoma (mRCC). This randomized phase II multicenter trial evaluated cabozantinib compared with sunitinib as first-line therapy in patients with mRCC. Patients and Methods Eligible patients had untreated clear cell mRCC and Eastern Cooperative Oncology Group performance status of 0 to 2 and were intermediate or poor risk per International Metastatic Renal Cell Carcinoma Database Consortium criteria. Patients were randomly assigned at a one-to-one ratio to cabozantinib (60 mg once per day) or sunitinib (50 mg once per day; 4 weeks on, 2 weeks off). Progression-free survival (PFS) was the primary end point. Objective response rate (ORR), overall survival, and safety were secondary end points. Results From July 2013 to April 2015, 157 patients were randomly assigned (cabozantinib, n = 79; sunitinib, n = 78). Compared with sunitinib, cabozantinib treatment significantly increased median PFS (8.2 v 5.6 months) and was associated with a 34% reduction in rate of progression or death (adjusted hazard ratio, 0.66; 95% CI, 0.46 to 0.95; one-sided P = .012). ORR was 33% (95% CI, 23 to 44) for cabozantinib versus 12% (95% CI, 5.4 to 21) for sunitinib. All-causality grade 3 or 4 adverse events were 67% for cabozantinib and 68% for sunitinib and included diarrhea (cabozantinib, 10% v sunitinib, 11%), fatigue (6% v 15%), hypertension (28% v 22%), palmar-plantar erythrodysesthesia (8% v 4%), and hematologic adverse events (3% v 22%). Conclusion Cabozantinib demonstrated a significant clinical benefit in PFS and ORR over standard-of-care sunitinib as first-line therapy in patients with intermediate- or poor-risk mRCC.
Collapse
Affiliation(s)
- Toni K Choueiri
- Toni K. Choueiri and Meghara K. Walsh, Dana-Farber Cancer Institute; M. Dror Michaelson, Massachusetts General Hospital Cancer Center, Boston, MA; Susan Halabi and Ben L. Sanford, Alliance Statistics and Data Center and Duke University; Daniel J. George, Duke Cancer Institute, Duke University Medical Center, Durham, NC; Olwen Hahn, Alliance Protocol Operations Office, Chicago, IL; Darren R. Feldman and Michael J. Morris, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University Medical Center, Columbus, OH; Joel Picus, Siteman Cancer Center, Washington University School of Medicine, St Louis, MO; Eric J. Small, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; and Shaker Dakhil, University of Kansas Wichita, Wichita, KS
| | - Susan Halabi
- Toni K. Choueiri and Meghara K. Walsh, Dana-Farber Cancer Institute; M. Dror Michaelson, Massachusetts General Hospital Cancer Center, Boston, MA; Susan Halabi and Ben L. Sanford, Alliance Statistics and Data Center and Duke University; Daniel J. George, Duke Cancer Institute, Duke University Medical Center, Durham, NC; Olwen Hahn, Alliance Protocol Operations Office, Chicago, IL; Darren R. Feldman and Michael J. Morris, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University Medical Center, Columbus, OH; Joel Picus, Siteman Cancer Center, Washington University School of Medicine, St Louis, MO; Eric J. Small, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; and Shaker Dakhil, University of Kansas Wichita, Wichita, KS
| | - Ben L Sanford
- Toni K. Choueiri and Meghara K. Walsh, Dana-Farber Cancer Institute; M. Dror Michaelson, Massachusetts General Hospital Cancer Center, Boston, MA; Susan Halabi and Ben L. Sanford, Alliance Statistics and Data Center and Duke University; Daniel J. George, Duke Cancer Institute, Duke University Medical Center, Durham, NC; Olwen Hahn, Alliance Protocol Operations Office, Chicago, IL; Darren R. Feldman and Michael J. Morris, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University Medical Center, Columbus, OH; Joel Picus, Siteman Cancer Center, Washington University School of Medicine, St Louis, MO; Eric J. Small, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; and Shaker Dakhil, University of Kansas Wichita, Wichita, KS
| | - Olwen Hahn
- Toni K. Choueiri and Meghara K. Walsh, Dana-Farber Cancer Institute; M. Dror Michaelson, Massachusetts General Hospital Cancer Center, Boston, MA; Susan Halabi and Ben L. Sanford, Alliance Statistics and Data Center and Duke University; Daniel J. George, Duke Cancer Institute, Duke University Medical Center, Durham, NC; Olwen Hahn, Alliance Protocol Operations Office, Chicago, IL; Darren R. Feldman and Michael J. Morris, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University Medical Center, Columbus, OH; Joel Picus, Siteman Cancer Center, Washington University School of Medicine, St Louis, MO; Eric J. Small, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; and Shaker Dakhil, University of Kansas Wichita, Wichita, KS
| | - M Dror Michaelson
- Toni K. Choueiri and Meghara K. Walsh, Dana-Farber Cancer Institute; M. Dror Michaelson, Massachusetts General Hospital Cancer Center, Boston, MA; Susan Halabi and Ben L. Sanford, Alliance Statistics and Data Center and Duke University; Daniel J. George, Duke Cancer Institute, Duke University Medical Center, Durham, NC; Olwen Hahn, Alliance Protocol Operations Office, Chicago, IL; Darren R. Feldman and Michael J. Morris, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University Medical Center, Columbus, OH; Joel Picus, Siteman Cancer Center, Washington University School of Medicine, St Louis, MO; Eric J. Small, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; and Shaker Dakhil, University of Kansas Wichita, Wichita, KS
| | - Meghara K Walsh
- Toni K. Choueiri and Meghara K. Walsh, Dana-Farber Cancer Institute; M. Dror Michaelson, Massachusetts General Hospital Cancer Center, Boston, MA; Susan Halabi and Ben L. Sanford, Alliance Statistics and Data Center and Duke University; Daniel J. George, Duke Cancer Institute, Duke University Medical Center, Durham, NC; Olwen Hahn, Alliance Protocol Operations Office, Chicago, IL; Darren R. Feldman and Michael J. Morris, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University Medical Center, Columbus, OH; Joel Picus, Siteman Cancer Center, Washington University School of Medicine, St Louis, MO; Eric J. Small, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; and Shaker Dakhil, University of Kansas Wichita, Wichita, KS
| | - Darren R Feldman
- Toni K. Choueiri and Meghara K. Walsh, Dana-Farber Cancer Institute; M. Dror Michaelson, Massachusetts General Hospital Cancer Center, Boston, MA; Susan Halabi and Ben L. Sanford, Alliance Statistics and Data Center and Duke University; Daniel J. George, Duke Cancer Institute, Duke University Medical Center, Durham, NC; Olwen Hahn, Alliance Protocol Operations Office, Chicago, IL; Darren R. Feldman and Michael J. Morris, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University Medical Center, Columbus, OH; Joel Picus, Siteman Cancer Center, Washington University School of Medicine, St Louis, MO; Eric J. Small, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; and Shaker Dakhil, University of Kansas Wichita, Wichita, KS
| | - Thomas Olencki
- Toni K. Choueiri and Meghara K. Walsh, Dana-Farber Cancer Institute; M. Dror Michaelson, Massachusetts General Hospital Cancer Center, Boston, MA; Susan Halabi and Ben L. Sanford, Alliance Statistics and Data Center and Duke University; Daniel J. George, Duke Cancer Institute, Duke University Medical Center, Durham, NC; Olwen Hahn, Alliance Protocol Operations Office, Chicago, IL; Darren R. Feldman and Michael J. Morris, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University Medical Center, Columbus, OH; Joel Picus, Siteman Cancer Center, Washington University School of Medicine, St Louis, MO; Eric J. Small, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; and Shaker Dakhil, University of Kansas Wichita, Wichita, KS
| | - Joel Picus
- Toni K. Choueiri and Meghara K. Walsh, Dana-Farber Cancer Institute; M. Dror Michaelson, Massachusetts General Hospital Cancer Center, Boston, MA; Susan Halabi and Ben L. Sanford, Alliance Statistics and Data Center and Duke University; Daniel J. George, Duke Cancer Institute, Duke University Medical Center, Durham, NC; Olwen Hahn, Alliance Protocol Operations Office, Chicago, IL; Darren R. Feldman and Michael J. Morris, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University Medical Center, Columbus, OH; Joel Picus, Siteman Cancer Center, Washington University School of Medicine, St Louis, MO; Eric J. Small, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; and Shaker Dakhil, University of Kansas Wichita, Wichita, KS
| | - Eric J Small
- Toni K. Choueiri and Meghara K. Walsh, Dana-Farber Cancer Institute; M. Dror Michaelson, Massachusetts General Hospital Cancer Center, Boston, MA; Susan Halabi and Ben L. Sanford, Alliance Statistics and Data Center and Duke University; Daniel J. George, Duke Cancer Institute, Duke University Medical Center, Durham, NC; Olwen Hahn, Alliance Protocol Operations Office, Chicago, IL; Darren R. Feldman and Michael J. Morris, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University Medical Center, Columbus, OH; Joel Picus, Siteman Cancer Center, Washington University School of Medicine, St Louis, MO; Eric J. Small, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; and Shaker Dakhil, University of Kansas Wichita, Wichita, KS
| | - Shaker Dakhil
- Toni K. Choueiri and Meghara K. Walsh, Dana-Farber Cancer Institute; M. Dror Michaelson, Massachusetts General Hospital Cancer Center, Boston, MA; Susan Halabi and Ben L. Sanford, Alliance Statistics and Data Center and Duke University; Daniel J. George, Duke Cancer Institute, Duke University Medical Center, Durham, NC; Olwen Hahn, Alliance Protocol Operations Office, Chicago, IL; Darren R. Feldman and Michael J. Morris, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University Medical Center, Columbus, OH; Joel Picus, Siteman Cancer Center, Washington University School of Medicine, St Louis, MO; Eric J. Small, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; and Shaker Dakhil, University of Kansas Wichita, Wichita, KS
| | - Daniel J George
- Toni K. Choueiri and Meghara K. Walsh, Dana-Farber Cancer Institute; M. Dror Michaelson, Massachusetts General Hospital Cancer Center, Boston, MA; Susan Halabi and Ben L. Sanford, Alliance Statistics and Data Center and Duke University; Daniel J. George, Duke Cancer Institute, Duke University Medical Center, Durham, NC; Olwen Hahn, Alliance Protocol Operations Office, Chicago, IL; Darren R. Feldman and Michael J. Morris, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University Medical Center, Columbus, OH; Joel Picus, Siteman Cancer Center, Washington University School of Medicine, St Louis, MO; Eric J. Small, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; and Shaker Dakhil, University of Kansas Wichita, Wichita, KS
| | - Michael J Morris
- Toni K. Choueiri and Meghara K. Walsh, Dana-Farber Cancer Institute; M. Dror Michaelson, Massachusetts General Hospital Cancer Center, Boston, MA; Susan Halabi and Ben L. Sanford, Alliance Statistics and Data Center and Duke University; Daniel J. George, Duke Cancer Institute, Duke University Medical Center, Durham, NC; Olwen Hahn, Alliance Protocol Operations Office, Chicago, IL; Darren R. Feldman and Michael J. Morris, Memorial Sloan Kettering Cancer Center, New York, NY; Thomas Olencki, Ohio State University Medical Center, Columbus, OH; Joel Picus, Siteman Cancer Center, Washington University School of Medicine, St Louis, MO; Eric J. Small, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA; and Shaker Dakhil, University of Kansas Wichita, Wichita, KS
| |
Collapse
|
49
|
Motzer RJ, Escudier B, Gannon A, Figlin RA. Sunitinib: Ten Years of Successful Clinical Use and Study in Advanced Renal Cell Carcinoma. Oncologist 2016; 22:41-52. [PMID: 27807302 DOI: 10.1634/theoncologist.2016-0197] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 08/03/2016] [Indexed: 01/07/2023] Open
Abstract
The oral multikinase inhibitor sunitinib malate was approved by the U.S. Food and Drug Administration in January 2006 for use in patients with advanced renal cell carcinoma (RCC). Since then, it has been approved globally for this indication and for patients with imatinib-resistant or -intolerant gastrointestinal stromal tumors and advanced pancreatic neuroendocrine tumors. As we mark the 10-year anniversary of the beginning of the era of targeted therapy, and specifically the approval of sunitinib, it is worthwhile to highlight the progress that has been made in advanced RCC as it relates to the study of sunitinib. We present the key trials and data for sunitinib that established it as a reference standard of care for first-line advanced RCC therapy and, along with other targeted agents, significantly altered the treatment landscape in RCC. Moreover, we discuss the research with sunitinib that has sought to refine its role via patient selection and prognostic markers, improve dosing and adverse event management, and identify predictive efficacy biomarkers, plus the extent to which this research has contributed to the overall understanding and management of RCC. We also explore the key learnings regarding study design and data interpretation from the sunitinib studies and how these findings and the sunitinib development program, in general, can be a model for successful development of other agents. Finally, ongoing research into the continued and future role of sunitinib in RCC management is discussed. THE ONCOLOGIST 2017;22:41-52 IMPLICATIONS FOR PRACTICE: Approved globally, sunitinib is established as a standard of care for first-line advanced renal cell carcinoma (RCC) therapy and, along with other targeted agents, has significantly altered the treatment landscape in RCC. Research with sunitinib that has sought to refine its role via patient selection and prognostic markers, improve dosing and adverse event management, and identify predictive efficacy biomarkers has contributed to the overall understanding and management of RCC. Key learnings regarding study design and data interpretation from the sunitinib studies and the sunitinib development program, in general, can be a model for the successful development of other agents.
Collapse
Affiliation(s)
- Robert J Motzer
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | | | - Robert A Figlin
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
50
|
Garcia CA, Wu S. Attributable Risk of Infection to mTOR Inhibitors Everolimus and Temsirolimus in the Treatment of Cancer. Cancer Invest 2016; 34:521-530. [DOI: 10.1080/07357907.2016.1242009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Christine A. Garcia
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Shenhong Wu
- Division of Hematology/Oncology, Department of Medicine, Stony Brook University Hospital, Stony Brook, New York, USA
- Northport VA Medical Center, Northport, New York, USA
| |
Collapse
|