1
|
Yin L, Mao L, Yin R, Lv C, Shi X, Yue C, Chen Y, Lu C, Wu Z, Xu K, Cao W. ACE Loss Drives Renal Cell Carcinoma Growth and Invasion by Modulating AKT-FOXO1. Biologics 2024; 18:397-412. [PMID: 39717370 PMCID: PMC11665188 DOI: 10.2147/btt.s485178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 12/12/2024] [Indexed: 12/25/2024]
Abstract
Purpose Emerging literature links the role of the renin-angiotensin-aldosterone system (RAAS) to the progression of cancers. However, the function of RAAS has not been verified in Clear-cell renal cell carcinoma (ccRCC). Methods ACE expression in ccRCC tissues was determined using RT-PCR, Western blot, and immunohistochemistry staining. The clinical significance of ACE was evaluated through Cox regression analysis. To assess the impact of ACE expression on ccRCC cell growth, metastasis, and glucose activity, CCK-8 assays, transwell assays, Seahorse detection, and xenograft models were utilized. The mechanisms of ACE and its upstream and downstream regulatory factors were investigated using RNA-seq, chromatin immunoprecipitation (ChIP), and luciferase reporter assays. Results RAAS-related gene Angiotensin-Converting Enzyme (ACE) was significantly under expressed in ccRCC cells and tissues. High ACE expression was positively associated with a favorable prognosis in ccRCC patients. Functional studies showed that ACE overexpression suppressed ccRCC cell line OS-RC-2 and A498 growth, metastasis, and glycolysis activities, while its knockdown had the opposite effect. Mechanistically, ACE inhibited ccRCC progression and epithelial-mesenchymal transition (EMT) by disrupting the AKT-FOXO1 signaling pathway. Furthermore, we provide evidence that ACE could enhance everolimus (approved agent for ccRCC) antitumor effect and ACE expression is transcriptionally regulated by ZBTB26. Conclusion Our findings investigated the roles and mechanisms of ACE in ccRCC. ACE inhibits the growth and metastasis of ccRCC cells in vitro and in vivo by promoting FOXO1 expression, which is the downstream target of PI3K-AKT pathway. Thus, this research suggests that ACE may be a promising target for new therapeutic strategy in ccRCC.
Collapse
Affiliation(s)
- Lei Yin
- Department of Urology, Shidong Hospital, Yangpu District, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, 200438, People’s Republic of China
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
- Department of Urology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201999, People’s Republic of China
| | - Lixin Mao
- Department of Urology, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| | - Rui Yin
- Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250012, People’s Republic of China
| | - Chengxun Lv
- Department of Urology, Shidong Hospital, Yangpu District, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, 200438, People’s Republic of China
| | - Xiaokai Shi
- Department of Urology, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| | - Chuang Yue
- Department of Urology, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| | - Yin Chen
- Department of Urology, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| | - Chao Lu
- Department of Urology, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| | - Zonglin Wu
- Department of Urology, Shidong Hospital, Yangpu District, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, 200438, People’s Republic of China
| | - Kai Xu
- Department of Urology, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| | - Wei Cao
- Department of Urology, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| |
Collapse
|
2
|
Li Z, You Y, Feng B, Chen J, Gao H, Li F. Construction methods and latest applications of kidney cancer organoids. Oncol Rev 2024; 18:1434981. [PMID: 39600908 PMCID: PMC11588466 DOI: 10.3389/or.2024.1434981] [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: 05/19/2024] [Accepted: 10/31/2024] [Indexed: 11/29/2024] Open
Abstract
Renal cell carcinoma (RCC) is one of the deadliest malignant tumors. Despite significant advances in RCC treatment over the past decade, complete remission is rarely achieved. Consequently, there is an urgent need to explore and develop new therapies to improve the survival rates and quality of life for patients. In recent years, the development of tumor organoid technology has attracted widespread attention as it can more accurately simulate the spatial structure and physiological characteristics of tumors within the human body. In this review, we summarize the main methods currently used to construct kidney cancer organoids, as well as their various biological and clinical applications. Furthermore, combining organoids with other technologies, such as co-culture techniques and microfluidic technologies, can further develop organoids and address their limitations, creating more practical models. This approach summarizes the interactions between different tissues or organs during tumor progression. Finally, we also provide an outlook on the construction and application of kidney cancer organoids. These rapidly evolving kidney cancer organoids may soon become a focal point in the development of in vitro clinical models and therapeutic research for kidney cancer.
Collapse
Affiliation(s)
- Zhiqiang Li
- Medical College of Guangxi University, Nan Ning, Guang Xi, China
| | - Yanqiu You
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nan Ning, China
| | - Bingzheng Feng
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nan Ning, China
| | - Jibing Chen
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nan Ning, China
| | - Hongjun Gao
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nan Ning, China
| | - Fujun Li
- Medical College of Guangxi University, Nan Ning, Guang Xi, China
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nan Ning, China
| |
Collapse
|
3
|
Yildirim HC, Bayram E, Chalabiyev E, Majidova N, Avci T, Güzel HG, Kapar C, Uzun M, Perkin P, Akgül F, Yildirim SS, Sali S, Yildiz A, Kazaz SN, Hendem E, Arcagok M, Tufan G, Yildirim U, Akgul OF, Arslan Ç, Taban H, Sahin E, Caglayan M, Esen R, Öksüzoğlu B, Guven DC, Kaplan MA, Araz M, Basaran M, Cubukcu E, Gokmen E, Cicin I, Algin E, Semiz HS, Tural D, Ozturk B, Erdogan AP, Sari M, Kara O, Erman M. Comparison of the efficacy of sunitinib and pazopanib in patients with advanced non-clear renal cell carcinoma. J Chemother 2024:1-12. [PMID: 39257075 DOI: 10.1080/1120009x.2024.2403051] [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: 07/29/2024] [Revised: 09/03/2024] [Accepted: 09/06/2024] [Indexed: 09/12/2024]
Abstract
Non-clear cell renal cell carcinoma (non-ccRCC) is a highly heterogeneous disease group, accounting for approximately 25% of all RCC cases. Due to its rarity and especially heterogeneity, phase III trial data is limited and treatment options generally follow those of clear cell RCC. In the literature, there exists a number of studies with sunitinib, cabozantinib, and everolimus, but data on the efficacy of pazopanib are limited. Our aim in this study was to compare the efficacy of pazopanib and sunitinib, in a multicenter retrospective cohort of non-ccRCC patients. Our study included patients diagnosed with non-ccRCC who received pazopanib or sunitinib treatment as first-line therapy from 22 tertiary hospitals. We compared the progression-free survival (PFS), overall survival (OS), and response rates of pazopanib and sunitinib treatments. Additionally, we investigated prognostic factors in non-ccRCC. PFS and response rates of sunitinib and pazopanib were found to be similar, while a numerical difference was observed in OS. Being 65 years and older, being in the intermediate or poor risk group according to the International Metastatic Renal Cell Carcinoma Database Consortium, having liver metastases, presence of a sarcomatoid component, and having de novo metastatic disease were found to be significantly associated with shorter PFS. Pazopanib treatment appears to have similar efficacy in the treatment of non-ccRCC compared to sunitinib. Though randomized controlled trials are lacking and will probably be never be available, we suggest that pazopanib could be a preferred agent like sunitinib and cabozantinib.
Collapse
Affiliation(s)
- Hasan Cagri Yildirim
- Department of Medical Oncology, Niğde Education and Research Hospital, Niğde, Turkey
| | - Ertuğrul Bayram
- Department of Medical Oncology, Çukurova University Faculty of Medicine, Adana, Turkey
| | - Elvin Chalabiyev
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Nargız Majidova
- Department of Medical Oncology, Marmara University Faculty of Medicine, Istanbul, Turkey
| | - Tugay Avci
- Department of Medical Oncology, Celal Bayar University Faculty of Medicine, Manisa, Turkey
| | - Halil Göksel Güzel
- Department of Medical Oncology, Antalya Education and Research Hospital, Antalya, Turkey
| | - Caner Kapar
- Department of Medical Oncology, Bakırkoy Dr. Sadi Konuk Education and Research Hospital, İstanbul, Turkey
| | - Mehmet Uzun
- Department of Medical Oncology, 9 Eylül University Faculty of Medicine, Izmir, Turkey
| | - Perihan Perkin
- Department of Medical Oncology, Yildirim Beyazit University, Bilkent State Hospital, Ankara, Turkey
| | - Fahri Akgül
- Department of Medical Oncology, Trakya University Faculty of Medicine, Edirne, Turkey
| | - Saadet Sim Yildirim
- Department of Medical Oncology, Ege University Faculty of Medicine, Izmir, Turkey
| | - Seda Sali
- Department of Medical Oncology, Uludag University Faculty of Medicine, Bursa, Turkey
| | - Anil Yildiz
- Department of Medical Oncology, Istanbul University Institute of Oncology, Istanbul, Turkey
| | - Seher Nazli Kazaz
- Department of Medical Oncology, Medical Park Hospital, Trabzon, Turkey
| | - Engin Hendem
- Department of Medical Oncology, Necmettin Erbakan University Faculty of Medicine, Konya, Turkey
| | - Murat Arcagok
- Department of Medical Oncology, Dicle University Faculty of Medicine, Diyarbakir, Turkey
| | - Gulnihal Tufan
- Department of Medical Oncology, Dr. A. Y. Ankara Oncology Research and Education Hospital, University of Health Sciences, Ankara, Turkey
| | - Umit Yildirim
- Department of Medical Oncology, Yuzuncu Yil University Faculty of Medicine, Van, Turkey
| | - Omer Faruk Akgul
- Department of Medical Oncology, Adnan Menderes University Faculty of Medicine, Aydin, Turkey
| | - Çagatay Arslan
- Department of Medical Oncology, Medicalpoint Hospital, Izmir, Turkey
| | - Hakan Taban
- Department of Medical Oncology, Samsun Training and Research Hospital, Samsun, Turkey
| | - Eren Sahin
- Department of Medical Oncology, Akdeniz University Faculty of Medicine, Antalya, Turkey
| | - Melek Caglayan
- Department of Medical Oncology, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Ramazan Esen
- Department of Medical Oncology, Yuzuncu Yil University Faculty of Medicine, Van, Turkey
| | - Berna Öksüzoğlu
- Department of Medical Oncology, Dr. A. Y. Ankara Oncology Research and Education Hospital, University of Health Sciences, Ankara, Turkey
| | - Deniz Can Guven
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Muhammet Ali Kaplan
- Department of Medical Oncology, Dicle University Faculty of Medicine, Diyarbakir, Turkey
| | - Murat Araz
- Department of Medical Oncology, Necmettin Erbakan University Faculty of Medicine, Konya, Turkey
| | - Mert Basaran
- Department of Medical Oncology, Istanbul University Institute of Oncology, Istanbul, Turkey
| | - Erdem Cubukcu
- Department of Medical Oncology, Uludag University Faculty of Medicine, Bursa, Turkey
| | - Erhan Gokmen
- Department of Medical Oncology, Ege University Faculty of Medicine, Izmir, Turkey
| | - Irfan Cicin
- Department of Medical Oncology, Trakya University Faculty of Medicine, Edirne, Turkey
| | - Efnan Algin
- Department of Medical Oncology, Yildirim Beyazit University, Bilkent State Hospital, Ankara, Turkey
| | - Huseyin Salih Semiz
- Department of Medical Oncology, 9 Eylül University Faculty of Medicine, Izmir, Turkey
| | - Deniz Tural
- Department of Medical Oncology, Bakırkoy Dr. Sadi Konuk Education and Research Hospital, İstanbul, Turkey
| | - Banu Ozturk
- Department of Medical Oncology, Antalya Education and Research Hospital, Antalya, Turkey
| | - Atike Pinar Erdogan
- Department of Medical Oncology, Celal Bayar University Faculty of Medicine, Manisa, Turkey
| | - Murat Sari
- Department of Medical Oncology, Marmara University Faculty of Medicine, Istanbul, Turkey
| | - Oguz Kara
- Department of Medical Oncology, Çukurova University Faculty of Medicine, Adana, Turkey
| | - Mustafa Erman
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| |
Collapse
|
4
|
Naffrichoux J, Poupin P, Pouillot W, Linassier C, Rioux-Leclercq N, De Vries-Brilland M, Mourey L, Laguerre B, Oudard S, Gross-Goupil M, Mousset C, Gravis G, Rolland F, Moise L, Emambux S, Vassal C, Zanetta S, Penel N, Albiges L, Fromont G, Cancel M. PD-L1 expression and its prognostic value in metastatic papillary renal cell carcinoma: Results from a GETUG multicenter retrospective cohort. Eur J Cancer 2024; 205:114121. [PMID: 38749111 DOI: 10.1016/j.ejca.2024.114121] [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: 03/05/2024] [Revised: 05/02/2024] [Accepted: 05/09/2024] [Indexed: 06/09/2024]
Abstract
INTRODUCTION Papillary renal cell carcinoma (pRCC) is a rare and aggressive cancer with no specifically established therapeutic strategy in the metastatic setting. Combinations of tyrosine kinase and immune checkpoint inhibitors (ICI) are a promising option. We aimed to study the immune landscape of metastatic pRCC, and its interactions with angiogenesis pathways, to search for potential therapeutic targets. METHODS The expression of immune markers (PD-L1, PD-1, PD-L2, LAG-3) and angiogenic pathways (CAIX, c-MET), was analyzed by immunohistochemistry on 68 metastatic pRCC retrieved from a retrospective multicenter GETUG cohort. Our primary endpoint was to estimate the prevalence of PD-L1 expression and its prognostic impact in metastatic pRCC. Secondary endpoints included the evaluation of other immune markers (PD-1, PD-L2, and LAG-3) and their association with PD-L1. We also assessed angiogenic markers and their association with PD-L1. RESULTS Overall, 27.9 % of tumors were PD-L1 positive. PD-L2 was more frequently expressed (45.6 %), PD-1 and LAG-3 were positive in 17.6 % and 19.1 % respectively. None of these markers was correlated with PD-L1 expression. 66 % (45/68) expressed at least one immune marker, and 43 % (29/68) were "double-positive", as they expressed both immune and angiogenic markers. OS was significantly shorter for patients with PD-L1 positive pRCC. A multivariate analysis confirmed a significant association between PD-L1 expression and shorter overall survival (HR = 4.0, p = 0.01). CONCLUSION These results reinforce clinical data on the expected benefit of ICI in metastatic pRCC treatment, as PD-L1 expression is a factor of poor prognosis in this multicenter cohort.
Collapse
Affiliation(s)
| | | | | | - Claude Linassier
- Department of Medical Oncology, University Hospital, Tours, France
| | | | | | - Loïc Mourey
- Department of Medical Oncology, IUCT Oncopole, Toulouse, France
| | - Brigitte Laguerre
- Department of Medical Oncology, Eugène Marquis Cancer Center, Rennes, France
| | - Stéphane Oudard
- Department of Medical Oncology, Georges Pompidou Hospital, University Paris Cité, Paris, France
| | - Marine Gross-Goupil
- Department of Medical Oncology, Saint-André University Hospital, Bordeaux, France
| | | | - Gwenaelle Gravis
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Frédéric Rolland
- Department of Medical Oncology, Institut de Cancérologie de L'Ouest, Saint Herblain, France
| | - Laura Moise
- Department of Medical Oncology, Centre François Baclesse, Caen, France
| | - Sheik Emambux
- Department of Medical Oncology, La Milétrie University Hospital, Poitiers, France
| | - Cécile Vassal
- Department of Medical Oncology, Institut de Cancérologie Lucien Neuwirth, Saint-Priest-en-Jarez, France
| | - Sylvie Zanetta
- Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France
| | - Nicolas Penel
- Lille University and Department of Medical Oncology, Centre Oscar Lambret, Lille, France
| | - Laurence Albiges
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Gaëlle Fromont
- Department of Pathology, University Hospital, Tours, France; INSERM UMR 1069, N2COx, Tours University, Tours, France
| | - Mathilde Cancel
- Department of Medical Oncology, University Hospital, Tours, France; INSERM UMR 1069, N2COx, Tours University, Tours, France.
| |
Collapse
|
5
|
Wang H, Zhang L, Liu H, Yang Y, Lu W, Cao X, Yang X, Qin Q, Song R, Feng D, Wang S, Bai T, He J. PDZK1 confers sensitivity to sunitinib in clear cell renal cell carcinoma by suppressing the PDGFR-β pathway. Br J Cancer 2024; 131:347-360. [PMID: 38822145 PMCID: PMC11263541 DOI: 10.1038/s41416-024-02725-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 05/10/2024] [Accepted: 05/14/2024] [Indexed: 06/02/2024] Open
Abstract
BACKGROUND Sunitinib has emerged as the primary treatment for advanced or metastatic clear cell renal cell carcinoma (ccRCC) due to its significant improvement in patients' average survival time. However, drug resistance and adverse effects of sunitinib pose challenges to its clinical benefits. METHODS The differentially expressed genes (DEGs) associated with sunitinib sensitivity and resistance in ccRCC were investigated. Cell counting kit-8, plate colony formation, flow cytometry and subcutaneous xenograft tumor model assays were employed to explore the effects of PDZK1 on ccRCC. Further research on the molecular mechanism was conducted through western blot, co-immunoprecipitation, immunofluorescence co-localization and immunohistochemical staining. RESULTS We elucidated that PDZK1 is significantly downregulated in sunitinib-resistant ccRCC specimens, and PDZK1 negatively regulates the phosphorylation of PDGFR-β and the activation of its downstream pathways through interaction with PDGFR-β. The dysregulated low levels of PDZK1 contribute to inadequate inhibition of cell proliferation, tumor growth, and insensitivity to sunitinib treatment. Notably, our preclinical investigations showed that miR-15b antagomirs enhance sunitinib cytotoxic effects against ccRCC cells by upregulating PDZK1 levels, suggesting their potential in overcoming sunitinib resistance. CONCLUSIONS Our findings establish the miR-15b/PDZK1/PDGFR-β axis as a promising therapeutic target and a novel predictor for ccRCC patients' response to sunitinib treatment.
Collapse
MESH Headings
- Sunitinib/pharmacology
- Sunitinib/therapeutic use
- Carcinoma, Renal Cell/drug therapy
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/metabolism
- Humans
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Receptor, Platelet-Derived Growth Factor beta/genetics
- Kidney Neoplasms/drug therapy
- Kidney Neoplasms/pathology
- Kidney Neoplasms/genetics
- Kidney Neoplasms/metabolism
- Animals
- Drug Resistance, Neoplasm/genetics
- Mice
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Xenograft Model Antitumor Assays
- MicroRNAs/genetics
- Signal Transduction/drug effects
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Male
- Mice, Nude
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
Collapse
Affiliation(s)
- Haibo Wang
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, People's Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Lijie Zhang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Hua Liu
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
| | - Yumeng Yang
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
| | - Wenxiu Lu
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
| | - Xuedi Cao
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
| | - Xiaomei Yang
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Qiong Qin
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Ran Song
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Duiping Feng
- Department of Interventional Radiology, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Songlin Wang
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, People's Republic of China
- Salivary Gland Disease Center and Molecular Laboratory for Gene Therapy and Tooth Regeneration, School of Stomatology, Capital Medical University, Beijing, People's Republic of China
| | - Tao Bai
- Department of Pathology, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China.
| | - Junqi He
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China.
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China.
| |
Collapse
|
6
|
Soares A, Monteiro FSM, da Trindade KM, Silva AGE, Cardoso APG, Sasse AD, Fay AP, Carneiro APCD, Alencar Junior AM, de Andrade Mota AC, Santucci B, da Motta Girardi D, Herchenhorn D, Araújo DV, Jardim DL, Bastos DA, Rosa DR, Schutz FA, Kater FR, da Silva Marinho F, Maluf FC, de Oliveira FNG, Vidigal F, Morbeck IAP, Rinck Júnior JA, Costa LAGA, Maia MCDF, Zereu M, Freitas MRP, Dias MSF, Tariki MS, Muniz P, Beato PMM, Lages PSM, Velho PI, de Carvalho RS, Mariano RC, de Araújo Cavallero SR, Oliveira TM, Souza VC, Smaletz O, de Cássio Zequi S. Advanced renal cell carcinoma management: the Latin American Cooperative Oncology Group (LACOG) and the Latin American Renal Cancer Group (LARCG) consensus update. J Cancer Res Clin Oncol 2024; 150:183. [PMID: 38594593 PMCID: PMC11003910 DOI: 10.1007/s00432-024-05663-z] [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: 12/27/2023] [Accepted: 02/22/2024] [Indexed: 04/11/2024]
Abstract
PURPOSE Renal cell carcinoma is an aggressive disease with a high mortality rate. Management has drastically changed with the new era of immunotherapy, and novel strategies are being developed; however, identifying systemic treatments is still challenging. This paper presents an update of the expert panel consensus from the Latin American Cooperative Oncology Group and the Latin American Renal Cancer Group on advanced renal cell carcinoma management in Brazil. METHODS A panel of 34 oncologists and experts in renal cell carcinoma discussed and voted on the best options for managing advanced disease in Brazil, including systemic treatment of early and metastatic renal cell carcinoma as well as nonclear cell tumours. The results were compared with the literature and graded according to the level of evidence. RESULTS Adjuvant treatments benefit patients with a high risk of recurrence after surgery, and the agents used are pembrolizumab and sunitinib, with a preference for pembrolizumab. Neoadjuvant treatment is exceptional, even in initially unresectable cases. First-line treatment is mainly based on tyrosine kinase inhibitors (TKIs) and immune checkpoint inhibitors (ICIs); the choice of treatment is based on the International Metastatic Database Consortium (IMCD) risk score. Patients at favourable risk receive ICIs in combination with TKIs. Patients classified as intermediate or poor risk receive ICIs, without preference for ICI + ICIs or ICI + TKIs. Data on nonclear cell renal cancer treatment are limited. Active surveillance has a place in treating favourable-risk patients. Either denosumab or zoledronic acid can be used for treating metastatic bone disease. CONCLUSION Immunotherapy and targeted therapy are the standards of care for advanced disease. The utilization and sequencing of these therapeutic agents hinge upon individual risk scores and responses to previous treatments. This consensus reflects a commitment to informed decision-making, drawn from professional expertise and evidence in the medical literature.
Collapse
Affiliation(s)
- Andrey Soares
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil.
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
- Centro Paulista de Oncologia/Oncoclínicas, São Paulo, SP, Brazil.
| | - Fernando Sabino Marques Monteiro
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Hospital Sírio-Libanês, Brasília, DF, Brazil
| | - Karine Martins da Trindade
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Oncologia D'Or, Fortaleza, CE, Brazil
| | - Adriano Gonçalves E Silva
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Instituto do Câncer e Transplante de Curitiba/PR (ICTr Curitiba), Curitiba, PR, Brazil
| | - Ana Paula Garcia Cardoso
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - André Deeke Sasse
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Grupo SONHE de Campinas, Campinas, SP, Brazil
| | - André P Fay
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Escola de Medicina da Pontifícia, Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - André Paternò Castello Dias Carneiro
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
- Hospital Municipal Vila Santa Catarina, São Paulo, SP, Brazil
| | - Antonio Machado Alencar Junior
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Hospital São Domingos, São Luís, MA, Brazil
- Dasa Oncologia, Brasília, DF, Brazil
- Hospital Universitário da Universidade Federal do Maranhão (UFMA), São Luís, MA, Brazil
| | - Augusto César de Andrade Mota
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Clínica AMO-DASA, Feira de Santana, BA, Brazil
| | - Bruno Santucci
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Instituto Paulista de Cancerologia, São Paulo, SP, Brazil
| | - Daniel da Motta Girardi
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Hospital Sírio-Libanês, Brasília, DF, Brazil
| | - Daniel Herchenhorn
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Oncologia D'Or, Rio de Janeiro, RJ, Brazil
| | - Daniel Vilarim Araújo
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Hospital de Base de São José do Rio Preto/SP, São José do Rio Preto, São Paulo, SP, Brazil
| | - Denis Leonardo Jardim
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Grupo Oncoclínicas, São Paulo, São Paulo, SP, Brazil
| | - Diogo Assed Bastos
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Hospital Sirio-Libanês de São Paulo, São Paulo, SP, Brazil
| | - Diogo Rodrigues Rosa
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Grupo Oncoclínicas, Rio de Janeiro, RJ, Brazil
| | - Fabio A Schutz
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Beneficência Portuguesa de São Paulo, São Paulo, SP, Brazil
| | - Fábio Roberto Kater
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Beneficência Portuguesa de São Paulo, São Paulo, SP, Brazil
| | - Felipe da Silva Marinho
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Grupo Oncoclínicas, Recife, PE, Brazil
| | - Fernando Cotait Maluf
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
- Beneficência Portuguesa de São Paulo, São Paulo, SP, Brazil
| | - Fernando Nunes Galvão de Oliveira
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Grupo Oncoclínicas, Salvador, BA, Brazil
| | - Fernando Vidigal
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Dasa Oncologia, Brasília, DF, Brazil
| | - Igor Alexandre Protzner Morbeck
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Grupo Oncoclínicas, Brasília, DF, Brazil
| | - Jose Augusto Rinck Júnior
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- AC Camargo Cancer Center, São Paulo, SP, Brazil
| | - Leonardo Atem G A Costa
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Oncologia D'Or, Fortaleza, CE, Brazil
| | - Manuel Caitano Dias Ferreira Maia
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Hospital do Câncer Porto Dias, Belém, PA, Brazil
| | - Manuela Zereu
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Santa Casa de Misericórdia de Porto Alegre, Porto Alegre, RS, Brazil
| | - Marcelo Roberto Pereira Freitas
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Centro Especializado de Oncologia de Florianópolis, Florianópolis, SC, Brazil
| | - Mariane Sousa Fontes Dias
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Grupo Oncoclínicas, Rio de Janeiro, RJ, Brazil
| | - Milena Shizue Tariki
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- AC Camargo Cancer Center, São Paulo, SP, Brazil
| | - Pamela Muniz
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Grupo Oncoclínicas, São Paulo, São Paulo, SP, Brazil
- Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Patrícia Medeiros Milhomem Beato
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Hospital Amaral Carvalho, Jaú, SP, Brazil
| | - Paulo Sérgio Moraes Lages
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Grupo Oncoclínicas, Brasília, DF, Brazil
| | - Pedro Isaacsson Velho
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Hospital Moinhos de Vento, Porto Alegre, RS, Brazil
- Johns Hopkins University, Baltimore, MD, USA
| | - Ricardo Saraiva de Carvalho
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Beneficência Portuguesa de São Paulo, São Paulo, SP, Brazil
| | - Rodrigo Coutinho Mariano
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Beneficência Portuguesa de São Paulo, São Paulo, SP, Brazil
| | - Sandro Roberto de Araújo Cavallero
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Hospital Adventista de Belém, Belém, PA, Brazil
| | - Thiago Martins Oliveira
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Hospital São Rafael, Salvador, BA, Brazil
| | - Vinicius Carrera Souza
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Instituto D'Or de Ensino e Pesquisa, Salvador, BA, Brazil
| | - Oren Smaletz
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Stênio de Cássio Zequi
- AC Camargo Cancer Center, São Paulo, SP, Brazil
- National Institute for Science and Technology in Oncogenomics and Therapeutic Innovation, AC Camargo Cancer Center, São Paulo, SP, Brazil
| |
Collapse
|
7
|
Huang JY, Xie XF, Chen XL, Zhang QY, Chen LP, Bai X, Lan XF, Song L, Guo JF, Du CW. A single-arm phase II clinical trial of anlotinib combined with chemotherapy for the treatment of metastatic triple-negative breast cancer. Front Oncol 2023; 13:1122294. [PMID: 37124484 PMCID: PMC10130368 DOI: 10.3389/fonc.2023.1122294] [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: 12/12/2022] [Accepted: 03/28/2023] [Indexed: 05/02/2023] Open
Abstract
Background Anlotinib is a novel oral small-molecule tyrosine kinase inhibitor (TKI), which can inhibit angiogenesis. The purpose of this study was to evaluate the efficacy and safety of anlotinib combined with chemotherapy in patients with metastatic triple-negative breast cancer (TNBC). Methods This phase II clinical trial included 40 patients with metastatic TNBC who had previously received anthracycline and/or taxane treatment. All patients received anlotinib combined with chemotherapy. The primary endpoint was progression-free survival (PFS). The secondary endpoints included overall survival (OS), objective response rate (ORR), clinical benefit rate (CBR), disease control rate (DCR) and safety. Results During May 1, 2019 and April 30, 2022, there were 40 patients enrolled in this study. The median PFS and median OS were 8.8 months (95% confidence interval [CI] 6.5-11.1 months) and 19.0 months (95% CI, 12.1-25.9 months), respectively. The ORR, CBR and DCR were 40.0% (16/40), 85.0% (34/40) and 95.0% (38/40), respectively. Cox univariate and multivariate analyses demonstrated that having more than 3 metastatic sites (p = 0.001; p = 0.020) was an independent and meaningful unfavorable prognostic factor for PFS. 37.5% of patients had grade 3 to 4 treatment-related adverse events (TRAEs). The grade 3 to 4 TRAEs included neutropenia (22.5%), leukopenia (20.0%), secondary hypertension (10.0%), hand-foot syndrome (5.0%), vomiting (5.0%), proteinuria (5.0%) and thrombocytopenia (2.5%). None of the patients withdrew from the study or died due to TRAEs. Conclusion In this single-arm study, the treatment of metastatic TNBC with anlotinib combined with chemotherapy showed certain efficacy, and its toxicity was acceptable.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Cai-Wen Du
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, China
| |
Collapse
|
8
|
Climent C, Soriano S, Bonfill T, Lopez N, Rodriguez M, Sierra M, Andreu P, Fragio M, Busquets M, Carrasco A, Cano O, Seguí MA, Gallardo E. The role of immunotherapy in non-clear cell renal cell carcinoma. Front Oncol 2023; 13:941835. [PMID: 36816976 PMCID: PMC9936973 DOI: 10.3389/fonc.2023.941835] [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: 05/11/2022] [Accepted: 01/13/2023] [Indexed: 02/05/2023] Open
Abstract
The category of non-clear cell renal cell carcinoma (nccRCC) includes several clinically, histologically, and molecularly diverse entities. Traditionally, they comprise type 1 and type 2 papillary, chromophobe, unclassified, and other histologies (medullary, collecting duct carcinoma, and translocation-associated). Molecular knowledge has allowed the identification of some other specific subtypes, such as fumarate hydratase-deficient renal cell carcinoma (RCC) or succinate dehydrogenase-associated RCC. In addition, it has recognized some alterations with a possible predictive role, e.g., MET proto-oncogene receptor tyrosine kinase (MET) alterations in papillary tumors. Standard therapies for the management of advanced clear cell RCC (ccRCC), i.e., vascular endothelial growth factor receptor (VEGFR) pathway inhibitors and mammalian target of rapamycin inhibitors, have shown poorer results in nccRCC patients. Therefore, there is a need to improve the efficacy of the treatment for advanced nccRCC. Immunotherapy, especially immune checkpoint inhibitors (ICIs) targeting programmed death 1/programmed death ligand 1 and cytotoxic T-lymphocyte associated protein 4 (CTLA-4), has demonstrated a significant survival benefit in several malignant neoplasias, including ccRCC, with a proportion of patients achieving long survival. The combinations of ICI or ICI + VEGFR tyrosine kinase inhibitors (TKIs) are the standard of care in advanced ccRCC. Unfortunately, major pivotal trials did not include specific nccRCC populations. In recent years, several studies have retrospectively or prospectively evaluated ICIs alone or in combination with another ICI or with TKIs in nccRCC patients. In this article, we review data from available trials in order to elucidate clinical and molecular profiles that could benefit from immunotherapy approaches.
Collapse
|
9
|
John A, Spain L, Hamid AA. Navigating the Current Landscape of Non-Clear Cell Renal Cell Carcinoma: A Review of the Literature. Curr Oncol 2023; 30:923-937. [PMID: 36661719 PMCID: PMC9858145 DOI: 10.3390/curroncol30010070] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/24/2022] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
Non-clear cell renal cell carcinoma (nccRCC) is an entity comprised of a heterogeneous constellation of RCC subtypes. Genomic profiling has broadened our understanding of molecular pathogenic mechanisms unique to individual nccRCC subtypes. To date, clinical trials evaluating the use of immunotherapies and targeted therapies have predominantly been conducted in patients with clear cell histology. A comprehensive review of the literature has been undertaken in order to describe molecular pathogenic mechanisms pertaining to each nccRCC subtype, and concisely summarise findings from therapeutic trials conducted in the nccRCC space.
Collapse
Affiliation(s)
- Alexius John
- Department of Medical Oncology, Eastern Health, Melbourne, VIC 3128, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Lavinia Spain
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Anis A. Hamid
- Department of Medical Oncology, Eastern Health, Melbourne, VIC 3128, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Eastern Health Clinical School, Monash University, Melbourne, VIC 3128, Australia
- Department of Surgery, University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
10
|
Current Options for Second-Line Systemic Therapy in Metastatic Renal Cell Carcinoma. J Kidney Cancer VHL 2022; 9:29-40. [PMID: 36310639 PMCID: PMC9551369 DOI: 10.15586/jkcvhl.v9i3.243] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/26/2022] [Indexed: 11/06/2022] Open
Abstract
Standard systemic therapy of advanced renal cell carcinoma (RCC) involves targeting angiogenesis, mainly through tyrosine kinase inhibitors (TKI) against the vascular endothelial growth factor receptor (VEGFR) pathway and targeting the immune checkpoints, namely, programmed death-1 (PD-1) or its ligand (PD-L1), and cytotoxic T-lymphocyte-associated protein 4 (CTLA4). With current strategies of combining these two approaches in the front-line setting, less is known about optimal selection of therapy upon development of resistance in the second and later lines of treatment for progressive disease. This review discusses currently available therapeutic options in patients who have progressive RCC after prior treatment with double immune check-point inhibitors (ICIs) or ICI-TKI combinations.
Collapse
|
11
|
Izarn F, Allignet B, Gille R, Boyle H, Neidhardt EM, Négrier S, Fléchon A. Real world data of diagnosis, survival, and treatment outcomes in patients with metastatic non clear cell renal cell carcinoma. Clin Genitourin Cancer 2022; 21:e35-e43. [PMID: 36272959 DOI: 10.1016/j.clgc.2022.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/03/2022]
Abstract
INTRODUCTION Metastatic non clear cell renal cell carcinoma (nccRCC) is an heterogenous group, usually excluded from phase 3 trials. We report real life data of prognosis and systemic management of those patients. METHODS We retrospectively included 102 metastatic nccRCC patients (unspecified papillary, n = 10; type 1 and 2 papillary n = 10 and n = 32; translocation RCC, n = 9; chromophobe, n = 14; collecting duct, n = 14) treated between 2006 and 2020. Objective response rate (ORR), progression-free survival (PFS) and overall survival (OS) were evaluated. RESULTS Among patients who underwent pathological review, 40.8% presented a complete histological discordance. First line treatments were mainly tyrosine kinase inhibitor (60.8%), combination including immunotherapy (7.8%) or combination of chemotherapy (13.7%). Median ORR ranged from 0% in unspecified papillary RCC to 42.9% in type 1 papillary RCC. Median PFS ranged from 2.9 months in collecting duct carcinoma to 10.9 months in type 1 papillary RCC. Median OS ranged from 6.8 months in collecting duct carcinoma to 29.1 months in MiT family translocation RCC. Thirty (29.4%) patients were included in a treatment trial during their treatment course. CONCLUSION Metastatic nccRCC patients have variable prognosis due to heterogeneity of histological subtypes. Their diagnosis and access to therapeutic innovation remain suboptimal. Dedicated prospective trials are needed.
Collapse
|
12
|
Kojima T, Kato R, Sazuka T, Yamamoto H, Fukuda S, Yamana K, Nakaigawa N, Sugino Y, Hamamoto S, Ito H, Murakami H, Obara W. Real-world effectiveness of nivolumab plus ipilimumab and second-line therapy in Japanese untreated patients with metastatic renal cell carcinoma: 2-year analysis from a multicenter retrospective clinical study (J-cardinal study). Jpn J Clin Oncol 2022; 52:1345-1352. [PMID: 35920793 PMCID: PMC9631464 DOI: 10.1093/jjco/hyac124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 07/14/2022] [Indexed: 11/15/2022] Open
Abstract
Background Nivolumab plus ipilimumab combination therapy is one of the standard therapies for untreated renal cell carcinoma patients with an International Metastatic Renal Cell Carcinoma Database Consortium intermediate/poor risk. We have previously reported the 1-year analysis results of the effectiveness and safety of nivolumab plus ipilimumab combination therapy in the real-world setting in Japan. Here, we report the effectiveness of nivolumab plus ipilimumab combination therapy and of second-line therapy, using 2-year analysis. Methods This retrospective observational study enrolled Japanese patients with previously untreated metastatic renal cell carcinoma who initiated nivolumab plus ipilimumab combination therapy between August 2018 and January 2019. Data were collected from patients’ medical records at baseline and at 3 months, 1 year and 2 years after the last enrollment. Results Of the 45 patients enrolled, 10 patients (22.2%) each had non-clear cell renal cell carcinoma and Eastern Cooperative Oncology Group performance status ≥2 at baseline. Median follow-up period was 24.0 months; objective response rate was 41.5%, with 6 patients achieving complete response; median progression-free survival was 17.8 months and 24-month progression-free survival and overall survival rates were 41.6 and 59.1%, respectively. Second-line therapy achieved an objective response rate of 20%; median progression-free survival was 9.8 months. Median progression-free survival 2 was 26.4 months. Conclusions The effectiveness of nivolumab plus ipilimumab combination therapy at 2-year analysis in the real-world setting in Japan was comparable to that reported in CheckMate 214. The current analysis also demonstrated the effectiveness of second-line therapy after nivolumab plus ipilimumab combination therapy.
Collapse
Affiliation(s)
- Takahiro Kojima
- Department of Urology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan.,Department of Urology, Aichi Cancer Center Hospital, Aichi, Japan
| | - Renpei Kato
- Department of Urology, Iwate Medical University, Iwate, Japan
| | - Tomokazu Sazuka
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Hayato Yamamoto
- Department of Urology, Hirosaki University Graduate School of Medicine, Aomori, Japan
| | - Shohei Fukuda
- Department of Urology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kazutoshi Yamana
- Department of Urology, Molecular Oncology, Graduate School of Medicine and Dental Sciences, Niigata University, Niigata, Japan
| | - Noboru Nakaigawa
- Department of Urology, Yokohama City University Graduate School of Medicine, Kanagawa, Japan
| | - Yusuke Sugino
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, Mie, Japan
| | - Shuzo Hamamoto
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| | - Hiroaki Ito
- Oncology Medical, Bristol-Myers Squibb K.K., Tokyo, Japan
| | - Hiroshi Murakami
- Oncology Medical Affairs, Ono Pharmaceutical Co., Ltd., Tokyo, Japan
| | - Wataru Obara
- Department of Urology, Iwate Medical University, Iwate, Japan
| |
Collapse
|
13
|
Serzan M, Atkins MB. Adjuvant therapy for patients with renal cell carcinoma following surgery: a focus on pembrolizumab. Expert Rev Anticancer Ther 2022; 22:565-574. [PMID: 35483033 DOI: 10.1080/14737140.2022.2072300] [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: 11/04/2022]
Abstract
INTRODUCTION Many patients with renal cell carcinoma (RCC) who undergo surgery with curative intent have a high risk of disease recurrence and until recently no palatable adjuvant systemic therapy options. Blocking the programmed death ligand (PD-1) immune checkpoint pathway with pembrolizumab has robust clinical efficacy in patients with metastatic RCC. Results from the KEYNOTE 564 trial demonstrate that adjuvant pembrolizumab significantly improves disease- free survival after nephrectomy or metastatectomy. AREAS COVERED We provide an overview of efforts to develop an adjuvant therapy in patients with high-risk RCC. This includes a critical review of efficacy, toxicity, and clinical implications from a large phase III trial leading to the FDA and EMA approvals of adjuvant pembrolizumab. EXPERT OPINION Pembrolizumab offers an effective and well-tolerated adjuvant therapy for patients with surgically resected RCC at high-risk of disease recurrence. Future research will focus on optimal patient selection and biomarkers that predict benefit and/or toxicity from therapy.
Collapse
Affiliation(s)
- Michael Serzan
- Department of Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Washington DC.,Department of Medicine, Medstar Georgetown University Hospital, Washington DC
| | - Michael B Atkins
- Department of Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Washington DC.,Department of Medicine, Medstar Georgetown University Hospital, Washington DC
| |
Collapse
|
14
|
Grande E, Alonso-Gordoa T, Reig O, Esteban E, Castellano D, Garcia-Del-Muro X, Mendez MJ, García-Donas J, González Rodríguez M, Arranz-Arija JA, Lopez-Criado P, Molina-Cerrillo J, Mellado B, Alvarez-Fernandez C, De Velasco G, Cuéllar-Rivas MA, Rodríguez-Alonso RM, Rodríguez-Moreno JF, Suarez-Rodriguez C. Results from the INMUNOSUN-SOGUG trial: a prospective phase II study of sunitinib as a second-line therapy in patients with metastatic renal cell carcinoma after immune checkpoint-based combination therapy. ESMO Open 2022; 7:100463. [PMID: 35405437 PMCID: PMC9058923 DOI: 10.1016/j.esmoop.2022.100463] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 02/14/2022] [Accepted: 03/04/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND The INMUNOSUN trial had the objective of prospectively evaluating the efficacy and safety of sunitinib as a pure second-line treatment in patients with metastatic renal cell carcinoma (mRCC) who have progressed to first-line immune checkpoint inhibitor (ICI)-based therapies. PATIENTS AND METHODS A multicenter, phase II, single-arm, open-label study was carried out in patients with a histologically confirmed diagnosis of mRCC with a clear-cell component who had progressed to a first-line regimen of ICI-based therapies. All patients received sunitinib 50 mg once daily orally for 4 weeks, followed by a 2-week rest period following package insert instructions. The primary outcome was the objective response rate. RESULTS Twenty-one assessable patients were included in the efficacy and safety analyses. Four patients [19.0%, 95% confidence interval (CI) 2.3% to 35.8%] showed an objective response (OR), and all of them had partial responses. Additionally, 14 (67%) patients showed a stable response, leading to clinical benefit in 18 patients (85.7%, 95% CI 70.7% to 100%). Among the four assessable patients who showed an OR, the median duration of the response was 7.1 months (interquartile range 4.2-12.0 months). The median progression-free survival (PFS) was 5.6 months (95% CI 3.1-8.0 months). The median overall survival (OS) was 23.5 months (95% CI 6.3-40.7 months). Patients who had better antitumor response to first-line ICI-based treatment showed a longer PFS and OS with sunitinib. The most frequent treatment-emergent adverse events were diarrhea (n = 11, 52%), dysgeusia (n = 8, 38%), palmar-plantar erythrodysesthesia (n = 8, 38%), and hypertension (n = 8, 38%). There was 1 patient who exhibited grade 5 pancytopenia, and 11 patients experienced grade 3 adverse events. Eight (38%) patients had serious adverse events, four of which were considered to be related to sunitinib. CONCLUSION Although the INMUNOSUN trial did not reach the pre-specified endpoint, it demonstrated that sunitinib is active and can be safely used as a second-line option in patients with mRCC who progress to new standard ICI-based regimens.
Collapse
Affiliation(s)
- E Grande
- Medical Oncology, MD Anderson Cancer Center Madrid, Madrid, Spain.
| | - T Alonso-Gordoa
- Medical Oncology, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - O Reig
- Medical Oncology, Hospital Clinic and Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - E Esteban
- Medical Oncology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - D Castellano
- Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - X Garcia-Del-Muro
- Medical Oncology, Institut Català d'Oncologia (ICO Bellvitge) Idibell, University of Barcelona, Barcelona, Spain
| | - M J Mendez
- Medical Oncology, Maimonides Institute for Biomedical Research of Córdoba (IMIBIC) Hospital Universitario Reina Sofia (HURS), Córdoba, Spain
| | - J García-Donas
- Medical Oncology, Clara Campal Comprehensive Cancer Center, Madrid, Spain
| | - M González Rodríguez
- Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - J A Arranz-Arija
- Medical Oncology, MD Anderson Cancer Center Madrid, Madrid, Spain
| | - P Lopez-Criado
- Medical Oncology, MD Anderson Cancer Center Madrid, Madrid, Spain
| | - J Molina-Cerrillo
- Medical Oncology, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - B Mellado
- Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain
| | | | - G De Velasco
- Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - M A Cuéllar-Rivas
- Medical Oncology, Institut Català d'Oncologia (ICO Bellvitge) Idibell, University of Barcelona, Barcelona, Spain
| | - R M Rodríguez-Alonso
- Medical Oncology, Maimonides Institute for Biomedical Research of Córdoba (IMIBIC) Hospital Universitario Reina Sofia (HURS), Córdoba, Spain
| | | | - C Suarez-Rodriguez
- Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Barcelona, Spain
| |
Collapse
|
15
|
Kasherman L, Siu DHW, Woodford R, Harris CA. Angiogenesis Inhibitors and Immunomodulation in Renal Cell Cancers: The Past, Present, and Future. Cancers (Basel) 2022; 14:1406. [PMID: 35326557 PMCID: PMC8946206 DOI: 10.3390/cancers14061406] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/07/2022] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis inhibitors have been adopted into the standard armamentarium of therapies for advanced-stage renal cell carcinomas (RCC), but more recently, combination regimens with immune checkpoint inhibitors have demonstrated better outcomes. Despite this, the majority of affected patients still eventually experience progressive disease due to therapeutic resistance mechanisms, and there remains a need to develop novel therapeutic strategies. This article will review the synergistic mechanisms behind angiogenesis and immunomodulation in the tumor microenvironment and discuss the pre-clinical and clinical evidence for both clear-cell and non-clear-cell RCC, exploring opportunities for future growth in this exciting area of drug development.
Collapse
Affiliation(s)
- Lawrence Kasherman
- Department of Medical Oncology, St. George Hospital, Kogarah, NSW 2217, Australia; (D.H.W.S.); (R.W.); (C.A.H.)
- St. George and Sutherland Clinical Schools, University of New South Wales, Sydney, NSW 2217, Australia
- Department of Medical Oncology, Illawarra Cancer Care Centre, Wollongong, NSW 2500, Australia
| | - Derrick Ho Wai Siu
- Department of Medical Oncology, St. George Hospital, Kogarah, NSW 2217, Australia; (D.H.W.S.); (R.W.); (C.A.H.)
- National Health Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW 2050, Australia
| | - Rachel Woodford
- Department of Medical Oncology, St. George Hospital, Kogarah, NSW 2217, Australia; (D.H.W.S.); (R.W.); (C.A.H.)
- Faculty of Medciine and Health, University of Sydney, Camperdown, NSW 2050, Australia
| | - Carole A. Harris
- Department of Medical Oncology, St. George Hospital, Kogarah, NSW 2217, Australia; (D.H.W.S.); (R.W.); (C.A.H.)
- St. George and Sutherland Clinical Schools, University of New South Wales, Sydney, NSW 2217, Australia
| |
Collapse
|
16
|
McGregor BA, Xie W, Adib E, Stadler WM, Zakharia Y, Alva A, Michaelson MD, Gupta S, Lam ET, Farah S, Nassar AH, Wei XX, Kilbridge KL, Harshman L, Signoretti S, Sholl L, Kwiatkowski DJ, McKay RR, Choueiri TK. Biomarker-Based Phase II Study of Sapanisertib (TAK-228): An mTORC1/2 Inhibitor in Patients With Refractory Metastatic Renal Cell Carcinoma. JCO Precis Oncol 2022; 6:e2100448. [PMID: 35171658 PMCID: PMC8865529 DOI: 10.1200/po.21.00448] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/06/2021] [Accepted: 01/10/2022] [Indexed: 12/16/2022] Open
Abstract
PURPOSE Sapanisertib is a kinase inhibitor that inhibits both mammalian target of rapamycin complex 1 (mTORC1) and mTORC2. In this multicenter, single-arm phase II trial, we evaluated the efficacy of sapanisertib in patients with treatment-refractory metastatic renal cell carcinoma (mRCC; NCT03097328). METHODS Patients with mRCC of any histology progressing through standard therapy (including prior mTOR inhibitors) had baseline biopsy and received sapanisertib 30 mg by mouth once weekly until unacceptable toxicity or disease progression. The primary end point was objective response rate by RECIST 1.1. Tissue biomarkers of mTOR pathway activation were explored. RESULTS We enrolled 38 patients with mRCC (clear cell = 28; variant histology = 10) between August 2017 and November 2019. Twenty-four (63%) had received ≥ 3 prior lines of therapy; 17 (45%) had received prior rapalog therapy. The median follow-up was 10.4 (range 1-27.4) months. Objective response rate was two of 38 (5.3%; 90% CI, 1 to 15.6); the median progression-free survival (PFS) was 2.5 months (95% CI, 1.8 to 3.7). Twelve patients (32%) developed treatment-related grade 3 adverse events, with no grade 4 or 5 toxicities. Alterations in the mTOR pathway genes were seen in 5 of 29 evaluable patients (MTOR n = 1, PTEN n = 3, and TSC1 n = 1) with no association with response or PFS. Diminished or loss of PTEN expression by immunohistochemistry was seen in 8 of 21 patients and trended toward shorter PFS compared with intact PTEN (median 1.9 v 3.7 months; hazard ratio 2.5; 95% CI, 0.9 to 6.7; P = .055). CONCLUSION Sapanisertib had minimal activity in treatment-refractory mRCC independent of mTOR pathway alterations. Additional therapeutic strategies are needed for patients with refractory mRCC.
Collapse
Affiliation(s)
| | | | - Elio Adib
- Dana-Farber Cancer Institute, Boston, MA
- Brigham and Women's Hospital, Boston, MA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
An J, Packiam VT, Chennamadhavuni A, Richards J, Jain J, Mott SL, Garje R. Patient Characteristics and Survival Outcomes of Non-Metastatic, Non-Clear Cell Renal Cell Carcinoma. Front Oncol 2022; 11:786307. [PMID: 35083144 PMCID: PMC8786111 DOI: 10.3389/fonc.2021.786307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/09/2021] [Indexed: 11/17/2022] Open
Abstract
Background Non-clear cell renal cell carcinoma (ccRCC) includes histologically and molecularly distinct subtypes such as papillary, chromophobe, collecting duct, and sarcomatoid RCC, with an incidence ranging from 20% to 25%. Oncologic outcomes and the role of adjuvant systemic therapy [vascular endothelial growth factor inhibitor (VEGFi) or immunotherapy] for non-ccRCC are not well-described. Objective To assess the incidence and survival outcomes of non-ccRCC subtypes in comparison to ccRCC. Methods The National Cancer Database was utilized to identify patients with non-metastatic RCC (T1–T4, N0–N1) between 2004 and 2015. The non-ccRCC cohort was further stratified by histologic subtype: papillary, chromophobe, sarcomatoid, and collecting duct RCC. Multivariable Cox regression models were used to compare overall survival (OS). Results The 5-year OS for chromophobe, papillary, clear cell, collecting duct, and sarcomatoid RCC was 91%, 82%, 81%, 44%, and 40%, respectively. After adjusting for clinicopathologic and treatment characteristics, there was no significant difference in OS between papillary RCC and ccRCC (p = 0.17). Patients with collecting duct and sarcomatoid subtypes were at over two times increased risk of death compared to patients with clear cell (p < 0.01 and p < 0.01, respectively). Conversely, patients with chromophobe RCC were at 36% decreased risk of death compared to ccRCC (p < 0.01). Conclusions This hospital-based analysis confirms that collecting duct and sarcomatoid histologic subtypes are uncommon and associated with poor survival after surgery when compared to the other RCC subtypes. Further studies are needed to evaluate the role of neoadjuvant and adjuvant systemic therapies in these subtypes to improve oncologic outcomes.
Collapse
Affiliation(s)
- Josiah An
- Division of Hematology, Oncology, Blood & Marrow Transplantation, Department of Internal Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Vignesh T Packiam
- Department of Urology, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Adithya Chennamadhavuni
- Division of Hematology, Oncology, Blood & Marrow Transplantation, Department of Internal Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Jordan Richards
- Department of Urology, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Jayanshu Jain
- Department of Internal Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Sarah L Mott
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Rohan Garje
- Division of Hematology, Oncology, Blood & Marrow Transplantation, Department of Internal Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| |
Collapse
|
18
|
Wang G, Beeraka NM, Xiao W, Zhang Y, Xue N, Chen G, Liu J, Liu Y. Comparative Clinical Efficacy of 'Concurrent Chemoradiotherapy (CCRT) and Anlotinib' Than CCRT in Patients with Locally Advanced ESCC. Technol Cancer Res Treat 2022; 21:15330338221080939. [PMID: 35235470 PMCID: PMC8894970 DOI: 10.1177/15330338221080939] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/14/2022] [Accepted: 01/31/2022] [Indexed: 12/24/2022] Open
Abstract
Objective: Radiotherapy or chemoradiotherapy has been preferred as the clinical therapeutic modalities to combat locally advanced esophageal squamous cell carcinoma (ESCC). The aim of this retrospective study is to ascertain combinatorial efficacy of anlotinib with concurrent radiotherapy (CCRT) rather than CCRT alone. Methods: Locally advanced ESCC patients registered between August 2018 to April 2019 in the third People's hospital of Zhengzhou, the First affiliated hospital of Zhengzhou University, Anyang Cancer Hospital, the Affiliated Hospital of Qingdao University were selected for this retrospective study; and these patients segregated into two groups subsequently who received combinatorial regimen with CCRT and anlotinib compared for treatment-related toxicity, response rates, safety, survival outcomes, than CCRT alone. Results: Progression free survival (PFS) was 0.577 (95% CI, 0.333-0.902, P = 0.014); the median overall survival time was 5 months (95% CI, 4.1-7.5) for the CCRT group, whereas 9 months (95% CI, 7.3-18.0) for the group received 'anlotinib with CCRT' (HR = 0.578, 95% CI, 0.337-0.924, P = 0.021). Overall objective response rates were considerable with a statistical difference between the two groups at 6 months (P1 = 0.027, P2 = 0.015) and 12 months (P1 = 0.012, P2 = 0.027). Overall adverse events are mitigated in combinatorial regimen than CCRT alone except the incidence of hypertension, which was higher in 'anlotinib with CCRT' group than CCRT group (P = 0.023). Total 13 patients exhibited hand-foot skin reactions in the group that received anlotinib in combination with CCRT. Anlotinib in combination with CCRT enhanced the overall survival (OS) rates, whereas incidence of treatment-related toxicity is minimized than CCRT alone. Conclusion: Combinatorial regimen of anlotinib with CCRT significantly enhanced clinical efficacy, safety and may benefit for treating the locally advanced ESCC patients.
Collapse
Affiliation(s)
- Gang Wang
- Department of Radiation Oncology, Zhengzhou Third People's Hospital & Zhengzhou Cancer Hospital Affiliated to Henan University, Zhengzhou, China
| | - Narasimha M. Beeraka
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Human Anatomy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Academy of Higher Education and Research (JSS AHER), Mysuru, Karnataka India
| | - Wenjing Xiao
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yaowen Zhang
- Department of Radiation Oncology, Anyang Cancer Hospital, The Fourth Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Anyang, China
| | - Nannan Xue
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Gongan Chen
- Department of Radiation Oncology, Zhengzhou Third People's Hospital & Zhengzhou Cancer Hospital Affiliated to Henan University, Zhengzhou, China
| | - Junqi Liu
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Liu
- Department of Radiotherapy, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
19
|
Zhou W, Peng C, Liu Z, Cao W. A novel clinical signature predicts the survival of elderly patients with oral squamous cell carcinoma. Eur Arch Otorhinolaryngol 2022; 279:391-398. [PMID: 33783598 DOI: 10.1007/s00405-021-06786-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/24/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND The risk factors for the survival of elderly patients with oral squamous cell carcinoma (OSCC) are multifarious. Here, we developed a novel clinical signature to serve as an indicator of prognosis in these patients. MATERIALS AND METHODS Clinicopathological data were collected for 554 patients aged ≥ 60 years who were treated for primary OSCC. Overall survival (OS), disease-specific survival, and disease-free survival were the primary outcomes. RESULTS Multivariate cox regression analysis showed that high N stage, low hemoglobin level, low body mass index (BMI), and high neutrophil-to-lymphocyte ratio (NLR) showed a poor survival (P < 0.05). A nomogram was constructed with a c-index of 0.702. CONCLUSION A novel clinical signature including hemoglobin level, BMI, and NLR, which are obtained through noninvasive examinations can be used as prognostic indicators in clinical practice for elderly patients with OSCC.
Collapse
Affiliation(s)
- Wenkai Zhou
- Department of Oral and Maxillofacial and Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, 200011, People's Republic of China
| | - Canbang Peng
- School and Hospital of Stomatology, Kunming Medical University, Kunming, China
| | - Zheqi Liu
- Department of Oral and Maxillofacial and Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, 200011, People's Republic of China.
| | - Wei Cao
- Department of Oral and Maxillofacial and Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, 200011, People's Republic of China.
| |
Collapse
|
20
|
Abstract
Mechanistic target of rapamycin (mTOR) inhibitors are macrocyclic lactone antibiotics derived from Streptomyces hygroscopicus that prevent T lymphocyte activation and B cell differentiation. Unlike calcineurin inhibitors (CNIs) that inhibit cytokine production, mTOR inhibitors block the cytokine signal transduction to arrest cells in the G1 to S phase. This class of drugs is commonly used for post-transplantation and cancer management because of its immunosuppressive and antiproliferative properties, respectively. The potential uses of mTOR inhibitors are heavily explored because of their impact on cell growth and proliferation. However, mTOR inhibitors have a broad range of effects that can result in adverse reactions, but side effects can occur with other immunosuppressive agents as well. Thus, the performance of mTOR inhibitors is compared to the outcomes and adverse effects of other immunosuppressive drugs or the combination of other immunosuppressants and mTOR inhibitors. Because mTOR regulates many downstream pathways, mTOR inhibitors can affect these pathways to manage various diseases. Sirolimus (rapamycin) is approved by the Food and Drug Administration (FDA) to treat post-renal transplantation and lymphangioleiomyomatosis (LAM). Everolimus is approved by the FDA to treat postmenopausal advanced hormone receptor-positive, HER2-negative breast cancer in women, progressive neuroendocrine tumors of pancreatic origin (PNET), advanced renal cell carcinoma (RCC), renal angiomyolipoma (AML) and tuberous sclerosis complex (TSC), and subependymal giant cell astrocytoma (SEGA) associated with TSC as well as renal and liver transplantation. Temsirolimus is approved by the FDA to treat advanced RCC. Opportunities to use mTOR inhibitors as therapy for other transplantation, metabolic disease, and cancer management are being researched. mTOR inhibitors are often called proliferation signal inhibitors (PSIs) because of their effects on proliferation pathways.
Collapse
Affiliation(s)
- Denise Wang
- Mount Sinai Icahn School of Medicine, New York, NY, USA.
| | - Howard J Eisen
- Pennsylvania State University College of Medicine/Milton S. Hershey Medical Center, Hershey, PA, USA
| |
Collapse
|
21
|
Cancel M, Fromont G, Blonz C, Chevreau C, Rioux-Leclercq N, Laguerre B, Oudard S, Gross-Goupil M, Gravis G, Goldwasser F, Rolland F, Delva R, Moise L, Emambux S, Vassal C, Zanetta S, Penel N, Fléchon A, Barthélémy P, Saldana C, Lefort F, Escudier B, Linassier C, Albiges L. Everolimus or sunitinib as first-line treatment of metastatic papillary renal cell carcinoma: A retrospective study of the GETUG group (Groupe d'Etude des Tumeurs Uro-Génitales). Eur J Cancer 2021; 158:1-11. [PMID: 34619467 DOI: 10.1016/j.ejca.2021.08.046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/24/2021] [Accepted: 08/31/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Two phase II trials (NCT00688753 and NCT00541008) reported efficacy data of sunitinib and everolimus in first-line treatment of metastatic papillary renal cell carcinoma (mpRCC). Although most patients receive sunitinib or a mammalian target of rapamycin (mTOR) inhibitor in first- and second-line treatment, the optimal strategy remained unknown. MATERIAL AND METHODS In 23 centres of the Groupe d'Etude des Tumeurs Urogénitales group, after centralised pathological review, we analysed retrospectively progression-free survival (PFS) of patients with mpRCC treated in first-line treatment (PFS-1) with sunitinib or everolimus (primary end-point), PFS in second-line treatment (PFS-2), overall survival (OS), objective response rate, disease control rate (DCR), overall sequence and prognostic factors for OS (secondary end-points). RESULTS One hundred thirty-eight patients (119 men and 19 women), median age 62.5 years, with mpRCC type 1 (n = 24) or non-type 1 (n = 114), received first-line sunitinib (n = 107) or everolimus (n = 31). With a median follow-up of 92 months, we found no significant difference between the treatment groups in terms of PFS-1 (5.5 versus 6.2 months) and DCR (69% versus 83%). Ninety-eight patients received a second-line treatment, 69% with mTOR inhibitors after sunitinib and 100% with tyrosine kinase inhibitors after everolimus, with similar DCR (64% versus 58%), median PFS-2 (3.4 versus 4.8 months) and OS (16.0 versus 20.3 months). No factor was prognostic for PFS-1, whereas leukocytosis, anaemia and the time from diagnosis to first systemic therapy < 1 year were prognostic for OS. We found no prognostic difference between both pRCC subtypes. The International Metastatic Renal Cell Database Consortium risk factors were prognostic for OS. CONCLUSION Sunitinib and everolimus had similar efficacy in first-line treatment of patients with mpRCC.
Collapse
Affiliation(s)
- Mathilde Cancel
- Department of Medical Oncology, CHU Bretonneau Tours, France
| | | | - Cyriac Blonz
- Department of Medical Oncology, Institut de Cancérologie de L'Ouest, Saint-Herblain, France
| | - Christine Chevreau
- Department of Medical Oncology, IUCT Oncopole - CLCC Institut Claudius Regaud, Toulouse, France
| | | | | | - Stéphane Oudard
- Department of Medical Oncology, CHU Hôpital Européen Georges Pompidou, Paris, France
| | | | - Gwenaelle Gravis
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | | | - Frédéric Rolland
- Department of Medical Oncology, Institut de Cancérologie de L'Ouest, Saint-Herblain, France
| | - Rémy Delva
- Department of Medical Oncology, Institut de Cancérologie de L'Ouest, Angers, France
| | - Laura Moise
- Department of Medical Oncology, Centre François Baclesse, Caen, France
| | - Sheik Emambux
- Department of Medical Oncology, CHU La Milétrie, Poitiers, France
| | - Cécile Vassal
- Department of Medical Oncology, Institut de Cancérologie Lucien Neuwirth, Saint-Priest-en-Jarez, France
| | - Sylvie Zanetta
- Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France
| | - Nicolas Penel
- Lille University and Department of Medical Oncology, Centre Oscar Lambret, Lille, France
| | - Aude Fléchon
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - Philippe Barthélémy
- Department of Medical Oncology, Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| | | | - Félix Lefort
- Department of Medical Oncology, CHU Saint-André, Bordeaux, France
| | - Bernard Escudier
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | | | - Laurence Albiges
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| |
Collapse
|
22
|
Yu EM, Linville L, Rosenthal M, Aragon-Ching JB. A Contemporary Review of Immune Checkpoint Inhibitors in Advanced Clear Cell Renal Cell Carcinoma. Vaccines (Basel) 2021; 9:919. [PMID: 34452045 PMCID: PMC8402652 DOI: 10.3390/vaccines9080919] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/06/2021] [Accepted: 08/10/2021] [Indexed: 12/20/2022] Open
Abstract
The use of checkpoint inhibitors in advanced and metastatic renal cell carcinomas (RCCs) has rapidly evolved over the past several years. While immune-oncology (IO) drug therapy has been successful at resulting in improved responses and survival, combination therapies with immune checkpoint inhibitors and vascular endothelial growth factor (VEGF) inhibitors have further improved outcomes. This article reviews the landmark trials that have led to the approval of IO therapies, including the Checkmate 214 trial and combination IO/VEGF TKI therapies with Checkmate 9ER, CLEAR, and Keynote-426, and it includes a discussion on promising therapies moving in the future.
Collapse
Affiliation(s)
- Eun-mi Yu
- GU Medical Oncology, Inova Schar Cancer Institute, Fairfax, VA 22031, USA;
| | - Laura Linville
- Department of Internal Medicine, George Washington University Medical Center, Washington, DC 20037, USA; (L.L.); (M.R.)
| | - Matthew Rosenthal
- Department of Internal Medicine, George Washington University Medical Center, Washington, DC 20037, USA; (L.L.); (M.R.)
| | | |
Collapse
|
23
|
Tomita Y, Kimura G, Fukasawa S, Numakura K, Sugiyama Y, Yamana K, Naito S, Kaneko H, Tajima Y, Oya M. Subgroup analysis of the AFTER I-O study: a retrospective study on the efficacy and safety of subsequent molecular targeted therapy after immune-oncology therapy in Japanese patients with metastatic renal cell carcinoma. Jpn J Clin Oncol 2021; 51:1656-1664. [PMID: 34350454 PMCID: PMC8558912 DOI: 10.1093/jjco/hyab114] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/05/2021] [Indexed: 11/13/2022] Open
Abstract
Background We performed subgroup analyses of the AFTER I-O study to clarify the association of time-to-treatment failure (TTF) and discontinuation reason of prior immune-oncology (I-O) therapy, and molecular targeted therapy (TT) regimen with the outcomes of TT after I-O. Methods The data of Japanese metastatic renal cell carcinoma patients treated with TT after nivolumab (NIVO) (CheckMate 025) or NIVO + ipilimumab (IPI) (CheckMate 214) were retrospectively analyzed. The objective response rates (ORRs), progression-free survival (PFS) and overall survival (OS) of TT after I-O were analyzed by subgroups: TTF (<6 or ≥6 months) and discontinuation reason of prior I-O (progression or adverse events), and TT regimen (sunitinib or axitinib). We also analyzed PFS2 of prior I-O and OS from first-line therapy. Results The ORR and median PFS of TT after NIVO and NIVO+IPI among the subgroups was 17–36% and 20–44%, and 7.1–11.6 months and 16.3-not reached (NR), respectively. The median OS of TT after NIVO was longer in patients with longer TTF of NIVO and treated with axitinib. Conversely, median OS of TT after NIVO+IPI was similar among subgroups. The median PFS2 of NIVO and NIVO+IPI was 36.7 and 32.0 months, respectively. The median OS from first-line therapy was 70.5 months for patients treated with NIVO and NR with NIVO+IPI. The safety profile of each TT after each I-O was similar to previous reports. Conclusions The efficacy of TT after NIVO or NIVO+IPI was favorable regardless of the TTF and discontinuation reason of prior I-O, and TT regimen.
Collapse
Affiliation(s)
- Yoshihiko Tomita
- Department of Urology, Molecular Oncology, Graduate School of Medicine and Dental Sciences, Niigata University, Niigata, Japan
| | - Go Kimura
- Department of Urology, Nippon Medical School Hospital, Tokyo, Japan
| | - Satoshi Fukasawa
- Prostate Center and Division of Urology, Chiba Cancer Center, Chiba, Japan
| | - Kazuyuki Numakura
- Department of Urology, Akita University Graduate School of Medicine, Akita, Japan
| | - Yutaka Sugiyama
- Department of Urology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kazutoshi Yamana
- Department of Urology, Molecular Oncology, Graduate School of Medicine and Dental Sciences, Niigata University, Niigata, Japan
| | - Sei Naito
- Department of Urology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | | | | | - Mototsugu Oya
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
24
|
Sharma R, Kadife E, Myers M, Kannourakis G, Prithviraj P, Ahmed N. Determinants of resistance to VEGF-TKI and immune checkpoint inhibitors in metastatic renal cell carcinoma. J Exp Clin Cancer Res 2021; 40:186. [PMID: 34099013 PMCID: PMC8183071 DOI: 10.1186/s13046-021-01961-3] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/25/2021] [Indexed: 01/03/2023] Open
Abstract
Vascular endothelial growth factor tyrosine kinase inhibitors (VEGF-TKIs) have been the mainstay of treatment for patients with advanced renal cell carcinoma (RCC). Despite its early promising results in decreasing or delaying the progression of RCC in patients, VEGF-TKIs have provided modest benefits in terms of disease-free progression, as 70% of the patients who initially respond to the treatment later develop drug resistance, with 30% of the patients innately resistant to VEGF-TKIs. In the past decade, several molecular and genetic mechanisms of VEGF-TKI resistance have been reported. One of the mechanisms of VEGF-TKIs is inhibition of the classical angiogenesis pathway. However, recent studies have shown the restoration of an alternative angiogenesis pathway in modulating resistance. Further, in the last 5 years, immune checkpoint inhibitors (ICIs) have revolutionized RCC treatment. Although some patients exhibit potent responses, a non-negligible number of patients are innately resistant or develop resistance within a few months to ICI therapy. Hence, an understanding of the mechanisms of VEGF-TKI and ICI resistance will help in formulating useful knowledge about developing effective treatment strategies for patients with advanced RCC. In this article, we review recent findings on the emerging understanding of RCC pathology, VEGF-TKI and ICI resistance mechanisms, and potential avenues to overcome these resistance mechanisms through rationally designed combination therapies.
Collapse
Affiliation(s)
- Revati Sharma
- Fiona Elsey Cancer Research Institute, Ballarat, Victoria, 3350, Australia
- Federation University Australia, Ballarat, Victoria, 3350, Australia
| | - Elif Kadife
- Fiona Elsey Cancer Research Institute, Ballarat, Victoria, 3350, Australia
| | - Mark Myers
- Federation University Australia, Ballarat, Victoria, 3350, Australia
| | - George Kannourakis
- Fiona Elsey Cancer Research Institute, Ballarat, Victoria, 3350, Australia
- Federation University Australia, Ballarat, Victoria, 3350, Australia
| | | | - Nuzhat Ahmed
- Fiona Elsey Cancer Research Institute, Ballarat, Victoria, 3350, Australia.
- Federation University Australia, Ballarat, Victoria, 3350, Australia.
- The Hudson Institute of Medical Research, Clayton, Victoria, 3168, Australia.
- Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, Victoria, 3052, Australia.
| |
Collapse
|
25
|
Patwardhan GA, Marczyk M, Wali VB, Stern DF, Pusztai L, Hatzis C. Treatment scheduling effects on the evolution of drug resistance in heterogeneous cancer cell populations. NPJ Breast Cancer 2021; 7:60. [PMID: 34040000 PMCID: PMC8154902 DOI: 10.1038/s41523-021-00270-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 04/19/2021] [Indexed: 12/12/2022] Open
Abstract
The effect of scheduling of targeted therapy combinations on drug resistance is underexplored in triple-negative breast cancer (TNBC). TNBC constitutes heterogeneous cancer cell populations the composition of which can change dynamically during treatment resulting in the selection of resistant clones with a fitness advantage. We evaluated crizotinib (ALK/MET inhibitor) and navitoclax (ABT-263; Bcl-2/Bcl-xL inhibitor) combinations in a large design consisting of 696 two-cycle sequential and concomitant treatment regimens with varying treatment dose, duration, and drug holiday length over a 26-day period in MDA-MB-231 TNBC cells and found that patterns of resistance depend on the schedule and sequence in which the drugs are given. Further, we tracked the clonal dynamics and mechanisms of resistance using DNA-integrated barcodes and single-cell RNA sequencing. Our study suggests that longer formats of treatment schedules in vitro screening assays are required to understand the effects of resistance and guide more realistically in vivo and clinical studies.
Collapse
Affiliation(s)
- Gauri A Patwardhan
- Breast Medical Oncology, Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Michal Marczyk
- Breast Medical Oncology, Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
- Department of Data Science and Engineering, Silesian University of Technology, Gliwice, Poland
| | - Vikram B Wali
- Breast Medical Oncology, Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - David F Stern
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Lajos Pusztai
- Breast Medical Oncology, Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Christos Hatzis
- Breast Medical Oncology, Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
26
|
Thrombospondin-2 and LDH Are Putative Predictive Biomarkers for Treatment with Everolimus in Second-Line Metastatic Clear Cell Renal Cell Carcinoma (MARC-2 Study). Cancers (Basel) 2021; 13:cancers13112594. [PMID: 34070677 PMCID: PMC8199288 DOI: 10.3390/cancers13112594] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/20/2021] [Accepted: 05/20/2021] [Indexed: 01/10/2023] Open
Abstract
Simple Summary Treatment of metastatic renal cell carcinoma (mRCC) remains a challenge due to the lack of biomarkers indicating the optimal drug for each patient. This study analyzed blood samples of patients with predominant clear cell mRCC who were treated with the mTOR inhibitor everolimus after failure of one prior tumor therapy. In an exploratory approach, predictive blood biomarkers were searched. We found lower levels of the protein thrombospondin-2 (TSP-2) at the start of the therapy and higher lactate dehydrogenase (LDH) levels in serum two weeks after therapy initiation to be associated with therapy response. Of note, these blood biomarkers had a higher predictive value than baseline patient parameters or risk classifications. Polymorphisms in the mTOR gene appeared to be associated with therapy response, but were not significant. To conclude, it seems feasible to identify patients showing longtime responses to everolimus and possible to increase tumor therapy response rates based on biomarkers for individual therapy selection. Abstract There is an unmet need for predictive biomarkers in metastatic renal cell carcinoma (mRCC) therapy. The phase IV MARC-2 trial searched for predictive blood biomarkers in patients with predominant clear cell mRCC who benefit from second-line treatment with everolimus. In an exploratory approach, potential biomarkers were assessed employing proteomics, ELISA, and polymorphism analyses. Lower levels of angiogenesis-related protein thrombospondin-2 (TSP-2) at baseline (≤665 parts per billion, ppb) identified therapy responders with longer median progression-free survival (PFS; ≤665 ppb at baseline: 6.9 months vs. 1.8, p = 0.005). Responders had higher lactate dehydrogenase (LDH) levels in serum two weeks after therapy initiation (>27.14 nmol/L), associated with a longer median PFS (3.8 months vs. 2.2, p = 0.013) and improved overall survival (OS; 31.0 months vs. 14.0 months, p < 0.001). Baseline TSP-2 levels had a stronger relation to PFS (HR 0.36, p = 0.008) than baseline patient parameters, including IMDC score. Increased serum LDH levels two weeks after therapy initiation were the best predictor for OS (HR 0.21, p < 0.001). mTOR polymorphisms appeared to be associated with therapy response but were not significant. Hence, we identified TSP-2 and LDH as promising predictive biomarkers for therapy response on everolimus after failure of one VEGF-targeted therapy in patients with clear cell mRCC.
Collapse
|
27
|
de Vries-Brilland M, McDermott DF, Suárez C, Powles T, Gross-Goupil M, Ravaud A, Flippot R, Escudier B, Albigès L. Checkpoint inhibitors in metastatic papillary renal cell carcinoma. Cancer Treat Rev 2021; 99:102228. [PMID: 34111642 DOI: 10.1016/j.ctrv.2021.102228] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/13/2021] [Accepted: 05/17/2021] [Indexed: 02/02/2023]
Abstract
Papillary Renal Cell Carcinoma (pRCC) is the most common non-clear cell RCC (nccRCC) and a distinct entity, although heterogenous, associated with poor outcomes. The treatment landscape of metastatic pRCC (mpRCC) relied so far on targeted therapies, mimicking previous developments in metastatic clear-cell renal cell carcinoma. However, antiangiogenics as well as mTOR inhibitors retain only limited activity in mpRCC. As development of immune checkpoint inhibitors (ICI) is now underway in patients with mpRCC, we aimed at discussing early activity data and potential for future therapeutic strategies in monotherapy or combination. Expression of immune checkpoints such as PD-L1 and infiltrative immune cells in pRCC could provide insights into their potential immunogenicity, although this is currently poorly described. Based on retrospective and prospective data, efficacy of ICI as single agent remains limited. Combinations with tyrosine-kinase inhibitors, notably with anti-MET inhibitors, harbor promising response rates and may enter the standard of care in untreated patients. Collaborative work is needed to refine the molecular and immune landscape of pRCC, and pursue efforts to set up predictive biomarker-driven clinical trials in these rare tumors.
Collapse
Affiliation(s)
- M de Vries-Brilland
- Department of Medical Oncology, Integrated Centers of Oncology (ICO) Paul Papin, Angers, France
| | - D F McDermott
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - C Suárez
- Medical Oncology, Vall d́Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d́Hebron, Vall d́Hebron Barcelona Hospital Campus, Spain
| | - T Powles
- The Royal Free NHS Trust and Barts Cancer Institute, Queen Mary University of London, London, UK
| | - M Gross-Goupil
- Department of Medical Oncology, Hôpital Saint-André, University of Bordeaux-CHU, Bordeaux, France
| | - A Ravaud
- Department of Medical Oncology, Hôpital Saint-André, University of Bordeaux-CHU, Bordeaux, France
| | - R Flippot
- Medical Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - B Escudier
- Medical Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - L Albigès
- Medical Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France.
| |
Collapse
|
28
|
Patel HV, Srivastava A, Srinivasan R, Singer EA. A challenging frontier - the genomics and therapeutics of nonclear cell renal cell carcinoma. Curr Opin Oncol 2021; 33:212-220. [PMID: 33818540 PMCID: PMC8244822 DOI: 10.1097/cco.0000000000000721] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW As molecular profiling of renal cell carcinoma (RCC) continues to elucidate novel targets for nonclear cell histologies, understanding the landscape of these targets is of utmost importance. In this review, we highlight the genomic landscape of nonclear cell RCC and its implications for current and future systemic therapies. RECENT FINDINGS Several genomic studies have described the mutational burden among nonclear cell histologies. These studies have highlighted the importance of MET in papillary RCC and led to several clinical trials evaluating the efficacy of MET inhibitors for papillary RCC. The success of immune checkpoint inhibitors, such as ipilimumab and nivolumab, in clear cell RCC has led to ongoing trials evaluating these novel therapeutics in nonclear cell RCC. SUMMARY Genomic profiling has allowed for the evaluation of novel targets for nonclear cell RCC. This evolving therapeutic landscape is being explored in promising, ongoing trials that have the potential for changing how nonclear cell RCC is managed.
Collapse
Affiliation(s)
- Hiren V. Patel
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Arnav Srivastava
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Ramaprasad Srinivasan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Eric A. Singer
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| |
Collapse
|
29
|
Huang JJ, Hsieh JJ. The Therapeutic Landscape of Renal Cell Carcinoma: From the Dark Age to the Golden Age. Semin Nephrol 2021; 40:28-41. [PMID: 32130964 DOI: 10.1016/j.semnephrol.2019.12.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Oncologic treatments for renal cell carcinoma (RCC) have undergone a major revolution in the past 2 decades, moving away from the pre-2004 Dark Age during which interleukin 2 and interferon-α were the only therapeutic options and induced treatment responses in only 5% to 10% of patients with metastatic disease. The development of anti-angiogenic tyrosine kinase inhibitors against vascular endothelial growth factor receptor 2 and inhibitors of mammalian target of rapamycin complex 1 in 2005 introduced the Modern Age with better overall and progression-free survival and a greater number of patients (30%-40%) responding to and (∼80%) benefiting from these targeted therapeutic agents. The coming of age of the immuno-oncology era with the use of immune checkpoint inhibitors (ICIs) have ushered us into the Golden Age of metastatic RCC care, in which combined administrations of two ICIs (anti-programmed cell death protein 1/programmed death-ligand 1 and anti-cytotoxic T-lymphocyte-associated protein 4 or one tyrosine kinase inhibitor plus one ICI (anti-programmed cell death protein 1/programmed death-ligand 1) have recast the treatment landscape of clear cell RCC, the most common RCC subtype, with an approximately 60% response rate and an approximately 90% disease control rate that further improves metastatic RCC survival. Exciting clinical trials are in the pipeline investigating complementary/synergistic molecular mechanisms, based on studies investigating the biology, pathology, and genomics of renal carcinoma and the respective treatment outcome. This will enable us to enter the Diamond Age of precision medicine in which a specific treatment can be tailored to the specific biological and pathologic circumstance of an individual kidney tumor to offer more effective yet less toxic therapy.
Collapse
Affiliation(s)
- Jennifer J Huang
- Molecular Oncology, Department of Medicine, Siteman Cancer Center, Washington University, St. Louis, MO
| | - James J Hsieh
- Molecular Oncology, Department of Medicine, Siteman Cancer Center, Washington University, St. Louis, MO.
| |
Collapse
|
30
|
Kajdasz A, Majer W, Kluzek K, Sobkowiak J, Milecki T, Derebecka N, Kwias Z, Bluyssen HAR, Wesoly J. Identification of RCC Subtype-Specific microRNAs-Meta-Analysis of High-Throughput RCC Tumor microRNA Expression Data. Cancers (Basel) 2021; 13:548. [PMID: 33535553 PMCID: PMC7867039 DOI: 10.3390/cancers13030548] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 11/17/2022] Open
Abstract
Renal cell carcinoma (RCC) is one of the most common cancers worldwide with a nearly non-symptomatic course until the advanced stages of the disease. RCC can be distinguished into three subtypes: papillary (pRCC), chromophobe (chRCC) and clear cell renal cell carcinoma (ccRCC) representing up to 75% of all RCC cases. Detection and RCC monitoring tools are limited to standard imaging techniques, in combination with non-RCC specific morphological and biochemical read-outs. RCC subtype identification relays mainly on results of pathological examination of tumor slides. Molecular, clinically applicable and ideally non-invasive tools aiding RCC management are still non-existent, although molecular characterization of RCC is relatively advanced. Hence, many research efforts concentrate on the identification of molecular markers that will assist with RCC sub-classification and monitoring. Due to stability and tissue-specificity miRNAs are promising candidates for such biomarkers. Here, we performed a meta-analysis study, utilized seven NGS and seven microarray RCC studies in order to identify subtype-specific expression of miRNAs. We concentrated on potentially oncocytoma-specific miRNAs (miRNA-424-5p, miRNA-146b-5p, miRNA-183-5p, miRNA-218-5p), pRCC-specific (miRNA-127-3p, miRNA-139-5p) and ccRCC-specific miRNAs (miRNA-200c-3p, miRNA-362-5p, miRNA-363-3p and miRNA-204-5p, 21-5p, miRNA-224-5p, miRNA-155-5p, miRNA-210-3p) and validated their expression in an independent sample set. Additionally, we found ccRCC-specific miRNAs to be differentially expressed in ccRCC tumor according to Fuhrman grades and identified alterations in their isoform composition in tumor tissue. Our results revealed that changes in the expression of selected miRNA might be potentially utilized as a tool aiding ccRCC subclass discrimination and we propose a miRNA panel aiding RCC subtype distinction.
Collapse
Affiliation(s)
- Arkadiusz Kajdasz
- Laboratory of Human Molecular Genetics, Faculty of Biology, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University Poznan, Uniwersytetu Poznanskiego 6, 61-614 Poznan, Poland; (K.K.); (H.A.R.B.)
| | - Weronika Majer
- Laboratory of High Throughput Technologies, Faculty of Biology, Adam Mickiewicz University Poznan, Uniwersytetu Poznanskiego 6, 61-614 Poznan, Poland; (W.M.); (N.D.)
| | - Katarzyna Kluzek
- Laboratory of Human Molecular Genetics, Faculty of Biology, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University Poznan, Uniwersytetu Poznanskiego 6, 61-614 Poznan, Poland; (K.K.); (H.A.R.B.)
| | - Jacek Sobkowiak
- Department of Urology, Poznan University of Medical Sciences, Szwajcarska 3, 61-285 Poznan, Poland; (J.S.); (T.M.); (Z.K.)
| | - Tomasz Milecki
- Department of Urology, Poznan University of Medical Sciences, Szwajcarska 3, 61-285 Poznan, Poland; (J.S.); (T.M.); (Z.K.)
| | - Natalia Derebecka
- Laboratory of High Throughput Technologies, Faculty of Biology, Adam Mickiewicz University Poznan, Uniwersytetu Poznanskiego 6, 61-614 Poznan, Poland; (W.M.); (N.D.)
| | - Zbigniew Kwias
- Department of Urology, Poznan University of Medical Sciences, Szwajcarska 3, 61-285 Poznan, Poland; (J.S.); (T.M.); (Z.K.)
| | - Hans A. R. Bluyssen
- Laboratory of Human Molecular Genetics, Faculty of Biology, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University Poznan, Uniwersytetu Poznanskiego 6, 61-614 Poznan, Poland; (K.K.); (H.A.R.B.)
| | - Joanna Wesoly
- Laboratory of High Throughput Technologies, Faculty of Biology, Adam Mickiewicz University Poznan, Uniwersytetu Poznanskiego 6, 61-614 Poznan, Poland; (W.M.); (N.D.)
| |
Collapse
|
31
|
Schmidt AL, Bain PA, McGregor BA. Tissue Based Biomarkers for Metastatic Clear Cell Renal Carcinoma: A Systematic Review. KIDNEY CANCER 2020. [DOI: 10.3233/kca-200103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background: Treatments for metastatic clear cell renal carcinoma (mccRCC) are evolving with multiple targeted and immune therapy drugs currently approved by regulatory agencies as single agents or in combination. Developing predictive biomarkers to determine which patients derive a differential benefit from a particular treatment is an area of ongoing clinical research. Objective: We sought to systematically evaluate the role of tumour tissue-based biomarkers that assist in selection of therapy for mccRCC. Methods: Literature addressing the role of biomarkers in mccRCC was identified through a search of the electronic databases MEDLINE, Embase, and the Web of Science and a hand search of major conference abstracts (from Jan 2010 –Sep 2020). Abstracts were screened to identify papers meriting full-text review. Studies with a comparison arm were included to assess biomarker relevance. A narrative review of studies was performed. Results: The literature search yielded 6784 potentially relevant articles. 133 articles met criteria for full text review, and 10 articles were identified by scanning bibliographies of relevant studies. A total of 33 articles (involving 13 studies) were selected for data extraction and subsequent review. Conclusions: Predictive biomarkers for immediate use in the clinic are lacking, and embedding their evaluation and validation in future clinical trials is needed to refine practice and patient selection.
Collapse
Affiliation(s)
- Andrew L. Schmidt
- Lark Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Paul A. Bain
- Countway Library, Harvard Medical School, Boston, MA, USA
| | - Bradley A. McGregor
- Lark Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
32
|
Khan Y, Slattery TD, Pickering LM. Individualizing Systemic Therapies in First Line Treatment and beyond for Advanced Renal Cell Carcinoma. Cancers (Basel) 2020; 12:E3750. [PMID: 33322163 PMCID: PMC7764621 DOI: 10.3390/cancers12123750] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/28/2020] [Accepted: 12/07/2020] [Indexed: 12/15/2022] Open
Abstract
Therapeutic options for treating advanced renal cell cancer (RCC) are rapidly evolving. Vascular endothelial growth factor (VEGF)-directed therapy, predominantly VEGF receptor (VEGFr) tyrosine kinase inhibitors (TKIs) had been the most effective first line treatment since 2005 irrespective of International Metastatic RCC Database Consortium (IMDC) risk stratification. However, immune checkpoint inhibitors (ICI) have recently changed the treatment paradigm for advanced RCC particularly as the first-line systemic treatment modality. The combination of Ipilimumab and Nivolumab provides better disease control and long-term outcomes compared with the anti-VEGFr TKI Sunitinib for IMDC intermediate- to poor-risk patients and we now have the option of using ICI with TKI upfront for all IMDC risk groups. This poses a challenge for physicians, both to select the most suitable first line regimen and the most suitable subsequent therapy given the lack of data about sequencing in this setting. This treatment landscape is expected to become more complex with the emerging treatment options. Moreover, these therapeutic options cannot be generalized as significant variability exists between individual's disease biologies and their physiologies for handling treatment adverse effects. Notable efforts are being made to identify promising predictive biomarkers ranging from neo-antigen load to gene expression profiling. These biomarkers need prospective validation to justify their utility in clinical practice and in treatment decision making. This review article discusses various clinicopathological characteristics that should be carefully evaluated to help select appropriate treatment and discusses the current status of biomarker-based selection.
Collapse
Affiliation(s)
| | | | - Lisa M. Pickering
- The Royal Marsden Hospital NHS Foundation Trust, Fulham Road, London SW3 6JJ, UK; (Y.K.); (T.D.S.)
| |
Collapse
|
33
|
Hofmann F, Hwang EC, Lam TB, Bex A, Yuan Y, Marconi LS, Ljungberg B. Targeted therapy for metastatic renal cell carcinoma. Cochrane Database Syst Rev 2020; 10:CD012796. [PMID: 33058158 PMCID: PMC8094280 DOI: 10.1002/14651858.cd012796.pub2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Several comparative randomised controlled trials (RCTs) have been performed including combinations of tyrosine kinase inhibitors (TKIs) and immune checkpoint inhibitors since the publication of a Cochrane Review on targeted therapy for metastatic renal cell carcinoma (mRCC) in 2008. This review represents an update of that original review. OBJECTIVES To assess the effects of targeted therapies for clear cell mRCC in patients naïve to systemic therapy. SEARCH METHODS We performed a comprehensive search with no restrictions on language or publication status. The date of the latest search was 18 June 2020. SELECTION CRITERIA We included randomised controlled trials, recruiting patients with clear cell mRCC naïve to previous systemic treatment. The index intervention was any TKI-based targeted therapy. DATA COLLECTION AND ANALYSIS Two review authors independently assessed the included studies and extracted data for the primary outcomes: progression-free survival (PFS), overall survival (OS) and serious adverse events (SAEs); and the secondary outcomes: health-related quality of life (QoL), response rate and minor adverse events (AEs). We performed statistical analyses using a random-effects model and rated the certainty of evidence according to the GRADE approach. MAIN RESULTS We included 18 RCTs reporting on 11,590 participants randomised across 18 comparisons. This abstract focuses on the primary outcomes of select comparisons. 1. Pazopanib versus sunitinib Pazopanib may result in little to no difference in PFS as compared to sunitinib (hazard ratio (HR) 1.05, 95% confidence interval (CI) 0.90 to 1.23; 1 study, 1110 participants; low-certainty evidence). Based on the control event risk of 420 per 1000 in this trial at 12 months, this corresponds to 18 fewer participants experiencing PFS (95% CI 76 fewer to 38 more) per 1000 participants. Pazopanib may result in little to no difference in OS compared to sunitinib (HR 0.92, 95% CI 0.80 to 1.06; 1 study, 1110 participants; low-certainty evidence). Based on the control event risk of 550 per 1000 in this trial at 12 months, this corresponds to 27 more OSs (95% CI 19 fewer to 70 more) per 1000 participants. Pazopanib may result in little to no difference in SAEs as compared to sunitinib (risk ratio (RR) 1.01, 95% CI 0.94 to 1.09; 1 study, 1102 participants; low-certainty evidence). Based on the control event risk of 734 per 1000 in this trial, this corresponds to 7 more participants experiencing SAEs (95% CI 44 fewer to 66 more) per 1000 participants. 2. Sunitinib versus avelumab and axitinib Sunitinib probably reduces PFS as compared to avelumab plus axitinib (HR 1.45, 95% CI 1.17 to 1.80; 1 study, 886 participants; moderate-certainty evidence). Based on the control event risk of 550 per 1000 in this trial at 12 months, this corresponds to 130 fewer participants experiencing PFS (95% CI 209 fewer to 53 fewer) per 1000 participants. Sunitinib may result in little to no difference in OS (HR 1.28, 95% CI 0.92 to 1.79; 1 study, 886 participants; low-certainty evidence). Based on the control event risk of 890 per 1000 in this trial at 12 months, this would result in 29 fewer OSs (95% CI 78 fewer to 8 more) per 1000 participants. Sunitinib may result in little to no difference in SAEs (RR 1.01, 95% CI 0.93 to 1.10; 1 study, 873 participants; low-certainty evidence). Based on the control event risk of 705 per 1000 in this trial, this corresponds to 7 more SAEs (95% CI 49 fewer to 71 more) per 1000 participants. 3. Sunitinib versus pembrolizumab and axitinib Sunitinib probably reduces PFS as compared to pembrolizumab plus axitinib (HR 1.45, 95% CI 1.19 to 1.76; 1 study, 861 participants; moderate-certainty evidence). Based on the control event risk of 590 per 1000 in this trial at 12 months, this corresponds to 125 fewer participants experiencing PFS (95% CI 195 fewer to 56 fewer) per 1000 participants. Sunitinib probably reduces OS (HR 1.90, 95% CI 1.36 to 2.65; 1 study, 861 participants; moderate-certainty evidence). Based on the control event risk of 880 per 1000 in this trial at 12 months, this would result in 96 fewer OSs (95% CI 167 fewer to 40 fewer) per 1000 participants. Sunitinib may reduce SAEs as compared to pembrolizumab plus axitinib (RR 0.90, 95% CI 0.81 to 1.02; 1 study, 854 participants; low-certainty evidence) although the CI includes the possibility of no effect. Based on the control event risk of 604 per 1000 in this trial, this corresponds to 60 fewer SAEs (95% CI 115 fewer to 12 more) per 1000 participants. 4. Sunitinib versus nivolumab and ipilimumab Sunitinib may reduce PFS as compared to nivolumab plus ipilimumab (HR 1.30, 95% CI 1.11 to 1.52; 1 study, 847 participants; low-certainty evidence). Based on the control event risk of 280 per 1000 in this trial at 30 months' follow-up, this corresponds to 89 fewer PFSs (95% CI 136 fewer to 37 fewer) per 1000 participants. Sunitinib reduces OS (HR 1.52, 95% CI 1.23 to 1.89; 1 study, 847 participants; high-certainty evidence). Based on the control event risk 600 per 1000 in this trial at 30 months, this would result in 140 fewer OSs (95% CI 219 fewer to 67 fewer) per 1000 participants. Sunitinib probably increases SAEs (RR 1.37, 95% CI 1.22 to 1.53; 1 study, 1082 participants; moderate-certainty evidence). Based on the control event risk of 457 per 1000 in this trial, this corresponds to 169 more SAEs (95% CI 101 more to 242 more) per 1000 participants. AUTHORS' CONCLUSIONS Based on the low to high certainty of evidence, several combinations of immune checkpoint inhibitors appear to be superior to single-agent targeted therapy in terms of PFS and OS, and with a favourable AE profile. Some single-agent targeted therapies demonstrated a similar or improved oncological outcome compared to others; minor differences were observed for AE within this group. The certainty of evidence was variable ranging from high to very low and all comparisons were based on single trials.
Collapse
Key Words
- adult
- humans
- antibodies, monoclonal, humanized
- antibodies, monoclonal, humanized/adverse effects
- antibodies, monoclonal, humanized/therapeutic use
- antineoplastic agents
- antineoplastic agents/adverse effects
- antineoplastic agents/therapeutic use
- antineoplastic agents, immunological
- antineoplastic agents, immunological/therapeutic use
- axitinib
- axitinib/adverse effects
- axitinib/therapeutic use
- bevacizumab
- bevacizumab/adverse effects
- bevacizumab/therapeutic use
- bias
- carcinoma, renal cell
- carcinoma, renal cell/drug therapy
- carcinoma, renal cell/mortality
- everolimus
- everolimus/adverse effects
- everolimus/therapeutic use
- indazoles
- ipilimumab
- ipilimumab/adverse effects
- ipilimumab/therapeutic use
- kidney neoplasms
- kidney neoplasms/drug therapy
- kidney neoplasms/mortality
- kidney neoplasms/pathology
- phenylurea compounds
- phenylurea compounds/adverse effects
- phenylurea compounds/therapeutic use
- progression-free survival
- protein kinase inhibitors
- protein kinase inhibitors/adverse effects
- protein kinase inhibitors/therapeutic use
- pyrimidines
- pyrimidines/adverse effects
- pyrimidines/therapeutic use
- quality of life
- quinolines
- quinolines/adverse effects
- quinolines/therapeutic use
- randomized controlled trials as topic
- receptors, vascular endothelial growth factor
- receptors, vascular endothelial growth factor/antagonists & inhibitors
- sirolimus
- sirolimus/adverse effects
- sirolimus/analogs & derivatives
- sirolimus/therapeutic use
- sorafenib
- sorafenib/adverse effects
- sorafenib/therapeutic use
- sulfonamides
- sulfonamides/adverse effects
- sulfonamides/therapeutic use
- sunitinib
- sunitinib/adverse effects
- sunitinib/therapeutic use
Collapse
Affiliation(s)
- Fabian Hofmann
- Department of Urology, Sunderby Sjukhus, Umeå University, Luleå, Sweden
| | - Eu Chang Hwang
- Department of Urology, Chonnam National University Medical School, Chonnam National University Hwasun Hospital, Hwasun, Korea, South
| | - Thomas Bl Lam
- Academic Urology Unit, University of Aberdeen, Aberdeen, UK
| | - Axel Bex
- Department of Urology and UCL Division of Surgery and Interventional Science, Royal Free London NHS Foundation Trust, London, UK
| | - Yuhong Yuan
- Department of Medicine, Division of Gastroenterology, McMaster University, Hamilton, Canada
| | - Lorenzo So Marconi
- Department of Urology and Renal Transplantation, Centro Hospitalar e Universitario de Coimbra, Coimbra, Portugal
| | - Börje Ljungberg
- Department of Surgical and Perioperative Sciences, Umeå University, Umeå, Sweden
| |
Collapse
|
34
|
Hadjipanteli A, Doolan P, Kyriacou E, Constantinidou A. Breast Cancer Brain Metastasis: The Potential Role of MRI Beyond Current Clinical Applications. Cancer Manag Res 2020; 12:9953-9964. [PMID: 33116852 PMCID: PMC7567538 DOI: 10.2147/cmar.s252801] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 06/29/2020] [Indexed: 12/30/2022] Open
Abstract
PURPOSE Breast cancer brain metastasis (BCBM) represents a major clinical challenge. Can MRI help in advancements in the management of BCBM? This review discusses MRI developments and the corresponding potential advancements in BCBM management. METHODS An exhaustive literature search was undertaken to identify studies which look into the potential of MRI in BCBM management. Seven hundred and eighty-four studies published from September 1984 to May 2020 were identified. Three topics are covered where MRI is not clinically established yet: 1) the prognosis of BCBM; 2) the screening of BC patients for BCBM development, and 3) the assessment of imaging features correlated to BC subtype. RESULTS Thirty-six studies were considered eligible for the purposes of this review. On-going progress is made with the identification of different BCBM characteristics and MRI metrics that might be related to prognosis. Progress has been made with the identification of different BCBM characteristics, including BCBM location, degree of edema, white matter disruption, tumor edge sharpness, and temporal muscle thickness. A more accurate prediction of prognosis could lead to more suitable patient management and treatment. The use of MRI in BCBM screening of the high-risk breast cancer population remains a controversial subject. To date, there are no results from clinical trials; however, there is a rising number of relatively small studies that show concern on this subject and support BCBM screening. It is important to oncologists to be able to assess the tumor subtype non-invasively. MRI features, which have shown some correlation with subtype, include the number of tumors, location, and their distribution in the brain. Advanced tools and metrics have been produced to carry out radiological characteristics analysis on MRI images. Assessing MRI features in more detail could provide a more personalized management of patients. CONCLUSION Developments in the use of MRI have the potential to improve BCBM management.
Collapse
Affiliation(s)
- Andria Hadjipanteli
- Medical School, University of Cyprus, Shacolas Educational Centre for Clinical Medicine, Aglantzia, Nicosia2029, Cyprus
- Bank of Cyprus Oncology Centre, Nicosia2006, Cyprus
| | - Paul Doolan
- German Oncology Center, Limassol, Agios Athanasios4108, Cyprus
| | | | - Anastasia Constantinidou
- Medical School, University of Cyprus, Shacolas Educational Centre for Clinical Medicine, Aglantzia, Nicosia2029, Cyprus
- Bank of Cyprus Oncology Centre, Nicosia2006, Cyprus
| |
Collapse
|
35
|
Huang JJ, Hsieh JJ. The Pan-Omics Landscape of Renal Cell Carcinoma and Its Implication on Future Clinical Practice. KIDNEY CANCER 2020; 4:121-129. [PMID: 33195887 PMCID: PMC7605346 DOI: 10.3233/kca-200085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Renal cell carcinoma has traditionally been classified based on histological features. Contemporary studies have identified genomic, transcriptomic, epigenomic, and metabolomic signatures that correspond to or even transcend histological subtypes. Much remains to be learned about improving the algorithm of pan-omics integration for precision oncology, which will not only advance our understanding of RCC pathobiology and treatment response but also result in novel therapeutic opportunities. Accordingly, this review focuses on recent RCC multi-omics literature. Encouragingly, a few reports on omics integration into routinely employed prognostic risk models have shown early promise that could lay the foundation for future development of precision kidney cancer therapies. Hence, this article serves as a primer on what we have learned and how we might better realize the clinical potential of the burgeoning pan-omics data.
Collapse
Affiliation(s)
- Jennifer J Huang
- Department of Medicine, Molecular Oncology, Washington University, St. Louis, MO, USA
| | - James J Hsieh
- Department of Medicine, Molecular Oncology, Washington University, St. Louis, MO, USA
| |
Collapse
|
36
|
Huelse J, Fridlyand D, Earp S, DeRyckere D, Graham DK. MERTK in cancer therapy: Targeting the receptor tyrosine kinase in tumor cells and the immune system. Pharmacol Ther 2020; 213:107577. [PMID: 32417270 PMCID: PMC9847360 DOI: 10.1016/j.pharmthera.2020.107577] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The receptor tyrosine kinase MERTK is aberrantly expressed in numerous human malignancies, and is a novel target in cancer therapeutics. Physiologic roles of MERTK include regulation of tissue homeostasis and repair, innate immune control, and platelet aggregation. However, aberrant expression in a wide range of liquid and solid malignancies promotes neoplasia via growth factor independence, cell cycle progression, proliferation and tumor growth, resistance to apoptosis, and promotion of tumor metastases. Additionally, MERTK signaling contributes to an immunosuppressive tumor microenvironment via induction of an anti-inflammatory cytokine profile and regulation of the PD-1 axis, as well as regulation of macrophage, myeloid-derived suppressor cell, natural killer cell and T cell functions. Various MERTK-directed therapies are in preclinical development, and clinical trials are underway. In this review we discuss MERTK inhibition as an emerging strategy for cancer therapy, focusing on MERTK expression and function in neoplasia and its role in mediating resistance to cytotoxic and targeted therapies as well as in suppressing anti-tumor immunity. Additionally, we review preclinical and clinical pharmacological strategies to target MERTK.
Collapse
Affiliation(s)
- Justus Huelse
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Diana Fridlyand
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Shelton Earp
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Deborah DeRyckere
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Douglas K. Graham
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University, Atlanta, Georgia
| |
Collapse
|
37
|
Huelse JM, Fridlyand DM, Earp S, DeRyckere D, Graham DK. MERTK in cancer therapy: Targeting the receptor tyrosine kinase in tumor cells and the immune system. Pharmacol Ther 2020. [PMID: 32417270 DOI: 10.1016/j.pharmthera.2020.107577107577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
The receptor tyrosine kinase MERTK is aberrantly expressed in numerous human malignancies, and is a novel target in cancer therapeutics. Physiologic roles of MERTK include regulation of tissue homeostasis and repair, innate immune control, and platelet aggregation. However, aberrant expression in a wide range of liquid and solid malignancies promotes neoplasia via growth factor independence, cell cycle progression, proliferation and tumor growth, resistance to apoptosis, and promotion of tumor metastases. Additionally, MERTK signaling contributes to an immunosuppressive tumor microenvironment via induction of an anti-inflammatory cytokine profile and regulation of the PD-1 axis, as well as regulation of macrophage, myeloid-derived suppressor cell, natural killer cell and T cell functions. Various MERTK-directed therapies are in preclinical development, and clinical trials are underway. In this review we discuss MERTK inhibition as an emerging strategy for cancer therapy, focusing on MERTK expression and function in neoplasia and its role in mediating resistance to cytotoxic and targeted therapies as well as in suppressing anti-tumor immunity. Additionally, we review preclinical and clinical pharmacological strategies to target MERTK.
Collapse
Affiliation(s)
- Justus M Huelse
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Diana M Fridlyand
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Shelton Earp
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Deborah DeRyckere
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Douglas K Graham
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University, Atlanta, GA, USA.
| |
Collapse
|
38
|
Gao Y, Liu P, Shi R. Anlotinib as a molecular targeted therapy for tumors. Oncol Lett 2020; 20:1001-1014. [PMID: 32724339 PMCID: PMC7377159 DOI: 10.3892/ol.2020.11685] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/30/2020] [Indexed: 12/24/2022] Open
Abstract
Angiogenesis has an essential role in tumor growth and metastasis, and blocking this pathway has been a successfully utilized strategy in the clinical treatment of cancer. Anlotinib (AL3818) is a novel oral receptor tyrosine kinase inhibitor targeting vascular endothelial growth factor receptor 2 and 3, fibroblast growth factor 1-4, platelet-derived growth factor receptor α and β, c-Kit and Ret. Anlotinib exerts inhibitory effects on tumor growth and angiogenesis and received its first approval as a third-line treatment for refractory advanced non-small-cell lung cancer in May 2018 and its second approval as a second-line treatment for advanced soft-tissue sarcoma in June 2019 in the People's Republic of China. Anlotinib has encouraging efficacy and a manageable and tolerable safety profile in a broad range of malignancies, including medullary thyroid cancer, renal cell cancer, gastric cancer and esophageal squamous cell carcinoma. In the present review, the preclinical and clinical trials of anlotinib were summarized with a focus on safety evaluation and adverse event management.
Collapse
Affiliation(s)
- Yi Gao
- School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
- Department of Gastroenterology, The Jiangyin Clinical College of Xuzhou Medical University, Jiangyin, Jiangsu 214400, P.R. China
- Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Pengfei Liu
- Department of Gastroenterology, The Jiangyin Clinical College of Xuzhou Medical University, Jiangyin, Jiangsu 214400, P.R. China
- State Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Ruihua Shi
- School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
- Department of Gastroenterology, Zhongda Hospital, Affiliated Hospital of Southeast University, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
39
|
Abstract
Variant histology renal cell carcinoma (vRCC) encompasses rare non-clear cell subtypes that have long been associated with poor prognosis and minimal response to therapies targeting vascular endothelial growth factor and its receptor. Molecular advances have helped classify vRCC into distinct entities and identify putative targetable driver alterations, such as MET in papillary subtypes. More have since been identified in other vRCC subtypes, including alterations of tumor metabolism, chromatin remodeling genes, cell-cycle genes, and inactivation of tumor suppressors such as TP53 or NF2. New targeted therapies, as well as immune checkpoint inhibitors, have been in development and yielded encouraging results. Collaborative clinical trials will be an essential step toward better implementation of these regimens in clinical practice.
Collapse
|
40
|
Les cancers du rein non à cellules claires : caractéristiques clinico-biologiques et prise en charge thérapeutique hors chirurgie. Bull Cancer 2020; 107:S56-S65. [DOI: 10.1016/s0007-4551(20)30279-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
41
|
Survival Outcomes After Adding Stereotactic Body Radiotherapy to Metastatic Renal Cell Carcinoma Patients Treated With Tyrosine Kinase Inhibitors. Am J Clin Oncol 2020; 43:58-63. [PMID: 31651452 PMCID: PMC6922069 DOI: 10.1097/coc.0000000000000622] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
OBJECTIVE Long-lasting control is rarely achieved with tyrosine kinase inhibitors (TKI) alone in metastatic renal cell carcinoma (mRCC). Our study aimed to investigate the survival outcomes of adding stereotactic body radiotherapy (SBRT) to TKI in mRCC. MATERIALS AND METHODS From September 2015 to September 2018, 56 patients treated with TKI received SBRT for 103 unresectable lesions. A total of 24 and 32 patients were irradiated before and after TKI failure, respectively. Overall survival (OS) was calculated from metastases. Progression-free survival (PFS) was calculated from SBRT. RESULTS Overall, 10, 32, and 12 patients had International Metastatic Renal Cell Carcinoma Database Consortium favorable, intermediate, and poor risk. Median follow-up was 21.7 months (range, 5.1 to 110.6 mo). Median OS was 61.2 months. The median PFS was 11.5 months, while the 2-year LC rate was 94%. Sixteen (34%) lesions achieved complete response (CR) in patients irradiated before TKI failure, whereas only 4 (7%) lesions yielded CR in those irradiated after TKI failure (P=0.001). The median PFS in CR group was significantly longer than that of non-CR group (18.9 vs. 7.1 mo; P=0.003). The 5-year OS in CR group was 86%, compared with 48% in non-CR group (P=0.010). Four (7%) patients experienced Grade 3 toxicity. CONCLUSIONS Adding SBRT to TKI is safe and seems to improve survival in mRCC. Patients irradiated before TKI failure have higher CR rate, and the favorable local response might turn into survival benefit.
Collapse
|
42
|
Gulati S, Philip E, Salgia S, Pal SK. Evolving treatment paradigm in metastatic non clear cell renal cell carcinoma. Cancer Treat Res Commun 2020; 23:100172. [PMID: 32252014 DOI: 10.1016/j.ctarc.2020.100172] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 12/18/2022]
Abstract
The treatment landscape for renal cell cancer (RCC) has evolved tremendously over the last two decades. Treatment algorithms have shifted from the highly toxic drugs with marginal benefit to better tolerated and more effective targeted therapy drugs. The latter include tyrosine kinase inhibitors, vascular endothelial growth factor inhibitors, MET inhibitors and, more recently, immunotherapy drugs alone and in various combinations. The majority of treatment algorithms for non-clear cell carcinoma have been based on extrapolating results from clear cell RCC trials and retrospective reviews. However, now that we understand that non-clear cell RCC is morphologically and clinically distinct from its clear cell counterpart, several collaborative clinical trials are underway for non-clear cell RCC. This review will delve into the historical aspects of treating non-clear cell RCC and the evolution of treatment paradigms over the last few decades with a focus on immunotherapy based trials.
Collapse
Affiliation(s)
- Shuchi Gulati
- Clinical Medicine, Division of Hematology and Oncology, University of Cincinnati Cancer Institute, Cincinnati, OH 45267-0562, United States
| | - Errol Philip
- University of California San Francisco School of Medicine, San Francisco, CA 94143, United States
| | - Sabrina Salgia
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, CA 91010, United States
| | - Sumanta K Pal
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, CA 91010, United States.
| |
Collapse
|
43
|
Abstract
PURPOSE OF REVIEW Due to the rapidly changing field of kidney cancer therapeutics, addressing the state of the art systemic therapy regimens, and sequencing with cytoreductive nephrectomy are the primary focus of this review. We will also discuss the role of biomarkers and novel therapeutic targets in the management of renal cell carcinoma. RECENT FINDINGS The management of metastatic renal cell cancer has undergone a paradigm shift with immune checkpoint inhibitors being used in the frontline setting. Over the last 4 years, programmed cell death-1 (PD-1) inhibitors as well as programmed cell death ligand-1 inhibitors have become available in various combinations with cytotoxic T lymphocyte-associated protein-4 (CTLA-4) inhibitors and tyrosine kinase inhibitors (TKIs). These drugs have improved outcomes in patients with renal cell cancer and more work is being done to refine these targets as well as discover newer ones. Despite the availability of several new treatment options, some questions that still need to be addressed in the management of kidney cancer include the sequencing of treatment options, treatment of patients who progress on immune checkpoint inhibitors, and role of biomarkers to ascertain the best treatment options to minimize costs and improve outcomes.
Collapse
|
44
|
Clinical Activity of Ipilimumab Plus Nivolumab in Patients With Metastatic Non-Clear Cell Renal Cell Carcinoma. Clin Genitourin Cancer 2019; 18:429-435. [PMID: 32800717 DOI: 10.1016/j.clgc.2019.11.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/26/2019] [Accepted: 11/27/2019] [Indexed: 01/24/2023]
Abstract
INTRODUCTION Ipilimumab plus nivolumab has been approved for intermediate- and poor-risk metastatic renal cell carcinoma (RCC). However, the activity in non-clear cell RCC (nccRCC) is unknown. PATIENTS AND METHODS Patients from Cleveland Clinic and the University of Texas Southwestern who had received ipilimumab plus nivolumab for metastatic nccRCC from October 2017 to May 2019 were retrospectively identified. Ipilimumab plus nivolumab was administered in accordance with the CHECKMATE 214 trial. Imaging was obtained at baseline and every 12 weeks. The baseline patient characteristics, objective response per Response Evaluation Criteria in Solid Tumors, version 1.1, and treatment-related adverse events (TRAEs) per Common Terminology Criteria for Adverse Events, version 5.0, were analyzed. RESULTS Eighteen patients were identified. The median age was 59 years (range, 32-81 years), 77.8% were men, and the Eastern Cooperative Oncology Group performance status was 0 (38%) or 1 (50%). The median treatment duration was 2.4 months (range, 0.7-12.3 months). The non-clear cell histologic types included 6 papillary, 5 chromophobe, 3 unclassified, 2 adenocarcinoma of renal origin, 1 translocation, and 1 medullary. Most had an intermediate (66%) or poor (22%) International Metastatic Database Consortium risk. The best objective response included 6 partial responses (PRs; 33.3%) and 3 with stable disease (16.7%). Of the patients with a PR, the median time to the best response was 3.0 months, and median duration of the PR was 4.3 months. The median progression-free survival was 7.1 months. All-grade TRAEs were noted in 11 patients (61.1%) and included colitis (22%), hepatotoxicity (16%), rash (11%), and fatigue (11%). Eleven patients (61%) had TRAEs requiring high-dose glucocorticoids (> 40 mg of prednisone equivalent daily). CONCLUSIONS Ipilimumab plus nivolumab demonstrated objective responses and notable toxicity in patients with nccRCC.
Collapse
|
45
|
Nandagopal L, Sonpavde GP, Agarwal N. Investigational MET inhibitors to treat Renal cell carcinoma. Expert Opin Investig Drugs 2019; 28:851-860. [DOI: 10.1080/13543784.2019.1673366] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
| | - Guru P. Sonpavde
- Dana Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Neeraj Agarwal
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
46
|
Zarrabi K, Paroya A, Wu S. Emerging therapeutic agents for genitourinary cancers. J Hematol Oncol 2019; 12:89. [PMID: 31484560 PMCID: PMC6727406 DOI: 10.1186/s13045-019-0780-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 08/21/2019] [Indexed: 12/11/2022] Open
Abstract
The treatment of genitourinary malignancies has dramatically evolved over recent years. Renal cell carcinoma, urothelial carcinoma of the bladder, and prostate adenocarcinoma are the most commonly encountered genitourinary malignancies and represent a heterogeneous population of cancers, in both histology and approach to treatment. However, all three cancers have undergone paradigm shifts in their respective therapeutic landscapes due to a greater understanding of their underlying molecular mechanisms and oncogenic drivers. The advance that has gained the most recent traction has been the advent of immunotherapies, particularly immune checkpoint inhibitors. Immunotherapy has increased overall survival and even provided durable responses in the metastatic setting in some patients. The early success of immune checkpoint inhibitors has led to further drug development with the emergence of novel agents which modulate the immune system within the tumor microenvironment. Notwithstanding immunotherapy, investigators are also developing novel agents tailored to a variety of targets including small-molecule tyrosine kinase inhibitors, mTOR inhibitors, and novel fusion proteins to name a few. Erdafitinib has become the first targeted therapy approved for metastatic bladder cancer. Moreover, the combination therapy of immune checkpoint inhibitors with targeted agents such as pembrolizumab or avelumab with axitinib has demonstrated both safety and efficacy and just received FDA approval for their use. We are in an era of rapid progression in drug development with multiple exciting trials and ongoing pre-clinical studies. We highlight many of the promising new emerging therapies that will likely continue to improve outcomes in patients with genitourinary malignancies.
Collapse
Affiliation(s)
- Kevin Zarrabi
- Department of Medicine, Stony Brook University Hospital, 9447 SUNY, Stony Brook, NY 11794-9447 USA
| | - Azzam Paroya
- Department of Medicine, Stony Brook University Hospital, 9447 SUNY, Stony Brook, NY 11794-9447 USA
| | - Shenhong Wu
- Department of Medicine, Stony Brook University Hospital, 9447 SUNY, Stony Brook, NY 11794-9447 USA
- Division of Hematology/Oncology, Department of Medicine, Northport VA Medical Center, Northport, NY USA
| |
Collapse
|
47
|
Lai Y, Zeng T, Liang X, Wu W, Zhong F, Wu W. Cell death-related molecules and biomarkers for renal cell carcinoma targeted therapy. Cancer Cell Int 2019; 19:221. [PMID: 31462894 PMCID: PMC6708252 DOI: 10.1186/s12935-019-0939-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 08/18/2019] [Indexed: 02/07/2023] Open
Abstract
Renal cell carcinoma (RCC) is not sensitive to conventional radio- and chemotherapies and is at least partially resistant to impairments in cell death-related signaling pathways. The hallmarks of RCC formation include diverse signaling pathways, such as maintenance of proliferation, cell death resistance, angiogenesis induction, immune destruction avoidance, and DNA repair. RCC diagnosed during the early stage has the possibility of cure with surgery. For metastatic RCC (mRCC), molecular targeted therapy, especially antiangiogenic therapy (e.g., tyrosine kinase inhibitors, TKIs, such as sunitinib), is one of the main partially effective therapeutics. Various forms of cell death that may be associated with the resistance to targeted therapy because of the crosstalk between targeted therapy and cell death resistance pathways were originally defined and differentiated into apoptosis, necroptosis, pyroptosis, ferroptosis and autophagic cell death based on cellular morphology. Particularly, as a new form of cell death, T cell-induced cell death by immune checkpoint inhibitors expands the treatment options beyond the current targeted therapy. Here, we provide an overview of cell death-related molecules and biomarkers for the progression, prognosis and treatment of mRCC by targeted therapy, with a focus on apoptosis and T cell-induced cell death, as well as other forms of cell death.
Collapse
Affiliation(s)
- Yongchang Lai
- Department of Urology, Minimally Invasive Surgery Center, Guangdong Key Laboratory of Urology, Guangzhou Urology Research Institute, The First Affiliated Hospital of Guangzhou Medical University, Kangda Road 1#, Haizhu District, Guangzhou, 510230 Guangdong China
| | - Tao Zeng
- Department of Urology, Minimally Invasive Surgery Center, Guangdong Key Laboratory of Urology, Guangzhou Urology Research Institute, The First Affiliated Hospital of Guangzhou Medical University, Kangda Road 1#, Haizhu District, Guangzhou, 510230 Guangdong China
| | - Xiongfa Liang
- Department of Urology, Minimally Invasive Surgery Center, Guangdong Key Laboratory of Urology, Guangzhou Urology Research Institute, The First Affiliated Hospital of Guangzhou Medical University, Kangda Road 1#, Haizhu District, Guangzhou, 510230 Guangdong China
| | - Weizou Wu
- Department of Urology, Minimally Invasive Surgery Center, Guangdong Key Laboratory of Urology, Guangzhou Urology Research Institute, The First Affiliated Hospital of Guangzhou Medical University, Kangda Road 1#, Haizhu District, Guangzhou, 510230 Guangdong China
| | - Fangling Zhong
- Department of Urology, Minimally Invasive Surgery Center, Guangdong Key Laboratory of Urology, Guangzhou Urology Research Institute, The First Affiliated Hospital of Guangzhou Medical University, Kangda Road 1#, Haizhu District, Guangzhou, 510230 Guangdong China
| | - Wenqi Wu
- Department of Urology, Minimally Invasive Surgery Center, Guangdong Key Laboratory of Urology, Guangzhou Urology Research Institute, The First Affiliated Hospital of Guangzhou Medical University, Kangda Road 1#, Haizhu District, Guangzhou, 510230 Guangdong China
| |
Collapse
|
48
|
The outcome to axitinib or everolimus after sunitinib in metastatic renal cell carcinoma. Anticancer Drugs 2019; 29:705-709. [PMID: 29846246 DOI: 10.1097/cad.0000000000000632] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
We aimed to investigate the different outcomes in patients with metastatic renal cell carcinoma treated with second-line axitinib or everolimus after sunitinib. Patients treated in 16 oncological centres in Italy were included, and those receiving axitinib or everolimus from January 2013 onwards were analysed for outcomes. Descriptive statistical tests were used to highlight differences between groups. The Kaplan-Meier method was used to estimate overall survival (OS) and progression-free survival (PFS). Data on 634 patients with metastatic renal cell carcinoma treated with first-line sunitinib have been obtained. A total of 182 patients received a second-line therapy with everolimus (79 patients, 43%) or axitinib (103 patients, 57%), respectively. The median PFS was 4.6 [95% confidence (CI): 2.6-6.5] months for patients treated with everolimus and 5.5 (95% CI: 4.3-6.7) months for patients treated with axitinib (P=0.7). The median OS was 13.9 (95% CI: 10.4-17.4) months for patients treated with everolimus and 12.0 (95% CI: 7.9-16.2) months for patients treated with axitinib (P=0.3). No differences were found based on length of first-line treatment. Major limitations are the retrospective nature of the study and the lack of a prospective evaluation of the progression. This study reports no significantly differences between everolimus and axitinib in terms of both PFS and OS. Furthermore, the length of first-line treatment cannot be used as such a predictive factor and cannot suggest the use of a molecule compared with another.
Collapse
|
49
|
Grunwitz C, Salomon N, Vascotto F, Selmi A, Bukur T, Diken M, Kreiter S, Türeci Ö, Sahin U. HPV16 RNA-LPX vaccine mediates complete regression of aggressively growing HPV-positive mouse tumors and establishes protective T cell memory. Oncoimmunology 2019; 8:e1629259. [PMID: 31428528 PMCID: PMC6685602 DOI: 10.1080/2162402x.2019.1629259] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 06/04/2019] [Accepted: 06/04/2019] [Indexed: 10/27/2022] Open
Abstract
HPV16 infections are associated with a variety of cancers and there is compelling evidence that the transforming activity of HPV16 critically depends on the expression of the viral oncoproteins E6 and E7. Therapeutic cancer vaccines capable of generating durable and specific immunity against these HPV16 antigens hold great promise to achieve long-term disease control. Here we show in mice that HPV16 E7 RNA-LPX, an intravenously administered cancer vaccine based on immuno-pharmacologically optimized antigen-encoding mRNA, efficiently primes and expands antigen-specific effector and memory CD8+ T cells. HPV-positive TC-1 and C3 tumors of immunized mice are heavily infiltrated with activated immune cells and HPV16-specific T cells and are polarized towards a proinflammatory, cytotoxic and less immune-suppressive contexture. E7 RNA-LPX immunization mediates complete and durable remission of progressing tumors. Circulating memory T cells are highly cytotoxic and protect from tumor rechallenge. Moreover, E7 RNA-LPX immunization sensitizes anti-PD-L1 refractory tumors to checkpoint blockade. In conclusion, our data highlight the potential of HPV16 RNA-LPX for the treatment of HPV-driven cancers.
Collapse
Affiliation(s)
- Christian Grunwitz
- Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany.,Research Center for Immunotherapy (FZI), University Medical Center Mainz, Mainz, Germany
| | - Nadja Salomon
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH, Mainz, Germany
| | - Fulvia Vascotto
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH, Mainz, Germany
| | - Abderaouf Selmi
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH, Mainz, Germany
| | - Thomas Bukur
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH, Mainz, Germany
| | - Mustafa Diken
- Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany.,TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH, Mainz, Germany
| | - Sebastian Kreiter
- Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany.,TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH, Mainz, Germany
| | - Özlem Türeci
- Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany
| | - Ugur Sahin
- Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany.,TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH, Mainz, Germany
| |
Collapse
|
50
|
Hua H, Kong Q, Zhang H, Wang J, Luo T, Jiang Y. Targeting mTOR for cancer therapy. J Hematol Oncol 2019; 12:71. [PMID: 31277692 PMCID: PMC6612215 DOI: 10.1186/s13045-019-0754-1] [Citation(s) in RCA: 597] [Impact Index Per Article: 99.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 06/14/2019] [Indexed: 02/05/2023] Open
Abstract
Mechanistic target of rapamycin (mTOR) is a protein kinase regulating cell growth, survival, metabolism, and immunity. mTOR is usually assembled into several complexes such as mTOR complex 1/2 (mTORC1/2). In cooperation with raptor, rictor, LST8, and mSin1, key components in mTORC1 or mTORC2, mTOR catalyzes the phosphorylation of multiple targets such as ribosomal protein S6 kinase β-1 (S6K1), eukaryotic translation initiation factor 4E binding protein 1 (4E-BP1), Akt, protein kinase C (PKC), and type-I insulin-like growth factor receptor (IGF-IR), thereby regulating protein synthesis, nutrients metabolism, growth factor signaling, cell growth, and migration. Activation of mTOR promotes tumor growth and metastasis. Many mTOR inhibitors have been developed to treat cancer. While some of the mTOR inhibitors have been approved to treat human cancer, more mTOR inhibitors are being evaluated in clinical trials. Here, we update recent advances in exploring mTOR signaling and the development of mTOR inhibitors for cancer therapy. In addition, we discuss the mechanisms underlying the resistance to mTOR inhibitors in cancer cells.
Collapse
Affiliation(s)
- Hui Hua
- State Key Laboratory of Biotherapy, Laboratory of Stem Cell Biology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qingbin Kong
- Laboratory of Oncogene, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hongying Zhang
- Laboratory of Oncogene, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jiao Wang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ting Luo
- Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yangfu Jiang
- Laboratory of Oncogene, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|