1
|
Choi SH, Chen YW, Panian J, Yuen K, McKay RR. Emerging innovative treatment strategies for advanced clear cell renal cell carcinoma. Oncologist 2024:oyae276. [PMID: 39401004 DOI: 10.1093/oncolo/oyae276] [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: 06/16/2024] [Accepted: 09/10/2024] [Indexed: 10/15/2024] Open
Abstract
Dramatic advances in biological discoveries, since the 1990s, have continued to reshape the treatment paradigm of metastatic renal cell carcinoma (RCC). Von Hippel Lindau (VHL) gene alterations are associated with pro-angiogenic activity and are central to the pathogenesis of clear cell RCC (ccRCC), the most predominant histologic subtype of RCC. Antiangiogenic strategies revolving around this VHL/HIF/VEGF axis have been shown to improve survival in metastatic ccRCC. The discovery of immune checkpoints and agents that target their inhibition introduced a new treatment paradigm for patients with RCC. While initially approved as monotherapy, studies investigating immune checkpoint inhibitor combinations have led to their approval as the new standard of care, providing durable responses and unprecedented improvements in clinical outcome. Despite these advances, the projected 14 390 deaths in 2024 from RCC underscore the need to continue efforts in expanding and optimizing treatment options for patients with metastatic RCC. This article reviews key findings that have transformed the way we understand and treat metastatic RCC, in addition to highlighting novel treatment strategies that are currently under development.
Collapse
Affiliation(s)
- Sharon H Choi
- Division of Hematology Oncology, University of California San Diego, San Diego, CA, United States
| | - Yu-Wei Chen
- Division of Hematology Oncology, University of California San Diego, San Diego, CA, United States
| | - Justine Panian
- Division of Hematology Oncology, University of California San Diego, San Diego, CA, United States
| | - Kit Yuen
- Department of Urology, University of California San Diego, San Diego, CA, United States
| | - Rana R McKay
- Division of Hematology Oncology, University of California San Diego, San Diego, CA, United States
- Department of Urology, University of California San Diego, San Diego, CA, United States
| |
Collapse
|
2
|
Lin YC, Ku CC, Wuputra K, Wu DC, Yokoyama KK. Vulnerability of Antioxidant Drug Therapies on Targeting the Nrf2-Trp53-Jdp2 Axis in Controlling Tumorigenesis. Cells 2024; 13:1648. [PMID: 39404411 PMCID: PMC11475825 DOI: 10.3390/cells13191648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/19/2024] Open
Abstract
Control of oxidation/antioxidation homeostasis is important for cellular protective functions, and disruption of the antioxidation balance by exogenous and endogenous ligands can lead to profound pathological consequences of cancerous commitment within cells. Although cancers are sensitive to antioxidation drugs, these drugs are sometimes associated with problems including tumor resistance or dose-limiting toxicity in host animals and patients. These problems are often caused by the imbalance between the levels of oxidative stress-induced reactive oxygen species (ROS) and the redox efficacy of antioxidants. Increased ROS levels, because of abnormal function, including metabolic abnormality and signaling aberrations, can promote tumorigenesis and the progression of malignancy, which are generated by genome mutations and activation of proto-oncogene signaling. This hypothesis is supported by various experiments showing that the balance of oxidative stress and redox control is important for cancer therapy. Although many antioxidant drugs exhibit therapeutic potential, there is a heterogeneity of antioxidation functions, including cell growth, cell survival, invasion abilities, and tumor formation, as well as the expression of marker genes including tumor suppressor proteins, cell cycle regulators, nuclear factor erythroid 2-related factor 2, and Jun dimerization protein 2; their effectiveness in cancer remains unproven. Here, we summarize the rationale for the use of antioxidative drugs in preclinical and clinical antioxidant therapy of cancer, and recent advances in this area using cancer cells and their organoids, including the targeting of ROS homeostasis.
Collapse
Affiliation(s)
- Ying-Chu Lin
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Chia-Chen Ku
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.K.); (K.W.)
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Kenly Wuputra
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.K.); (K.W.)
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Deng-Chyang Wu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Kazunari K. Yokoyama
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.K.); (K.W.)
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| |
Collapse
|
3
|
Ming Y, Gong Y, Fu X, Ouyang X, Peng Y, Pu W. Small-molecule-based targeted therapy in liver cancer. Mol Ther 2024; 32:3260-3287. [PMID: 39113358 PMCID: PMC11489561 DOI: 10.1016/j.ymthe.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 03/13/2024] [Accepted: 08/02/2024] [Indexed: 08/23/2024] Open
Abstract
Liver cancer is one of the most prevalent malignant tumors worldwide. According to the Barcelona Clinic Liver Cancer staging criteria, clinical guidelines provide tutorials to clinical management of liver cancer at their individual stages. However, most patients diagnosed with liver cancer are at advanced stage; therefore, many researchers conduct investigations on targeted therapy, aiming to improve the overall survival of these patients. To date, small-molecule-based targeted therapies are highly recommended (first line: sorafenib and lenvatinib; second line: regorafenib and cabozantinib) by current the clinical guidelines of the American Society of Clinical Oncology, European Society for Medical Oncology, and National Comprehensive Cancer Network. Herein, we summarize the small-molecule-based targeted therapies in liver cancer, including the approved and preclinical therapies as well as the therapies under clinical trials, and introduce their history of discovery, clinical trials, indications, and molecular mechanisms. For drug resistance, the revealed mechanisms of action and the combination therapies are also discussed. In fact, the known small-molecule-based therapies still have limited clinical benefits to liver cancer patients. Therefore, we analyze the current status and give our ideas for the urgent issues and future directions in this field, suggesting clues for novel techniques in liver cancer treatment.
Collapse
Affiliation(s)
- Yue Ming
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Yanqiu Gong
- National Clinical Research Center for Geriatrics and Department of General Practice, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xuewen Fu
- Jinhua Huanke Environmental Technology Co., Ltd., Jinhua 321000, China
| | - Xinyu Ouyang
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China; West China School of Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yong Peng
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China; Frontier Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, China.
| | - Wenchen Pu
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China; West China School of Medicine, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
4
|
Daher R, Ruplin A, Gupta S, Spiess PE, Kamat AM, Cigliola A, Tateo V, Mercinelli C, Grivas P, Necchi A. The spectrum of cutaneous toxicities related to novel genitourinary cancer therapies. Crit Rev Oncol Hematol 2024; 200:104420. [PMID: 38906514 DOI: 10.1016/j.critrevonc.2024.104420] [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: 03/15/2024] [Revised: 04/29/2024] [Accepted: 06/07/2024] [Indexed: 06/23/2024] Open
Abstract
CONTEXT Genitourinary cancers (GUCs) encompass malignancies affecting the urinary and reproductive systems, including renal cell carcinoma (RCC), urothelial carcinoma (UC), and prostate cancer (PC). With the rapidly evolving therapeutic domain of these cancers, cutaneous adverse events (AEs) remain among the most observed toxicities. OBJECTIVE To explore the dermatologic AEs linked to novel GUC treatments, their underlying pathophysiology, clinical presentations, and risk factors. EVIDENCE ACQUISITION A narrative review of the literature from PubMed and Embase databases was conducted. The search strategy included dermatologic/cutaneous adverse events, risk factors, and pathophysiology in conjunction with the following classes of therapies; immune checkpoint inhibitors (ICIs), antiangiogenic therapies, enfortumab vedotin (EV), erdafitinib, and androgen receptor antagonists (ARAs). EVIDENCE SYNTHESIS Maculopapular rash, pruritus, and alopecia are present among the five classes of therapies. ICIs demonstrate the highest incidence of severe drug AEs including Steven Johnson syndrome/toxic epidermal necrolysis. Unique cutaneous AEs present with specific therapies including hand-foot skin reaction and subungual splinter hemorrhage with antiangiogenic drugs, stomatitis/mucositis and onycholysis with erdafitinib. Incidence and type of cutaneous AE also differed within therapies in the same class as seen with apalutamide displaying the highest risk of cutaneous AEs within ARAs. Risk factors for development of cutaneous AEs can be general to therapies, or specific, and include age, immune status, BMI, and gender. CONCLUSIONS Dermatologic AEs may impact patients' quality of life and increase the tendency to dose reduce, hold or discontinue life-saving therapies, underscoring the need for vigilant monitoring, early recognition, and collaborative management between medical oncologists, pharmacists, dermatologists and other specialists.
Collapse
Affiliation(s)
- Rachel Daher
- Vita-Salute San Raffaele University, Milan, Italy.
| | - Andrew Ruplin
- University of Washington, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Shilpa Gupta
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Philippe E Spiess
- Department of Genitourinary Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Ashish M Kamat
- Department of Urology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Antonio Cigliola
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Valentina Tateo
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Chiara Mercinelli
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Petros Grivas
- University of Washington, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Andrea Necchi
- Vita-Salute San Raffaele University, Milan, Italy; Department of Medical Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| |
Collapse
|
5
|
Collinson F, Royle KL, Swain J, Ralph C, Maraveyas A, Eisen T, Nathan P, Jones R, Meads D, Min Wah T, Martin A, Bestall J, Kelly-Morland C, Linsley C, Oughton J, Chan K, Theodoulou E, Arias-Pinilla G, Kwan A, Daverede L, Handforth C, Trainor S, Salawu A, McCabe C, Goh V, Buckley D, Hewison J, Gregory W, Selby P, Brown J, Brown J. Temporary treatment cessation compared with continuation of tyrosine kinase inhibitors for adults with renal cancer: the STAR non-inferiority RCT. Health Technol Assess 2024; 28:1-171. [PMID: 39250424 PMCID: PMC11403377 DOI: 10.3310/jwtr4127] [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] [Indexed: 09/11/2024] Open
Abstract
Background There is interest in using treatment breaks in oncology, to reduce toxicity without compromising efficacy. Trial design A Phase II/III multicentre, open-label, parallel-group, randomised controlled non-inferiority trial assessing treatment breaks in patients with renal cell carcinoma. Methods Patients with locally advanced or metastatic renal cell carcinoma, starting tyrosine kinase inhibitor as first-line treatment at United Kingdom National Health Service hospitals. Interventions At trial entry, patients were randomised (1 : 1) to a drug-free interval strategy or a conventional continuation strategy. After 24 weeks of treatment with sunitinib/pazopanib, drug-free interval strategy patients took up a treatment break until disease progression with additional breaks dependent on disease response and patient choice. Conventional continuation strategy patients continued on treatment. Both trial strategies continued until treatment intolerance, disease progression on treatment, withdrawal or death. Objective To determine if a drug-free interval strategy is non-inferior to a conventional continuation strategy in terms of the co-primary outcomes of overall survival and quality-adjusted life-years. Co-primary outcomes For non-inferiority to be concluded, a margin of ≤ 7.5% in overall survival and ≤ 10% in quality-adjusted life-years was required in both intention-to-treat and per-protocol analyses. This equated to the 95% confidence interval of the estimates being above 0.812 and -0.156, respectively. Quality-adjusted life-years were calculated using the utility index of the EuroQol-5 Dimensions questionnaire. Results Nine hundred and twenty patients were randomised (461 conventional continuation strategy vs. 459 drug-free interval strategy) from 13 January 2012 to 12 September 2017. Trial treatment and follow-up stopped on 31 December 2020. Four hundred and eighty-eight (53.0%) patients [240 (52.1%) vs. 248 (54.0%)] continued on trial post week 24. The median treatment-break length was 87 days. Nine hundred and nineteen patients were included in the intention-to-treat analysis (461 vs. 458) and 871 patients in the per-protocol analysis (453 vs. 418). For overall survival, non-inferiority was concluded in the intention-to-treat analysis but not in the per-protocol analysis [hazard ratio (95% confidence interval) intention to treat 0.97 (0.83 to 1.12); per-protocol 0.94 (0.80 to 1.09) non-inferiority margin: 95% confidence interval ≥ 0.812, intention to treat: 0.83 > 0.812 non-inferior, per-protocol: 0.80 < 0.812 not non-inferior]. Therefore, a drug-free interval strategy was not concluded to be non-inferior to a conventional continuation strategy in terms of overall survival. For quality-adjusted life-years, non-inferiority was concluded in both the intention-to-treat and per-protocol analyses [marginal effect (95% confidence interval) intention to treat -0.05 (-0.15 to 0.05); per-protocol 0.04 (-0.14 to 0.21) non-inferiority margin: 95% confidence interval ≥ -0.156]. Therefore, a drug-free interval strategy was concluded to be non-inferior to a conventional continuation strategy in terms of quality-adjusted life-years. Limitations The main limitation of the study is the fewer than expected overall survival events, resulting in lower power for the non-inferiority comparison. Future work Future studies should investigate treatment breaks with more contemporary treatments for renal cell carcinoma. Conclusions Non-inferiority was shown for the quality-adjusted life-year end point but not for overall survival as pre-defined. Nevertheless, despite not meeting the primary end point of non-inferiority as per protocol, the study suggested that a treatment-break strategy may not meaningfully reduce life expectancy, does not reduce quality of life and has economic benefits. Although the treating clinicians' perspectives were not formally collected, the fact that clinicians recruited a large number of patients over a long period suggests support for the study and provides clear evidence that a treatment-break strategy for patients with renal cell carcinoma receiving tyrosine kinase inhibitor therapy is feasible. Trial registration This trial is registered as ISRCTN06473203. Funding This award was funded by the National Institute for Health and Care Research (NIHR) Health Technology Assessment Programme (NIHR award ref: 09/91/21) and is published in full in Health Technology Assessment; Vol. 28, No. 45. See the NIHR Funding and Awards website for further award information.
Collapse
Affiliation(s)
- Fiona Collinson
- Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - Kara-Louise Royle
- Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - Jayne Swain
- Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - Christy Ralph
- Leeds Institute of Medical Research, St James's University Hospital, University of Leeds, Leeds, UK
| | - Anthony Maraveyas
- Academic Oncology, Faculty of Health Sciences, Hull York Medical School, Queens Centre Oncology and Haematology, Hull, UK
| | - Tim Eisen
- Department of Oncology, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge, UK
| | - Paul Nathan
- Department of Oncology, Mount Vernon Cancer Centre, East and North Hertfordshire NHS Trust, Hertfordshire, UK
| | - Robert Jones
- School of Cancer Sciences, University of Glasgow, Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - David Meads
- Academic Unit of Health Economics, University of Leeds, Leeds, UK
| | - Tze Min Wah
- Department of Diagnostic and Interventional Radiology, Leeds Teaching Hospitals Trust, Leeds, UK
| | - Adam Martin
- Academic Unit of Health Economics, University of Leeds, Leeds, UK
| | - Janine Bestall
- Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | | | | | - Jamie Oughton
- Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - Kevin Chan
- Medical Oncology, Weston Park Cancer Hospital, Sheffield, UK
| | - Elisavet Theodoulou
- Division of Clinical Medicine, University of Sheffield, Weston Park Hospital, Sheffield, UK
| | - Gustavo Arias-Pinilla
- Division of Clinical Medicine, University of Sheffield, Weston Park Hospital, Sheffield, UK
| | - Amy Kwan
- Academic Unit of Clinical Oncology, University of Sheffield, Sheffield, UK
| | - Luis Daverede
- Department of Clinical Oncology, Austral University Hospital, Buenos Aires, Argentina
| | - Catherine Handforth
- Division of Clinical Medicine, University of Sheffield, Weston Park Hospital, Sheffield, UK
| | - Sebastian Trainor
- St James's Institute of Oncology, St James's University Hospital, Leeds, UK
| | - Abdulazeez Salawu
- Academic Unit of Clinical Oncology, University of Sheffield, Sheffield, UK
| | | | - Vicky Goh
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - David Buckley
- Faculty of Medicine and Health, School of Medicine, University of Leeds, Leeds, UK
| | - Jenny Hewison
- Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - Walter Gregory
- Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - Peter Selby
- Leeds Institute of Medical Research, St James's University Hospital, University of Leeds, Leeds, UK
| | - Julia Brown
- Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - Janet Brown
- Division of Clinical Medicine, University of Sheffield, Weston Park Hospital, Sheffield, UK
| |
Collapse
|
6
|
Buchler T, Poprach A. Planned Discontinuation of Tyrosine Kinase Inhibitor Therapy in Metastatic Renal Cell Carcinoma: Lessons for the Era of Immunotherapy. Target Oncol 2024; 19:175-180. [PMID: 38308662 PMCID: PMC10963498 DOI: 10.1007/s11523-023-01031-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2023] [Indexed: 02/05/2024]
Abstract
Several regimens combining immunotherapy and tyrosine kinase inhibitors (TKIs) have recently been validated for the first-line treatment of patients with metastatic renal cell carcinoma (mRCC). While immunotherapy is typically discontinued after 2 years in patients who neither progress nor experience limiting toxicity, according to the protocols of most recent phase III clinical trials, TKIs are to be continued until disease progression or the emergence of limiting toxicity. However, the prolonged use of TKIs is associated with significant toxicity and financial costs. This has sparked considerable debate about whether TKIs can be safely discontinued, particularly in mRCC patients who have achieved a verified complete response. This concise review examines the available evidence on TKI discontinuation in the context of mRCC management.
Collapse
Affiliation(s)
- Tomas Buchler
- Department of Oncology, Second Faculty of Medicine, Charles University and Motol University Hospital, V Uvalu 84, 150 06, Prague, Czech Republic.
- Department of Comprehensive Cancer Care and Faculty of Medicine, Masaryk Memorial Cancer Institute and Masaryk University, Brno, Czech Republic.
| | - Alexandr Poprach
- Department of Oncology, Second Faculty of Medicine, Charles University and Motol University Hospital, V Uvalu 84, 150 06, Prague, Czech Republic
| |
Collapse
|
7
|
Sakata Y, Nomura H, Nakajima H, Kitajima T, Ito Y, Yamamoto Y. Impact of telephone follow-up on hepatocellular carcinoma patients receiving oral chemotherapy from an ambulatory-care setting. J Oncol Pharm Pract 2023:10781552231215427. [PMID: 38043932 DOI: 10.1177/10781552231215427] [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: 12/05/2023]
Abstract
INTRODUCTION In recent years, most molecular target drugs have been administered orally, as prescribed at ambulatory services in hospitals and at patients' homes. Telephone follow-up is increasingly being used in clinical practice for patients needing additional support post-discharge and for the prevention of hospital readmissions. The purpose of this study was to clarify the clinical benefits of telephone follow-up while administering oral anticancer drugs. METHODS This was a single-center, observational, retrospective study. We evaluated hepatocellular carcinoma patients who received sorafenib or lenvatinib between March 2010 and February 2018. The primary endpoint was the incidence of adverse events. RESULTS From the total of 130 patients, 83 patients received telephone follow-up and 47 did not. The incidence of hand-foot skin reactions significantly reduced in patients with telephone follow-up (odds ratio (OR) 3.69, 95% confidence interval (CI) 1.16-11.8, p = 0.020). The median durations (ranges) of adherence to oral chemotherapy were 259 days (15-1730) for the telephone follow-up group and 121 days (14-1105) for the no-telephone follow-up group (p < 0.001). Moreover, the disease control rate was significantly higher in the telephone follow-up group (OR 2.52, 95% CI 1.15-5.53, p = 0.020). CONCLUSIONS Remote interventions, such as telephone follow-up, are useful means of managing adverse events in patients receiving oral anticancer drugs and can lead to improved treatment results.
Collapse
Affiliation(s)
- Yukio Sakata
- Department of Pharmacy, Hakodate Municipal Hospital, Hakodate, Hokkaido, Japan
| | - Hisanaga Nomura
- Department of Pharmacy, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
- Clinical Research Support Office, Seeds Development Promotion Department, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Hirofumi Nakajima
- Department of Pharmacy, Hakodate Municipal Hospital, Hakodate, Hokkaido, Japan
| | - Tomomi Kitajima
- Department of Nursing, Hakodate Municipal Hospital, Hakodate, Hokkaido, Japan
| | - Yukiko Ito
- Department of Nursing, Hakodate Municipal Hospital, Hakodate, Hokkaido, Japan
| | - Yoshiya Yamamoto
- Department of Gastroenterology and Hepatology, Hakodate Municipal Hospital, Hakodate, Hakodate, Japan
| |
Collapse
|
8
|
Trédan O, Toulmonde M, Le Tourneau C, Montane L, Italiano A, Ray-Coquard I, De La Fouchardière C, Bertucci F, Gonçalves A, Gomez-Roca C, You B, Attignon V, Boyault S, Cassier PA, Dufresne A, Tabone-Eglinger S, Viari A, Sohier E, Kamal M, Garin G, Blay JY, Pérol D. Sorafenib in Molecularly Selected Cancer Patients: Final Analysis of the MOST-Plus Sorafenib Cohort. Cancers (Basel) 2023; 15:3441. [PMID: 37444551 DOI: 10.3390/cancers15133441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/20/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND MOST-plus is a multicenter, randomized, open-label, adaptive Phase II trial evaluating the clinical benefit of targeted treatments matched to molecular alteration in advanced/metastatic solid tumors. Sorafenib was tested on patients with tumors harboring sorafenib-targeted genes. METHODS The MOST-plus trial used a randomized discontinuation design. After 12 weeks of sorafenib (400 mg, po BID), patients with progressive disease discontinued study, patients with objective response were proposed to continue sorafenib, whereas patients with stable disease (SD) were randomly assigned (1:1) to the maintenance or interruption of treatment. The primary endpoint was RECIST version 1.1 progression-free rate at 16 weeks after randomization (PFR-16w). Secondary endpoints included progression-free survival (PFS), overall survival (OS), and toxicity. Statistical analyses used a sequential Bayesian approach with interim efficacy analyses. The enrolment could be stopped in the case of a 95% probability for the estimated PFR-16w to be higher in the maintenance than in the interruption arm (NCT02029001). RESULTS 151 patients were included, of whom 35 had SD at 12 weeks of Sorafenib. For the 35 patients with SD on sorafenib, the PFR-16w was 65% [95% credibility interval 43.4-83.7] in the continuation arm and 25% [7.8-48.1] in the interruption arm. Median PFS and OS were improved in the maintenance versus the interruption arm (mPFS: 5.6 [95%CI 1.97-6.77] months versus 2.0 [95%CI 1.61-3.91] months (p = 0.0231) and mOS: 14.3 [95%CI 8.9-23.8] versus 8.0 months [95%CI 3.5-15.2] (p = 0.0857)). CONCLUSION Sorafenib showed activity in progressive patients with solid tumors harboring somatic genomic alterations in sorafenib-targeted genes. Continuing sorafenib when SD is achieved improves PFR compared to interruption.
Collapse
Affiliation(s)
- Olivier Trédan
- Medical Oncology Department, Centre Léon Bérard, 69008 Lyon, France
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France
| | - Maud Toulmonde
- Medical Oncology Department, Institut Bergonié, 33076 Bordeaux, France
| | - Christophe Le Tourneau
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris-Saclay University, 75005 Paris, France
| | - Laure Montane
- Department of Clinical Research and Innovation, Léon Bérard Cancer Center, 69008 Lyon, France
| | - Antoine Italiano
- Medical Oncology Department, Institut Bergonié, 33076 Bordeaux, France
| | | | | | - François Bertucci
- Department of Medical Oncology, Institut Paoli-Calmettes, 13009 Marseille, France
| | - Anthony Gonçalves
- Department of Medical Oncology, Institut Paoli-Calmettes, 13009 Marseille, France
| | - Carlos Gomez-Roca
- Medical Oncology Department, Institute Universitaire de Cancérologie de Toulouse, 31037 Toulouse, France
| | - Benoit You
- Department of Medical Oncology, Lyon Sud Hospital Center, CITOHL, Institute of Cancerology, Hospices Civils de Lyon (IC-HCL), 69495 Lyon, France
| | - Valéry Attignon
- Genomic Platform, Léon Bérard Cancer Center, 69008 Lyon, France
| | | | | | - Armelle Dufresne
- Medical Oncology Department, Centre Léon Bérard, 69008 Lyon, France
| | | | - Alain Viari
- Gilles Thomas Bioinformatic Platform, Léon Bérard Cancer Center, 69008 Lyon, France
| | - Emilie Sohier
- Gilles Thomas Bioinformatic Platform, Léon Bérard Cancer Center, 69008 Lyon, France
| | - Maud Kamal
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris-Saclay University, 75005 Paris, France
| | - Gwenaël Garin
- Department of Clinical Research and Innovation, Léon Bérard Cancer Center, 69008 Lyon, France
| | - Jean-Yves Blay
- Medical Oncology Department, Centre Léon Bérard, 69008 Lyon, France
| | - David Pérol
- Department of Clinical Research and Innovation, Léon Bérard Cancer Center, 69008 Lyon, France
| |
Collapse
|
9
|
Franczyk B, Rysz J, Ławiński J, Ciałkowska-Rysz A, Gluba-Brzózka A. Cardiotoxicity of Selected Vascular Endothelial Growth Factor Receptor Tyrosine Kinase Inhibitors in Patients with Renal Cell Carcinoma. Biomedicines 2023; 11:181. [PMID: 36672689 PMCID: PMC9855533 DOI: 10.3390/biomedicines11010181] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/28/2022] [Accepted: 01/03/2023] [Indexed: 01/13/2023] Open
Abstract
Renal cell carcinoma (RCC) is one of the most frequent malignant neoplasms of the kidney. The therapeutic options available for the treatment of advanced or metastatic RCC include vascular endothelial growth factor receptor (VEGFR)-targeted molecules, for example, tyrosine kinase inhibitors (TKI). Various VEGFR-TKIs proved to be effective in the treatment of patients with solid tumours. The combination of two drugs may prove most beneficial in the treatment of metastatic RCC; however, it also enhances the risk of toxicity compared to monotherapy. Specific VEGFR-TKIs (e.g., sunitinib, sorafenib or pazopanib) may increase the rate of cardiotoxicity in metastatic settings. VEGF inhibitors modulate multiple signalling pathways; thus, the identification of the mechanism underlying cardiotoxicity appears challenging. VEGF signalling is vital for the maintenance of cardiomyocyte homeostasis and cardiac function; therefore, its inhibition can be responsible for the reported adverse effects. Disturbed growth factor signalling pathways may be associated with endothelial dysfunction, impaired revascularization, the development of dilated cardiomyopathy, cardiac hypertrophies and altered peripheral vascular load. Patients at high cardiovascular risk at baseline could benefit from clinical follow-up in the first 2-4 weeks after the introduction of targeted molecular therapy; however, there is no consensus concerning the surveillance strategy.
Collapse
Affiliation(s)
- Beata Franczyk
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 113 Żeromskiego Street, 90-549 Lodz, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 113 Żeromskiego Street, 90-549 Lodz, Poland
| | - Janusz Ławiński
- Department of Urology, Institute of Medical Sciences, Medical College of Rzeszow University, 35-055 Rzeszow, Poland
| | | | - Anna Gluba-Brzózka
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 113 Żeromskiego Street, 90-549 Lodz, Poland
| |
Collapse
|
10
|
Abdallah AE, Mabrouk RR, Al Ward MMS, Eissa SI, Elkaeed EB, Mehany ABM, Abo-Saif MA, El-Feky OA, Alesawy MS, El-Zahabi MA. Synthesis, biological evaluation, and molecular docking of new series of antitumor and apoptosis inducers designed as VEGFR-2 inhibitors. J Enzyme Inhib Med Chem 2022; 37:573-591. [PMID: 35012403 PMCID: PMC8757611 DOI: 10.1080/14756366.2021.2017911] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 11/20/2021] [Accepted: 11/22/2021] [Indexed: 02/07/2023] Open
Abstract
Based on quinazoline, quinoxaline, and nitrobenzene scaffolds and on pharmacophoric features of VEGFR-2 inhibitors, 17 novel compounds were designed and synthesised. VEGFR-2 IC50 values ranged from 60.00 to 123.85 nM for the new derivatives compared to 54.00 nM for sorafenib. Compounds 15a, 15b, and 15d showed IC50 from 17.39 to 47.10 µM against human cancer cell lines; hepatocellular carcinoma (HepG2), prostate cancer (PC3), and breast cancer (MCF-7). Meanwhile, the first in terms of VEGFR-2 inhibition was compound 15d which came second with regard to antitumor assay with IC50 = 24.10, 40.90, and 33.40 µM against aforementioned cell lines, respectively. Furthermore, Compound 15d increased apoptosis rate of HepG2 from 1.20 to 12.46% as it significantly increased levels of Caspase-3, BAX, and P53 from 49.6274, 40.62, and 42.84 to 561.427, 395.04, and 415.027 pg/mL, respectively. Moreover, 15d showed IC50 of 253 and 381 nM against HER2 and FGFR, respectively.
Collapse
Affiliation(s)
- Abdallah E. Abdallah
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Reda R. Mabrouk
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Maged Mohammed Saleh Al Ward
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Sally I. Eissa
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, Riyadh, Saudi Arabia
| | - Eslam B. Elkaeed
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, Riyadh, Saudi Arabia
| | - Ahmed B. M. Mehany
- Zoology Department, Faculty of Science, Al-Azhar University, Cairo, Egypt
| | - Mariam A. Abo-Saif
- Biochemistry Department, Faculty of Pharmacy, Tanta university, Tanta, Egypt
| | - Ola A. El-Feky
- Biochemistry Department, Faculty of Pharmacy, Tanta university, Tanta, Egypt
| | - Mohamed S. Alesawy
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Mohamed Ayman El-Zahabi
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| |
Collapse
|
11
|
Biostatistics of VHL-Gene Transfection in the Health Informatics Analysis of Renal Cell Carcinoma. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:5297580. [PMID: 35035522 PMCID: PMC8759873 DOI: 10.1155/2022/5297580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/26/2021] [Accepted: 09/28/2021] [Indexed: 11/18/2022]
Abstract
Objective In this paper, we study the role of the VHL gene in regulating the proliferation and apoptosis of renal cell carcinoma, as well as the safety and transfection efficiency of ultrasound microbubble gene transfection technology. Method We use kidney cancer cell lines as an in vitro research object and apply ultrasound microbubble gene transfection technology to transfect the VHL gene into kidney cancer cell line (786-0). The proliferation and apoptosis of cells were measured to clarify the inhibitory effect of the VHL gene in renal cell carcinoma. After that, pEGFP-VHL was transfected using ultrasonic microbubble and liposome gene transfection techniques, respectively, and the transfection efficiency was measured by immunofluorescence. Results Compared with untreated and 786-0 cells that are transfected with empty vector, the expression level of VHL gene mRNA in 786-0 cells that are transfected with pcDNA3.1-VHL was significantly increased, and the cell growth inhibition rate was significantly higher. The rate of apoptosis increased significantly. Transfection efficiency of the pEGFP-VHL gene after transfection of 786-0 cells for 48 h: control group 0, liposome group (35.55 ± 2.77) %, ultrasound microbubble group (18.27 ± 2.83) %, and two transfection methods on cells. There is no significant difference in the impact of vitality. Conclusion VHL gene expression can significantly inhibit the proliferation ability of renal cancer cell line 786-0 and promote its apoptosis. VHL gene is a potential target for gene therapy of kidney cancer.
Collapse
|
12
|
Han G, Pusztai L, Hatzis C. Data augmentation based on waterfall plots to increase value of response data generated by small single arm Phase II trials. Contemp Clin Trials 2021; 110:106589. [PMID: 34634476 DOI: 10.1016/j.cct.2021.106589] [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: 05/20/2021] [Revised: 08/30/2021] [Accepted: 09/29/2021] [Indexed: 11/17/2022]
Abstract
Waterfall plots have been increasingly used to visualize tumor response measurements in Phase II clinical trials. Despite the growing popularity of waterfall plots, quantitative summaries and distribution features of the data indicating antitumor activities are typically not reported. Statistical summaries from the raw and augmented data may provide valuable information for understanding such features. This issue has not been discussed adequately in the literature or fully recognized within the oncology community. In this article, we propose to augment the data using a statistical distribution system. Summary statistics of the data set corresponding to waterfall plot can be calculated using the original sample of the tumor changes or the augmentation sample, which may lead to additional insights into the treatment effect. We demonstrate the proposed method in numerical studies and in a Phase II clinical trial investigating the efficacy of a treatment for ovarian carcinoma. We recommend the proposed statistical analyses for making inferences in addition to the waterfall plot visualization.
Collapse
Affiliation(s)
- Gang Han
- Department of Epidemiology and Biostatistics, School of Public Health, Texas A&M University, College Station, TX, United States of America.
| | - Lajos Pusztai
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, United States of America.
| | - Christos Hatzis
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, United States of America.
| |
Collapse
|
13
|
Boissel JP, Pérol D, Décousus H, Klingmann I, Hommel M. Using numerical modeling and simulation to assess the ethical burden in clinical trials and how it relates to the proportion of responders in a trial sample. PLoS One 2021; 16:e0258093. [PMID: 34634062 PMCID: PMC8504716 DOI: 10.1371/journal.pone.0258093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 09/21/2021] [Indexed: 01/24/2023] Open
Abstract
In order to propose a more precise definition and explore how to reduce ethical losses in randomized controlled clinical trials (RCTs), we set out to identify trial participants who do not contribute to demonstrating that the treatment in the experimental arm is superior to that in the control arm. RCTs emerged mid-last century as the gold standard for assessing efficacy, becoming the cornerstone of the value of new therapies, yet their ethical grounds are a matter of debate. We introduce the concept of unnecessary participants in RCTs, the sum of non-informative participants and non-responders. The non-informative participants are considered not informative with respect to the efficacy measured in the trial in contrast to responders who carry all the information required to conclude on the treatment's efficacy. The non-responders present the event whether or not they are treated with the experimental treatment. The unnecessary participants carry the burden of having to participate in a clinical trial without benefiting from it, which might include experiencing side effects. Thus, these unnecessary participants carry the ethical loss that is inherent to the RCT methodology. On the contrary, responders to the experimental treatment bear its entire efficacy in the RCT. Starting from the proportions observed in a real placebo-controlled trial from the literature, we carried out simulations of RCTs progressively increasing the proportion of responders up to 100%. We show that the number of unnecessary participants decreases steadily until the RCT's ethical loss reaches a minimum. In parallel, the trial sample size decreases (presumably its cost as well), although the trial's statistical power increases as shown by the increase of the chi-square comparing the event rates between the two arms. Thus, we expect that increasing the proportion of responders in RCTs would contribute to making them more ethically acceptable, with less false negative outcomes.
Collapse
Affiliation(s)
| | - David Pérol
- Department of Biostatistics, Centre Léon Bérard, Lyon, France
| | - Hervé Décousus
- INSERM, CIC 1408—F Crin, INNOVTE, CHU Saint-Etienne, Hôpital Nord, Service Médecine Vasculaire et Thérapeutique, Saint Etienne, France
| | - Ingrid Klingmann
- European Forum for Good Clinical Practice (EFGCP), Brussels, Belgium
| | - Marc Hommel
- Novadiscovery, Lyon, France
- University Hospital Grenoble, Grenoble, EA 4407 AGEIS UGA, France
| |
Collapse
|
14
|
Ayala-Aguilera CC, Valero T, Lorente-Macías Á, Baillache DJ, Croke S, Unciti-Broceta A. Small Molecule Kinase Inhibitor Drugs (1995-2021): Medical Indication, Pharmacology, and Synthesis. J Med Chem 2021; 65:1047-1131. [PMID: 34624192 DOI: 10.1021/acs.jmedchem.1c00963] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The central role of dysregulated kinase activity in the etiology of progressive disorders, including cancer, has fostered incremental efforts on drug discovery programs over the past 40 years. As a result, kinase inhibitors are today one of the most important classes of drugs. The FDA approved 73 small molecule kinase inhibitor drugs until September 2021, and additional inhibitors were approved by other regulatory agencies during that time. To complement the published literature on clinical kinase inhibitors, we have prepared a review that recaps this large data set into an accessible format for the medicinal chemistry community. Along with the therapeutic and pharmacological properties of each kinase inhibitor approved across the world until 2020, we provide the synthesis routes originally used during the discovery phase, many of which were only available in patent applications. In the last section, we also provide an update on kinase inhibitor drugs approved in 2021.
Collapse
Affiliation(s)
- Cecilia C Ayala-Aguilera
- Cancer Research UK Edinburgh Centre, Institute of Genetics & Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, United Kingdom
| | - Teresa Valero
- Cancer Research UK Edinburgh Centre, Institute of Genetics & Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, United Kingdom
| | - Álvaro Lorente-Macías
- Cancer Research UK Edinburgh Centre, Institute of Genetics & Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, United Kingdom
| | - Daniel J Baillache
- Cancer Research UK Edinburgh Centre, Institute of Genetics & Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, United Kingdom
| | - Stephen Croke
- Cancer Research UK Edinburgh Centre, Institute of Genetics & Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, United Kingdom
| | - Asier Unciti-Broceta
- Cancer Research UK Edinburgh Centre, Institute of Genetics & Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, United Kingdom
| |
Collapse
|
15
|
Moon J, LeBlanc M, Othus M. Accounting for All Patients in Waterfall Plots. JCO Clin Cancer Inform 2021; 5:414-420. [PMID: 33830787 DOI: 10.1200/cci.20.00150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- James Moon
- Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Michael LeBlanc
- Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Megan Othus
- Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| |
Collapse
|
16
|
Abdallah AE, Eissa SI, Al Ward MMS, Mabrouk RR, Mehany ABM, El-Zahabi MA. Design, synthesis and molecular modeling of new quinazolin-4(3H)-one based VEGFR-2 kinase inhibitors for potential anticancer evaluation. Bioorg Chem 2021; 109:104695. [PMID: 33647743 DOI: 10.1016/j.bioorg.2021.104695] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/26/2020] [Accepted: 01/24/2021] [Indexed: 01/03/2023]
Abstract
Globally cancer is the second leading cause of death. So that this work is an attempt to develop new effective anti-cancer agents. In line with pharmacophoric features of VEGFR-2 kinase inhibitors, new nineteen quinazolin-4-one derivatives were designed, synthesized and biologically evaluated for their potential anticancer activity. All target compounds were evaluated in vitro for VEGFR-2 tyrosine kinase inhibition. Then, nine compounds of best results were further investigated by in vitro assay against three human cancer cell lines, namely HepG2, PC3 and MCF. N'-{2-](3-Ethyl-6-nitro-4-oxo-3,4-dihydroquinazoline-2-yl)thio[acetyl}benzohydrazide (36) was found to be the most potent candidate as it showed IC50 = 4.6 ± 0.06 µM against VEGFR-2 kinase. It also exhibited IC50 = 17.23 ± 1.5, 26.10 ± 2.2 and 30.85 ± 2.3 µg/mL against HepG2, PC3 and MCF, respectively. At the same time it showed IC50 = 145.93 ± 1.1 µg/mL against the normal human lung fibroblasts cell line (WI-38), indicating good selectivity index. Further investigation into HepG2 cell cycle showed the ability of compound 36 to induce apoptosis and arrest cell growth at G2/M phase. Moreover, docking studies demonstrated the ability of compound 36 to bind VEGFR-2 in a correct manner making three essential hydrogen bonds with the key residues Glu885, Asp1046 and Cys919. In sum, this work suggests that compound 36 can serve as a lead for development of effective anticancer agents targeting VEGFR-2.
Collapse
Affiliation(s)
- Abdallah E Abdallah
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt.
| | - Sally I Eissa
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11884, Egypt; Faculty of Pharmacy Al-Maareffa University for Science and Technology, Riyadh, Saudi Arabia
| | - Maged Mohammed Saleh Al Ward
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt
| | - Reda R Mabrouk
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt
| | - Ahmed B M Mehany
- Zoology Department, Faculty of Science, Al-Azhar University, Cairo 11884, Egypt.
| | - Mohamed Ayman El-Zahabi
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt.
| |
Collapse
|
17
|
Ahiwale RJ, Chellampillai B, Pawar AP. Investigation of novel sorafenib tosylate loaded biomaterial based nano-cochleates dispersion system for treatment of hepatocellular carcinoma. J DISPER SCI TECHNOL 2021. [DOI: 10.1080/01932691.2021.1878034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Raj J. Ahiwale
- Department of Pharmaceutics, Poona College of Pharmacy, Bharati Vidyapeeth University, Pune, Maharashtra, India
| | - Bothiraja Chellampillai
- Department of Pharmaceutics, Poona College of Pharmacy, Bharati Vidyapeeth University, Pune, Maharashtra, India
| | - Atmaram P. Pawar
- Department of Pharmaceutics, Poona College of Pharmacy, Bharati Vidyapeeth University, Pune, Maharashtra, India
| |
Collapse
|
18
|
Sevryukov FS, Borodavina EV, Isaev РА, Polkin VV, Ilyin АА, Vasilkov SV, Panaseykin YА, Derbugov DN, Ivanov SА, Podvyaznikov SО, Kaprin АD. Side skin reactions of multikinase inhibitors in treatment of thyroid cancer. HEAD AND NECK TUMORS (HNT) 2021; 10:44-49. [DOI: 10.17650/2222-1468-2020-10-4-44-49] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/11/2024]
Abstract
Dermal toxicity of anti-cancer drugs apparent in patients with thyroid cancer. This side effect appears, in particular, in relation to increased administration of targeted anti-cancer treatment, especially tyrosinkinase inhibitors, towards various receptors of growth factors which are applied in the ethiopathogenesis of a tumor cell. Our article focuses on the dermatotoxity, designated also as skin reaction, which most frequently occurs in patients treated by tyrosinkinase inhibitors – sorafenib, vandetonib, cabozantinib and lenvantinib. High prevalence of dermatotoxity, reported by patients, treated with these drugs underscores the need for greater understanding of the pathogenesis and management of this syndrome.
Collapse
Affiliation(s)
- F. S. Sevryukov
- National Medical Research Radiology Center, Ministry of Health of Russia
| | - E. V. Borodavina
- National Medical Research Radiology Center, Ministry of Health of Russia
| | - Р. А. Isaev
- National Medical Research Radiology Center, Ministry of Health of Russia
| | - V. V. Polkin
- National Medical Research Radiology Center, Ministry of Health of Russia
| | - А. А. Ilyin
- National Medical Research Radiology Center, Ministry of Health of Russia
| | - S. V. Vasilkov
- National Medical Research Radiology Center, Ministry of Health of Russia
| | - Yu. А. Panaseykin
- National Medical Research Radiology Center, Ministry of Health of Russia
| | - D. N. Derbugov
- National Medical Research Radiology Center, Ministry of Health of Russia
| | - S. А. Ivanov
- National Medical Research Radiology Center, Ministry of Health of Russia
| | - S. О. Podvyaznikov
- Russian Medical Academy of Continuing Professional Education, Ministry of Health of Russia
| | - А. D. Kaprin
- National Medical Research Radiology Center, Ministry of Health of Russia
| |
Collapse
|
19
|
Abdelalim LR, Elnaggar YSR, Abdallah OY. Oleosomes Encapsulating Sildenafil Citrate as Potential Topical Nanotherapy for Palmar Plantar Erythrodysesthesia with High Ex vivo Permeation and Deposition. AAPS PharmSciTech 2020; 21:310. [PMID: 33164131 DOI: 10.1208/s12249-020-01862-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 10/24/2020] [Indexed: 12/19/2022] Open
Abstract
Palmar plantar erythrodysesthesia (PPE) is a commonly reported skin toxicity of chemotherapeutic agents that significantly affects patients' quality of life. PPE is described as inflammation, swelling, and even cracks and ulcers in the skin of palms and soles of the feet. Conventional treatment includes topical creams, analgesics, or corticosteroids. However, serious cases are not responding to these medications. PPE has been reported to cause drug cessation or dose reduction if not properly treated. Sildenafil citrate (SC) has a well-documented activity in wound healing through improving blood supply to the affected area. However, SC has poor physicochemical properties limiting its transdermal permeation and deposition. This research endeavored to elaborate novel vesicular system with natural components, phospholipids and oleic acid, loaded with sildenafil citrate for topical management of PPE. Sildenafil-loaded oleosomes were prepared using modified ethanol injection method. Optimized oleosomes had nanometric particle size (157.6 nm), negative zeta potential (- 85.2 mv), and high entrapment efficiency (95.56%). Ex vivo studies on human skin revealed that oleosomes displayed 2.3-folds higher permeation and 4.5-folds more deposition through the human skin compared to drug suspension. Results endorsed SC oleosomes as suitable topical treatment of PPE providing ameliorated sildenafil permeability in addition to acting as a reservoir for gradual release of the drug. Graphical abstract.
Collapse
|
20
|
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: 14] [Impact Index Per Article: 3.5] [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
|
21
|
Tumor response criteria for assessing pazopanib first-line therapy in patients with metastatic renal cell carcinoma (mRCC). Abdom Radiol (NY) 2020; 45:1872-1882. [PMID: 31822966 DOI: 10.1007/s00261-019-02354-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
PURPOSE To identify the optimum response criteria for first-line targeted pazopanib therapy in patients with mRCC using solid tumor response criteria and clinical risk factors. METHODS Pre- and post-treatment CTs of patients (n = 43) with mRCC treated with first-line pazopanib therapy were analyzed retrospectively. Treatment response was evaluated with Response Evaluation Criteria in Solid Tumors (RECIST) v1.1, Choi, modified Choi (mChoi), revised Choi (rChoi), MASS (Morphology, Attenuation, Size, and Structure), 10% threshold criteria, and subjective assessment. Memorial Sloan-Kettering Cancer Center and International Metastatic Renal Cell Carcinoma Database Consortium clinical risk factors were also used to define response groups. Response evaluations were correlated with overall survival (OS) and progression-free survival (PFS) using log-rank test. RESULTS Patients with partial response (PR) by mChoi and rChoi had longer OS than those with stable disease (SD) (P < 0.001). Responders by 10% threshold criteria also had better OS, without or combined with clinical criteria. Patients with PR by rChoi, mChoi, RECIST v1.1, MASS criteria, and by subjective assessment had longer PFS than those with SD. rChoi and mChoi criteria best delineated the difference in PFS for patients with PR versus SD, without or combined with clinical criteria. CONCLUSION For mRCC patients treated with pazopanib, OS and PFS for PR and SD groups were best predicted by rChoi and mChoi criteria, without or combined with clinical risk factors. 10% threshold criteria also predicted OS and PFS, whereas RECIST did so only in a limited number of patients.
Collapse
|
22
|
s-HBEGF/SIRT1 circuit-dictated crosstalk between vascular endothelial cells and keratinocytes mediates sorafenib-induced hand-foot skin reaction that can be reversed by nicotinamide. Cell Res 2020; 30:779-793. [PMID: 32296111 PMCID: PMC7608389 DOI: 10.1038/s41422-020-0309-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 03/13/2020] [Indexed: 12/11/2022] Open
Abstract
Hand-foot skin reaction (HFSR), among the most significant adverse effects of sorafenib, has been limiting the clinical benefits of this frontline drug in treating various malignant tumors. The mechanism underlying such toxicity remains poorly understood, hence the absence of effective intervention strategies. In the present study, we show that vascular endothelial cells are the primary cellular target of sorafenib-induced HFSR wherein soluble heparin-binding epidermal growth factor (s-HBEGF) mediates the crosstalk between vascular endothelial cells and keratinocytes. Mechanistically, s-HBEGF released from vascular endothelial cells activates the epidermal growth factor receptor (EGFR) on keratinocytes and promotes the phosphorylation of c-Jun N-terminal kinase 2 (JNK2), which stabilizes sirtuin 1 (SIRT1), an essential keratinization inducer, and ultimately gives rise to HFSR. The administration of s-HBEGF in vivo could sufficiently induce hyper-keratinization without sorafenib treatment. Furthermore, we report that HBEGF neutralization antibody, Sirt1 knockdown, and a classic SIRT1 inhibitor nicotinamide could all significantly reduce the sorafenib-induced HFSR in the mouse model. It is noteworthy that nicotinic acid, a prodrug of nicotinamide, could substantially reverse the sorafenib-induced HFSR in ten patients in a preliminary clinical study. Collectively, our findings reveal the mechanism of vascular endothelial cell-promoted keratinization in keratinocytes and provide a potentially promising therapeutic strategy for the treatment of sorafenib-induced HFSR.
Collapse
|
23
|
Şirin N, Elmas L, Seçme M, Dodurga Y. Investigation of possible effects of apigenin, sorafenib and combined applications on apoptosis and cell cycle in hepatocellular cancer cells. Gene 2020; 737:144428. [PMID: 32045658 DOI: 10.1016/j.gene.2020.144428] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/16/2020] [Accepted: 01/30/2020] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is the most common type of liver tumors. There is only one chemodrug for treatment called sorafenib that is an effective multikinase inhibitor. However, most of the patients gain resistance to sorafenib treatment in six months. Thus, there is a limitation for treatment of HCC. Apigenin is a natural flavonoid that has been used for many years as an antioxidant and anti-inflammatory agent. The aim of this study is to investigate the combined therapeutic effects of sorafenib and apigenin upon apoptosis and cell cycle on HepG2 cell line. Cytotoxic effects of sorafenib and apigenin on HepG2 cells were determined by XTT assay. Effects of single and combined treatment on cell migration, invasion and colony formation were analysed by wound healing, transwell matrigel invasion assay and colony formation assay, respectively. TUNEL assay was performed for analyse apoptosis rates. Expression changes of genes related with apoptosis and cell cycle were analysed by quantitative real-time PCR. Combined treatment of sorafenib and apigenin has more decreasing effects on cell viability than single treatment groups. Also, combination group caused significant increase of apoptotic cells. Migration and invasion capability of cells in combined treatment group are decreased. Lastly, quantitative real-time PCR results showed that combination of both drugs arrested cell cycle and increased apoptotic gene expressions more than single treatment groups. This is the first study that investigating the combined treatment of sorafenib and apigenin on HCC in vitro. By combined treatment, apigenin potentiates sorafenib cytotoxicity on HepG2 cells. Effects of combined treatment on migration, invasion, apoptosis and gene expressions showed that may sorafenib and apigenin have synergistic effect.
Collapse
Affiliation(s)
- Nazlı Şirin
- Department of Medical Biology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Levent Elmas
- Department of Medical Biology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Mücahit Seçme
- Department of Medical Biology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Yavuz Dodurga
- Department of Medical Biology, Faculty of Medicine, Pamukkale University, Denizli, Turkey.
| |
Collapse
|
24
|
Rubin DL, Ugur Akdogan M, Altindag C, Alkim E. ePAD: An Image Annotation and Analysis Platform for Quantitative Imaging. ACTA ACUST UNITED AC 2020; 5:170-183. [PMID: 30854455 PMCID: PMC6403025 DOI: 10.18383/j.tom.2018.00055] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Medical imaging is critical for assessing the response of patients to new cancer therapies. Quantitative lesion assessment on images is time-consuming, and adopting new promising quantitative imaging biomarkers of response in clinical trials is challenging. The electronic Physician Annotation Device (ePAD) is a freely available web-based zero-footprint software application for viewing, annotation, and quantitative analysis of radiology images designed to meet the challenges of quantitative evaluation of cancer lesions. For imaging researchers, ePAD calculates a variety of quantitative imaging biomarkers that they can analyze and compare in ePAD to identify potential candidates as surrogate endpoints in clinical trials. For clinicians, ePAD provides clinical decision support tools for evaluating cancer response through reports summarizing changes in tumor burden based on different imaging biomarkers. As a workflow management and study oversight tool, ePAD lets clinical trial project administrators create worklists for users and oversee the progress of annotations created by research groups. To support interoperability of image annotations, ePAD writes all image annotations and results of quantitative imaging analyses in standardized file formats, and it supports migration of annotations from various propriety formats. ePAD also provides a plugin architecture supporting MATLAB server-side modules in addition to client-side plugins, permitting the community to extend the ePAD platform in various ways for new cancer use cases. We present an overview of ePAD as a platform for medical image annotation and quantitative analysis. We also discuss use cases and collaborations with different groups in the Quantitative Imaging Network and future directions.
Collapse
Affiliation(s)
- Daniel L Rubin
- Department of Biomedical Data Science, Radiology, and Medicine (Biomedical Informatics Research), Stanford University, Stanford, CA
| | - Mete Ugur Akdogan
- Department of Biomedical Data Science, Radiology, and Medicine (Biomedical Informatics Research), Stanford University, Stanford, CA
| | - Cavit Altindag
- Department of Biomedical Data Science, Radiology, and Medicine (Biomedical Informatics Research), Stanford University, Stanford, CA
| | - Emel Alkim
- Department of Biomedical Data Science, Radiology, and Medicine (Biomedical Informatics Research), Stanford University, Stanford, CA
| |
Collapse
|
25
|
Whiting D, Sriprasad S. Molecular biology and targeted therapy in metastatic renal cell carcinoma. JOURNAL OF CLINICAL UROLOGY 2020. [DOI: 10.1177/2051415819849322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The treatment of metastatic renal cell carcinoma is challenging as it has proven to be relatively resistant to conventional oncological treatments. An improved understanding of the molecular biology of renal cell carcinoma has led to the development of a number of targeted therapies in metastatic renal cell carcinoma. This includes vascular endothelial growth factor inhibitors, tyrosine kinase inhibitors, mammalian target of rapamycin inhibitors and most recently immune checkpoint inhibitors. This article will review the mechanisms of development and progression of renal cell carcinoma as well as the mechanisms of current approved treatments in metastatic disease.Level of evidence: Not applicable for this multicentre audit.
Collapse
Affiliation(s)
- D Whiting
- Department of Urology, Darent Valley Hospital, UK
| | - S Sriprasad
- Department of Urology, Darent Valley Hospital, UK
| |
Collapse
|
26
|
Abstract
The urea functionality is inherent to numerous bioactive compounds, including a variety of clinically approved therapies. Urea containing compounds are increasingly used in medicinal chemistry and drug design in order to establish key drug-target interactions and fine-tune crucial drug-like properties. In this perspective, we highlight physicochemical and conformational properties of urea derivatives. We provide outlines of traditional reagents and chemical procedures for the preparation of ureas. Also, we discuss newly developed methodologies mainly aimed at overcoming safety issues associated with traditional synthesis. Finally, we provide a broad overview of urea-based medicinally relevant compounds, ranging from approved drugs to recent medicinal chemistry developments.
Collapse
Affiliation(s)
- Arun K Ghosh
- Department of Chemistry and Department of Medicinal Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Margherita Brindisi
- Department of Chemistry and Department of Medicinal Chemistry, Purdue University, West Lafayette, Indiana 47907, United States.,Department of Excellence of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
27
|
Zhao Y, Liu F, He G, Li K, Zhu C, Yu W, Zhang C, Xie M, Lin J, Zhang J, Jin Y. Discovery of arylamide-5-anilinoquinazoline-8-nitro derivatives as VEGFR-2 kinase inhibitors: Synthesis, in vitro biological evaluation and molecular docking. Bioorg Med Chem Lett 2019; 29:126711. [PMID: 31668972 DOI: 10.1016/j.bmcl.2019.126711] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/23/2019] [Accepted: 09/19/2019] [Indexed: 01/07/2023]
Abstract
Herein, we embarked on a structural optimization campaign aiming at the discovery of novel anticancer agents with our previously reported XL-6f as a lead compound. A library of 23 compounds has been synthesized based on the highly conserved active site of VEGFR-2. Several title compounds exhibited selective inhibitory activities against VEGFR-2, which also displayed selective anti-proliferation potency against HepG2 cell. All synthesized compounds were evaluated for anti-angiogenesis capability. Compound 7o showed the most potent anti-angiogenesis ability, the efficient cytotoxic activities (in vitro against HUVEC and HepG2 cell lines with IC50 values of 0.58 and 0.23 µM, respectively). The molecular docking analysis revealed 7o is a Type-II inhibitor of VEGFR-2 kinase. In general, these results indicated these arylamide-5-anilinoquinazoline-8-nitro derivatives are promising inhibitors of VEGFR-2 for the potential treatment of anti-angiogenesis.
Collapse
Affiliation(s)
- Yongqiang Zhao
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650091, PR China
| | - Feifei Liu
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Guojing He
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650091, PR China
| | - Ke Li
- Biomedical Department, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, PR China.
| | - Changcheng Zhu
- Institute of Drug Research and Development, Kunming Pharmaceutical Corporation, Kunming 650100, PR China
| | - Wei Yu
- Pharmaceutical Department, Kunming General Hospital of Chengdu Military Command, Kunming 650118, PR China
| | - Conghai Zhang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650091, PR China
| | - Mingjin Xie
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650091, PR China
| | - Jun Lin
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650091, PR China.
| | - Jihong Zhang
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming 650500, PR China.
| | - Yi Jin
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650091, PR China.
| |
Collapse
|
28
|
Jones RL, Ratain MJ, O'Dwyer PJ, Siu LL, Jassem J, Medioni J, DeJonge M, Rudin C, Sawyer M, Khayat D, Awada A, de Vos-Geelen JMPGM, Evans TRJ, Obel J, Brockstein B, DeGreve J, Baurain JF, Maki R, D'Adamo D, Dickson M, Undevia S, Geary D, Janisch L, Bedard PL, Abdul Razak AR, Kristeleit R, Vitfell-Rasmussen J, Walters I, Kaye SB, Schwartz G. Phase II randomised discontinuation trial of brivanib in patients with advanced solid tumours. Eur J Cancer 2019; 120:132-139. [PMID: 31522033 PMCID: PMC8852771 DOI: 10.1016/j.ejca.2019.07.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/20/2019] [Accepted: 07/23/2019] [Indexed: 01/29/2023]
Abstract
BACKGROUND Brivanib is a selective inhibitor of vascular endothelial growth factor and fibroblast growth factor (FGF) signalling. We performed a phase II randomised discontinuation trial of brivanib in 7 tumour types (soft-tissue sarcomas [STS], ovarian cancer, breast cancer, pancreatic cancer, non-small-cell lung cancer [NSCLC], gastric/esophageal cancer and transitional cell carcinoma [TCC]). PATIENTS AND METHODS During a 12-week open-label lead-in period, patients received brivanib 800 mg daily and were evaluated for FGF2 status by immunohistochemistry. Patients with stable disease at week 12 were randomised to brivanib or placebo. A study steering committee evaluated week 12 response to determine if enrolment in a tumour type would continue. The primary objective was progression-free survival (PFS) for brivanib versus placebo in patients with FGF2-positive tumours. RESULTS A total of 595 patients were treated, and stable disease was observed at the week 12 randomisation point in all tumour types. Closure decisions were made for breast cancer, pancreatic cancer, NSCLC, gastric cancer and TCC. Criteria for expansion were met for STS and ovarian cancer. In 53 randomised patients with STS and FGF2-positive tumours, the median PFS was 2.8 months for brivanib and 1.4 months for placebo (hazard ratio [HR]: 0.58, p = 0.08). For all randomised patients with sarcomas, the median PFS was 2.8 months (95% confidence interval [CI]: 1.4-4.0) for those treated with brivanib compared with 1.4 months (95% CI: 1.3-1.6) for placebo (HR = 0.64, 95% CI: 0.38-1.07; p = 0.09). In the 36 randomised patients with ovarian cancer and FGF2-positive tumours, the median PFS was 4.0 (95% CI: 2.6-4.2) months for brivanib and 2.0 months (95% CI: 1.2-2.7) for placebo (HR: 0.56, 95% CI: 0.26-1.22). For all randomised patients with ovarian cancer, the median PFS in those randomised to brivanib was 4.0 months (95% CI: 2.6-4.2) and was 2.0 months (95% CI: 1.2-2.7) in those randomised to placebo (HR = 0.54, 95% CI: 0.25-1.17; p = 0.11). CONCLUSION Brivanib demonstrated activity in STS and ovarian cancer with an acceptable safety profile. FGF2 expression, as defined in the protocol, is not a predictive biomarker of the efficacy of brivanib.
Collapse
Affiliation(s)
- Robin L Jones
- Royal Marsden Hospital, Institute of Cancer Research, London, United Kingdom.
| | | | | | | | | | - Jacques Medioni
- Hôpital Européen Georges Pompidou, Paris, France; Paris-Descartes University, Paris, France
| | - Maja DeJonge
- Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | | | | | | | - Judith M P G M de Vos-Geelen
- Department of Internal Medicine, Division of Medical Oncology, GROW - School for Oncology and Developmental Biology, Maastricht UMC+, Maastricht, the Netherlands
| | - T R Jeffry Evans
- Beatson West of Scotland Cancer Centre, University of Glasgow, Glasgow, United Kingdom
| | - Jennifer Obel
- North Shore University Health System, Evanston, IL, USA
| | | | | | | | | | - David D'Adamo
- Eisai Inc, Woodcliff Lake, NJ Previously Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Mark Dickson
- Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | | | | | | | | | | | - Rebecca Kristeleit
- Royal Marsden Hospital, Institute of Cancer Research, London, United Kingdom
| | | | - Ian Walters
- Intensity Therapeutics Inc, Westport, CT Previously BMS, USA
| | - Stan B Kaye
- Royal Marsden Hospital, Institute of Cancer Research, London, United Kingdom
| | | |
Collapse
|
29
|
Luo K, Bao Y, Liu F, Xiao C, Li K, Zhang C, Huang R, Lin J, Zhang J, Jin Y. Synthesis and biological evaluation of novel benzylidene-succinimide derivatives as noncytotoxic antiangiogenic inhibitors with anticolorectal cancer activity in vivo. Eur J Med Chem 2019; 179:805-827. [DOI: 10.1016/j.ejmech.2019.06.094] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 06/05/2019] [Accepted: 06/30/2019] [Indexed: 02/07/2023]
|
30
|
Yalcin S, Lacin S. Impact of tivozanib on patient outcomes in treatment of advanced renal cell carcinoma. Cancer Manag Res 2019; 11:7779-7785. [PMID: 31496820 PMCID: PMC6701608 DOI: 10.2147/cmar.s206105] [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] [Received: 02/20/2019] [Accepted: 06/13/2019] [Indexed: 11/23/2022] Open
Abstract
Renal cell carcinoma (RCC) is the most common type of kidney malignancy, and the clear-cell subtype represents the majority of RCCs. RCC is a heterogeneous disease in terms of genetic and histological features which determine the behavior of the disease. The von Hippel-Lindau (VHL) is a tumor suppressor gene and mutations of this gene are seen in 95% of clear-cell RCCs. Inactivation of VHL causes the accumulation of hypoxia-inducible factor-1 (HIF-1), and in turn, accumulation of HIF-1 induces overexpression of vascular endothelial growth factor (VEGF); the increase in VEGF expression makes RCC a highly vascularized tumor, and forms the rationale for antiVEGF treatment. In the past decade, improvement in the survival of RCC patients has been observed due to new effective therapies, such as antiVEGF and mammalian target of rapamycin (mTOR) targeting agents and immune checkpoint inhibitors. The majority of VEGF targeted agents are not just selective to VEGF receptors, but usually also have inhibitory effects on other kinases, such as c-KIT and FLT3. Tivozanib is an extremely potent and selective tyrosine kinase inhibitor (TKI) of VEGFR-1, 2, and 3, with a relatively long half-life, that is approved by the European Commission for the treatment of advanced/metastatic RCC. Tivozanib, at very low serum concentration can inhibit phosphorylation of VEGFR -1, -2, and -3 tyrosine kinase activity. This article summarizes the clinical data on tivozanib in the treatment of advanced/metastatic RCC.
Collapse
Affiliation(s)
- Suayib Yalcin
- Hacettepe University Institute of Cancer, Department of Medical Oncology, Ankara, Turkey
| | - Sahin Lacin
- University of Health Sciences, Diyarbakir Gazi Yasargil Training and Research Hospital, Department of Medical Oncology, Diyarbakir, Turkey
| |
Collapse
|
31
|
Zi M, Liu F, Wu D, Li K, Zhang D, Zhu C, Zhang Z, Li L, Zhang C, Xie M, Lin J, Zhang J, Jin Y. Discovery of 6-Arylurea-2-arylbenzoxazole and 6-Arylurea-2-arylbenzimidazole Derivatives as Angiogenesis Inhibitors: Design, Synthesis and in vitro Biological Evaluation. ChemMedChem 2019; 14:1291-1302. [PMID: 31131561 DOI: 10.1002/cmdc.201900216] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/22/2019] [Indexed: 12/15/2022]
Abstract
We embarked on a structural optimization campaign aimed at the discovery of novel anti-angiogenesis agents with previously reported imidazole kinase inhibitors as a lead compound. A library of 29 compounds was synthesized. Several title compounds exhibited selective inhibitory activities against vascular endothelial growth factor receptor 2 (VEGFR-2) over epidermal growth factor receptor (EGFR) kinase; these compounds also displayed selective and potent antiproliferative activity against three cancer cell lines. The newly synthesized compounds were evaluated for anti-angiogenesis activity by chick chorioallantoic membrane (CAM) assay. Among them, 1-(2-(2-chlorophenyl)benzo[d]oxazol-5-yl)-3-(4-(trifluoromethoxy)phenyl)urea (compound 5 n) showed the most potent anti-angiogenesis capacity, efficient cytotoxic activities (in vitro against human umbilical vein endothelial cells (HUVEC), H1975, A549, and HeLa cell lines, with respective IC50 values of 8.46, 1.40, 7.61, and 0.28 μm), and an acceptable level of VEGFR-2 kinase inhibition (IC50 =0.25 μm). Molecular docking analysis revealed 5 n to be a type II inhibitor of VEGFR-2 kinase. In general, these results indicate that these 6-arylurea-2-arylbenzoxazole/benzimidazole derivatives are promising inhibitors of VEGFR-2 kinase for potential development into anti-angiogenesis drugs.
Collapse
Affiliation(s)
- Mengli Zi
- Key Laboratory of Medicinal Chemistry for Natural Resources, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, P.R. China
| | - Feifei Liu
- Laboratory of Molecular Genetics of Aging and Tumors, Medical School, Kunming University of Science and Technology, Kunming, 650500, P.R. China
| | - Di Wu
- Key Laboratory of Medicinal Chemistry for Natural Resources, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, P.R. China
| | - Ke Li
- Biomedical Department, Yunnan Cancer Hospital, Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, P.R. China
| | - Da Zhang
- Key Laboratory of Medicinal Chemistry for Natural Resources, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, P.R. China
| | - Changcheng Zhu
- Institute of Drug Research and Development, Kunming Pharmaceutical Corporation, Kunming, 650100, P.R. China
| | - Zhiyun Zhang
- Department of Anorectal, Kunming Municipal Hospital of Traditional Chinese Medicine, Kunming, 650011, P.R. China
| | - Linghua Li
- Department of Anorectal, Kunming Municipal Hospital of Traditional Chinese Medicine, Kunming, 650011, P.R. China
| | - Conghai Zhang
- Key Laboratory of Medicinal Chemistry for Natural Resources, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, P.R. China
| | - Mingjin Xie
- Key Laboratory of Medicinal Chemistry for Natural Resources, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, P.R. China
| | - Jun Lin
- Key Laboratory of Medicinal Chemistry for Natural Resources, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, P.R. China
| | - Jihong Zhang
- Laboratory of Molecular Genetics of Aging and Tumors, Medical School, Kunming University of Science and Technology, Kunming, 650500, P.R. China
| | - Yi Jin
- Key Laboratory of Medicinal Chemistry for Natural Resources, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, P.R. China
| |
Collapse
|
32
|
Niinivirta M, Enblad G, Lindskog C, Pontén F, Dragomir A, Ullenhag GJ. Tumoral Pyruvate Kinase L/R as a Predictive Marker for the Treatment of Renal Cancer Patients with Sunitinib and Sorafenib. J Cancer 2019; 10:3224-3231. [PMID: 31289593 PMCID: PMC6603390 DOI: 10.7150/jca.30130] [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: 09/22/2018] [Accepted: 04/26/2019] [Indexed: 01/04/2023] Open
Abstract
Background and aims: Treatment with tyrosine kinase inhibitors (TKI) like sunitinib and sorafenib has improved the prognosis of patients with metastatic renal cell cancer (mRCC). No predictive marker is available to select patients who will gain from these treatments. Tumoral pyruvate kinase L/R (PKLR) is a membrane protein with highly specific expression in the renal tubule. We have previously shown that the tumoral expression of cubilin (CUBN) is associated with progression free survival (PFS) in mRCC patients treated with sunitinib and sorafenib. The aim of the present study was to investigate if PKLR can predict response in these patients, alone and/or in combination with CUBN. Methods: A tissue microarray (TMA) was constructed of tumor samples from 139 mRCC patients. One hundred and thirty-six of these patients had been treated with sunitinib or sorafenib in the first or second-line setting. Thirty patients suffered from early severe toxicity leading to the termination of treatment. The remaining patients (n=106) were selected for the current study. Results: Fifty-five (52%) of the tumors expressed membranous PKLR. Patients with PKLR tumor expression experienced a significantly longer PFS compared to patients with no expression (eight versus five months, p = 0.019). Overall survival (OS) was also significantly better for patients with PKLR expression. In addition, the combined expression of PKLR and CUBN resulted in a higher predictive value than either marker alone. Conclusions: In this real world study we show that tumoral PKLR membrane expression is a positive predictive biomarker for sunitinib and sorafenib treatment in patients suffering from mRCC. Our results also indicate that the combined expression with cubilin more accurately than PKLR alone can select patients with no benefit from treatment.
Collapse
Affiliation(s)
- Marjut Niinivirta
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Department of Oncology, Uppsala University Hospital, Entrance 78, 751 85 Uppsala, Sweden
| | - Gunilla Enblad
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Department of Oncology, Uppsala University Hospital, Entrance 78, 751 85 Uppsala, Sweden
| | - Cecilia Lindskog
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden
| | - Fredrik Pontén
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden
| | - Anca Dragomir
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden.,Department of Surgical Pathology, Uppsala University Hospital, 75185 Uppsala, Sweden
| | - Gustav J Ullenhag
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Department of Oncology, Uppsala University Hospital, Entrance 78, 751 85 Uppsala, Sweden
| |
Collapse
|
33
|
Mikami S, Mizuno R, Kondo T, Shinohara N, Nonomura N, Ozono S, Eto M, Tatsugami K, Takayama T, Matsuyama H, Kishida T, Oya M. Clinical significance of programmed death-1 and programmed death-ligand 1 expression in the tumor microenvironment of clear cell renal cell carcinoma. Cancer Sci 2019; 110:1820-1828. [PMID: 30972888 PMCID: PMC6550131 DOI: 10.1111/cas.14019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 03/24/2019] [Accepted: 03/27/2019] [Indexed: 12/31/2022] Open
Abstract
Recently, immunotherapy based on blocking immune checkpoints with programmed death-1 (PD-1) or PD-ligand 1 (PD-L1) Abs has been introduced for the treatment of advanced clear cell renal cell carcinoma (ccRCC), especially tumors resistant to vascular endothelial growth factor-tyrosine kinase inhibitors (VEGF-TKIs), but the significance of their expression in the tumor microenvironment is unclear. We investigated these immune checkpoint markers in tumor cells and tumor-infiltrating immune cells (TIIC) in the tumor microenvironment of 100 untreated and 25 VEGF-TKI-treated primary ccRCC tissues. Upregulated expression of PD-1 and PD-L1 by TIIC, and PD-L1 by tumor cells was associated with the histological grade and unfavorable prognosis of RCC patients. High PD-1 and PD-L1 expression by TIIC was associated with a poorer response to VEGF-TKI, whereas PD-L1 expression by tumor cells did not affect the efficacy of the treatment. Furthermore, increased PD-1-positive TIIC and PD-L1-positive TIIC were observed in tumors treated with VEGF-TKIs compared with those in untreated tumors. Our data suggest that PD-1 and PD-L1 expression by TIIC in the tumor microenvironment is involved in treatment resistance, and that sequential therapy with immune checkpoint inhibitors could be a promising therapeutic strategy for ccRCC resistant to VEGF-TKI treatment.
Collapse
Grants
- 15H04977 Ministry of Education, Culture, Sports, Science and Technology
- 16K08657 Ministry of Education, Culture, Sports, Science and Technology
- 17K11159 Ministry of Education, Culture, Sports, Science and Technology
- 19K07468 Ministry of Education, Culture, Sports, Science and Technology
Collapse
Affiliation(s)
- Shuji Mikami
- Department of Diagnostic PathologyKeio University School of MedicineTokyoJapan
| | - Ryuichi Mizuno
- Department of UrologyKeio University School of MedicineTokyoJapan
| | - Tsunenori Kondo
- Department of UrologyTokyo Women's Medical University, Medical Center EastTokyoJapan
| | - Nobuo Shinohara
- Department of Genitourinary SurgeryHokkaido University Graduate School of MedicineSapporoJapan
| | - Norio Nonomura
- Department of UrologyOsaka University School of MedicineOsakaJapan
| | - Seiichiro Ozono
- Department of UrologyHamamatsu University School of MedicineHamamatsuJapan
| | - Masatoshi Eto
- Department of UrologyGraduate School of Medical ScienceKyushu UniversityFukuokaJapan
- Department of UrologyFaculty of Life SciencesKumamoto UniversityKumamotoJapan
| | - Katsunori Tatsugami
- Department of UrologyGraduate School of Medical ScienceKyushu UniversityFukuokaJapan
| | | | - Hideyasu Matsuyama
- Department of UrologyGraduate School of MedicineYamaguchi UniversityUbeJapan
| | | | - Mototsugu Oya
- Department of UrologyKeio University School of MedicineTokyoJapan
| | | |
Collapse
|
34
|
D'Adamo DR, Dickson MA, Keohan ML, Carvajal RD, Hensley ML, Hirst CM, Ezeoke MO, Ahn L, Qin LX, Antonescu CR, Lefkowitz RA, Maki RG, Schwartz GK, Tap WD. A Phase II Trial of Sorafenib and Dacarbazine for Leiomyosarcoma, Synovial Sarcoma, and Malignant Peripheral Nerve Sheath Tumors. Oncologist 2019; 24:857-863. [PMID: 30126857 PMCID: PMC6656505 DOI: 10.1634/theoncologist.2018-0160] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 05/31/2018] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Sorafenib and dacarbazine have low single-agent response rates in metastatic sarcomas. As angiogenesis inhibitors can enhance the efficacy of chemotherapy, we investigated the combination of sorafenib and dacarbazine in select sarcoma subtypes. MATERIALS AND METHODS Patients with leiomyosarcoma (LMS), synovial sarcoma (SS), or malignant peripheral nerve sheath tumors (MPNST) with up to two previous lines of therapy and adequate hepatic, renal, and marrow function received 3-week cycles of sorafenib at 400 mg oral twice daily and dacarbazine 1,000 mg/m2 intravenously (later reduced to 850 mg/m2). Patients were evaluated for response every 6 weeks. The primary objective was to determine the disease control rate (DCR) of sorafenib plus dacarbazine in the selected sarcoma subtypes. RESULTS The study included 37 patients (19 female); median age was 55 years (range 26-87); and histologies included LMS (22), SS (11), and MPNST (4). The DCR was 46% (17/37). Median progression-free survival was 13.4 weeks. The RECIST response rate was 14% (5/37). The Choi response rate was 51% (19/37). Median overall survival was 13.2 months. Of the first 25 patients, 15 (60%) required dacarbazine dose reductions for hematologic toxicity, with one episode of grade 5 neutropenic fever. After reducing the starting dose of dacarbazine to 850 mg/m2, only 3 of the final 12 (25%) patients required dose reduction. CONCLUSION This phase II study met its primary endpoint with an 18-week DCR of 46%. The clinical activity of dacarbazine plus sorafenib in patients with these diagnoses is modest. IMPLICATIONS FOR PRACTICE Metastatic soft tissue sarcomas are a heterogeneous group of relatively rare malignancies. Most patients are treated with cytotoxic chemotherapy or targeted therapy in the form of tyrosine kinase inhibitors. Response rates are relatively low, and there is a need for better therapies. This clinical trial demonstrates that combining a cytotoxic therapy (dacarbazine) with an antiangiogenic small molecule (sorafenib) is feasible and associated with favorable disease-control rates; however, it also increases the potential for significant toxicity.
Collapse
Affiliation(s)
| | - Mark A Dickson
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York, USA
| | - Mary L Keohan
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York, USA
| | | | - Martee L Hensley
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York, USA
| | - Catherine M Hirst
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York, USA
| | - Marietta O Ezeoke
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York, USA
| | - Linda Ahn
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York, USA
| | - Li-Xuan Qin
- Department of Biostatistics, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York, USA
| | - Cristina R Antonescu
- Department of Pathology, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York, USA
| | - Robert A Lefkowitz
- Department of Radiology, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York, USA
| | | | - Gary K Schwartz
- Department of Medicine, Columbia University, New York, New York, USA
| | - William D Tap
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
35
|
Ornstein MC, Wood LS, Hobbs BP, Allman KD, Martin A, Bevan M, Gilligan TD, Garcia JA, Rini BI. A phase II trial of intermittent nivolumab in patients with metastatic renal cell carcinoma (mRCC) who have received prior anti-angiogenic therapy. J Immunother Cancer 2019; 7:127. [PMID: 31097024 PMCID: PMC6524207 DOI: 10.1186/s40425-019-0615-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 05/08/2019] [Indexed: 12/28/2022] Open
Abstract
Background Nivolumab is approved for mRCC patients who have received prior anti-angiogenic therapy but the duration of therapy required for sustained clinical benefit is unknown. A phase II clinical trial to investigate the feasibility of intermittent nivolumab dosing was conducted. Methods Patients ≥18 years of age with mRCC who were previously treated with at least one antiangiogenic therapy were eligible. Patients were treated with nivolumab for twelve weeks. Patients who had RECIST PD were removed from the trial. Patients who did not initially achieve ≥10% reduction in tumor burden (TB) continued nivolumab per standard of care. Patients with ≥10% TB reduction entered a treatment-free observation phase with re-imaging every 12 weeks. Nivolumab was restarted in patients with a ≥ 10% TB increase and again held with TB reduction ≥10%. This intermittent nivolumab dosing continued until RECIST PD while on nivolumab. The primary objective was feasibility of intermittent nivolumab, defined as the proportion of patients eligible for intermittent therapy who elect to receive intermittent nivolumab. Intermittent nivolumab would be considered “feasible” if the acceptance rate was ≥80%. Forty patients provides > 95% power with 0.05 type I error, assuming a null acceptance rate of 50%. With the approval of the combination of ipilimumab/nivolumab (April 2018) in front-line mRCC, this cohort was closed prior to completed pre-planned approval. Results Of the 14 patients enrolled, 13 (93%) were male with a median age 65. All had a prior nephrectomy and 12 (86%) were intermediate-risk by IMDC criteria. Five patients (36%) met the criteria for the intermittent phase of the trial (median TB decrease 46%) and all agreed to intermittent therapy. With a median follow-up of 48 weeks, only one patient restarted therapy. The four remaining patients have a sustained response for a median of 34 weeks (range, 16–53) off therapy. No patients developed RECIST PD while off therapy. Conclusions This prospective experience of intermittent nivolumab dosing in mRCC supports further investigation of intermittent immunotherapy dosing strategies in RCC. Trial registration NCT03126331 (Intermittent Nivolumab in Metastatic Renal Cell Carcinoma Patients; Date of registration 4/27/2017; https://clinicaltrials.gov/ct2/show/NCT03126331).
Collapse
Affiliation(s)
- Moshe C Ornstein
- Department of Hematology and Medical Oncology, Cleveland Clinic Taussig Cancer Institute, 9500 Euclid Avenue, CA-60, Cleveland, OH, 44195, USA.
| | - Laura S Wood
- Department of Hematology and Medical Oncology, Cleveland Clinic Taussig Cancer Institute, 9500 Euclid Avenue, CA-60, Cleveland, OH, 44195, USA
| | - Brian P Hobbs
- Quantitative Health Sciences and Taussig Cancer Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Kimberly D Allman
- Department of Hematology and Medical Oncology, Cleveland Clinic Taussig Cancer Institute, 9500 Euclid Avenue, CA-60, Cleveland, OH, 44195, USA
| | - Allison Martin
- Department of Hematology and Medical Oncology, Cleveland Clinic Taussig Cancer Institute, 9500 Euclid Avenue, CA-60, Cleveland, OH, 44195, USA
| | - Michael Bevan
- Department of Hematology and Medical Oncology, Cleveland Clinic Taussig Cancer Institute, 9500 Euclid Avenue, CA-60, Cleveland, OH, 44195, USA
| | - Timothy D Gilligan
- Department of Hematology and Medical Oncology, Cleveland Clinic Taussig Cancer Institute, 9500 Euclid Avenue, CA-60, Cleveland, OH, 44195, USA
| | - Jorge A Garcia
- Department of Hematology and Medical Oncology, Cleveland Clinic Taussig Cancer Institute, 9500 Euclid Avenue, CA-60, Cleveland, OH, 44195, USA
| | - Brian I Rini
- Department of Hematology and Medical Oncology, Cleveland Clinic Taussig Cancer Institute, 9500 Euclid Avenue, CA-60, Cleveland, OH, 44195, USA
| |
Collapse
|
36
|
Wang L, Chen S, Zhang J, Mao S, Mao W, Zhang W, Guo Y, Wu Y, Wang R, Yan Y, Yao X. Suppressed OGT expression inhibits cell proliferation and modulates EGFR expression in renal cell carcinoma. Cancer Manag Res 2019; 11:2215-2223. [PMID: 30962710 PMCID: PMC6433112 DOI: 10.2147/cmar.s190642] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Purpose O-linked N-acetylglucosamine (O-GlcNAc or O-GlcNAcylation) is a post-translational modification, which plays a vital role in the progression of various cancers. The purpose of the present study was to assess O-GlcNAcylation in human renal cell carcinoma (RCC). Methods O-GlcNAcylation levels and O-GlcNAc-transferase (OGT) expression in human RCC cell lines and 10 paired clinical tissues were detected by Western blot and Immunohistochemis-try. Then, the effects of O-GlcNAcylation on RCC cell proliferation in vitro were investigated by Cell Counting Kit-8 assay. A xenograft assay was performed to assess the in vivo effects of OGT knockdown in RCC cells. Cell apoptosis and cell cycle assays were performed by flow cytometry. Co-immunoprecipitation assays were used to assess epidermal growth factor receptor (EGFR) O-GlcNAcylation and the interaction between OGT and EGFR. Results O-GlcNAcylation levels and OGT expression were increased in RCC, and the high amounts correlated with poor patient prognosis. OGT knockdown significantly suppressed RCC cell proliferation in vitro and in vivo. Notably, EGFR was modulated by O-GlcNAcylation and directly interacted with OGT. Conclusion These findings provide novel insights into the oncogenic roles of O-GlcNAcylation and OGT in the development of RCC, indicating that OGT might be used as a target for RCC therapy in the future.
Collapse
Affiliation(s)
- Longsheng Wang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai 200072, China,
| | - Shaojun Chen
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 200080, China
| | - Junfeng Zhang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai 200072, China,
| | - Shiyu Mao
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai 200072, China,
| | - Weipu Mao
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai 200072, China,
| | - Wentao Zhang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai 200072, China,
| | - Yadong Guo
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai 200072, China,
| | - Yuan Wu
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai 200072, China,
| | - Ruiliang Wang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai 200072, China,
| | - Yang Yan
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai 200072, China,
| | - Xudong Yao
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai 200072, China,
| |
Collapse
|
37
|
Shirotake S, Kondo H, Okabe T, Makino S, Araki R, Komatsuda A, Kaneko G, Nishimoto K, Oyama M. Early tumor shrinkage as a predictive factor of metastatic renal cell carcinoma in molecular targeted therapy: A single institutional study. Mol Clin Oncol 2019; 10:125-131. [PMID: 30655987 DOI: 10.3892/mco.2018.1762] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 10/11/2018] [Indexed: 01/05/2023] Open
Abstract
The aim of the present study was to investigate the impact of metastatic sites and early tumor shrinkage (eTS) as prognostic predictive factors of metastatic renal cell carcinoma (mRCC) in molecular targeted therapy. A total of 209 advanced RCC cases treated with sorafenib, sunitinib, axitinib, pazopanib, temsirolimus and everolimus from our single institution were included in the present study. Several known prognostic predictive factors, including metastatic sites and the rate of eTS, were analyzed by Kaplan-Meier survival estimate analysis followed by Cox's proportional hazards model analysis. eTS was measured by three independent physicians. Four metastatic sites in the liver, bone, lymph nodes and brain as well as greater eTS were identified as potential independent predictors of overall survival (OS) in several cohorts: i) Metastatic RCC (n=194); ii) metastatic clear cell RCC (n=119); and iii) mRCC patients with eTS data (n=127). In sub-analyses of patients treated with each 1st line tyrosine kinase inhibitor, eTS was identified as a potentially potent predictor of OS in patients treated with axitinib. The liver, bone, lymph nodes, brain metastases and eTS were identified as independent predictive factors of OS by analyzing a limited Japanese cohort.
Collapse
Affiliation(s)
- Suguru Shirotake
- Department of Uro-Oncology, Saitama Medical University International Medical Center, Hidaka, Saitama 350-1298, Japan
| | - Hideyuki Kondo
- Department of Uro-Oncology, Saitama Medical University International Medical Center, Hidaka, Saitama 350-1298, Japan
| | - Takashi Okabe
- Department of Uro-Oncology, Saitama Medical University International Medical Center, Hidaka, Saitama 350-1298, Japan
| | - Soichi Makino
- Department of Radiation Oncology, Saitama Medical University International Medical Center, Hidaka, Saitama 350-1298, Japan
| | - Ryuichiro Araki
- Saitama Medical University, Community Health Science Center, Moroyama, Saitama 350-0495, Japan
| | - Akari Komatsuda
- Department of Uro-Oncology, Saitama Medical University International Medical Center, Hidaka, Saitama 350-1298, Japan
| | - Go Kaneko
- Department of Uro-Oncology, Saitama Medical University International Medical Center, Hidaka, Saitama 350-1298, Japan
| | - Koshiro Nishimoto
- Department of Uro-Oncology, Saitama Medical University International Medical Center, Hidaka, Saitama 350-1298, Japan
| | - Masafumi Oyama
- Department of Uro-Oncology, Saitama Medical University International Medical Center, Hidaka, Saitama 350-1298, Japan
| |
Collapse
|
38
|
Bains P, Sharma S. Sorafenib-Induced Cutaneous Toxicities: Four Different Presentations. INDIAN JOURNAL OF DRUGS IN DERMATOLOGY 2019. [DOI: 10.4103/ijdd.ijdd_36_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
39
|
Wang HT, Xia M. A meta-analysis of efficacy and safety of sorafenib versus other targeted agents for metastatic renal cell carcinoma. Medicine (Baltimore) 2019; 98:e13779. [PMID: 30608388 PMCID: PMC6344165 DOI: 10.1097/md.0000000000013779] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Molecular targeted therapies were found to be efficacious and safer in the treatment of metastatic renal cell carcinoma (mRCC). Sorafenib is the first target agent (TA) to report a benefit in this disease and has largely established a prominent role in progression-free survival (PFS). However, there have been conflicting results across the trials that evaluated the efficacy of sorafenib. OBJECTIVE The aim of the study was to perform a meta-analysis to compare the efficacy and safety of sorafenib in first-line treatments of mRCC. METHODS We searched online electronic databases: PubMed, Embase, and the Cochrane Library updated on September 2017. Trials on the efficacy of sorafenib in first-line treatments of advanced RCC were included, of which the primary outcomes were objective response rate (ORR), PFS, overall survival (OS), and grade 3/4 adverse events (AEs). RESULTS A total of 5 trials were included in this analysis. The group of AEs showed significantly improved PFS (odds ratio [OR] = 0.78, 95% confidence interval [CI] = 0.70-0.86, P < .001), as well with the ORR (OR = 1.89, 95%CI = 1.38-2.59, P < .0001) compared with sorafenib. However, there was no significant difference in OS (OR = 0.97, 95%CI = 0.78-1.22, P = .82). CONCLUSION Sorafenib did not achieve efficacy and safety benefit in patients with mRCC compared with those treated with TAs. The role of sorafenib in first-line treatments of mRCC may change in favor of newer drugs. More research is needed to confirm whether these new TAs could replace sorafenib as the gold standard in the future.
Collapse
|
40
|
Crona DJ, Skol AD, Leppänen VM, Glubb DM, Etheridge AS, Hilliard E, Peña CE, Peterson YK, Klauber-DeMore N, Alitalo KK, Innocenti F. Genetic Variants of VEGFA and FLT4 Are Determinants of Survival in Renal Cell Carcinoma Patients Treated with Sorafenib. Cancer Res 2019; 79:231-241. [PMID: 30385613 PMCID: PMC6541205 DOI: 10.1158/0008-5472.can-18-1089] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 09/03/2018] [Accepted: 10/29/2018] [Indexed: 12/30/2022]
Abstract
Molecular markers of sorafenib efficacy in patients with metastatic renal cell carcinoma (mRCC) are not available. The purpose of this study was to discover genetic markers of survival in patients with mRCC treated with sorafenib. Germline variants from 56 genes were genotyped in 295 patients with mRCC. Variant-overall survival (OS) associations were tested in multivariate regression models. Mechanistic studies were conducted to validate clinical associations. VEGFA rs1885657, ITGAV rs3816375, and WWOX rs8047917 (sorafenib arm), and FLT4 rs307826 and VEGFA rs3024987 (sorafenib and placebo arms combined) were associated with shorter OS. FLT4 rs307826 increased VEGFR-3 phosphorylation, membrane trafficking, and receptor activation. VEGFA rs1885657 and rs58159269 increased transcriptional activity of the constructs containing these variants in endothelial and RCC cell lines, and VEGFA rs58159269 increased endothelial cell proliferation and tube formation. FLT4 rs307826 and VEGFA rs58159269 led to reduced sorafenib cytotoxicity. Genetic variation in VEGFA and FLT4 could affect survival in sorafenib-treated patients with mRCC. These markers should be examined in additional malignancies treated with sorafenib and in other angiogenesis inhibitors used in mRCC. SIGNIFICANCE: Clinical and mechanistic data identify germline genetic variants in VEGFA and FLT4 as markers of survival in patients with metastatic renal cell carcinoma.
Collapse
Affiliation(s)
- Daniel J Crona
- Division of Pharmacotherapy and Experimental Therapeutics, Center for Pharmacogenomics and Individualized Therapy, The University of North Carolina Eshelman School of Pharmacy, Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Andrew D Skol
- The University of Chicago, Department of Medicine, Chicago, Illinois
| | | | - Dylan M Glubb
- Division of Pharmacotherapy and Experimental Therapeutics, Center for Pharmacogenomics and Individualized Therapy, The University of North Carolina Eshelman School of Pharmacy, Chapel Hill, North Carolina
- The Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Amy S Etheridge
- Division of Pharmacotherapy and Experimental Therapeutics, Center for Pharmacogenomics and Individualized Therapy, The University of North Carolina Eshelman School of Pharmacy, Chapel Hill, North Carolina
| | - Eleanor Hilliard
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Carol E Peña
- Bayer HealthCare Pharmaceuticals, Montville, New Jersey
| | - Yuri K Peterson
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Nancy Klauber-DeMore
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Kari K Alitalo
- Wihuri Research Institute and University of Helsinki, Helsinki, Finland
| | - Federico Innocenti
- Division of Pharmacotherapy and Experimental Therapeutics, Center for Pharmacogenomics and Individualized Therapy, The University of North Carolina Eshelman School of Pharmacy, Chapel Hill, North Carolina.
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
41
|
Escudier B, Worden F, Kudo M. Sorafenib: key lessons from over 10 years of experience. Expert Rev Anticancer Ther 2018; 19:177-189. [PMID: 30575405 DOI: 10.1080/14737140.2019.1559058] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Introduction: In 2005, sorafenib was the first targeted therapy approved for advanced renal cell carcinoma (RCC), transforming treatment. In hepatocellular carcinoma (HCC), for more than a decade, sorafenib remained the only approved systemic therapy to have demonstrated a survival benefit in first-line unresectable HCC. In 2013, sorafenib was the first targeted agent approved for patients with differentiated thyroid cancer (DTC) refractory to radioactive iodine treatment. Areas covered: This review discusses the development, advances, and challenges associated with sorafenib use in RCC, HCC, and DTC over the past decade. A search was performed on PubMed and key congresses as required, with no time limits. Expert commentary: Sorafenib has had a lasting impact on the therapeutic landscape of RCC, HCC, and DTC, and remains an important treatment option despite a rapidly evolving treatment landscape. Extensive clinical and real-world experience has been invaluable in improving patient management and maximizing benefit from treatment. Ongoing clinical trials continue to evaluate sorafenib in different settings, and in combination with other therapies in HCC and DTC. We have no doubt that sorafenib will continue to be an important treatment option in the coming years.
Collapse
Affiliation(s)
- Bernard Escudier
- a Department of Medical Oncology , Institut Gustave Roussy , Paris , France
| | - Francis Worden
- b Department of Internal Medicine, Division of Hematology and Oncology, Comprehensive Cancer Center , University of Michigan , Ann Arbor , MI , USA
| | - Masatoshi Kudo
- c Department of Gastroenterology and Hepatology , Kindai University Faculty of Medicine , Osaka , Japan
| |
Collapse
|
42
|
Hellerstedt BA, Vogelzang NJ, Kluger HM, Yasenchak CA, Aftab DT, Ramies DA, Gordon MS, Lara P. Results of a Phase II Placebo-controlled Randomized Discontinuation Trial of Cabozantinib in Patients with Non-small-cell Lung Carcinoma. Clin Lung Cancer 2018; 20:74-81.e1. [PMID: 30528315 DOI: 10.1016/j.cllc.2018.10.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 10/01/2018] [Accepted: 10/13/2018] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Cabozantinib, an orally bioavailable tyrosine kinase inhibitor with activity against MET, vascular endothelial growth factor receptor 2, AXL, ROS1, and RET was assessed in patients with non-small-cell lung carcinoma (NSCLC) as part of a phase II randomized discontinuation trial with cohorts from 9 tumor types. PATIENTS AND METHODS Patients received cabozantinib 100 mg/day during a 12-week open-label lead-in stage. Those with stable disease per Response Evaluation Criteria in Solid Tumors version 1.0 at week 12 were randomized to cabozantinib or placebo. Primary endpoints were objective response rate (ORR) at week 12 and progression-free survival (PFS) after randomization. RESULTS Sixty patients with NSCLC who had received a median of 2 prior lines of therapy were enrolled. ORR at week 12 was 10%; 6 patients had a confirmed partial response, and no patients had a complete response. Overall disease-control rate (ORR + stable disease) at week 12 was 38%. Tumor regression was observed in 30 (64%) of 47 patients with post-baseline radiographic tumor assessments, including 3 or 4 patients with KRAS or epidermal growth factor receptor mutations, respectively. Median PFS after randomization was 2.4 months for both the cabozantinib and placebo arms. Median PFS from first dose for the entire cohort was 4.2 months. The most common grade 3/4 adverse events were fatigue (13%), palmar-plantar erythrodysesthesia (10%), diarrhea (7%), hypertension (7%), and asthenia (5%); 1 treatment-related grade 5 adverse event (hemorrhage) was reported during the lead-in stage. CONCLUSION Cabozantinib exhibited clinical activity based on ORR and regression of tumor lesions in pretreated patients with NSCLC, including in patients with KRAS mutations.
Collapse
Affiliation(s)
- Beth A Hellerstedt
- US Oncology Research, LLC, McKesson Specialty Health, The Woodlands, TX; Texas Oncology, Central Austin Cancer Center, Austin, TX.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Imaoka H, Sasaki M, Takahashi H, Hashimoto Y, Ohno I, Mitsunaga S, Watanabe K, Umemoto K, Kimura G, Suzuki Y, Kan M, Ikeda M. Alternate Endpoints for Phase II Trials in Advanced Neuroendocrine Tumors. Oncologist 2018; 24:47-53. [PMID: 30072388 DOI: 10.1634/theoncologist.2017-0651] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 05/07/2018] [Accepted: 05/16/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND In phase II trials for neuroendocrine tumors (NETs), the objective response rate (ORR) is traditionally used as a primary endpoint. However, the validity of the ORR as a primary endpoint has never been systematically examined. Therefore, a literature-based analysis of phase II trials for NETs was performed to identify valid alternative endpoints for predicting median progression-free survival (PFS) in clinical trials for NETs. MATERIALS AND METHODS Phase II trials of medical treatment for advanced NETs were identified based on a systematic search using MEDLINE, EMBASE, and the Cochrane Central Register of Controlled Trials. RESULTS A total of 22 trials were identified, and 1,310 patients and 27 treatment arms were included in the analysis. There was no significant relationship between the ORR and median PFS (r = .374; 95% confidence interval [CI], -0.051 to 0.800; p = .085). Conversely, 12-month PFS rates showed very strong correlations with median PFS (r = .929; 95% CI, 0.831-1.027; p < .001). CONCLUSION The results of the present analysis indicate that the ORR is not significantly correlated with median PFS and suggest that 12-month PFS rates are good alternate endpoints for screening phase II trials for NETs. IMPLICATIONS FOR PRACTICE Phase II trials are screening trials that seek to identify agents with sufficient activity to continue development. Thus, earlier endpoints are preferable, and the objective response rate (ORR) has been traditionally used as a surrogate endpoint in phase II trials for neuroendocrine tumors (NETs). However, the present study showed that the ORR was not significantly correlated with median progression-free survival (PFS). On the other hand, the 12-month PFS rate showed very strong correlation with median PFS and is considered a good alternate endpoint for screening phase II trials for NETs.
Collapse
Affiliation(s)
- Hiroshi Imaoka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Mitsuhito Sasaki
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hideaki Takahashi
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yusuke Hashimoto
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Izumi Ohno
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shuichi Mitsunaga
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kazuo Watanabe
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kumiko Umemoto
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Gen Kimura
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yuko Suzuki
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Motoyasu Kan
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Masafumi Ikeda
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
44
|
Selby PJ, Banks RE, Gregory W, Hewison J, Rosenberg W, Altman DG, Deeks JJ, McCabe C, Parkes J, Sturgeon C, Thompson D, Twiddy M, Bestall J, Bedlington J, Hale T, Dinnes J, Jones M, Lewington A, Messenger MP, Napp V, Sitch A, Tanwar S, Vasudev NS, Baxter P, Bell S, Cairns DA, Calder N, Corrigan N, Del Galdo F, Heudtlass P, Hornigold N, Hulme C, Hutchinson M, Lippiatt C, Livingstone T, Longo R, Potton M, Roberts S, Sim S, Trainor S, Welberry Smith M, Neuberger J, Thorburn D, Richardson P, Christie J, Sheerin N, McKane W, Gibbs P, Edwards A, Soomro N, Adeyoju A, Stewart GD, Hrouda D. Methods for the evaluation of biomarkers in patients with kidney and liver diseases: multicentre research programme including ELUCIDATE RCT. PROGRAMME GRANTS FOR APPLIED RESEARCH 2018. [DOI: 10.3310/pgfar06030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BackgroundProtein biomarkers with associations with the activity and outcomes of diseases are being identified by modern proteomic technologies. They may be simple, accessible, cheap and safe tests that can inform diagnosis, prognosis, treatment selection, monitoring of disease activity and therapy and may substitute for complex, invasive and expensive tests. However, their potential is not yet being realised.Design and methodsThe study consisted of three workstreams to create a framework for research: workstream 1, methodology – to define current practice and explore methodology innovations for biomarkers for monitoring disease; workstream 2, clinical translation – to create a framework of research practice, high-quality samples and related clinical data to evaluate the validity and clinical utility of protein biomarkers; and workstream 3, the ELF to Uncover Cirrhosis as an Indication for Diagnosis and Action for Treatable Event (ELUCIDATE) randomised controlled trial (RCT) – an exemplar RCT of an established test, the ADVIA Centaur® Enhanced Liver Fibrosis (ELF) test (Siemens Healthcare Diagnostics Ltd, Camberley, UK) [consisting of a panel of three markers – (1) serum hyaluronic acid, (2) amino-terminal propeptide of type III procollagen and (3) tissue inhibitor of metalloproteinase 1], for liver cirrhosis to determine its impact on diagnostic timing and the management of cirrhosis and the process of care and improving outcomes.ResultsThe methodology workstream evaluated the quality of recommendations for using prostate-specific antigen to monitor patients, systematically reviewed RCTs of monitoring strategies and reviewed the monitoring biomarker literature and how monitoring can have an impact on outcomes. Simulation studies were conducted to evaluate monitoring and improve the merits of health care. The monitoring biomarker literature is modest and robust conclusions are infrequent. We recommend improvements in research practice. Patients strongly endorsed the need for robust and conclusive research in this area. The clinical translation workstream focused on analytical and clinical validity. Cohorts were established for renal cell carcinoma (RCC) and renal transplantation (RT), with samples and patient data from multiple centres, as a rapid-access resource to evaluate the validity of biomarkers. Candidate biomarkers for RCC and RT were identified from the literature and their quality was evaluated and selected biomarkers were prioritised. The duration of follow-up was a limitation but biomarkers were identified that may be taken forward for clinical utility. In the third workstream, the ELUCIDATE trial registered 1303 patients and randomised 878 patients out of a target of 1000. The trial started late and recruited slowly initially but ultimately recruited with good statistical power to answer the key questions. ELF monitoring altered the patient process of care and may show benefits from the early introduction of interventions with further follow-up. The ELUCIDATE trial was an ‘exemplar’ trial that has demonstrated the challenges of evaluating biomarker strategies in ‘end-to-end’ RCTs and will inform future study designs.ConclusionsThe limitations in the programme were principally that, during the collection and curation of the cohorts of patients with RCC and RT, the pace of discovery of new biomarkers in commercial and non-commercial research was slower than anticipated and so conclusive evaluations using the cohorts are few; however, access to the cohorts will be sustained for future new biomarkers. The ELUCIDATE trial was slow to start and recruit to, with a late surge of recruitment, and so final conclusions about the impact of the ELF test on long-term outcomes await further follow-up. The findings from the three workstreams were used to synthesise a strategy and framework for future biomarker evaluations incorporating innovations in study design, health economics and health informatics.Trial registrationCurrent Controlled Trials ISRCTN74815110, UKCRN ID 9954 and UKCRN ID 11930.FundingThis project was funded by the NIHR Programme Grants for Applied Research programme and will be published in full inProgramme Grants for Applied Research; Vol. 6, No. 3. See the NIHR Journals Library website for further project information.
Collapse
Affiliation(s)
- Peter J Selby
- Clinical and Biomedical Proteomics Group, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
- Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Rosamonde E Banks
- Clinical and Biomedical Proteomics Group, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Walter Gregory
- Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - Jenny Hewison
- Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - William Rosenberg
- Institute for Liver and Digestive Health, Division of Medicine, University College London, London, UK
| | - Douglas G Altman
- Centre for Statistics in Medicine, University of Oxford, Oxford, UK
| | - Jonathan J Deeks
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Christopher McCabe
- Department of Emergency Medicine, University of Alberta Hospital, Edmonton, AB, Canada
| | - Julie Parkes
- Primary Care and Population Sciences Academic Unit, University of Southampton, Southampton, UK
| | | | | | - Maureen Twiddy
- Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - Janine Bestall
- Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | | | - Tilly Hale
- LIVErNORTH Liver Patient Support, Newcastle upon Tyne, UK
| | - Jacqueline Dinnes
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Marc Jones
- Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | | | | | - Vicky Napp
- Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - Alice Sitch
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Sudeep Tanwar
- Institute for Liver and Digestive Health, Division of Medicine, University College London, London, UK
| | - Naveen S Vasudev
- Clinical and Biomedical Proteomics Group, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
- Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Paul Baxter
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Sue Bell
- Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - David A Cairns
- Clinical and Biomedical Proteomics Group, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | | | - Neil Corrigan
- Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - Francesco Del Galdo
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Peter Heudtlass
- Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - Nick Hornigold
- Clinical and Biomedical Proteomics Group, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Claire Hulme
- Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - Michelle Hutchinson
- Clinical and Biomedical Proteomics Group, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Carys Lippiatt
- Department of Specialist Laboratory Medicine, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | | | - Roberta Longo
- Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - Matthew Potton
- Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - Stephanie Roberts
- Clinical and Biomedical Proteomics Group, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Sheryl Sim
- Clinical and Biomedical Proteomics Group, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Sebastian Trainor
- Clinical and Biomedical Proteomics Group, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Matthew Welberry Smith
- Clinical and Biomedical Proteomics Group, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
- Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - James Neuberger
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | | | - Paul Richardson
- Royal Liverpool and Broadgreen University Hospitals NHS Trust, Liverpool, UK
| | - John Christie
- Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - Neil Sheerin
- Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - William McKane
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Paul Gibbs
- Portsmouth Hospitals NHS Trust, Portsmouth, UK
| | | | - Naeem Soomro
- Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | | | - Grant D Stewart
- NHS Lothian, Edinburgh, UK
- Academic Urology Group, University of Cambridge, Cambridge, UK
| | - David Hrouda
- Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
45
|
Wei C, Wang S, Ye Z, Chen Z. Efficacy of targeted therapy for advanced renal cell carcinoma: a systematic review and meta-analysis of randomized controlled trials. Int Braz J Urol 2018; 44:219-237. [PMID: 29211397 PMCID: PMC6051488 DOI: 10.1590/s1677-5538.ibju.2017.0315] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 07/18/2017] [Indexed: 12/28/2022] Open
Abstract
We conducted a systematic review and meta-analysis of the literature on the efficacy of the targeted therapies in the treatment of advanced RCC and, via an indirect comparison, to provide an optimal treatment among these agents. A systematic search of Medline, Scopus, Cochrane Library and Clinical Trials unpublished was performed up to Jan 1, 2015 to identify eligible randomized trials. Outcomes of interest assessing a targeted agent included progression free survival (PFS), overall survival (OS) and objective response rate (ORR). Thirty eligible randomized controlled studies, total twentyfourth trails (5110 cases and 4626 controls) were identified. Compared with placebo and IFN-α, single vascular epithelial growth factor (receptor) tyrosine kinase inhibitor and mammalian target of rapamycin agent (VEGF(r)-TKI & mTOR inhibitor) were associated with improved PFS, improved OS and higher ORR, respectively. Comparing sorafenib combination vs sorafenib, there was no significant difference with regard to PFS and OS, but with a higher ORR. Comparing single or combination VEGF(r)-TKI & mTOR inhibitor vs BEV + IFN-α, there was no significant difference with regard to PFS, OS, or ORR. Our network ITC meta-analysis also indicated a superior PFS of axitinib and everolimus compared to sorafenib. Our data suggest that targeted therapy with VEGF(r)-TKI & mTOR inhibitor is associated with superior efficacy for treating advanced RCC with improved PFS, OS and higher ORR compared to placebo and IFN-α. In summary, here we give a comprehensive overview of current targeted therapies of advanced RCC that may provide evidence for the adequate targeted therapy selecting.
Collapse
Affiliation(s)
- Chao Wei
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shen Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhangqun Ye
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiqiang Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
46
|
Zahoor H, Rini BI, Ornstein MC. Extended therapy breaks from VEGFR TKI therapy in renal cell carcinoma: Sometimes less is more. Oncotarget 2018; 9:14036-14037. [PMID: 29581822 PMCID: PMC5865648 DOI: 10.18632/oncotarget.23005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 12/04/2017] [Indexed: 11/25/2022] Open
Affiliation(s)
- Haris Zahoor
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Brian I Rini
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | | |
Collapse
|
47
|
Khan MA, Raza A, Ovais M, Sohail MF, Ali S. Current state and prospects of nano-delivery systems for sorafenib. INT J POLYM MATER PO 2018. [DOI: 10.1080/00914037.2018.1429434] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Muhammad Adeeb Khan
- School of Material Science and Engineering (MSE), Nanyang Technological University, Nanyang, Singapore
- Department of Zoology, University of Azad Jammu and Kashmir, Muzaffarabad, Pakistan
- NILOP Nanomedicine Research Labs, National Institute of Lasers & Optronics (NILOP), Islamabad, Pakistan
| | - Abida Raza
- NILOP Nanomedicine Research Labs, National Institute of Lasers & Optronics (NILOP), Islamabad, Pakistan
| | - Muhammad Ovais
- Department of Biotechnology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Muhammad Farhan Sohail
- Department of Medicine, Biomaterials Innovation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA, USA
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore Campus, Lahore, Pakistan
| | - Shaukat Ali
- Department of Zoology, University of Azad Jammu and Kashmir, Muzaffarabad, Pakistan
| |
Collapse
|
48
|
Sartore-Bianchi A, Ricotta R, Cerea G, Maugeri M, Siena S. Rationale and Clinical Results of Multi-target Treatments in Oncology. Int J Biol Markers 2018; 22:77-87. [DOI: 10.1177/17246008070221s410] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
During the last 10 years, the concept of targeted biological therapy for the treatment of cancer has emerged. Targeted agents entered clinical practice only recently, and the first drugs with demonstrated clinical efficacy were mainly inhibitors of the ErbB family of receptors (i.e., EGFR and HER-2), either monoclonal antibodies (MAbs) or tyrosine kinase inhibitors (TKIs). After the proof of concept for the clinical efficacy and tolerability of these selective agents, it was conceived that most tumors will depend on more than one signaling pathway for their growth and survival. As a consequence, different strategies were pursued to inhibit multiple signaling pathways or multiple steps in the same pathway, either by the development of multi-targeted agents or the combination of single targeted drugs. The recent FDA and EMEA approval of sorafenib and sunitinib, both multi-targeted TKIs, marked the coming of age of this new generation of drugs. Now a whole new wave of multi-targeted compounds is moving into clinical trials, raising in the minds of investigators important questions about the best strategies to pursue in their use and many doubts about their differences and the seeming redundancies in the pipelines of pharmaceutical companies. This review will deal with the rationale underlying the multi-targeted approach and with the available clinical experience with multi-targeted agents, especially focusing on molecules with anti-EGFR mechanisms of action.
Collapse
Affiliation(s)
- A. Sartore-Bianchi
- The Falck Division of Medical Oncology, Ospedale Niguarda Ca’ Granda, Milan - Italy
| | - R. Ricotta
- The Falck Division of Medical Oncology, Ospedale Niguarda Ca’ Granda, Milan - Italy
| | - G. Cerea
- The Falck Division of Medical Oncology, Ospedale Niguarda Ca’ Granda, Milan - Italy
| | - M.R. Maugeri
- The Falck Division of Medical Oncology, Ospedale Niguarda Ca’ Granda, Milan - Italy
| | - S. Siena
- The Falck Division of Medical Oncology, Ospedale Niguarda Ca’ Granda, Milan - Italy
| |
Collapse
|
49
|
Bajetta E, Procopio G, Verzoni E, Catena L, De Dosso S, Platania M, Gevorgyan A. Renal Cell Cancer and Sorafenib: Skin Toxicity and Treatment Outcome. TUMORI JOURNAL 2018; 93:201-3. [PMID: 17557570 DOI: 10.1177/030089160709300216] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
We describe the case of a young man with refractory renal cell carcinoma who achieved an objective response in a metastatic lesion after biotherapy with the multikinase inhibitor sorafenib and also developed a severe skin reaction. The patient had been previously treated with various combinations of immunochemotherapy without any clinical benefit. We performed a brief review of the literature where similar cases were documented with the use of various anti-EGFR agents. The hypothesis of the correlation of skin toxicity with disease response is not new, but in the absence of any strong evidence remains controversial.
Collapse
Affiliation(s)
- Emilio Bajetta
- Unit of Medical Oncology 2, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| | | | | | | | | | | | | |
Collapse
|
50
|
Qin F, Yu H, Xu CR, Chen HH, Bai JL. Safety of axitinib and sorafenib monotherapy for patients with renal cell carcinoma: a meta-analysis. J Biomed Res 2018; 32:30-38. [PMID: 29353818 PMCID: PMC5956256 DOI: 10.7555/jbr.32.20170080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
We sought to investigate safety of axitinib or sorafenib in renal cell carcinoma (RCC)
patients and compare toxicity of these two vascular endothelial growth factor receptor
inhibitors. Databases of PubMed and Embase were searched. We included phase II and III
prospective trials, as well as retrospective studies, in which patients diagnosed with RCC
were treated with axitinib or sorafenib monotherapy at a starting dose of 5 mg and 400 mg
twice daily, respectively. The overall incidence of high grade hypertension, fatigue,
gastrointestinal toxicity and hand-foot syndrome, along with their 95% confidence
intervals (CI), were calculated using fixed- or random- effects model according to
heterogeneity test results. A total of 26 trials, including 4790 patients, were included
in our meta-analysis. Among them, 6 arms were related to axitinib and 22 were associated
with sorafenib. The incidences of hypertension (24.9% vs. 7.9%), fatigue
(8.2% vs. 6.6%), and gastrointestinal toxicity (17.6%
vs. 11.3%) were higher in patients receiving axitinib versus
those receiving sorafenib, while the incidence of hand-foot syndrome was lower in
patients receiving axitinib versus those receiving sorafenib (9.5%
vs. 13.3%). In conclusion, axitinib showed noticeably higher risks of
toxicity versus sorafenib. Close monitoring and effective measures for
adverse events are recommended during therapy.
Collapse
Affiliation(s)
- Fei Qin
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Hao Yu
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Chang-Rong Xu
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Hui-Hui Chen
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jian-Ling Bai
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|