1
|
Koudonas A, Dimitriadis G, Anastasiadis A, Papaioannou M. DNA Methylation as Drug Sensitivity Marker in RCC: A Systematic Review. EPIGENOMES 2024; 8:28. [PMID: 39051186 PMCID: PMC11270435 DOI: 10.3390/epigenomes8030028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/16/2024] [Accepted: 06/07/2024] [Indexed: 07/27/2024] Open
Abstract
Patient response after treatment of renal cell cancer (RCC) with systemic agents, which include various drug categories, is generally poor and unpredictable. In this context, the ideal drug administration includes tools to predict the sensitivity of the disease to therapy. The aim of this study was to systematically summarize the reports on the predictive value of the methylation status in the systemic therapy of RCC. Only original articles reporting on the association of promoter methylation with the response of patients or cell lines to systemic agents were included in this review. We applied PRISMA recommendations to the structure and methodology of this systematic review. Our literature search concluded with 31 articles conducted on RCC cell lines and patient tissues. The majority of the studies demonstrated a methylation-dependent response to systemic agents. This correlation suggests that the methylation pattern can be used as a predictive tool in the management of RCC with various classes of systemic agents. However, although methylation biomarkers show promise for predicting response, the evidence of such correlation is still weak. More studies on the gene methylation pattern in patients under systemic therapy and its correlation with different degrees of response are needed.
Collapse
Affiliation(s)
- Antonios Koudonas
- First Department of Urology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (A.K.); (G.D.); (A.A.)
- Laboratory of Biological Chemistry, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
- Department of Urology, 424 Military Hospital, 564 29 Thessaloniki, Greece
| | - Georgios Dimitriadis
- First Department of Urology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (A.K.); (G.D.); (A.A.)
| | - Anastasios Anastasiadis
- First Department of Urology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (A.K.); (G.D.); (A.A.)
| | - Maria Papaioannou
- First Department of Urology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (A.K.); (G.D.); (A.A.)
- Laboratory of Biological Chemistry, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| |
Collapse
|
2
|
Demurtas S, Frascaroli M, Sottotetti F, Rametta A, Procopio G, Locati LD. Impressive and Prolonged Response with Lenvatinib in a Highly Pretreated Patient with Metastatic Clear Cell Renal Cancer: A Case Report. J Kidney Cancer VHL 2024; 11:1-6. [PMID: 38628557 PMCID: PMC11017133 DOI: 10.15586/jkcvhl.v11i2.317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 03/27/2024] [Indexed: 04/19/2024] Open
Abstract
Clear cell renal carcinoma (ccRCC) can occur in young people and could be associated with an aggressive behavior. While for the first-line treatment in metastatic disease, there is an agreement to rely on an immunotherapy (IO)-based combination regimen, no standard second-line regimens exist. Generally, tyrosine kinase inhibitors (TKIs) are employed, even in sequence, although no trials have demonstrated yet the best succession. Herein, we present the case of a 39-year-old male, with a very aggressive ccRCC with somatic VHL mutation and distant metastases at diagnosis. He was treated with four different lines of therapies, including TKIs, with progressive multiple tumor deposits. Lenvatinib alone as the fifth line was able to induce a remarkable and prolonged tumor shrinkage with manageable toxicities.
Collapse
Affiliation(s)
- Sara Demurtas
- Internal Medicine and Therapeutics Department, University of Pavia, Pavia, Italy
- Medical Oncology, Maugeri Clinical Research Institutes IRCCS, Pavia; Italy
| | - Mara Frascaroli
- Medical Oncology, Maugeri Clinical Research Institutes IRCCS, Pavia; Italy
| | | | - Alessandro Rametta
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giuseppe Procopio
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Laura Deborah Locati
- Internal Medicine and Therapeutics Department, University of Pavia, Pavia, Italy
- Medical Oncology, Maugeri Clinical Research Institutes IRCCS, Pavia; Italy
| |
Collapse
|
3
|
Renal Carcinoma and Angiogenesis: Therapeutic Target and Biomarkers of Response in Current Therapies. Cancers (Basel) 2022; 14:cancers14246167. [PMID: 36551652 PMCID: PMC9776425 DOI: 10.3390/cancers14246167] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
Due to the aberrant hypervascularization and the high immune infiltration of renal tumours, current therapeutic regimens of renal cell carcinoma (RCC) target angiogenic or immunosuppressive pathways or both. Tumour angiogenesis plays an essential role in tumour growth and immunosuppression. Indeed, the aberrant vasculature promotes hypoxia and can also exert immunosuppressive functions. In addition, pro-angiogenic factors, including VEGF-A, have an immunosuppressive action on immune cells. Despite the progress of treatments in RCC, there are still non responders or acquired resistance. Currently, no biomarkers are used in clinical practice to guide the choice between the different available treatments. Considering the role of angiogenesis in RCC, angiogenesis-related markers are interesting candidates. They have been studied in the response to antiangiogenic drugs (AA) and show interest in predicting the response. They have been less studied in immunotherapy alone or combined with AA. In this review, we will discuss the role of angiogenesis in tumour growth and immune escape and the place of angiogenesis-targeted biomarkers to predict response to current therapies in RCC.
Collapse
|
4
|
Sharma R, Kannourakis G, Prithviraj P, Ahmed N. Precision Medicine: An Optimal Approach to Patient Care in Renal Cell Carcinoma. Front Med (Lausanne) 2022; 9:766869. [PMID: 35775004 PMCID: PMC9237320 DOI: 10.3389/fmed.2022.766869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 04/11/2022] [Indexed: 12/24/2022] Open
Abstract
Renal cell cancer (RCC) is a heterogeneous tumor that shows both intra- and inter-heterogeneity. Heterogeneity is displayed not only in different patients but also among RCC cells in the same tumor, which makes treatment difficult because of varying degrees of responses generated in RCC heterogeneous tumor cells even with targeted treatment. In that context, precision medicine (PM), in terms of individualized treatment catered for a specific patient or groups of patients, can shift the paradigm of treatment in the clinical management of RCC. Recent progress in the biochemical, molecular, and histological characteristics of RCC has thrown light on many deregulated pathways involved in the pathogenesis of RCC. As PM-based therapies are rapidly evolving and few are already in current clinical practice in oncology, one can expect that PM will expand its way toward the robust treatment of patients with RCC. This article provides a comprehensive background on recent strategies and breakthroughs of PM in oncology and provides an overview of the potential applicability of PM in RCC. The article also highlights the drawbacks of PM and provides a holistic approach that goes beyond the involvement of clinicians and encompasses appropriate legislative and administrative care imparted by the healthcare system and insurance providers. It is anticipated that combined efforts from all sectors involved will make PM accessible to RCC and other patients with cancer, making a tremendous positive leap on individualized treatment strategies. This will subsequently enhance the quality of life of patients.
Collapse
Affiliation(s)
- Revati Sharma
- Fiona Elsey Cancer Research Institute, Ballarat Central Technology Central Park, Ballarat Central, VIC, Australia
- School of Science, Psychology and Sport, Federation University, Mt Helen, VIC, Australia
| | - George Kannourakis
- Fiona Elsey Cancer Research Institute, Ballarat Central Technology Central Park, Ballarat Central, VIC, Australia
- School of Science, Psychology and Sport, Federation University, Mt Helen, VIC, Australia
| | - Prashanth Prithviraj
- Fiona Elsey Cancer Research Institute, Ballarat Central Technology Central Park, Ballarat Central, VIC, Australia
- School of Science, Psychology and Sport, Federation University, Mt Helen, VIC, Australia
| | - Nuzhat Ahmed
- Fiona Elsey Cancer Research Institute, Ballarat Central Technology Central Park, Ballarat Central, VIC, Australia
- School of Science, Psychology and Sport, Federation University, Mt Helen, VIC, Australia
- Centre for Reproductive Health, Hudson Institute of Medical Research and Department of Translational Medicine, Monash University, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
5
|
Komiyama T, Kim H, Tanaka M, Isaki S, Yokoyama K, Miyajima A, Kobayashi H. RNA-seq and Mitochondrial DNA Analysis of Adrenal Gland Metastatic Tissue in a Patient with Renal Cell Carcinoma. BIOLOGY 2022; 11:biology11040589. [PMID: 35453788 PMCID: PMC9030821 DOI: 10.3390/biology11040589] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/02/2022] [Accepted: 04/11/2022] [Indexed: 01/27/2023]
Abstract
This study aimed to clarify whether genetic mutations participate in renal cell carcinoma (RCC) metastasis to the adrenal gland (AG). Our study analyzed whole mitochondrial gene and ribonucleic acid sequencing (RNA-seq) data from a male patient in his 60s with metastatic RCC. We confirmed common mutation sites in the mitochondrial gene and carried out Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis using RNA-seq data for RCC and adrenal carcinoma. Furthermore, we confirmed the common mutation sites of mitochondrial genes in which the T3394Y (p.H30Y) site transitioned from histidine (His.; H) to tyrosine (Tyr.; Y) in the NADH dehydrogenase subunit 1 (ND1) gene. The R11,807G (p.T350A) site transitioned from threonine (Thr.; T) to alanine (Ala.; A). Additionally, the G15,438R or A (p.G231D) site transitioned from glycine (Gly.; G) to aspartic acid (Asp.; D) in cytochrome b (CYTB). Furthermore, pathway analysis, using RNA-seq, confirmed the common mutant pathway between RCC and adrenal carcinoma as cytokine–cytokine receptor (CCR) interaction. Confirmation of the original mutation sites suggests that transfer to AG may be related to the CCR interaction. Thus, during metastasis to the AG, mitochondria DNA mutation may represent the initial origin of the metastasis, followed by the likely mutation of the nuclear genes.
Collapse
Affiliation(s)
- Tomoyoshi Komiyama
- Department of Clinical Pharmacology, Tokai University School of Medicine, Isehara 259-1193, Kanagawa, Japan;
- Correspondence: (T.K.); (H.K.); Tel.: +81-463-93-1121 (T.K.)
| | - Hakushi Kim
- Department of Urology, Tokai University Hachioji Hospital, Tokyo 192-0032, Japan
- Correspondence: (T.K.); (H.K.); Tel.: +81-463-93-1121 (T.K.)
| | - Masayuki Tanaka
- Medical Science College Office, Tokai University, Isehara 259-1193, Kanagawa, Japan; (M.T.); (S.I.); (K.Y.)
| | - Sanae Isaki
- Medical Science College Office, Tokai University, Isehara 259-1193, Kanagawa, Japan; (M.T.); (S.I.); (K.Y.)
| | - Keiko Yokoyama
- Medical Science College Office, Tokai University, Isehara 259-1193, Kanagawa, Japan; (M.T.); (S.I.); (K.Y.)
| | - Akira Miyajima
- Department of Urology, Tokai University School of Medicine, Isehara 259-1193, Kanagawa, Japan;
| | - Hiroyuki Kobayashi
- Department of Clinical Pharmacology, Tokai University School of Medicine, Isehara 259-1193, Kanagawa, Japan;
| |
Collapse
|
6
|
Molecular biomarkers of prognosis in advanced renal cell carcinoma patients treated with Pazopanib plus interferon alpha (INF-2A) in a phase I/IIstudy by the Spanish Oncology Genitourinary Group. Clin Genitourin Cancer 2022; 20:388.e1-388.e10. [DOI: 10.1016/j.clgc.2022.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/14/2022] [Accepted: 03/14/2022] [Indexed: 11/21/2022]
|
7
|
Genome-wide association study of serum tumor markers in Southern Chinese Han population. BMC Cancer 2022; 22:160. [PMID: 35144566 PMCID: PMC8832811 DOI: 10.1186/s12885-022-09236-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 01/13/2022] [Indexed: 11/10/2022] Open
Abstract
Background Serum indicators AFP, CA50, CA125, CA153, CA19-9, CEA, f-PSA, SCC-Ag have been confirmed as tumor markers (TMs). We conducted a genome-wide association study on 8 tumor markers of our 427 Han population in southern China, in order to identify genetic loci that are significantly associated with the level of 8 tumor markers. Methods We use Gene Titan multi-channel instrument and Axiom Analysis Suite 6.0 software for genotyping. We used IMPUTE2 software for imputation, and 1000 Genomes Project (Phase 3) was used as haplotype reference. After necessary quality control and statistical analysis, genetic loci genome-wide associated with TMs (p < 5E-8) will be identified. Finally, we selected Top SNPs (p < 5E-7) from the GWAS results for replication test. We used SPSS software to draw the distribution box plots of serum TMs under different genotypes of significant loci. Results The results showed that there were only MUC1 (mucin 1)-rs4072037 significantly genome-wide associated with CA153 (p = 1.28E-18). However, we found that a total of 30 genetic loci have a suggestively significant genome-wide association with the level of 8 serum tumor markers (p < 5E-6). Then 3 Top SNPs (p < 5E-7) were selected for replication verification. The results showed that MUC1-rs4072037 was still significantly associated with CA153 in another population (p = 3.73E-08). Comparing with the TT genotype of rs4072037, the CA153 level was higher under CC or CT genotype of rs4072037. Conclusion MUC1-rs4072037 is significantly genome-wide associated with CA153 level. There are 30 genetic loci suggestively genome-wide associated with level of tumor markers among the Han population from Southern China. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09236-6.
Collapse
|
8
|
Nanotechnology-based approaches for effective detection of tumor markers: A comprehensive state-of-the-art review. Int J Biol Macromol 2022; 195:356-383. [PMID: 34920057 DOI: 10.1016/j.ijbiomac.2021.12.052] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 02/08/2023]
Abstract
As well-appreciated biomarkers, tumor markers have been spotlighted as reliable tools for predicting the behavior of different tumors and helping clinicians ascertain the type of molecular mechanism of tumorigenesis. The sensitivity and specificity of these markers have made them an object of even broader interest for sensitive detection and staging of various cancers. Enzyme-linked immunosorbent assay (ELISA), fluorescence-based, mass-based, and electrochemical-based detections are current techniques for sensing tumor markers. Although some of these techniques provide good selectivity, certain obstacles, including a low sample concentration or difficulty carrying out the measurement, limit their application. With the advent of nanotechnology, many studies have been carried out to synthesize and employ nanomaterials (NMs) in sensing techniques to determine these tumor markers at low concentrations. The fabrication, sensitivity, design, and multiplexing of sensing techniques have been uplifted due to the attractive features of NMs. Various NMs, such as magnetic and metal nanoparticles, up-conversion NPs, carbon nanotubes (CNTs), carbon-based NMs, quantum dots (QDs), and graphene-based nanosensors, hyperbranched polymers, optical nanosensors, piezoelectric biosensors, paper-based biosensors, microfluidic-based lab-on-chip sensors, and hybrid NMs have proven effective in detecting tumor markers with great sensitivity and selectivity. This review summarizes various categories of NMs for detecting these valuable markers, such as prostate-specific antigen (PSA), human carcinoembryonic antigen (CEA), alpha-fetoprotein (AFP), human chorionic gonadotropin (hCG), human epidermal growth factor receptor-2 (HER2), cancer antigen 125 (CA125), cancer antigen 15-3 (CA15-3, MUC1), and cancer antigen 19-9 (CA19-9), and highlights recent nanotechnology-based advancements in detection of these prognostic biomarkers.
Collapse
|
9
|
Kim TJ, Lee YH, Koo KC. Current and future perspectives on CAR-T cell therapy for renal cell carcinoma: A comprehensive review. Investig Clin Urol 2022; 63:486-498. [PMID: 36067994 PMCID: PMC9448669 DOI: 10.4111/icu.20220103] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 05/22/2022] [Accepted: 07/06/2022] [Indexed: 01/02/2023] Open
Abstract
In the clinical setting of renal cell carcinoma (RCC), immune reactions such as tumor-specific T cell responses can be spontaneous events or can be elicited by checkpoint inhibitors, cytokines, and other immunotherapy modalities. The results from immunotherapy have led to significant advances in treatment methods and patient outcomes. The approval of nivolumab primarily as a second-line monotherapy and the latest approval of novel combination therapies as first-line treatment have established the significance of immunotherapy in the treatment of RCC. In this perspective, chimeric antigen receptor (CAR)-T cell therapy represents a major advance in the developing field of immunotherapy. This treatment modality facilitates T cells to express specific CARs on the cell surface which are reinfused to the patient to treat the analogous tumor cells. After showing treatment potential in hematological malignancies, this new therapeutic approach has become a strong candidate as a therapeutic modality for solid neoplasms. Although CAR-T cell therapy has shown promise and clinical benefit compared to previous T-cell modulated immunotherapies, further studies are warranted to overcome unfavorable physiological settings and hindrances such as the lack of specific molecular targets, depletion of CAR-T cells, a hostile tumor microenvironment, and on/off-tumor toxicities. Several approaches are being considered and research is ongoing to overcome these problems. In this comprehensive review, we provide the rationale and preliminary results of CAR-T cell therapy in RCC and discuss emerging novel strategies and future directions.
Collapse
Affiliation(s)
- Tae Jin Kim
- Department of Urology, CHA University College of Medicine, CHA Bundang Medical Center, Seongnam, Korea
| | - Young Hwa Lee
- Department of Urology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Kyo Chul Koo
- Department of Urology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
10
|
Mikkelsen ML, Marcussen N, Rabjerg M. Surgically induced ischemia has no impact on protein expression levels of HIF-1α and related biomarkers in renal cell carcinoma. APMIS 2021; 129:535-547. [PMID: 33948984 DOI: 10.1111/apm.13131] [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: 06/11/2020] [Accepted: 03/08/2020] [Indexed: 11/27/2022]
Abstract
The increasing demands for personalized targeted therapy directed against renal cell carcinoma have driven a search for predictive markers. Novel therapies targeting HIF-1α in renal cell carcinoma have been developed, and HIF-1α has been suggested as a novel predictive marker of response to therapy. The surgical resection of a kidney tumor induces tissue ischemia, and HIF-1α is an oxygen-sensitive transcription factor, which is known to be upregulated during hypoxia. This study investigated the impact of intra-surgical and post-surgical ischemia on protein expression levels of HIF-1α and three related biomarkers (VEGF, GLUT-1, and CAIX) in 20 patients with renal cell carcinoma with immunohistochemistry and Western blotting. Surgical ischemia did not have a significant impact on protein expression levels of any of the investigated markers. Long-post-surgical ischemia resulted in reduced expression levels of HIF-1α, probably due to autolysis. Our results suggest that HIF-1α is a stable protein, with expression levels not affected by intra-surgical ischemia, and hence, HIF-1α is suited for marker analysis.
Collapse
Affiliation(s)
| | - Niels Marcussen
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Maj Rabjerg
- Department of Pathology, Odense University Hospital, Odense, Denmark
| |
Collapse
|
11
|
Correlative serum biomarker analyses in the phase 2 trial of lenvatinib-plus-everolimus in patients with metastatic renal cell carcinoma. Br J Cancer 2020; 124:237-246. [PMID: 33024271 PMCID: PMC7782770 DOI: 10.1038/s41416-020-01092-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 07/02/2020] [Accepted: 08/27/2020] [Indexed: 12/17/2022] Open
Abstract
Background No biomarkers have been established to predict treatment efficacy in renal cell carcinoma (RCC). In an exploratory retrospective analysis of a Phase 2 study, we constructed composite biomarker scores (CBSs) to predict progression-free survival (PFS) and overall survival (OS) in patients with metastatic RCC randomised to receive lenvatinib-plus-everolimus. Methods Of 40 biomarkers tested, the 5 most strongly associated with PFS (HGF, MIG, IL-18BP, IL-18, ANG-2) or OS (TIMP-1, M-CSF, IL-18BP, ANG-2, VEGF) were used to make a 5-factor PFS-CBS or OS-CBS, respectively. A 2-factor CBS was generated with biomarkers common to PFS-CBS and OS-CBS. Patients were divided into groups accordingly (5-factor-CBS high: 3−5, CBS-low: 0–2; 2-factor-CBS high: 1–2, CBS-low: 0). Results PFS/OS with lenvatinib-plus-everolimus were significantly longer in the 5-factor CBS-high group versus the CBS-low group (P = 0.0022/P < 0.0001, respectively). In the CBS-high group, PFS/OS were significantly longer with lenvatinib-plus-everolimus versus everolimus (P < 0.001/P = 0.0079, respectively); PFS was also significantly longer with lenvatinib-plus-everolimus versus lenvatinib (P = 0.0046). The 5-factor-CBS had a predictive role in PFS and OS after multivariate analysis. Similar trends were observed with the 2-factor-CBS for PFS (i.e., lenvatinib-plus-everolimus versus everolimus). Conclusions The 5-factor CBS may identify patients with metastatic RCC who would benefit from lenvatinib-plus-everolimus versus everolimus; additional validation is required. Clinical trial registration The clinical trial registration number is NCT01136733.
Collapse
|
12
|
Beuselinck B, Van Brussel T, Verbiest A, Vanmechelen M, Couchy G, Oudard S, Elaïdi R, Roussel E, Albersen M, Debruyne P, Baldewijns M, Machiels JP, Richard V, Verschaeve V, Wolter P, Rioux-Leclercq N, Laguerre B, Zucman-Rossi J, Lambrechts D. Validation of the Correlation Between Single Nucleotide Polymorphism rs307826 in VEGFR3 and Outcome in Metastatic Clear-Cell Renal Cell Carcinoma Patients Treated with Sunitinib. KIDNEY CANCER 2020. [DOI: 10.3233/kca-200086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND: Previously, we have shown a correlation between single nucleotide polymorphism (SNP) rs307826 in vascular endothelial growth factor receptor-3 (VEGFR3) and outcome in metastatic clear-cell renal cell carcinoma (m-ccRCC) patients treated with sunitinib. OBJECTIVE: We aimed to validate this finding in an independent patient series. METHODS: m-ccRCC patients receiving sunitinib as first-line targeted therapy were included in a validation cohort. Endpoints were response rate (RR), progression-free survival (PFS) and overall survival (OS). We also updated survival data of our discovery cohort as described previously. RESULTS: Eighty-four patients were included in the validation cohort. rs307826 AG/GG-carriers had a shorter PFS (8 versus 12 months, p = 0.04) and a trend towards a shorter OS (18 versus 27 months, p = 0.22) compared to AA-carriers. In the total series of 168 patients (from the discovery cohort, as described previously, and the validation cohort), rs307826 AG/GG-carriers had a poorer RR (29% versus 53%, p = 0.008), PFS (8 versus 15 months, p = 0.0002) and OS (22 versus 31 months, p = 0.004) compared to AA-carriers. rs307826 was independently associated with PFS and OS on multivariate analysis. CONCLUSION: VEGFR3 rs307826 seems to be associated with outcome on sunitinib in m-ccRCC. Its impact highlights the role of VEGFR3 in ccRCC pathogenesis and as a target of sunitinib.
Collapse
Affiliation(s)
- Benoit Beuselinck
- Department of General Medical Oncology and Laboratory for Experimental Oncology, University Hospitals Leuven, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Thomas Van Brussel
- Department of Oncology, Laboratory for Translational Genetics, KU Leuven, Leuven, Belgium
- Vesalius Research Center, VIB, Leuven, Belgium
| | - Annelies Verbiest
- Department of General Medical Oncology and Laboratory for Experimental Oncology, University Hospitals Leuven, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Maxime Vanmechelen
- Department of General Medical Oncology and Laboratory for Experimental Oncology, University Hospitals Leuven, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | | | - Stéphane Oudard
- Department of Medical Oncology, Georges Pompidou European Hospital, Université Paris-5 René Descartes, Paris, France
| | - Reza Elaïdi
- Department of Medical Oncology, Georges Pompidou European Hospital, Université Paris-5 René Descartes, Paris, France
| | - Eduard Roussel
- Department of Urology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Maarten Albersen
- Department of Urology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Philip Debruyne
- Department of Medical Oncology, AZ Groeninge, Kortrijk, Belgium, and Faculty of Health, Education, Medicine & Social Care, Anglia Ruskin University, Chelmsford, UK
| | - Marcella Baldewijns
- Department of Pathology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Jean-Pascal Machiels
- Department of Medical Oncology, Cliniques Universitaires Saint-Luc, UCLouvain, Brussels, Belgium
| | - Vincent Richard
- Department of Medical Oncology, CHU Ambroise Paré, Mons, Belgium
| | - Vincent Verschaeve
- Department of Medical Oncology, Grand Hôpital de Charleroi, Charleroi, Belgium
| | - Pascal Wolter
- Department of Medical Oncology, St. Nikolaus-Hospital Eupen, Eupen, Belgium
| | | | | | | | - Diether Lambrechts
- Department of Oncology, Laboratory for Translational Genetics, KU Leuven, Leuven, Belgium
- Vesalius Research Center, VIB, Leuven, Belgium
| |
Collapse
|
13
|
Anti-angiogenesis and Immunotherapy: Novel Paradigms to Envision Tailored Approaches in Renal Cell-Carcinoma. J Clin Med 2020; 9:jcm9051594. [PMID: 32456352 PMCID: PMC7291047 DOI: 10.3390/jcm9051594] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/18/2020] [Accepted: 05/20/2020] [Indexed: 12/16/2022] Open
Abstract
Although decision making strategy based on clinico-histopathological criteria is well established, renal cell carcinoma (RCC) represents a spectrum of biological ecosystems characterized by distinct genetic and molecular alterations, diverse clinical courses and potential specific therapeutic vulnerabilities. Given the plethora of drugs available, the subtype-tailored treatment to RCC subtype holds the potential to improve patient outcome, shrinking treatment-related morbidity and cost. The emerging knowledge of the molecular taxonomy of RCC is evolving, whilst the antiangiogenic and immunotherapy landscape maintains and reinforces their potential. Although several prognostic factors of survival in patients with RCC have been described, no reliable predictive biomarkers of treatment individual sensitivity or resistance have been identified. In this review, we summarize the available evidence able to prompt more precise and individualized patient selection in well-designed clinical trials, covering the unmet need of medical choices in the era of next-generation anti-angiogenesis and immunotherapy.
Collapse
|
14
|
Kovacova J, Juracek J, Poprach A, Kopecky J, Fiala O, Svoboda M, Fabian P, Radova L, Brabec P, Buchler T, Slaby O. MiR-376b-3p Is Associated With Long-term Response to Sunitinib in Metastatic Renal Cell Carcinoma Patients. Cancer Genomics Proteomics 2020; 16:353-359. [PMID: 31467229 DOI: 10.21873/cgp.20140] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 06/10/2019] [Accepted: 06/11/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND/AIM Sunitinib is a tyrosine kinase inhibitor routinely used as first-line therapy in metastatic renal cell carcinoma (mRCC). Emerging evidence suggests that microRNAs (miRNAs) could be suitable biomarkers with predictive potential in mRCC. The aim of this study was to identify miRNA-based predictive biomarkers of therapy response to avoid unnecessary therapy to non-responding patients. PATIENTS AND METHODS High-throughput miRNA microarray profiling was performed on a cohort of 47 patients treated with sunitinib. Validation of candidate miRNAs was carried out on an independent cohort of 132 mRCC patients using qRT-PCR. RESULTS Out of 158 miRNAs (65 down-regulated, 93 up-regulated), six miRNAs were chosen for independent validation and miR-376b-3p was confirmed to be differentially expressed in tumors of patients with primary resistance versus long-term response (p<0.0002). CONCLUSION A predictive miRNA associated with progression-free survival in metastatic renal cell carcinoma patients treated with sunitinib was identified.
Collapse
Affiliation(s)
- Julia Kovacova
- Masaryk University, Central European Institute of Technology, Brno, Czech Republic
| | - Jaroslav Juracek
- Masaryk University, Central European Institute of Technology, Brno, Czech Republic
| | - Alexandr Poprach
- Masaryk Memorial Cancer Institute, Department of Comprehensive Cancer Care, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jindrich Kopecky
- Department of Clinical Oncology and Radiotherapy, University Hospital in Hradec Kralove, Hradec Kralove, Czech Republic
| | - Ondrej Fiala
- Department of Oncology and Radiotherapeutics, Medical School and Teaching Hospital in Pilsen, Charles University, Pilsen, Czech Republic.,Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Marek Svoboda
- Masaryk Memorial Cancer Institute, Department of Comprehensive Cancer Care, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Pavel Fabian
- Masaryk Memorial Cancer Institute, Department of Oncological and Experimental Pathology Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Lenka Radova
- Masaryk University, Central European Institute of Technology, Brno, Czech Republic
| | - Petr Brabec
- Institute for Biostatistics and Analyses, Masaryk University, Brno, Czech Republic
| | - Tomas Buchler
- Department of Oncology, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
| | - Ondrej Slaby
- Masaryk Memorial Cancer Institute, Department of Comprehensive Cancer Care, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| |
Collapse
|
15
|
Cao DL, Dai WX, Huang YQ, Yu LJ, Wu JL, Shi GH, Zhang HL, Zhu Y, Dai B, Ye DW. Development and validation of a robust multigene signature as an aid to predict early relapse in stage I-III clear cell and papillary renal cell cancer. J Cancer 2020; 11:997-1007. [PMID: 31956346 PMCID: PMC6959077 DOI: 10.7150/jca.38274] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 11/14/2019] [Indexed: 01/07/2023] Open
Abstract
Background and objectives: Multi-gene signature can be used as prognostic indicator in many types of cancer, but the association with early-relapse in patients with stage I-III clear cell and papillary renal cell cancer (RCC) is unknown. We aim to establish a mRNAs signature for improving prediction of early-relapse in patients with stage I-III clear cell and papillary RCC. Methods: The data of 610 patients with stage I-III RCC from The Cancer Genome Atlas (TCGA) and 270 patients from Fudan University Shanghai Cancer Center (FUSCC) were extracted. Propensity score matching analysis, linear models for microarray data VOOM method, least absolute shrinkage and selection operation Cox regression modeling analysis was conducted in turn for selecting multi-mRNA signature. Survival differences were assessed by Kaplan-Meier estimate and compared using log-rank test. Multivariable Cox regression and time-dependent receiver operating characteristic curves were used to evaluate the association of mRNAs signature with relapse-free survival (RFS). Results: Seventeen mRNAs were identified to constitute the early-relapse signature. Among patients with stage I-III RCC, those with high-risk score calculated from 17 mRNAs signature showed shorter RFS than those with low-risk score, both in TCGA discovery and internal validation sets, and in FUSCC discovery and internal validation sets (all p < 0.05). In multivariable Cox regression analysis, the 17 mRNAs signature remained an independent prognostic factor both in TCGA discovery (HR 2.43, 95%CI 1.98-2.96) and internal validation sets (HR 1.66, 95%CI 1.19-2.30), and FUSCC discovery (HR 1.28, 95%CI 1.13-1.43) and internal validation sets (HR 1.65, 95%CI 1.11-2.48). Additionally, the 17 mRNAs signature achieved a higher accuracy for RFS estimation beyond clinical indicator. Conclusion: The 17 mRNAs signature could classify stage I-III RCC patients into low- or high-risk of early-relapse, and will help to guide interventions to optimize survival outcomes.
Collapse
Affiliation(s)
- Da-Long Cao
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Wei-Xing Dai
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yong-Qiang Huang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Lei-Jun Yu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jun-Long Wu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Guo-Hai Shi
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Hai-Liang Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yao Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Bo Dai
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ding-Wei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
16
|
D'Aniello C, Berretta M, Cavaliere C, Rossetti S, Facchini BA, Iovane G, Mollo G, Capasso M, Pepa CD, Pesce L, D'Errico D, Buonerba C, Di Lorenzo G, Pisconti S, De Vita F, Facchini G. Biomarkers of Prognosis and Efficacy of Anti-angiogenic Therapy in Metastatic Clear Cell Renal Cancer. Front Oncol 2019; 9:1400. [PMID: 31921657 PMCID: PMC6917607 DOI: 10.3389/fonc.2019.01400] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 11/27/2019] [Indexed: 12/30/2022] Open
Abstract
In the last decades, the prognosis of metastatic renal cell carcinoma (mRCC) has remarkably improved following the advent of the "targeted therapy" era. The expanding knowledge on the prominent role played by angiogenesis in RCC pathogenesis has led to approval of multiple anti-angiogenic agents such as sunitinib, pazopanib, axitinib, cabozantinib, sorafenib, and bevacizumab. These agents can induce radiological responses and delay cancer progression for months or years before onset of resistance, with a clinically meaningful activity. The need for markers of prognosis and efficacy of anti-angiogenic agents has become more compelling as novel systemic immunotherapy agents have also been approved in RCC and can be administered as an alternative to angiogenesis inhibitors. Anti PD-1 monoclonal antibody nivolumab has been approved in the second-line setting after tyrosine kinase inhibitors failure, while combination of nivolumab plus anti CTLA-4 monoclonal antibody ipilimumab has been approved as first-line therapy of RCC patients at intermediate or poor prognosis. In this review article, biomarkers of prognosis and efficacy of antiangiogenic therapies are summarized with a focus on those that have the potential to affect treatment decision-making in RCC. Biomarkers predictive of toxicity of anti-angiogenic agents have also been discussed.
Collapse
Affiliation(s)
- Carmine D'Aniello
- Division of Medical Oncology, A.O.R.N. dei COLLI “Ospedali Monaldi-Cotugno-CTO,”Naples, Italy
| | - Massimiliano Berretta
- Division of Medical Oncology, Istituto Nazionale Tumori, IRCCS CRO Aviano (PN), Milan, Italy
| | - Carla Cavaliere
- UOC of Medical Oncology, ASL NA 3 SUD, Ospedali Riuniti Area Nolana, Nola, Italy
| | - Sabrina Rossetti
- Departmental Unit of Experimental Uro-Andrologic Clinical Oncology, Istituto Nazionale Tumori Fondazione G. Pascale—IRCCS, Naples, Italy
| | - Bianca Arianna Facchini
- Division of Medical Oncology, Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Gelsomina Iovane
- Departmental Unit of Experimental Uro-Andrologic Clinical Oncology, Istituto Nazionale Tumori Fondazione G. Pascale—IRCCS, Naples, Italy
| | - Giovanna Mollo
- Departmental Unit of Experimental Uro-Andrologic Clinical Oncology, Istituto Nazionale Tumori Fondazione G. Pascale—IRCCS, Naples, Italy
| | - Mariagrazia Capasso
- Departmental Unit of Experimental Uro-Andrologic Clinical Oncology, Istituto Nazionale Tumori Fondazione G. Pascale—IRCCS, Naples, Italy
| | | | - Laura Pesce
- Oncology Unit, San Luca Hospital, Vallo Della Lucania, Italy
| | - Davide D'Errico
- Departmental Unit of Experimental Uro-Andrologic Clinical Oncology, Istituto Nazionale Tumori Fondazione G. Pascale—IRCCS, Naples, Italy
| | - Carlo Buonerba
- CRTR Rare Tumors Reference Center, AOU Federico II, Naples, Italy
- Environment & Health Operational Unit, Zoo-Prophylactic Institute of Southern Italy, Portici, Italy
| | - Giuseppe Di Lorenzo
- Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy
- Department of Medicine, University of Molise, Campobasso, Italy
| | - Salvatore Pisconti
- Department of Onco-Hematology, Medical Oncology, S.G. Moscati Hospital, Taranto, Italy
| | - Ferdinando De Vita
- Division of Medical Oncology, Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Gaetano Facchini
- Departmental Unit of Experimental Uro-Andrologic Clinical Oncology, Istituto Nazionale Tumori Fondazione G. Pascale—IRCCS, Naples, Italy
| |
Collapse
|
17
|
Buller DM, Ristau BT. Improving patient selection for adjuvant therapy in high-risk renal cell carcinoma. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:S104. [PMID: 31576311 DOI: 10.21037/atm.2019.04.75] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Dylan M Buller
- University of Connecticut School of Medicine, Farmington, CT, USA
| | | |
Collapse
|
18
|
Vanmechelen M, Lambrechts D, Van Brussel T, Verbiest A, Couchy G, Schöffski P, Dumez H, Debruyne PR, Lerut E, Machiels JP, Richard V, Albersen M, Verschaeve V, Oudard S, Méjean A, Wolter P, Zucman-Rossi J, Beuselinck B. Fibroblast Growth Factor Receptor-2 Polymorphism rs2981582 is Correlated With Progression-free Survival and Overall Survival in Patients With Metastatic Clear-cell Renal Cell Carcinoma Treated With Sunitinib. Clin Genitourin Cancer 2019; 17:e235-e246. [DOI: 10.1016/j.clgc.2018.11.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 11/08/2018] [Accepted: 11/08/2018] [Indexed: 02/06/2023]
|
19
|
Abstract
Tumor blood vessel formation (angiogenesis) is essential for tumor growth and metastasis. Two main endothelial ligand–receptor pathways regulating angiogenesis are vascular endothelial growth factor (VEGF) receptor and angiopoietin-TIE receptor pathways. The angiopoietin-TIE pathway is required for the remodeling and maturation of the blood and lymphatic vessels during embryonic development after VEGF and VEGF-C mediated development of the primary vascular plexus. Angiopoietin-1 (ANGPT1) stabilizes the vasculature after angiogenic processes, via tyrosine kinase with immunoglobulin-like and EGF-like domains 2 (TIE2) activation. In contrast, ANGPT2 is upregulated at sites of vascular remodeling. ANGPT2 is secreted by activated endothelial cells in inflammation, promoting vascular destabilization. ANGPT2 has been found to be expressed in many human cancers. Intriguingly, in preclinical models inhibition of ANGPT2 has provided promising results in preventing tumor angiogenesis, tumor growth, and metastasis, making it an attractive candidate to target in tumors. However, until now the first ANGPT2 targeting therapies have been less effective in clinical trials than in experimental models. Additionally, in preclinical models combined therapy against ANGPT2 and VEGF or immune checkpoint inhibitors has been superior to monotherapies, and these pathways are also targeted in early clinical trials. In order to improve current anti-angiogenic therapies and successfully exploit ANGPT2 as a target for cancer treatment, the biology of the angiopoietin-TIE pathway needs to be profoundly clarified.
Collapse
Affiliation(s)
- Dieter Marmé
- Tumor Biology Center, Freiburg, Baden-Württemberg Germany
| |
Collapse
|
20
|
Jiménez-Fonseca P, Martín MN, Carmona-Bayonas A, Calvo A, Fernández-Mateos J, Redrado M, Capdevila J, Lago NM, Lacasta A, Muñarriz J, Segura Á, Fuster J, Barón F, Llanos M, Serrano R, Castillo A, Cruz Hernández JJ, Grande E. Biomarkers and polymorphisms in pancreatic neuroendocrine tumors treated with sunitinib. Oncotarget 2018; 9:36894-36905. [PMID: 30651923 PMCID: PMC6319342 DOI: 10.18632/oncotarget.26380] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 10/31/2018] [Indexed: 12/21/2022] Open
Abstract
Several circulating biomarkers and single nucleotide polymorphisms (SNPs) have been correlated with efficacy and tolerability to antiangiogenic agents. These associations remain unexplored in well-differentiated, metastatic pancreatic neuroendocrine tumors treated with the multitargeted tyrosine kinase inhibitor sunitinib. We have assessed the effect on tumor response at 6 months, overall survival, progression-free survival and safety of 14 SNPs, and 6 soluble proteins. Forty-three patients were recruited. Two SNPs in the vascular endothelial growth factor receptor 3 (VEGFR-3) gene predicted lower overall survival: rs307826 with hazard ratio (HR) 3.67 (confidence interval [CI] 95%, 1.35-10.00) and rs307821 with HR 3.84 (CI 95%, 1.47-10.0). Interleukin-6 was associated with increased mortality: HR 1.06 (CI 95%, 1.01-1.12), and osteopontin was associated with shorter PFS: HR 1.087 (1.01-1.16), independently of Ki-67. Furthermore, levels of osteopontin remained higher at the end of the study in patients considered non-responders: 38.5 ng/mL vs. responders: 18.7 ng/mL, p-value=0.039. Dynamic upward variations were also observed with respect to IL-8 levels in sunitinib-refractory individuals: 28.5 pg/mL at baseline vs. 38.3 pg/mL at 3 months, p-value=0.024. In conclusion, two VEGFR-3 SNPs as well as various serum biomarkers were associated with diverse clinical outcomes in patients with well-differentiated pancreatic neuroendocrine tumors treated with sunitinib.
Collapse
Affiliation(s)
- Paula Jiménez-Fonseca
- Medical Oncology Department, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Miguel Navarro Martín
- Medical Oncology Department, Hospital Universitario de Salamanca, IBSAL, Salamanca, Spain
| | - Alberto Carmona-Bayonas
- Hematology and Medical Oncology Department, Hospital Universitario Morales Meseguer, UMU, IMIB, Murcia, Spain
| | - Alfonso Calvo
- IDISNA and Program in Solid Tumors and Biomarkers, Center for Applied Medical Research (CIMA), Department of Histology and Pathology, University of Navarra, CIBERONC, ISC-II, Pamplona, Spain
| | - Javier Fernández-Mateos
- Molecular Medicine Unit, IBSAL, Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Miriam Redrado
- IDISNA and Program in Solid Tumors and Biomarkers, Center for Applied Medical Research (CIMA), Department of Histology and Pathology, University of Navarra, Pamplona, Navarra, Spain
| | - Jaume Capdevila
- Medical Oncology Department, Hospital Universitario Vall d'Hebron, Autonomous University of Barcelona, Barcelona, Spain
| | - Nieves Martínez Lago
- Medical Oncology Department, Hospital Universitario de A Coruña, La Coruña, Spain
| | - Adelaida Lacasta
- Medical Oncology Department, Hospital Universitario Donostia, Guipúzcoa, Spain
| | - Javier Muñarriz
- Medical Oncology Department, Hospital General Universitario de Castellón, Castellón, Spain
| | - Ángel Segura
- Medical Oncology Department, Hospital Universitario La Fe, Valencia, Spain
| | - Josep Fuster
- Medical Oncology Department, Hospital Universitario Son Espases, Palma de Mallorca, Spain
| | - Francisco Barón
- Medical Oncology Department, Hospital Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Marta Llanos
- Medical Oncology Department, Hospital Universitario de Canarias, Santa Cruz de Tenerife, Spain
| | - Raquel Serrano
- Medical Oncology Department, Hospital Universitario Reina Sofia, Cordoba, Spain
| | - Alfredo Castillo
- Medical Oncology Department, Hospital Universitario Central de Asturias, Oviedo, Spain
| | | | - Enrique Grande
- Medical Oncology Department, MD Anderson Cancer Center, Madrid, Spain
| |
Collapse
|
21
|
Marisi G, Scarpi E, Passardi A, Nanni O, Pagan F, Valgiusti M, Casadei Gardini A, Neri LM, Frassineti GL, Amadori D, Ulivi P. IL-8 and thrombospondin-1 as prognostic markers in patients with metastatic colorectal cancer receiving bevacizumab. Cancer Manag Res 2018; 10:5659-5666. [PMID: 30532588 PMCID: PMC6241685 DOI: 10.2147/cmar.s181570] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Purpose Bevacizumab (B) plus chemotherapy (CT) is a common choice for first-line treatment of metastatic colorectal cancer. Molecular predictors of B efficacy have still not been identified. We analyzed the role of 22 angiogenesis-associated proteins in patient outcome. Patients and methods Serum samples collected at baseline and at the first clinical evaluation were available for 58 patients enrolled in the randomized multicenter ITACa trial and who received CT+ B. Serum protein levels were determined using multiplex ELISA. Results Patients with baseline ≥145 pg/mL IL-8 showed shorter median progression-free survival and overall survival (OS) than those with lower levels (6.5 vs 6. 12.6 months; HR 7.39, P<0.0001 and 8.7 vs 28.8 months, HR 7.68, P<0.001, respectively). Moreover, patients with baseline thrombospondin-1 levels ≥12,000 ng/mL had a better median OS than those with lower levels (34.5 vs 13.1 months, HR 0.43, P=0.007). Patients with a ≥20% reduction in IL-8 levels from baseline to first clinical evaluation showed a better progression-free survival and OS than the others (HR 0.41, P=0.005 and HR 0.43, P=0.007, respectively). Conclusion Baseline IL-8 and thrombospondin-1 levels and reduced IL-8 during B treatment could represent potential prognostic markers in metastatic colorectal cancer.
Collapse
Affiliation(s)
- Giorgia Marisi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola 47014, Italy,
| | - Emanuela Scarpi
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola 47014, Italy
| | - Alessandro Passardi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola 47014, Italy
| | - Oriana Nanni
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola 47014, Italy
| | - Flavia Pagan
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola 47014, Italy
| | - Martina Valgiusti
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola 47014, Italy
| | - Andrea Casadei Gardini
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola 47014, Italy
| | - Luca Maria Neri
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara 44100, Italy
| | - Giovanni Luca Frassineti
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola 47014, Italy
| | - Dino Amadori
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola 47014, Italy
| | - Paola Ulivi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola 47014, Italy,
| |
Collapse
|
22
|
George DJ, Martini JF, Staehler M, Motzer RJ, Magheli A, Donskov F, Escudier B, Li S, Casey M, Valota O, Laguerre B, Pantuck AJ, Pandha HS, Patel A, Lechuga M, Ravaud A. Phase III Trial of Adjuvant Sunitinib in Patients with High-Risk Renal Cell Carcinoma: Exploratory Pharmacogenomic Analysis. Clin Cancer Res 2018; 25:1165-1173. [PMID: 30401688 DOI: 10.1158/1078-0432.ccr-18-1724] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 08/28/2018] [Accepted: 11/02/2018] [Indexed: 01/26/2023]
Abstract
PURPOSE In the S-TRAC trial, adjuvant sunitinib prolonged disease-free survival (DFS) versus placebo in patients with loco-regional renal cell carcinoma at high risk of recurrence after nephrectomy. An exploratory analysis evaluated associations between SNPs in several angiogenesis- or hypoxia-related genes and clinical outcomes in S-TRAC. PATIENTS AND METHODS Blood samples were genotyped for 10 SNPs and one insertion/deletion mutation using TaqMan assays. DFS was compared using log-rank tests for each genotype in sunitinib versus placebo groups and between genotypes within each of three (sunitinib, placebo, and combined sunitinib plus placebo) treatment groups. P values were unadjusted. RESULTS In all, 286 patients (sunitinib, n = 142; placebo, n = 144) were genotyped. Longer DFS [HR; 95% confidence interval (CI)] was observed with sunitinib versus placebo for VEGFR1 rs9554320 C/C (HR 0.44; 95% CI, 0.21-0.91; P = 0.023), VEGFR2 rs2071559 T/T (HR 0.46; 95% CI, 0.23-0.90; P = 0.020), and eNOS rs2070744 T/T (HR 0.53; 95% CI, 0.30-0.94; P = 0.028). Shorter DFS was observed for VEGFR1 rs9582036 C/A versus C/C with sunitinib, placebo, and combined therapies (P ≤ 0.05), and A/A versus C/C with sunitinib (P = 0.022). VEGFR1 rs9554320 A/C versus A/A was associated with shorter DFS in the placebo (P = 0.038) and combined (P = 0.006) groups. CONCLUSIONS Correlations between VEGFR1 and VEGFR2 SNPs and longer DFS with sunitinib suggest germline SNPs are predictive of improved outcomes with adjuvant sunitinib in patients with renal cell carcinoma. Independent validation studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Daniel J George
- Department of Medical Oncology, Duke Cancer Center, Durham, North Carolina.
| | | | - Michael Staehler
- Department of Urology, University Hospital of Munich, Munich, Germany
| | - Robert J Motzer
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ahmed Magheli
- Department of Urology, Charité Universitaetsmedizin Berlin, Berlin, Germany
| | - Frede Donskov
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Bernard Escudier
- Department of Urology, Institut Gustave Roussy, Villejuif, France
| | - Sherry Li
- Global Product Development, Pfizer Inc, La Jolla, California
| | - Michelle Casey
- Global Product Development, Pfizer Inc., Collegeville, Pennsylvania
| | - Olga Valota
- Global Product Development, Pfizer S.r.L, Milan, Italy
| | - Brigitte Laguerre
- Department of Medical Oncology, Centre Eugene Marquis, Rennes, France
| | - Allan J Pantuck
- Department of Urology, Ronald Reagan UCLA Medical Center, Los Angeles, California
| | - Hardev S Pandha
- Department of Medical Oncology, University of Surrey, Surrey, United Kingdom
| | | | - Maria Lechuga
- Global Product Development, Pfizer S.r.L, Milan, Italy
| | - Alain Ravaud
- Department of Medical Oncology, Bordeaux University Hospital, Bordeaux, France
| |
Collapse
|
23
|
Evaluation of KDR rs34231037 as a predictor of sunitinib efficacy in patients with metastatic renal cell carcinoma. Pharmacogenet Genomics 2018; 27:227-231. [PMID: 28430711 DOI: 10.1097/fpc.0000000000000280] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The identification of biomarkers able to predict clinical benefit from vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors is urgently needed. Recently, Maitland and colleagues described an association between KDR-rs34231037 and soluble VEGFR2 levels as well as pazopanib pharmacodynamics. We investigated in a well-characterized series of metastatic clear cell renal cell carcinoma patients whether rs34231037 could influence sunitinib response. Clinical data and DNA were available from an international series of 276 patients. KDR-rs34231037 association with sunitinib response, clinical benefit, and progression-free survival was analyzed using logistic and Cox regression analyses. We found that G-allele carriers were over-represented among patients with clinical benefit during sunitinib treatment compared with those refractory to the treatment (odds ratio: 3.78; 95% confidence interval: 1.02-14.06; P=0.047, multivariable analysis). In conclusion, rs34231037 variant carriers seemed to have better sunitinib response than wild-type patients. Moreover, the association with tumor size reduction suggests that this single nucleotide polymorphism might also identify patients with successful tumor downsizing under anti-VEGFR therapy.
Collapse
|
24
|
|
25
|
The tyrosine-kinase inhibitor sunitinib targets vascular endothelial (VE)-cadherin: a marker of response to antitumoural treatment in metastatic renal cell carcinoma. Br J Cancer 2018; 118:1179-1188. [PMID: 29563634 PMCID: PMC5943344 DOI: 10.1038/s41416-018-0054-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 02/02/2018] [Accepted: 02/20/2018] [Indexed: 12/19/2022] Open
Abstract
Background Vascular endothelial (VE)-cadherin is an endothelial cell-specific protein responsible for endothelium integrity. Its adhesive properties are regulated by post-translational processing, such as tyrosine phosphorylation at site Y685 in its cytoplasmic domain, and cleavage of its extracellular domain (sVE). In hormone-refractory metastatic breast cancer, we recently demonstrated that sVE levels correlate to poor survival. In the present study, we determine whether kidney cancer therapies had an effect on VE-cadherin structural modifications and their clinical interest to monitor patient outcome. Methods The effects of kidney cancer biotherapies were tested on an endothelial monolayer model mimicking the endothelium lining blood vessels and on a homotypic and heterotypic 3D cell model mimicking tumour growth. sVE was quantified by ELISA in renal cell carcinoma patients initiating sunitinib (48 patients) or bevacizumab (83 patients) in the first-line metastatic setting (SUVEGIL and TORAVA trials). Results Human VE-cadherin is a direct target for sunitinib which inhibits its VEGF-induced phosphorylation and cleavage on endothelial monolayer and endothelial cell migration in the 3D model. The tumour cell environment modulates VE-cadherin functions through MMPs and VEGF. We demonstrate the presence of soluble VE-cadherin in the sera of mRCC patients (n = 131) which level at baseline, is higher than in a healthy donor group (n = 96). Analysis of sVE level after 4 weeks of treatment showed that a decrease in sVE level discriminates the responders vs. non-responders to sunitinib, but not bevacizumab. Conclusions These data highlight the interest for the sVE bioassay in future follow-up of cancer patients treated with targeted therapies such as tyrosine-kinase inhibitors.
Collapse
|
26
|
Serova M, Tijeras-Raballand A, Dos Santos C, Martinet M, Neuzillet C, Lopez A, Mitchell DC, Bryan BA, Gapihan G, Janin A, Bousquet G, Riveiro ME, Bieche I, Faivre S, Raymond E, de Gramont A. Everolimus affects vasculogenic mimicry in renal carcinoma resistant to sunitinib. Oncotarget 2018; 7:38467-86. [PMID: 27509260 PMCID: PMC5122404 DOI: 10.18632/oncotarget.9542] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 05/04/2016] [Indexed: 01/04/2023] Open
Abstract
Angiogenesis is hallmark of clear cell renal cell carcinogenesis. Anti-angiogenic therapies have been successful in improving disease outcome; however, most patients treated with anti-angiogenic agents will eventually progress. In this study we report that clear cell renal cell carcinoma was associated with vasculogenic mimicry in both mice and human with tumor cells expressing endothelial markers in the vicinity of tumor vessels. We show that vasculogenic mimicry was efficiently targeted by sunitinib but eventually associated with tumor resistance and a more aggressive phenotype both in vitro and in vivo. Re-challenging these resistant tumors in mice, we showed that second-line treatment with everolimus particularly affected vasculogenic mimicry and tumor cell differentiation compared to sorafenib and axitinib. Finally, our results highlighted the phenotypic and genotypic changes at the tumor cell and microenvironment levels during sunitinib response and progression and the subsequent improvement second-line therapies bring to the current renal cell carcinoma treatment paradigm.
Collapse
Affiliation(s)
- Maria Serova
- AAREC Filia Research, Boulogne-Billancourt, France.,Department of Medical Oncology, Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), Clichy, France.,INSERM, Paris France
| | - Annemilaï Tijeras-Raballand
- AAREC Filia Research, Boulogne-Billancourt, France.,Department of Medical Oncology, Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), Clichy, France.,INSERM, Paris France
| | - Celia Dos Santos
- AAREC Filia Research, Boulogne-Billancourt, France.,Department of Medical Oncology, Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), Clichy, France.,INSERM, Paris France
| | | | - Cindy Neuzillet
- Department of Medical Oncology, Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), Clichy, France.,INSERM, Paris France
| | - Alfred Lopez
- Department of Biomedical Sciences, Center of Emphasis in Cancer Research at the Paul Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| | - Dianne C Mitchell
- Department of Biomedical Sciences, Center of Emphasis in Cancer Research at the Paul Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| | - Brad A Bryan
- Department of Biomedical Sciences, Center of Emphasis in Cancer Research at the Paul Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| | - Guillaume Gapihan
- INSERM, Paris France.,Department of Pathology Saint Louis University Hospital (AP-HP - PRES Paris 7 Diderot), Paris France
| | - Anne Janin
- INSERM, Paris France.,Department of Pathology Saint Louis University Hospital (AP-HP - PRES Paris 7 Diderot), Paris France
| | - Guilhem Bousquet
- Department of Pathology Saint Louis University Hospital (AP-HP - PRES Paris 7 Diderot), Paris France.,Department of Medical Oncology, Avicenne University Hospital (APHP- PRES Paris 13 University), Bobigny, France
| | - Maria Eugenia Riveiro
- Department of Medical Oncology, Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), Clichy, France.,INSERM, Paris France
| | - Ivan Bieche
- Laboratory of Oncogenetics, Institut Curie, Hôpital René Huguenin, St-Cloud, France
| | - Sandrine Faivre
- Department of Medical Oncology, Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), Clichy, France.,INSERM, Paris France
| | - Eric Raymond
- Department of Medical Oncology, Beaujon University Hospital (AP-HP - PRES Paris 7 Diderot), Clichy, France.,INSERM, Paris France
| | - Armand de Gramont
- AAREC Filia Research, Boulogne-Billancourt, France.,INSERM, Paris France.,Department of Pathology Saint Louis University Hospital (AP-HP - PRES Paris 7 Diderot), Paris France
| |
Collapse
|
27
|
Zhang Y, Mai H, Guo G, Bi G, Hao G, Li Y, Wang X, Cheng L, Wang J, Dong R, Liu Z, Chen L, Qu H. Association analysis of SNPs present in plasma with adverse events and population pharmacokinetics in Chinese sunitinib treated patients with renal cell carcinoma. Oncotarget 2018; 9:14109-14123. [PMID: 29581831 PMCID: PMC5865657 DOI: 10.18632/oncotarget.23881] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 11/11/2017] [Indexed: 12/16/2022] Open
Abstract
Background Sunitinib is a tyrosine kinase inhibitor with effective therapeutic outcomes in patients with renal-cell carcinoma. The study were to analyze the association of single-nucleotide polymorphisms present in cell-free DNA and pharmacokinetics with sunitinib treatment-emergent adverse events in Chinese patients with renal-cell carcinoma. Materials and Methods We genotyped 8 keys SNPs in 6 candidate genes. The plasma concentrations of sunitinib and N-desethyl sunitinib were measured using a high performance liquid chromatography-tandam mass spectrometry method. Correlations between the single-nucleotide polymorphisms and adverse events were investigated by univariate and multivariate logistic regression and we quantitatively evaluated the effect of single-nucleotide polymorphisms on the pharmacokinetics of sunitinib by using a population PK model. Results Necessary dose reductions of sunitinib were significantly correlated with SNP rs1933437 in FLT3. A higher severity of AEs were collected with SNP rs2032582 in ABCB1 and rs1800812 in PDGFRα. Thrombocytopenia was collected with rs1800812 in PDGFRα. Our study provides a population PK model of sunitinib with the ABCB1 genotype as a predictive covariate for apparent oral clearance. Conclusions Our study preliminarily confirmed the hypothesis that the pharmacokinetics of sunitinib is affected by the SNPs of enzyme in Chinese renal-cell carcinoma patients, and this affects the different distribution and severity of adverse events of sunitinib.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Department of Clinical Pharmacology, Academy of Military Medical Sciences Affiliated Hospital, 307 Clinical College, Anhui Medical University, Beijing 100071, China
| | - Haixing Mai
- Department of Urology Department, Academy of Military Medical Sciences Affiliated Hospital, Beijing 100071, China
| | - Gang Guo
- Department of Urology Department, The General Hospital of the People's Liberation Army, Beijing 100853, China
| | - Guofang Bi
- Department of Clinical Pharmacology, Academy of Military Medical Sciences Affiliated Hospital, 307 Clinical College, Anhui Medical University, Beijing 100071, China
| | - Guangtao Hao
- Department of Clinical Pharmacology, Academy of Military Medical Sciences Affiliated Hospital, 307 Clinical College, Anhui Medical University, Beijing 100071, China
| | - Yuanyuan Li
- Department of Clinical Pharmacology, Academy of Military Medical Sciences Affiliated Hospital, 307 Clinical College, Anhui Medical University, Beijing 100071, China
| | - Xiaofang Wang
- Department of Clinical Pharmacology, Academy of Military Medical Sciences Affiliated Hospital, 307 Clinical College, Anhui Medical University, Beijing 100071, China
| | - Longmei Cheng
- Department of Clinical Pharmacology, Academy of Military Medical Sciences Affiliated Hospital, 307 Clinical College, Anhui Medical University, Beijing 100071, China
| | - Jing Wang
- Department of Clinical Pharmacology, Academy of Military Medical Sciences Affiliated Hospital, 307 Clinical College, Anhui Medical University, Beijing 100071, China
| | - Ruihua Dong
- Department of Clinical Pharmacology, Academy of Military Medical Sciences Affiliated Hospital, 307 Clinical College, Anhui Medical University, Beijing 100071, China
| | - Zeyuan Liu
- Department of Clinical Pharmacology, Academy of Military Medical Sciences Affiliated Hospital, 307 Clinical College, Anhui Medical University, Beijing 100071, China
| | - Lijun Chen
- Department of Urology Department, Academy of Military Medical Sciences Affiliated Hospital, Beijing 100071, China
| | - Hengyan Qu
- Department of Clinical Pharmacology, Academy of Military Medical Sciences Affiliated Hospital, 307 Clinical College, Anhui Medical University, Beijing 100071, China
| |
Collapse
|
28
|
Hidalgo M, Martinez-Garcia M, Le Tourneau C, Massard C, Garralda E, Boni V, Taus A, Albanell J, Sablin MP, Alt M, Bahleda R, Varga A, Boetsch C, Franjkovic I, Heil F, Lahr A, Lechner K, Morel A, Nayak T, Rossomanno S, Smart K, Stubenrauch K, Krieter O. First-in-Human Phase I Study of Single-agent Vanucizumab, A First-in-Class Bispecific Anti-Angiopoietin-2/Anti-VEGF-A Antibody, in Adult Patients with Advanced Solid Tumors. Clin Cancer Res 2017; 24:1536-1545. [PMID: 29217526 DOI: 10.1158/1078-0432.ccr-17-1588] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 10/23/2017] [Accepted: 11/30/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Vanucizumab is an investigational antiangiogenic, first-in-class, bispecific mAb targeting VEGF-A and angiopoietin-2 (Ang-2). This first-in-human study evaluated the safety, pharmacokinetics, pharmacodynamics, and antitumor activity of vanucizumab in adults with advanced solid tumors refractory to standard therapies.Experimental Design: Patients received escalating biweekly (3-30 mg/kg) or weekly (10-30 mg/kg) intravenous doses guided by a Bayesian logistic regression model with overdose control.Results: Forty-two patients were treated. One dose-limiting toxicity, a fatal pulmonary hemorrhage from a large centrally located mediastinal mass judged possibly related to vanucizumab, occurred with the 19 mg/kg biweekly dose. Arterial hypertension (59.5%), asthenia (42.9%), and headache (31%) were the most common toxicities. Seventeen (41%) patients experienced treatment-related grade ≥3 toxicities. Toxicity was generally higher with weekly than biweekly dosing. A MTD of vanucizumab was not reached in either schedule. Pharmacokinetics were dose-linear with an elimination half-life of 6-9 days. All patients had reduced plasma levels of free VEGF-A and Ang-2; most had reductions in KTRANS (measured by dynamic contrast-enhanced MRI). Two patients (renal cell and colon cancer) treated with 30 mg/kg achieved confirmed partial responses. Ten patients were without disease progression for ≥6 months. A flat-fixed 2,000 mg biweekly dose (phamacokinetically equivalent to 30 mg/kg biweekly) was recommended for further investigation.Conclusions: Biweekly vanucizumab had an acceptable safety and tolerability profile consistent with single-agent use of selective inhibitors of the VEGF-A and Ang/Tie2 pathway. Vanucizumab modulated its angiogenic targets, impacted tumor vascularity, and demonstrated encouraging antitumor activity in this heterogeneous population. Clin Cancer Res; 24(7); 1536-45. ©2017 AACR.
Collapse
Affiliation(s)
- Manuel Hidalgo
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain. .,START Madrid-CIOCC, HM Sanchinarro, Madrid, Spain
| | | | | | | | | | | | - Alvaro Taus
- Medical Oncology Department, Hospital del Mar., Barcelona, Spain
| | - Joan Albanell
- Medical Oncology Department, Hospital del Mar., Barcelona, Spain
| | - Marie-Paule Sablin
- Department of Medical Oncology, Institut Curie, Saint-Cloud and Paris, France
| | - Marie Alt
- Department of Medical Oncology, Institut Curie, Saint-Cloud and Paris, France
| | - Ratislav Bahleda
- Department of Drug Development, Gustave Roussy, Villejuif, France
| | - Andrea Varga
- Department of Drug Development, Gustave Roussy, Villejuif, France
| | | | | | - Florian Heil
- Roche Innovation Center Munich, Penzberg, Germany
| | | | | | | | - Tapan Nayak
- Roche Innovation Center Basel, Basel, Switzerland
| | | | - Kevin Smart
- Roche Innovation Center Welwyn, Welwyn Garden City, United Kingdom
| | | | | |
Collapse
|
29
|
Motzer RJ, Figlin RA, Martini JF, Hariharan S, Agarwal N, Li CX, Williams JA, Hutson TE. Germline Genetic Biomarkers of Sunitinib Efficacy in Advanced Renal Cell Carcinoma: Results From the RENAL EFFECT Trial. Clin Genitourin Cancer 2017; 15:526-533. [DOI: 10.1016/j.clgc.2017.02.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 02/10/2017] [Accepted: 02/19/2017] [Indexed: 10/20/2022]
|
30
|
Miao C, Cao J, Wang Y, Liu B, Wang Z. Effects of VEGF and VEGFR polymorphisms on the outcome of patients with metastatic renal cell carcinoma treated with sunitinib: a systematic review and meta-analysis. Oncotarget 2017; 8:68854-68862. [PMID: 28978162 PMCID: PMC5620302 DOI: 10.18632/oncotarget.19924] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 07/26/2017] [Indexed: 12/15/2022] Open
Abstract
To summarize and clarify the association between vascular endothelial growth factor (VEGF) and vascular endothelial growth factor receptor (VEGFR) polymorphisms and the outcome in patients with metastatic renal cell carcinoma (mRCC) treated with sunitinib. A total of 8 studies including 900 patients were analyzed in this systematic review after screening the database of PubMed, EMBASE and Web of Science. Hazard ratios (HRs) with 95% confidence interval (CI) were used to evaluate the strength of the association. VEGFR1 rs9582036 AA/AC carriers and rs9554320 CC/AC carriers had more favorable overall survival (OS) in patients with mRCC treated with sunitinib (n = 3), but not in progression-free survival (PFS). In addition, VEGFA rs2010963 was associated with poorer PFS of mRCC (n = 1). VEGFA rs699947 was significant in predicting PFS by univariate analysis, but showed no statistical significance in OS (n = 1). VEGFR2 rs1870377 was verified to be associated with sunitinib OS (n = 1). Furthermore, patients with VEGFR3 rs307826 and rs307821 had shorter PFS and OS during sunitinib therapy (n = 2, respectively). Our results suggested that VEGF and VEGFR polymorphisms were associated with outcomes in sunitinib treated mRCC patients, especially VEGFR1 polymorphisms. However, considering the limited study numbers, its clinical application in sunitinib treated mRCC still needs further confirmation.
Collapse
Affiliation(s)
- Chenkui Miao
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jingyi Cao
- Department of Urology, Xuzhou Cancer Hospital, Xuzhou, China
| | - Yuhao Wang
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Bianjiang Liu
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zengjun Wang
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
31
|
Saharinen P, Eklund L, Alitalo K. Therapeutic targeting of the angiopoietin-TIE pathway. Nat Rev Drug Discov 2017; 16:635-661. [PMID: 28529319 DOI: 10.1038/nrd.2016.278] [Citation(s) in RCA: 409] [Impact Index Per Article: 51.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The endothelial angiopoietin (ANG)-TIE growth factor receptor pathway regulates vascular permeability and pathological vascular remodelling during inflammation, tumour angiogenesis and metastasis. Drugs that target the ANG-TIE pathway are in clinical development for oncological and ophthalmological applications. The aim is to complement current vascular endothelial growth factor (VEGF)-based anti-angiogenic therapies in cancer, wet age-related macular degeneration and macular oedema. The unique function of the ANG-TIE pathway in vascular stabilization also renders this pathway an attractive target in sepsis, organ transplantation, atherosclerosis and vascular complications of diabetes. This Review covers key aspects of the function of the ANG-TIE pathway in vascular disease and describes the recent development of novel therapeutics that target this pathway.
Collapse
Affiliation(s)
- Pipsa Saharinen
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, P.O. Box 63, FI-00014 Helsinki, Finland
| | - Lauri Eklund
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, Aapistie 5A, University of Oulu, 90220 Oulu, Finland
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, P.O. Box 63, FI-00014 Helsinki, Finland
| |
Collapse
|
32
|
Serum level of ANGPTL4 as a potential biomarker in renal cell carcinoma. Urol Oncol 2017; 35:279-285. [PMID: 28110976 DOI: 10.1016/j.urolonc.2016.12.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 12/18/2016] [Accepted: 12/20/2016] [Indexed: 11/23/2022]
Abstract
OBJECTIVES This study aimed to determine the serum levels of angiopoietin-like 4 (ANGPTL4) in patients with renal cell carcinoma (RCC) and explore its potential as a biomarker. MATERIALS AND METHODS Blood samples were taken from 110 patients with RCC, 66 healthy controls, and patients with other solid tumors. Serum ANGPTL4 levels were measured using the enzyme-linked immunosorbent assay, and their correlation with clinical characteristics was further analyzed. Received operating characteristic (ROC) curves, Kaplan-Meier curves, and log-rank analyses were used to evaluate diagnostic and prognostic significance. RESULTS Serum ANGPTL4 levels were significantly higher in patients with RCC compared with healthy controls and patients with other types of cancers (P<0.0001) and associated with sex, Fuhrman grades, metastasis states, and tumor node metastasis stages (P<0.05), but not with age, tumor size, and histological types (P>0.05). The ROCs/area under the ROC curve analysis indicated an area under the ROC curve of 0.844 (sensitivity = 0.691; specificity = 0.939) and 0.725 (sensitivity = 0.909; specificity = 0.568), respectively, to distinguish patients with RCC from healthy controls and those with metastasis from those without metastasis. The survival analysis revealed that patients with low serum ANGPTL4 had longer progression-free survival compared with those with high serum ANGPTL4 (P = 0.033). CONCLUSION The present study suggested that the elevated serum ANGPTL4 level might be a novel diagnostic and prognostic biomarker for patients with RCC.
Collapse
|
33
|
Liu X, Swen JJ, Boven E, Castellano D, Gelderblom H, Mathijssen RH, Rodríguez-Antona C, García-Donas J, Rini BI, Guchelaar HJ. Meta-analysis on the association of VEGFR1 genetic variants with sunitinib outcome in metastatic renal cell carcinoma patients. Oncotarget 2017; 8:1204-1212. [PMID: 27901483 PMCID: PMC5352048 DOI: 10.18632/oncotarget.13597] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 11/07/2016] [Indexed: 11/25/2022] Open
Abstract
VEGFR1 rs9582036 and rs9554320 were previously reported the association with sunitinib progression-free survival (PFS) and overall survival (OS) in patients with metastatic renal cell carcinoma (mRCC). Hereafter, the association of both single nucleotide polymorphisms (SNPs) with PFS/OS was confirmed in two independent mRCC cohorts. The aim of the current study was to validate the associations of both SNPs with sunitinib outcome in three independent well-characterized cohorts (SUTOX, CCF and SOGUG) including 286 sunitinib-treated mRCC patients, as well as to perform a meta-analysis of current and published data combined. We found that rs9582036 and rs9554320 showed a significant association with sunitinib PFS in the CCF cohort (HR: 0.254, 95%CI: 0.092-0.703; P=0.008 and HR: 0.430, 95%CI: 0.200-0.927; P=0.031, respectively). Patients with the variant genotype of rs9582036 and rs9554320 had a shorter median PFS. No significant association of both SNPs with sunitinib PFS or OS was detected in either the SUTOX or SOGUG cohort. After the combination of all available data into a meta-analysis, the association of both SNPs with sunitinib PFS or OS did not achieve the threshold for statistical significance. Our findings suggest that, although VEGFR1 rs9582036 and rs9554320 are involved in sunitinib therapy outcome, its clinical use as biomarkers for prediction of sunitinib outcome in mRCC patients is limited, due to inconsistent findings when analyzing all existing studies together.
Collapse
Affiliation(s)
- Xiaoyan Liu
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
- Institute of Clinical Pharmacology, Qilu Hospital of Shandong University, Jinan, China
| | - Jesse J. Swen
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Epie Boven
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Daniel Castellano
- Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
- Spanish Oncology Genitourinary Group (SOGUG), Madrid, Spain
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ron H.J. Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Cristina Rodríguez-Antona
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- ISCIII Center for Biomedical Research on Rare Diseases (CIBERER), Madrid, Spain
| | - Jesus García-Donas
- Spanish Oncology Genitourinary Group (SOGUG), Madrid, Spain
- Oncology Unit, Clara Campal Comprehensive Cancer Center, Madrid, Spain
| | - Brian I. Rini
- Department of Solid Tumor Oncology, Cleveland Clinic Taussig Cancer Institute (CCF), Cleveland, Ohio, USA
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
34
|
Nassif E, Thibault C, Vano Y, Fournier L, Mauge L, Verkarre V, Timsit MO, Mejean A, Tartour E, Oudard S. Sunitinib in kidney cancer: 10 years of experience and development. Expert Rev Anticancer Ther 2016; 17:129-142. [PMID: 27967249 DOI: 10.1080/14737140.2017.1272415] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Sunitinib is a multi-target, anti-angiogenic tyrosine kinase inhibitor and a key molecule in the treatment of metastatic renal cell carcinoma (mRCC). Since it first demonstrated its efficacy ten years ago, overall survival of mRCC has more than doubled, in part due to sunitinib. In most recent years, progress has been made in the comprehension of its mechanism of action and resistance. Areas Covered: In this article, clinical trials involving sunitinib in kidney cancer have been reviewed, defining its different indications in metastatic and localized RCC. The rationale of sunitinib's efficacy, preclinical trials, past-clinical trials and ongoing clinical trials are summarized. Dose and scheme base are discussed, as the recommended dosage is frequently not well tolerated. Combination therapies appear to be toxic. Novel immunotherapies are changing the landscape of mRCC treatment and challenging sunitinib. Special attention has been paid towards cancer cell biology and immunity involved in treatment response. Expert Commentary: Sunitinib's place in the therapeutic arsenal is being redefined with the arrival of major challengers. Dosage and scheduling of sunitinib remains a major challenge.
Collapse
Affiliation(s)
- Elise Nassif
- a Oncology Department , Georges Pompidou European Hospital , Paris , France
| | - Constance Thibault
- a Oncology Department , Georges Pompidou European Hospital , Paris , France.,g Université Paris Descartes Sorbonne Paris-Cité , Paris 5 , France
| | - Yann Vano
- a Oncology Department , Georges Pompidou European Hospital , Paris , France.,b Cordeliers Research Center, UMRS1138 Team 13 Cancer, Immune Control and Escape , Paris , France .,g Université Paris Descartes Sorbonne Paris-Cité , Paris 5 , France
| | - Laure Fournier
- c Radiology Department , Georges Pompidou European Hospital , Paris , France.,g Université Paris Descartes Sorbonne Paris-Cité , Paris 5 , France
| | - Laetitia Mauge
- d Biological Hematology Department , Georges Pompidou European Hospital , Paris , France.,g Université Paris Descartes Sorbonne Paris-Cité , Paris 5 , France
| | - Virginie Verkarre
- d Biological Hematology Department , Georges Pompidou European Hospital , Paris , France.,g Université Paris Descartes Sorbonne Paris-Cité , Paris 5 , France
| | - Marc-Olivier Timsit
- e Urology Department , Georges Pompidou European Hospital , Paris , France.,g Université Paris Descartes Sorbonne Paris-Cité , Paris 5 , France
| | - Arnaud Mejean
- e Urology Department , Georges Pompidou European Hospital , Paris , France.,g Université Paris Descartes Sorbonne Paris-Cité , Paris 5 , France
| | - Eric Tartour
- f Immunology Department , Georges Pompidou European Hospital , Paris , France.,g Université Paris Descartes Sorbonne Paris-Cité , Paris 5 , France
| | - Stéphane Oudard
- a Oncology Department , Georges Pompidou European Hospital , Paris , France.,g Université Paris Descartes Sorbonne Paris-Cité , Paris 5 , France
| |
Collapse
|
35
|
Motzer RJ, Escudier B, Gannon A, Figlin RA. Sunitinib: Ten Years of Successful Clinical Use and Study in Advanced Renal Cell Carcinoma. Oncologist 2016; 22:41-52. [PMID: 27807302 DOI: 10.1634/theoncologist.2016-0197] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 08/03/2016] [Indexed: 01/07/2023] Open
Abstract
The oral multikinase inhibitor sunitinib malate was approved by the U.S. Food and Drug Administration in January 2006 for use in patients with advanced renal cell carcinoma (RCC). Since then, it has been approved globally for this indication and for patients with imatinib-resistant or -intolerant gastrointestinal stromal tumors and advanced pancreatic neuroendocrine tumors. As we mark the 10-year anniversary of the beginning of the era of targeted therapy, and specifically the approval of sunitinib, it is worthwhile to highlight the progress that has been made in advanced RCC as it relates to the study of sunitinib. We present the key trials and data for sunitinib that established it as a reference standard of care for first-line advanced RCC therapy and, along with other targeted agents, significantly altered the treatment landscape in RCC. Moreover, we discuss the research with sunitinib that has sought to refine its role via patient selection and prognostic markers, improve dosing and adverse event management, and identify predictive efficacy biomarkers, plus the extent to which this research has contributed to the overall understanding and management of RCC. We also explore the key learnings regarding study design and data interpretation from the sunitinib studies and how these findings and the sunitinib development program, in general, can be a model for successful development of other agents. Finally, ongoing research into the continued and future role of sunitinib in RCC management is discussed. THE ONCOLOGIST 2017;22:41-52 IMPLICATIONS FOR PRACTICE: Approved globally, sunitinib is established as a standard of care for first-line advanced renal cell carcinoma (RCC) therapy and, along with other targeted agents, has significantly altered the treatment landscape in RCC. Research with sunitinib that has sought to refine its role via patient selection and prognostic markers, improve dosing and adverse event management, and identify predictive efficacy biomarkers has contributed to the overall understanding and management of RCC. Key learnings regarding study design and data interpretation from the sunitinib studies and the sunitinib development program, in general, can be a model for the successful development of other agents.
Collapse
Affiliation(s)
- Robert J Motzer
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | | | - Robert A Figlin
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
36
|
Pompas-Veganzones N, Sandonis V, Perez-Lanzac A, Beltran M, Beardo P, Juárez A, Vazquez F, Cozar JM, Alvarez-Ossorio JL, Sanchez-Carbayo M. Myopodin methylation is a prognostic biomarker and predicts antiangiogenic response in advanced kidney cancer. Tumour Biol 2016; 37:14301-14310. [PMID: 27592258 DOI: 10.1007/s13277-016-5267-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 07/15/2016] [Indexed: 10/21/2022] Open
Abstract
Myopodin is a cytoskeleton protein that shuttles to the nucleus depending on the cellular differentiation and stress. It has shown tumor suppressor functions. Myopodin methylation status was useful for staging bladder and colon tumors and predicting clinical outcome. To our knowledge, myopodin has not been tested in kidney cancer to date. The purpose of this study was to evaluate whether myopodin methylation status could be clinically useful in renal cancer (1) as a prognostic biomarker and 2) as a predictive factor of response to antiangiogenic therapy in patients with metastatic disease. Methylation-specific polymerase chain reactions (MS-PCR) were used to evaluate myopodin methylation in 88 kidney tumors. These belonged to patients with localized disease and no evidence of disease during follow-up (n = 25) (group 1), and 63 patients under antiangiogenic therapy (sunitinib, sorafenib, pazopanib, and temsirolimus), from which group 2 had non-metastatic disease at diagnosis (n = 32), and group 3 showed metastatic disease at diagnosis (n = 31). Univariate and multivariate Cox analyses were utilized to assess outcome and response to antiangiogenic agents taking progression, disease-specific survival, and overall survival as clinical endpoints. Myopodin was methylated in 50 out of the 88 kidney tumors (56.8 %). Among the 88 cases analyzed, 10 of them recurred (11.4 %), 51 progressed (57.9 %), and 40 died of disease (45.4 %). Myopodin methylation status correlated to MSKCC Risk score (p = 0.050) and the presence of distant metastasis (p = 0.039). Taking all patients, an unmethylated myopodin identified patients with shorter progression-free survival, disease-specific survival, and overall survival. Using also in univariate and multivariate models, an unmethylated myopodin predicted response to antiangiogenic therapy (groups 2 and 3) using progression-free survival, disease-specific, and overall survival as clinical endpoints. Myopodin was revealed hypermethylated in kidney cancer. Myopodin methylation status identified which patients showed a more aggressive clinical behavior and predicted antiangiogenic response. These observations support the clinical utility of an unmethylated myopodin as a prognostic and predictive biomarker in kidney cancer.
Collapse
Affiliation(s)
- N Pompas-Veganzones
- Translational Oncology Laboratory, Ikerbasque Research Program, Medicine Department, University of the Basque Country, Vitoria-Gasteiz, Spain
| | - V Sandonis
- Translational Oncology Laboratory, Ikerbasque Research Program, Medicine Department, University of the Basque Country, Vitoria-Gasteiz, Spain
| | | | - M Beltran
- Pathology Department, Hospital Puerta Del Mar, Cadiz, Spain
| | - P Beardo
- Urology Department, Hospital De Jerez, Jerez, Spain
| | - A Juárez
- Urology Department, Hospital De Jerez, Jerez, Spain
| | - F Vazquez
- Urology Department, Hospital Virgen De Las Nieves, Granada, Spain
| | - J M Cozar
- Urology Department, Hospital Virgen De Las Nieves, Granada, Spain
| | | | - Marta Sanchez-Carbayo
- Translational Oncology Laboratory, Ikerbasque Research Program, Medicine Department, University of the Basque Country, Vitoria-Gasteiz, Spain.
| |
Collapse
|
37
|
Liontos M, Trigka EA, Korkolopoulou P, Tzannis K, Lainakis G, Koutsoukos K, Kostouros E, Lykka M, Papandreou CN, Karavasilis V, Christodoulou C, Papatsoris A, Skolarikos A, Varkarakis I, Adamakis I, Alamanis C, Stravodimos K, Mitropoulos D, Deliveliotis C, Constantinidis CA, Saetta A, Patsouris E, Dimopoulos MΑ, Bamias A. Expression and prognostic significance of VEGF and mTOR pathway proteins in metastatic renal cell carcinoma patients: a prognostic immunohistochemical profile for kidney cancer patients. World J Urol 2016; 35:411-419. [PMID: 27395374 DOI: 10.1007/s00345-016-1890-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 06/29/2016] [Indexed: 01/13/2023] Open
Abstract
PURPOSE To identify prognostic molecular profiles in patients with mRCC treated with sunitinib, we performed immunohistochemical analysis for VEGF and PI3K/Akt/mTOR pathway components. METHODS The immunohistochemical expression of VEGF, p85α, p110γ, PTEN, p-Akt, p-mTOR, p-4E-BP1 and p-p70S6K was studied in 79 patients with mRCC who received first-line treatment with sunitinib. Expression was correlated with clinicopathological features and survival. RESULTS VEGF was highly expressed (median H-Score 150), while positivity for the markers of the PI3K/Akt/mTOR pathway was: p85α 43/66 (65 %), p110γ41/60 (68 %), PTEN 32/64 (50 %), p-Akt57/63 (90 %), p-mTOR48/64 (75 %), p-4E-BP1 58/64 (90 %) and p-p70S6K 60/65 (92 %). No single immunohistochemical marker was found to have prognostic significance. Instead, the combination of increased p-mTOR and low VEGF expression was adversely correlated with overall survival (OS) (3.2 vs. 16.9 months, P = 0.001). CONCLUSION Immunohistochemistry for VEGF and p-mTOR proteins may discriminate patients refractory to first-line sunitinib with poor prognosis. Prospective validation of our findings is needed.
Collapse
Affiliation(s)
- Michalis Liontos
- Oncology Unit, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, 80 V.Sofias Ave, 11528, Athens, Greece.
| | - Eleni-Andriana Trigka
- 1st Department of Pathology, Laiko Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Penelope Korkolopoulou
- 1st Department of Pathology, Laiko Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Kimon Tzannis
- Oncology Unit, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, 80 V.Sofias Ave, 11528, Athens, Greece.,Hellenic GenitoUrinary Cancer Group, Athens, Greece
| | - Giorgos Lainakis
- Oncology Unit, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, 80 V.Sofias Ave, 11528, Athens, Greece
| | - Konstantinos Koutsoukos
- Oncology Unit, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, 80 V.Sofias Ave, 11528, Athens, Greece
| | - Efthymios Kostouros
- Oncology Unit, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, 80 V.Sofias Ave, 11528, Athens, Greece
| | - Maria Lykka
- Oncology Unit, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, 80 V.Sofias Ave, 11528, Athens, Greece
| | - Christos N Papandreou
- Department of Medical Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Vassilis Karavasilis
- Department of Medical Oncology, "Papageorgiou" Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Athanasios Papatsoris
- 2nd Department of Urology, Sismanoglio Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.,Hellenic GenitoUrinary Cancer Group, Athens, Greece
| | - Andreas Skolarikos
- 2nd Department of Urology, Sismanoglio Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.,Hellenic GenitoUrinary Cancer Group, Athens, Greece
| | - Ioannis Varkarakis
- 2nd Department of Urology, Sismanoglio Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.,Hellenic GenitoUrinary Cancer Group, Athens, Greece
| | - Ioannis Adamakis
- 1st Department of Urology, Laiko Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Christos Alamanis
- 1st Department of Urology, Laiko Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Stravodimos
- Hellenic GenitoUrinary Cancer Group, Athens, Greece.,1st Department of Urology, Laiko Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Dionysios Mitropoulos
- Hellenic GenitoUrinary Cancer Group, Athens, Greece.,1st Department of Urology, Laiko Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Charalambos Deliveliotis
- 2nd Department of Urology, Sismanoglio Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.,Hellenic GenitoUrinary Cancer Group, Athens, Greece
| | - Constantinos A Constantinidis
- Hellenic GenitoUrinary Cancer Group, Athens, Greece.,1st Department of Urology, Laiko Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Angelica Saetta
- 1st Department of Pathology, Laiko Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Efstratios Patsouris
- 1st Department of Pathology, Laiko Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Meletios Α Dimopoulos
- Oncology Unit, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, 80 V.Sofias Ave, 11528, Athens, Greece.,Hellenic GenitoUrinary Cancer Group, Athens, Greece
| | - Aristotelis Bamias
- Oncology Unit, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, 80 V.Sofias Ave, 11528, Athens, Greece.,Hellenic GenitoUrinary Cancer Group, Athens, Greece
| |
Collapse
|
38
|
Kummar S, Do K, Coyne GO, Chen A, Ji J, Rubinstein L, Doroshow JH. Establishing proof of mechanism: Assessing target modulation in early-phase clinical trials. Semin Oncol 2016; 43:446-52. [PMID: 27663476 DOI: 10.1053/j.seminoncol.2016.06.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Since modulation of the putative target and the observed anti-tumor effects form the basis for the clinical development of a molecularly targeted therapy, early-phase clinical trials should be designed to demonstrate proof-of-mechanism in tissues of interest. In addition to establishing safety and the maximum tolerated dose, first-in-human clinical trials should be designed to demonstrate target modulation, define the proposed mechanism of action, and evaluate pharmacokinetic-pharmacodynamic relationships of a new anti-cancer agent. Assessing target modulation in paired tumor biopsies in patients with solid tumors presents multiple challenges, including procedural issues such as patient safety, ethical considerations, and logistics of sample handling and processing. In addition, the availability of qualified biomarker assay technologies, resources to conduct such studies, and real-time analysis of samples to detect inter-species differences that may affect the determination of optimal sampling time points must be taken into account. This article provides a discussion of the challenges that confront the practical application of pharmacodynamic studies in early-phase clinical trials of anti-cancer agents.
Collapse
Affiliation(s)
- Shivaani Kummar
- National Cancer Institute, National Institutes of Health, Bethesda, MD.
| | - Khanh Do
- National Cancer Institute, National Institutes of Health, Bethesda, MD
| | | | - Alice Chen
- National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jiuping Ji
- Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Larry Rubinstein
- National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - James H Doroshow
- National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
39
|
Dornbusch J, Walter M, Gottschalk A, Obaje A, Junker K, Ohlmann CH, Meinhardt M, Zacharis A, Zastrow S, Schoffer O, Grimm MO, Klug SJ, Wirth MP, Fuessel S. Evaluation of polymorphisms in angiogenesis-related genes as predictive and prognostic markers for sunitinib-treated metastatic renal cell carcinoma patients. J Cancer Res Clin Oncol 2016; 142:1171-82. [PMID: 26935927 DOI: 10.1007/s00432-016-2137-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 02/18/2016] [Indexed: 12/31/2022]
Abstract
PURPOSE Single nucleotide polymorphisms (SNPs) in angiogenesis-associated genes might play an important role in activity of the tyrosine kinase inhibitor sunitinib and could affect survival of cancer patients treated with this drug. The aim of this retrospective study was to elucidate the role of 10 known SNPs in VEGFA, VEGFR1, VEGFR2 and VEGFR3 as potential prognostic and predictive markers in an independent cohort of patients with metastatic renal cell carcinoma (mRCC). METHODS DNA from 121 mRCC patients treated with sunitinib was used to analyze SNPs by TaqMan genotyping assays. Disease control rate was evaluated according to RECIST. Adverse effects of sunitinib were registered from medical records. The results of Cox and logistic regression were verified by correction for multiple testing. RESULTS Kaplan-Meier analysis revealed a reduced progression-free survival in patients with the wild-type (WT) allele of the VEGFA SNP rs699947 compared to variant alleles. Patients with the AA/AC-alleles of the VEGFR1 SNP rs9582036 had an improved median overall survival compared to those with the CC-WT allele what could be confirmed by multivariable Cox proportional hazard regression analyses. No statistically significant associations between the analyzed SNPs and higher risk for adverse effects were observed. CONCLUSIONS The results of this study suggest that most of the selected SNPs in angiogenesis-related genes are not associated with survival of mRCC patients after sunitinib therapy or with adverse effects. Only the VEGFR1 SNP rs9582036 showed a statistically significant association with overall survival. The potential of SNPs as prognostic and predictive markers for sunitinib-treated mRCC patients should be finally assessed by prospective studies.
Collapse
Affiliation(s)
- Juana Dornbusch
- Department of Urology, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Martina Walter
- Department of Urology, University Hospital of Jena, Lessingstr. 1, 07743, Jena, Germany
| | - Andrea Gottschalk
- Institute for Medical Informatics and Biometry, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Alice Obaje
- Department of Urology, University Hospital of Jena, Lessingstr. 1, 07743, Jena, Germany
| | - Kerstin Junker
- Department of Urology, Saarland University Medical Center, Kirrberger Str. 1, 66424, Homburg, Germany
| | - Carsten-Henning Ohlmann
- Department of Urology, Saarland University Medical Center, Kirrberger Str. 1, 66424, Homburg, Germany
| | - Matthias Meinhardt
- Institute of Pathology, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Aristeidis Zacharis
- Department of Urology, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Stefan Zastrow
- Department of Urology, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Olaf Schoffer
- Cancer Epidemiology, University Cancer Center Dresden, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Marc-Oliver Grimm
- Department of Urology, University Hospital of Jena, Lessingstr. 1, 07743, Jena, Germany
| | - Stefanie J Klug
- Cancer Epidemiology, University Cancer Center Dresden, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Manfred P Wirth
- Department of Urology, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Susanne Fuessel
- Department of Urology, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany.
| |
Collapse
|
40
|
Rodriguez-Vida A, Strijbos M, Hutson T. Predictive and prognostic biomarkers of targeted agents and modern immunotherapy in renal cell carcinoma. ESMO Open 2016; 1:e000013. [PMID: 27843601 PMCID: PMC5070260 DOI: 10.1136/esmoopen-2015-000013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 04/08/2016] [Indexed: 12/26/2022] Open
Abstract
In the past decade, several agents targeting angiogenesis and signal transduction pathways have replaced the use of cytokines as standard of care treatment for metastatic renal cell carcinoma (RCC) after showing improved clinical benefit and survival. Currently, several novel immunotherapy agents targeting the programmed cell death-1 (PD-1) and cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) pathways are being tested in metastatic RCC and are bound to revolutionise the management of this disease. However, the success of both antiangiogenic drugs and new immunotherapy agents still depends on our ability to select patients most likely to respond to treatment. This article will review the current available evidence on prognostic and predictive biomarkers of response to signal transduction pathways-targeted agents and modern immunotherapy in metastatic RCC.
Collapse
Affiliation(s)
- Alejo Rodriguez-Vida
- Department of Medical Oncology, IMIM Hospital del Mar Medical Research Institute, Barcelona, Spain
| | - Michiel Strijbos
- Department of Medical Oncology, AZ Klina, Brasschaat, Antwerp, Belgium
| | - Thomas Hutson
- Texas Oncology-Baylor Charles A Sammons Cancer Center, Dallas, Texas, USA
| |
Collapse
|
41
|
Rautiola J, Lampinen A, Mirtti T, Ristimäki A, Joensuu H, Bono P, Saharinen P. Association of Angiopoietin-2 and Ki-67 Expression with Vascular Density and Sunitinib Response in Metastatic Renal Cell Carcinoma. PLoS One 2016; 11:e0153745. [PMID: 27100185 PMCID: PMC4839598 DOI: 10.1371/journal.pone.0153745] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Accepted: 04/04/2016] [Indexed: 12/12/2022] Open
Abstract
The Angiopoietin-2 (Ang2, Angpt2) growth factor is a context-dependent antagonist/agonist ligand of the endothelial Tie2 receptor tyrosine kinase and known to promote tumour angiogenesis and metastasis. Angiopoietin antagonists have been tested in clinical cancer trials in combination with VEGF-based anti-angiogenic therapy, including sunitinib, which is widely used as a first-line therapy for metastatic renal cell carcinoma (mRCC). However, little is known about Ang2 protein expression in human tumours and the correlation of tumour Ang2 expression with tumour vascularization, tumour cell proliferation and response to anti-angiogenic therapies. Here, we evaluated, using immunohistochemistry, the expression of Ang2, CD31 and the cell proliferation marker Ki-67 in the primary kidney cancer from 136 mRCC patients, who received first-line sunitinib after nephrectomy. Ang2 protein expression was restrained to RCC tumour vessels, and correlated with tumour vascularization and response to sunitinib. High pre-therapeutic Ang2 expression, and more strongly, combined high expression of both Ang2 and CD31, were associated with a high clinical benefit rate (CBR). Low cancer Ki-67 expression, but not Ang2 or CD31 expression, was associated with favourable progression-free (PFS) and overall survival (OS) as compared to patients with high Ki-67 expression (PFS 6.5 vs. 10.6 months, P = 0.009; OS, 15.7 vs. 28.5 months, P = 0.015). In summary, in this study to investigate endothelial Ang2 in mRCC patients treated with first-line sunitinib, high cancer Ang2 expression was associated with the CBR, but not PFS or OS, whereas low Ki-67 expression was significantly associated with long PFS and OS.
Collapse
Affiliation(s)
- Juhana Rautiola
- Comprehensive Cancer Center, Helsinki University Hospital, P.O.B. 180, 00029 HUS, Finland and University of Helsinki, Finland
| | - Anita Lampinen
- Translational Cancer Biology Program, Research Programs Unit, and Department of Virology, Haartman Institute, Biomedicum Helsinki, Haartmaninkatu 8, P.O.B. 63, FI-00014, University of Helsinki, Finland
| | - Tuomas Mirtti
- Institute for Molecular Medicine Finland, Haartmaninkatu 8, P.O.B. 63, FI-00014, University of Helsinki, Finland.,Pathology, Research Programs Unit and HUSLAB, University of Helsinki and Helsinki University Hospital, P.O.B. 400, FI-00029, HUS, Helsinki, Finland
| | - Ari Ristimäki
- Pathology, Research Programs Unit and HUSLAB, University of Helsinki and Helsinki University Hospital, P.O.B. 400, FI-00029, HUS, Helsinki, Finland
| | - Heikki Joensuu
- Comprehensive Cancer Center, Helsinki University Hospital, P.O.B. 180, 00029 HUS, Finland and University of Helsinki, Finland
| | - Petri Bono
- Comprehensive Cancer Center, Helsinki University Hospital, P.O.B. 180, 00029 HUS, Finland and University of Helsinki, Finland
| | - Pipsa Saharinen
- Translational Cancer Biology Program, Research Programs Unit, and Department of Virology, Haartman Institute, Biomedicum Helsinki, Haartmaninkatu 8, P.O.B. 63, FI-00014, University of Helsinki, Finland.,Wihuri Research Institute, Biomedicum Helsinki, Haartmaninkatu 8, FI-00290, Helsinki, Finland
| |
Collapse
|
42
|
Voss MH, Chen D, Marker M, Hakimi AA, Lee CH, Hsieh JJ, Knox JJ, Voi M, Motzer RJ. Circulating biomarkers and outcome from a randomised phase II trial of sunitinib vs everolimus for patients with metastatic renal cell carcinoma. Br J Cancer 2016; 114:642-9. [PMID: 26908330 PMCID: PMC4800293 DOI: 10.1038/bjc.2016.21] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 12/22/2016] [Accepted: 01/05/2016] [Indexed: 01/19/2023] Open
Abstract
Background: RECORD-3 assessed non-inferiority of progression-free survival (PFS) with everolimus vs sunitinib in previously untreated patients with metastatic renal cell carcinoma. Baseline plasma sample collection and randomised design enabled correlation of circulating biomarkers with efficacy. Methods: Samples were analysed for 121 cancer-related biomarkers. Analyses of biomarkers categorised patients as high or low (vs median) to assess association with first-line PFS (PFS1L) for each treatment arm. A composite biomarker score (CBS) incorporated biomarkers potentially predictive of PFS1L with everolimus. Results: Plasma samples from 442 of the 471 randomised patients were analysed. Biomarkers were associated with PFS1L for everolimus alone (29), sunitinib alone (9) or both (12). Everolimus-specific biomarkers (CSF1, ICAM1, IL-18BP, KIM1, TNFRII) with hazard ratio ⩾1.8 were integrated into a CBS (range 0–5). For CBS low (0–3, n=291) vs high (4–5, n=151), PFS1L differed significantly for everolimus but not for sunitinib. There was no significant difference in PFS1L between everolimus and sunitinib in the high CBS patient cohort. Conclusions: Baseline levels of multiple soluble biomarkers correlated with benefit from everolimus and/or sunitinib, independent of clinical risk factors. A similar PFS1L was observed for both treatments among patients with high CBS score.
Collapse
Affiliation(s)
- Martin H Voss
- Memorial Sloan Kettering Cancer Center, 353 East 68th Street, New York, NY 10065, USA
| | - David Chen
- Novartis Oncology, One Health Plaza, East Hanover, NJ 07936-1080, USA
| | - Mahtab Marker
- Novartis Oncology, One Health Plaza, East Hanover, NJ 07936-1080, USA
| | - A Ari Hakimi
- Memorial Sloan Kettering Cancer Center, 353 East 68th Street, New York, NY 10065, USA
| | - Chung-Han Lee
- Memorial Sloan Kettering Cancer Center, 353 East 68th Street, New York, NY 10065, USA
| | - James J Hsieh
- Memorial Sloan Kettering Cancer Center, 353 East 68th Street, New York, NY 10065, USA
| | - Jennifer J Knox
- Princess Margaret Cancer Center, University of Toronto, 5-210, 610 University Avenue, Toronto, Ontario M5G-2M9, Canada
| | - Maurizio Voi
- Novartis Oncology, One Health Plaza, East Hanover, NJ 07936-1080, USA
| | - Robert J Motzer
- Memorial Sloan Kettering Cancer Center, 353 East 68th Street, New York, NY 10065, USA
| |
Collapse
|
43
|
Diekstra MHM, Swen JJ, Gelderblom H, Guchelaar HJ. A decade of pharmacogenomics research on tyrosine kinase inhibitors in metastatic renal cell cancer: a systematic review. Expert Rev Mol Diagn 2016; 16:605-18. [PMID: 26837796 DOI: 10.1586/14737159.2016.1148601] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE The individual response to targeted tyrosine kinase inhibitors (TKIs) in the treatment of metastatic renal cell cancer (mRCC) is highly variable. Outlined in this article are findings on potential biomarkers for TKI treatment outcome in mRCC and an evaluation of the status of clinical implementation. METHODS Articles were selected by two independent reviewers using a systematic search in five medical databases on renal cell carcinoma, TKIs, and pharmacogenetics. RESULTS Many researchers have focused on predictive biomarkers for treatment outcome of targeted therapies in mRCC patients. Attempts to explain differences in efficacy and toxicity of TKIs by use of genetic variants in genes related to the pharmacokinetics and pharmacodynamics of the drug have been successful. CONCLUSION Most findings on potential biomarkers have not been validated and therefore biomarker testing to guide choice of therapy and dose in mRCC is not yet feasible.
Collapse
Affiliation(s)
- Meta H M Diekstra
- a Department of Clinical Pharmacy and Toxicology , Leiden University Medical Center , Leiden , Netherlands
| | - Jesse J Swen
- a Department of Clinical Pharmacy and Toxicology , Leiden University Medical Center , Leiden , Netherlands
| | - Hans Gelderblom
- b Department of Medical Oncology , Leiden University Medical Center , Leiden , Netherlands
| | - Henk-Jan Guchelaar
- a Department of Clinical Pharmacy and Toxicology , Leiden University Medical Center , Leiden , Netherlands
| |
Collapse
|
44
|
Rabjerg M, Bjerregaard H, Halekoh U, Jensen BL, Walter S, Marcussen N. Molecular characterization of clear cell renal cell carcinoma identifies CSNK2A1, SPP1 and DEFB1 as promising novel prognostic markers. APMIS 2016; 124:372-83. [DOI: 10.1111/apm.12519] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 12/23/2015] [Indexed: 12/23/2022]
Affiliation(s)
- Maj Rabjerg
- Department of Clinical Pathology; Odense University Hospital; Odense Denmark
- Department of Urology; Odense University Hospital; Odense Denmark
| | | | - Ulrich Halekoh
- Department of Epidemiology, Biostatistics and Biodemography; Faculty of Health Sciences; University of Southern Denmark; Odense Denmark
| | - Boye L. Jensen
- Institute of Molecular Medicine, Cardiovascular and Renal Research; University of Southern Denmark; Odense Denmark
| | - Steen Walter
- Department of Urology; Odense University Hospital; Odense Denmark
| | - Niels Marcussen
- Department of Clinical Pathology; Odense University Hospital; Odense Denmark
- Department of Urology; Odense University Hospital; Odense Denmark
| |
Collapse
|
45
|
Carlisle B, Demko N, Freeman G, Hakala A, MacKinnon N, Ramsay T, Hey S, London AJ, Kimmelman J. Benefit, Risk, and Outcomes in Drug Development: A Systematic Review of Sunitinib. J Natl Cancer Inst 2016; 108:djv292. [PMID: 26547927 PMCID: PMC5943825 DOI: 10.1093/jnci/djv292] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 05/19/2015] [Accepted: 09/22/2015] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Little is known about the total patient burden associated with clinical development and where burdens fall most heavily during a drug development program. Our goal was to quantify the total patient burden/benefit in developing a new drug. METHODS We measured risk using drug-related adverse events that were grade 3 or higher, benefit by objective response rate, and trial outcomes by whether studies met their primary endpoint with acceptable safety. The differences in risk (death rate) and benefit (overall response rate) between industry and nonindustry trials were analyzed with an inverse-variance weighted fixed effects meta-analysis implemented as a weighted regression analysis. All statistical tests were two-sided. RESULTS We identified 103 primary publications of sunitinib monotherapy, representing 9092 patients and 3991 patient-years of involvement over 10 years and 32 different malignancies. In total, 1052 patients receiving sunitinib monotherapy experienced objective tumor response (15.7% of intent-to-treat population, 95% confidence interval [CI] = 15.3% to 16.0%), 98 died from drug-related toxicities (1.08%, 95% CI = 1.02% to 1.14%), and at least 1245 experienced grade 3-4 drug-related toxicities (13.7%, 95% CI = 13.3% to 14.1%). Risk/benefit worsened as the development program matured, with several instances of replicated negative studies and almost no positive trials after the first responding malignancies were discovered. CONCLUSIONS Even for a successful drug, the risk/benefit balance of trials was similar to phase I cancer trials in general. Sunitinib monotherapy development showed worsening risk/benefit, and the testing of new indications responded slowly to evidence that sunitinib monotherapy would not extend to new malignancies. Research decision-making should draw on evidence from whole research programs rather than a narrow band of studies in the same indication.
Collapse
Affiliation(s)
- Benjamin Carlisle
- Studies of Translation, Ethics and Medicine (STREAM), Biomedical Ethics Unit, McGill University, Montréal, QC, Canada (BC, ND, GF, AH, NM, SH, JK); University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada (TR); Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SH); Department of Philosophy and Center for Ethics and Policy, Carnegie Mellon University, Pittsburgh, PA (AJL)
| | - Nadine Demko
- Studies of Translation, Ethics and Medicine (STREAM), Biomedical Ethics Unit, McGill University, Montréal, QC, Canada (BC, ND, GF, AH, NM, SH, JK); University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada (TR); Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SH); Department of Philosophy and Center for Ethics and Policy, Carnegie Mellon University, Pittsburgh, PA (AJL)
| | - Georgina Freeman
- Studies of Translation, Ethics and Medicine (STREAM), Biomedical Ethics Unit, McGill University, Montréal, QC, Canada (BC, ND, GF, AH, NM, SH, JK); University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada (TR); Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SH); Department of Philosophy and Center for Ethics and Policy, Carnegie Mellon University, Pittsburgh, PA (AJL)
| | - Amanda Hakala
- Studies of Translation, Ethics and Medicine (STREAM), Biomedical Ethics Unit, McGill University, Montréal, QC, Canada (BC, ND, GF, AH, NM, SH, JK); University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada (TR); Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SH); Department of Philosophy and Center for Ethics and Policy, Carnegie Mellon University, Pittsburgh, PA (AJL)
| | - Nathalie MacKinnon
- Studies of Translation, Ethics and Medicine (STREAM), Biomedical Ethics Unit, McGill University, Montréal, QC, Canada (BC, ND, GF, AH, NM, SH, JK); University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada (TR); Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SH); Department of Philosophy and Center for Ethics and Policy, Carnegie Mellon University, Pittsburgh, PA (AJL)
| | - Tim Ramsay
- Studies of Translation, Ethics and Medicine (STREAM), Biomedical Ethics Unit, McGill University, Montréal, QC, Canada (BC, ND, GF, AH, NM, SH, JK); University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada (TR); Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SH); Department of Philosophy and Center for Ethics and Policy, Carnegie Mellon University, Pittsburgh, PA (AJL)
| | - Spencer Hey
- Studies of Translation, Ethics and Medicine (STREAM), Biomedical Ethics Unit, McGill University, Montréal, QC, Canada (BC, ND, GF, AH, NM, SH, JK); University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada (TR); Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SH); Department of Philosophy and Center for Ethics and Policy, Carnegie Mellon University, Pittsburgh, PA (AJL)
| | - Alex John London
- Studies of Translation, Ethics and Medicine (STREAM), Biomedical Ethics Unit, McGill University, Montréal, QC, Canada (BC, ND, GF, AH, NM, SH, JK); University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada (TR); Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SH); Department of Philosophy and Center for Ethics and Policy, Carnegie Mellon University, Pittsburgh, PA (AJL)
| | - Jonathan Kimmelman
- Studies of Translation, Ethics and Medicine (STREAM), Biomedical Ethics Unit, McGill University, Montréal, QC, Canada (BC, ND, GF, AH, NM, SH, JK); University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada (TR); Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SH); Department of Philosophy and Center for Ethics and Policy, Carnegie Mellon University, Pittsburgh, PA (AJL).
| |
Collapse
|
46
|
English PA, Williams JA, Martini JF, Motzer RJ, Valota O, Buller RE. A case for the use of receiver operating characteristic analysis of potential clinical efficacy biomarkers in advanced renal cell carcinoma. Future Oncol 2015; 12:175-82. [PMID: 26674983 PMCID: PMC5549778 DOI: 10.2217/fon.15.290] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
AIM Assess patient-level utility of suggested pretreatment biomarkers of sunitinib in advanced renal cell carcinoma. PATIENTS & METHODS Kaplan-Meier analysis of data from a randomized, Phase II study (n = 292) suggested baseline predictive value for circulating soluble Ang-2 and MMP-2 and HIF-1α percentage of tumor expression. Using this dataset, the sensitivity, specificity and area under the curve (AUC) were calculated, using receiver operating characteristic (ROC) curves. RESULTS Based on a ROC (sensitivity vs 1 - specificity) threshold AUC value of >0.8, neither Ang-2 (0.67) nor MMP-2 (0.65), nor HIF-1α percentage of tumor expression (0.65), performed appropriately from a patient-selection standpoint. CONCLUSION To properly assess potential biomarkers, sensitivity and specificity characteristics should be obtained by ROC analysis.
Collapse
Affiliation(s)
| | - J Andrew Williams
- Pfizer Oncology, 10646 Science Center Drive, San Diego, CA 92121, USA
| | | | - Robert J Motzer
- Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA
| | - Olga Valota
- Pfizer Oncology, via AM Mozzoni 12, Milan, 20152, Italy
| | - Richard E Buller
- Pfizer Oncology, 10646 Science Center Drive, San Diego, CA 92121, USA
| |
Collapse
|
47
|
Albini A, Bertolini F, Bassani B, Bruno A, Gallo C, Caraffi SG, Maramotti S, Noonan DM. Biomarkers of cancer angioprevention for clinical studies. Ecancermedicalscience 2015; 9:600. [PMID: 26635904 PMCID: PMC4664506 DOI: 10.3332/ecancer.2015.600] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Indexed: 12/24/2022] Open
Abstract
With the great advances made in the treatment and prevention of infectious diseases over the last century, chronic degenerative diseases-cardiovascular, cerebrovascular, and cancer-represent the major causes of death in the developed world. Although massive efforts and investments have been made in cancer therapy, the progress made towards reducing mortality has been more successful for cardiovascular disease than for tumours. This can be attributable largely to an active prevention approach implemented for cardiovascular disease. Cardiologists treat their patients before the overt disease becomes life threatening, performing early interventions in phenotypically healthy patients, by using several markers that predict risk. If the concept of prevention could be applied to cancer in a more extensive way, a significant number of tumours could be avoided through preventive measures. Prevention approaches range from avoiding tobacco exposure to dietary strategies to active pharmacological approaches in higher risk groups. Host targets rather than the tumour cells themselves are attractive for chemoprevention, in particular endothelial and immune cells. Angioprevention i.e. preventing cancer angiogenesis is a key concept that we introduced; yet one of the major current challenges for anti-angiogenesis in therapy and prevention is finding the right biomarkers. Here we discuss the importance of angioprevention and the potential use of VEGF, PlGF, CD31, Ang and Tie, circulating vascular cell precursors, and microRNA as potential biomarkers.
Collapse
Affiliation(s)
- Adriana Albini
- Laboratory of Translational Oncology, Department of Research and Statistics, IRCCS Arcispedale Santa Maria Nuova, Reggio Emilia 42123, Italy ; These authors share equal contribution
| | - Francesco Bertolini
- Laboratory of Haematology-Oncology, European Institute of Oncology, Milan 20141, Italy ; These authors share equal contribution
| | - Barbara Bassani
- Scientific and Technology Park, IRCCS MultiMedica, Milan 20138, Italy
| | - Antonino Bruno
- Scientific and Technology Park, IRCCS MultiMedica, Milan 20138, Italy
| | - Cristina Gallo
- Laboratory of Translational Oncology, Department of Research and Statistics, IRCCS Arcispedale Santa Maria Nuova, Reggio Emilia 42123, Italy
| | - Stefano Giuseppe Caraffi
- Laboratory of Translational Oncology, Department of Research and Statistics, IRCCS Arcispedale Santa Maria Nuova, Reggio Emilia 42123, Italy
| | - Sally Maramotti
- Laboratory of Translational Oncology, Department of Research and Statistics, IRCCS Arcispedale Santa Maria Nuova, Reggio Emilia 42123, Italy
| | - Douglas M Noonan
- Scientific and Technology Park, IRCCS MultiMedica, Milan 20138, Italy ; Department of Biotechnology and Life Sciences, University of Insubria, Varese 21100, Italy
| |
Collapse
|
48
|
Park I, Cho YM, Lee JL, Ahn JH, Lee DH. Prognostic tissue biomarker exploration for patients with metastatic renal cell carcinoma receiving vascular endothelial growth factor receptor tyrosine kinase inhibitors. Tumour Biol 2015; 37:4919-27. [PMID: 26526582 DOI: 10.1007/s13277-015-4339-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 10/28/2015] [Indexed: 01/04/2023] Open
Abstract
In metastatic renal cell carcinoma (mRCC), the prognostic role of several tumor tissue biomarkers has been evaluated, but the results were controversial. This study aims to verify the prognostic importance of selected tumor tissue biomarkers in patients with mRCC. The clinicopathological features, immunohistochemical staining and scoring for select tissue biomarkers, treatment, and outcome of patients with mRCC treated with vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors (TKIs) between July 2006 and March 2011 at Asan Medical Center in Seoul, South Korea, were reviewed. In total, 123 patients met the inclusion criteria. Most patients had clear-cell carcinoma (107 patients, 87.0 %). First-line VEGFR TKIs were sunitinib (97 patients, 78.9 %), sorafenib (23 patients, 18.7 %), and pazopanib (3 patients, 2.4 %). With a median follow-up period of 60.0 months (95 % confidence interval (CI), 56.3-63.6), median overall survival (OS) and progression-free survival (PFS) were 25.6 months (95 % CI, 19.2-32.0) and 12.2 months (95 % CI, 8.1-16.3), respectively. In the multivariable analysis for OS, carbonic anhydrase IX (CAIX; 47.5 % or less vs. more than 47.5 %, p = 0.014), sarcomatoid change (40 % or less vs. more than 40 %, p < 0.001), tumor necrosis (20 % or less vs. more than 20 %, p = 0.006), and Heng's risk group (good vs. intermediate vs. poor, p = 0.011) were identified as independent prognostic factors. In the multivariable analysis for PFS, CAIX (p < 0.001), phosphatase and tensin homolog (PTEN; 45 % or less vs. more than 45 %, p = 0.004), sarcomatoid change (p = 0.002), and tumor necrosis (p = 0.001) were identified as independent factors affecting PFS. CAIX and PTEN had prognostic importance for mRCC patients receiving first-line VEGFR TKI. Future validation and mechanistic studies are required.
Collapse
Affiliation(s)
- Inkeun Park
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 138-736, Republic of Korea.,Department of Internal Medicine, Gachon University Gil Medical Centre, 1198, Guwol-Dong, Namdong-Gu, Incheon, 405-760, Republic of Korea
| | - Yong Mee Cho
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jae-Lyun Lee
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 138-736, Republic of Korea.
| | - Jin-Hee Ahn
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 138-736, Republic of Korea
| | - Dae-Ho Lee
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 138-736, Republic of Korea
| |
Collapse
|
49
|
Winer AG, Motzer RJ, Hakimi AA. Prognostic Biomarkers for Response to Vascular Endothelial Growth Factor-Targeted Therapy for Renal Cell Carcinoma. Urol Clin North Am 2015; 43:95-104. [PMID: 26614032 DOI: 10.1016/j.ucl.2015.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Improved understanding of renal carcinoma disease biology has led to the discovery and approval of five novel therapies targeting specific molecules in the vascular endothelial growth factor (VEGF) biochemical pathway. Biomarker studies attempting to predict response to VEGF-targeted therapies have largely focused on circulating proteins, tissue-based molecules, and germline polymorphisms. Thus far studies have yielded conflicting results that require prospective validation; therefore no definitive biomarker has yet been integrated into the clinician's armamentarium. However, early analyses featuring genomic biomarkers have generated promising findings. This article provides an overview of available biomarkers evaluated with respect to VEGF-targeted therapies in patients with advanced renal cell carcinoma.
Collapse
Affiliation(s)
- Andrew G Winer
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 353 East 68th Street, New York, NY 10065, USA
| | - Robert J Motzer
- Genitourinary Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065, USA
| | - A Ari Hakimi
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 353 East 68th Street, New York, NY 10065, USA.
| |
Collapse
|
50
|
Arnason T, Harkness T. Development, Maintenance, and Reversal of Multiple Drug Resistance: At the Crossroads of TFPI1, ABC Transporters, and HIF1. Cancers (Basel) 2015; 7:2063-82. [PMID: 26501324 PMCID: PMC4695877 DOI: 10.3390/cancers7040877] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 10/10/2015] [Indexed: 12/21/2022] Open
Abstract
Early detection and improved therapies for many cancers are enhancing survival rates. Although many cytotoxic therapies are approved for aggressive or metastatic cancer; response rates are low and acquisition of de novo resistance is virtually universal. For decades; chemotherapeutic treatments for cancer have included anthracyclines such as Doxorubicin (DOX); and its use in aggressive tumors appears to remain a viable option; but drug resistance arises against DOX; as for all other classes of compounds. Our recent work suggests the anticoagulant protein Tissue Factor Pathway Inhibitor 1α (TFPI1α) plays a role in driving the development of multiple drug resistance (MDR); but not maintenance; of the MDR state. Other factors; such as the ABC transporter drug efflux pumps MDR-1/P-gp (ABCB1) and BCRP (ABCG2); are required for MDR maintenance; as well as development. The patient population struggling with therapeutic resistance specifically requires novel treatment options to resensitize these tumor cells to therapy. In this review we discuss the development, maintenance, and reversal of MDR as three distinct phases of cancer biology. Possible means to exploit these stages to reverse MDR will be explored. Early molecular detection of MDR cancers before clinical failure has the potential to offer new approaches to fighting MDR cancer.
Collapse
Affiliation(s)
- Terra Arnason
- Department of Medicine, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada
- Correspondence: ; Tel.:+1-306-844-1119; Fax: +1-306-844-1512
| | - Troy Harkness
- Department of Anatomy and Cell Biology, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada;
| |
Collapse
|