1
|
Bai Y, You Y, Chen D, Chen Y, Yin Z, Liao S, You B, Lu D, Sun Y, Wu L, Wu Y. Amiloride reduces fructosamine-3-kinase expression to restore sunitinib sensitivity in renal cell carcinoma. iScience 2024; 27:109997. [PMID: 38868177 PMCID: PMC11166694 DOI: 10.1016/j.isci.2024.109997] [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: 10/09/2023] [Revised: 04/10/2024] [Accepted: 05/14/2024] [Indexed: 06/14/2024] Open
Abstract
The kidney is a vital organ responsible for water and sodium metabolism, while the primary function of amiloride is to promote the excretion of water and sodium. We investigated amiloride enhanced the sunitinib sensitivity in RCC. We found both sunitinib and amiloride displayed cytotoxicity and exerted the synergy effect in RCC cells in vivo and in vitro arrays. Protein expression profiles were screened via MS/TMT, revealing that FN3K was upregulated in the sunitinib group, and rescued in amiloride and the combination administration. Exogenous FN3K could promote proliferation, invasion and metastasis and decrease the sensitivity of Caki-1 cells to sunitinib, also, exogenous FN3K up-regulated VEGFR2 expression and activated AKT/mTOR signal pathway. More FN3K and VEGFR2 accumulated in R-Caki-1 cells and rescued by amiloride treatment. Co-IP and IF confirmed the interaction between FN3K and VEGFR2. In conclusion, FN3K depletion mediated VEGFR2 disruption promotes amiloride synergized the anti-RCC activity of sunitinib.
Collapse
Affiliation(s)
- Yuanyuan Bai
- Department of Urology, Affiliated Sanming First Hospital, Fujian Medical University, Sanming, Fujian 365100, P.R. China
| | - Yiqing You
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Daoxun Chen
- Department of Urology, Affiliated Sanming First Hospital, Fujian Medical University, Sanming, Fujian 365100, P.R. China
| | - Yongmei Chen
- Department of Urology, Affiliated Sanming First Hospital, Fujian Medical University, Sanming, Fujian 365100, P.R. China
| | - Zhenjie Yin
- Department of Urology, Affiliated Sanming First Hospital, Fujian Medical University, Sanming, Fujian 365100, P.R. China
| | - Shangfan Liao
- Department of Urology, Affiliated Sanming First Hospital, Fujian Medical University, Sanming, Fujian 365100, P.R. China
| | - Bingyong You
- Department of Urology, Affiliated Sanming First Hospital, Fujian Medical University, Sanming, Fujian 365100, P.R. China
| | - Dongming Lu
- Department of Urology, Affiliated Sanming First Hospital, Fujian Medical University, Sanming, Fujian 365100, P.R. China
| | - Yingming Sun
- Department of Oncology, Affiliated Sanming First Hospital, Fujian Medical University, Sanming, Fujian 365100, P.R. China
| | - Lixian Wu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou, China
| | - Yongyang Wu
- Department of Urology, Affiliated Sanming First Hospital, Fujian Medical University, Sanming, Fujian 365100, P.R. China
| |
Collapse
|
2
|
Hong SH, Hwang HJ, Son DH, Kim ES, Park SY, Yoon YE. Inhibition of EZH2 exerts antitumorigenic effects in renal cell carcinoma via LATS1. FEBS Open Bio 2023; 13:724-735. [PMID: 36808829 PMCID: PMC10068324 DOI: 10.1002/2211-5463.13579] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 02/01/2023] [Accepted: 02/20/2023] [Indexed: 02/23/2023] Open
Abstract
The most common type of kidney cancer in adults is renal cell carcinoma (RCC), which accounts for approximately 90% of cases. RCC is a variant disease with numerous subtypes; the most common subtype is clear cell RCC (ccRCC, 75%), followed by papillary RCC (pRCC, 10%) and chromophobe RCC (chRCC, 5%). To identify a genetic target for all subtypes, we analyzed The Cancer Genome Atlas (TCGA) databases of ccRCC, pRCC, and chromophobe RCC. Enhancer of zeste homolog 2 (EZH2), which encodes a methyltransferase, was observed to be significantly upregulated in tumors. The EZH2 inhibitor tazemetostat induced anticancer effects in RCC cells. TCGA analysis revealed that large tumor suppressor kinase 1 (LATS1), a key tumor suppressor of the Hippo pathway, was significantly downregulated in tumors; the expression of LATS1 was increased by tazemetostat. Through additional experiments, we confirmed that LATS1 plays a crucial role in EZH2 inhibition and has a negative association with EZH2. Therefore, we suggest that epigenetic control could be a novel therapeutic strategy for three subtypes of RCC.
Collapse
Affiliation(s)
- Seong Hwi Hong
- Department of Urology, Hanyang University College of Medicine, Seoul, Korea
| | - Hyun Ji Hwang
- Department of Urology, Hanyang University College of Medicine, Seoul, Korea
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul, Korea
| | - Da Hyeon Son
- Department of Urology, Hanyang University College of Medicine, Seoul, Korea
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul, Korea
| | - Eun Song Kim
- Department of Urology, Hanyang University College of Medicine, Seoul, Korea
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul, Korea
| | - Sung Yul Park
- Department of Urology, Hanyang University College of Medicine, Seoul, Korea
| | - Young Eun Yoon
- Department of Urology, Hanyang University College of Medicine, Seoul, Korea
| |
Collapse
|
3
|
Ding H, Wang G, Yu Z, Sun H, Wang L. Role of interferon-gamma (IFN-γ) and IFN-γ receptor 1/2 (IFNγR1/2) in regulation of immunity, infection, and cancer development: IFN-γ-dependent or independent pathway. Biomed Pharmacother 2022; 155:113683. [PMID: 36095965 DOI: 10.1016/j.biopha.2022.113683] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/27/2022] [Accepted: 09/07/2022] [Indexed: 11/02/2022] Open
Abstract
IFN-γ, a soluble cytokine being produced by T lymphocytes, macrophages, mucosal epithelial cells, or natural killer cells, is able to bind to the IFN-γ receptor (IFNγR) and in turn activate the Janus kinase (JAK)-signal transducer and transcription protein (STAT) pathway and induce expression of IFN-γ-stimulated genes. IFN-γ is critical for innate and adaptive immunity and aberrant IFN-γ expression and functions have been associated with different human diseases. However, the IFN-γ/IFNγR signaling could be a double-edged sword in cancer development because the tissue microenvironments could determine its anti- or pro-tumorigenic activities. The IFNγR protein consists of two IFNγR1 and IFNγR2 chains, subunits of which play different roles under certain conditions. This review assessed IFNγR polymorphisms, expression and functions in development and progression of various human diseases in an IFN-γ-dependent or independent manner. This review also discussed tumor microenvironment, microbial infection, and vital molecules in the IFN-γ upstream signaling that might regulate IFNγR expression, drug resistance, and druggable strategy, to provide evidence for further application of IFNγR.
Collapse
Affiliation(s)
- Huihui Ding
- School of Pharmacy, Shandong First Medical University, Jinan, Shandong, China.
| | - Gongfu Wang
- Center for Drug Evaluation, China Food and Drug Administration (CFDA), Beijing, China.
| | - Zhen Yu
- Department of Pharmacy, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| | - Huimin Sun
- School of Pharmacy, Shandong First Medical University, Jinan, Shandong, China.
| | - Lu Wang
- School of Pharmacy, Shandong First Medical University, Jinan, Shandong, China; Department of Pharmacy, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
4
|
Characterization and function of biomarkers in sunitinib-resistant renal carcinoma cells. Gene 2022; 832:146514. [PMID: 35550407 DOI: 10.1016/j.gene.2022.146514] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 03/21/2022] [Accepted: 04/14/2022] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Sunitinib is a first-line drug in the treatment of metastatic renal cell carcinoma, but patients will inevitably develop drug resistance after 6-15 months of systematic treatment, which seriously affects the prognosis in KIRC. METHODS During the study, the Gene Expression Omnibus (GEO) database was used to perform a systematic bioinformatics analysis,so that we could determine the genes (DEGs) which are differentially expressed between sunitinib-sensitive and sunitinib-resistant RCC (SRRC) cells. RESULTS A total of 31 DEGs were identified. Gene ontology (GO) was used to analyze the function of DEGS. These DEGs were found mainly enriched in organic aniontransmembrane transporter. The Cytohubba plug-in, STRING database and Cytoscape software were involved to construct a protein-protein interaction (PPI) network, and the pivot genes were identified by single-gene and multi-gene Cox regression analysis. Finally, DDX58 and MX2 were identified as prognostic genes. Survival analysis was performed by using prognostic nomogram, prognostic histogram and GEPIA database to verify the relationship between DDX58 and MX2 expression and survival. The relationship between the two pivot genes and the prognosis of patients was further verified by using the KM survival analyses and Time Dependency ROC curve analyses from TCGA database. Immunohistochemical analyses confirmed that, in tumor tissues and normal tissues, DDX58 and MX2 were differentially expressed. The expression of these two genes have relationship with the immune checkpoint. CONCLUSIONS This study provides insights into the molecular mechanisms of SRRC, as well as the selection of therapeutic and prognostic biomarkers for SRRC.
Collapse
|
5
|
Denize T, Farah S, Cimadamore A, Flaifel A, Walton E, Sticco-Ivins MA, Labaki C, Braun DA, Sun M, Wang E, Xie W, Choueiri TK, Signoretti S. Biomarkers of Angiogenesis and Clinical Outcomes to Cabozantinib and Everolimus in Patients with Metastatic Renal Cell Carcinoma from the Phase III METEOR Trial. Clin Cancer Res 2021; 28:748-755. [DOI: 10.1158/1078-0432.ccr-21-3088] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/03/2021] [Accepted: 12/13/2021] [Indexed: 11/16/2022]
|
6
|
Feasibility Study on Using Dynamic Contrast Enhanced MRI to Assess the Effect of Tyrosine Kinase Inhibitor Therapy within the STAR Trial of Metastatic Renal Cell Cancer. Diagnostics (Basel) 2021; 11:diagnostics11071302. [PMID: 34359384 PMCID: PMC8306403 DOI: 10.3390/diagnostics11071302] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/06/2021] [Accepted: 07/16/2021] [Indexed: 01/04/2023] Open
Abstract
Objective: To identify dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) parameters predictive of early disease progression in patients with metastatic renal cell cancer (mRCC) treated with anti-angiogenic tyrosine kinase inhibitors (TKI). Methods: The study was linked to a phase II/III randomised control trial. Patients underwent DCE-MRI before, at 4- and 10-weeks after initiation of TKI. DCE-MRI parameters at each time-point were derived from a single-compartment tracer kinetic model, following semi-automated tumour segmentation by two independent readers. Primary endpoint was correlation of DCE-MRI parameters with disease progression at 6-months. Receiver operating characteristic (ROC) curve analysis and area under the curve (AUC) values were calculated for parameters associated with disease progression at 6 months. Inter-observer agreement was assessed using the intraclass correlation coefficient (ICC). Results: 23 tumours in 14 patients were measurable. Three patients had disease progression at 6 months. The percentage (%) change in perfused tumour volume between baseline and 4-week DCE-MRI (p = 0.016), mean transfer constant Ktrans change (p = 0.038), and % change in extracellular volume (p = 0.009) between 4- and 10-week MRI, correlated with early disease progression (AUC 0.879 for each parameter). Inter-observer agreement was excellent for perfused tumour volume, Ktrans and extracellular volume (ICC: 0.928, 0.949, 0.910 respectively). Conclusions: Early measurement of DCE-MRI biomarkers of tumour perfusion at 4- and 10-weeks predicts disease progression at 6-months following TKI therapy in mRCC.
Collapse
|
7
|
Khanna P, Soh HJ, Chen CH, Saxena R, Amin S, Naughton M, Joslin PN, Moore A, Bakouny Z, O'Callaghan C, Catalano P, Signoretti S, McKay R, Choueiri TK, Bhasin M, Walther T, Bhatt RS. ACE2 abrogates tumor resistance to VEGFR inhibitors suggesting angiotensin-(1-7) as a therapy for clear cell renal cell carcinoma. Sci Transl Med 2021; 13:13/577/eabc0170. [PMID: 33472951 DOI: 10.1126/scitranslmed.abc0170] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 09/24/2020] [Indexed: 12/24/2022]
Abstract
Angiotensin converting enzyme 2 (ACE2) is an enzyme that belongs to the renin-angiotensin system (RAS) and antagonizes the classical angiotensin (Ang) II/angiotensin II receptor type 1 (AT1) receptor pathway. Here, we report that higher ACE2 expression correlates with better overall survival in patients with clear cell renal cell carcinoma (ccRCC). Moreover, ACE2 has inhibitory effects on tumor proliferation in ccRCC in vitro and in preclinical animal models of ccRCC. We further show that Ang-(1-7), a heptapeptide generated by ACE2, is the likely mediator of this effect. Vascular endothelial growth factor receptor-tyrosine kinase inhibitor (VEGFR-TKI) treatment of ccRCC xenografts decreased ACE2 expression, and combination treatment with VEGFR-TKI and Ang-(1-7) generated additive suppression of tumor growth and improved survival outcomes. Last, the addition of Ang-(1-7) to programmed death-ligand 1 (PD-L1) pathway inhibitor and VEGFR-TKI showed further growth suppression in an immunocompetent RCC model. Together, these results suggest that targeting the ACE2/Ang-(1-7) axis is a promising therapeutic strategy against ccRCC.
Collapse
Affiliation(s)
- Prateek Khanna
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Department of Medicine, Mount Auburn Hospital, Harvard Medical School, Cambridge, MA 02138, USA
| | - Hong Jie Soh
- Department of Pharmacology and Therapeutics, School of Medicine and School of Pharmacy, University College Cork, Cork T12 K8AF, Ireland
| | - Chun-Hau Chen
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ruchi Saxena
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Seema Amin
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Maura Naughton
- Department of Pharmacology and Therapeutics, School of Medicine and School of Pharmacy, University College Cork, Cork T12 K8AF, Ireland
| | - Patrick Neset Joslin
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Andrew Moore
- Department of Pharmacology and Therapeutics, School of Medicine and School of Pharmacy, University College Cork, Cork T12 K8AF, Ireland
| | - Ziad Bakouny
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Carol O'Callaghan
- Department of Pharmacology and Therapeutics, School of Medicine and School of Pharmacy, University College Cork, Cork T12 K8AF, Ireland
| | - Paul Catalano
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Rana McKay
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Toni K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Manoj Bhasin
- Division of Interdisciplinary Medicine and Biotechnology and Genomics, Proteomics, Bioinformatics and Systems Biology Center, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Thomas Walther
- Department of Pharmacology and Therapeutics, School of Medicine and School of Pharmacy, University College Cork, Cork T12 K8AF, Ireland. .,Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald 17489, Germany
| | - Rupal S Bhatt
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
8
|
Marchetti A, Rosellini M, Mollica V, Rizzo A, Tassinari E, Nuvola G, Cimadamore A, Santoni M, Fiorentino M, Montironi R, Massari F. The Molecular Characteristics of Non-Clear Cell Renal Cell Carcinoma: What's the Story Morning Glory? Int J Mol Sci 2021; 22:6237. [PMID: 34207825 PMCID: PMC8226484 DOI: 10.3390/ijms22126237] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/06/2021] [Accepted: 06/08/2021] [Indexed: 01/05/2023] Open
Abstract
Non-clear cell renal cell carcinomas are a miscellaneous group of tumors that include different histological subtypes, each one characterized by peculiarity in terms of genetic alteration, clinical behavior, prognosis, and treatment response. Because of their low incidence and poor enrollment in clinical trials, alongside their heterogeneity, additional efforts are required to better unveil the pathogenetic mechanisms and, consequently, to improve the treatment algorithm. Nowadays, tyrosine kinase inhibitors, mTOR and MET inhibitors, and even cisplatin-based chemotherapy and immunotherapy are potential weapons that are still under evaluation in this setting. Various biomarkers have been evaluated for detecting progression and monitoring renal cell carcinoma, but more studies are necessary to improve this field. In this review, we provide an overview on the molecular characteristics of this group of tumors and the recently published trials, giving an insight into what might become the future therapeutic standard in this complex world of non-clear cell kidney cancers.
Collapse
Affiliation(s)
- Andrea Marchetti
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni—15, 40138 Bologna, Italy; (A.M.); (M.R.); (V.M.); (A.R.); (E.T.); (G.N.)
| | - Matteo Rosellini
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni—15, 40138 Bologna, Italy; (A.M.); (M.R.); (V.M.); (A.R.); (E.T.); (G.N.)
| | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni—15, 40138 Bologna, Italy; (A.M.); (M.R.); (V.M.); (A.R.); (E.T.); (G.N.)
| | - Alessandro Rizzo
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni—15, 40138 Bologna, Italy; (A.M.); (M.R.); (V.M.); (A.R.); (E.T.); (G.N.)
| | - Elisa Tassinari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni—15, 40138 Bologna, Italy; (A.M.); (M.R.); (V.M.); (A.R.); (E.T.); (G.N.)
| | - Giacomo Nuvola
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni—15, 40138 Bologna, Italy; (A.M.); (M.R.); (V.M.); (A.R.); (E.T.); (G.N.)
| | - Alessia Cimadamore
- Section of Pathological Anatomy, School of Medicine, Polytechnic University of the Marche Region, United Hospitals, 60126 Ancona, Italy; (A.C.); (R.M.)
| | - Matteo Santoni
- Oncology Unit, Macerata Hospital, 62100 Macerata, Italy;
| | - Michelangelo Fiorentino
- Department of Specialistic Diagnostic and Experimental Medicine, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy;
| | - Rodolfo Montironi
- Section of Pathological Anatomy, School of Medicine, Polytechnic University of the Marche Region, United Hospitals, 60126 Ancona, Italy; (A.C.); (R.M.)
| | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni—15, 40138 Bologna, Italy; (A.M.); (M.R.); (V.M.); (A.R.); (E.T.); (G.N.)
| |
Collapse
|
9
|
Bond KH, Fetting JL, Lary CW, Emery IF, Oxburgh L. FOXD1 regulates cell division in clear cell renal cell carcinoma. BMC Cancer 2021; 21:312. [PMID: 33761914 PMCID: PMC7988646 DOI: 10.1186/s12885-021-07957-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 02/23/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Forkhead transcription factors control cell growth in multiple cancer types. Foxd1 is essential for kidney development and mitochondrial metabolism, but its significance in renal cell carcinoma (ccRCC) has not been reported. METHODS Transcriptome data from the TCGA database was used to correlate FOXD1 expression with patient survival. FOXD1 was knocked out in the 786-O cell line and known targets were analyzed. Reduced cell growth was observed and investigated in vitro using growth rate and Seahorse XF metabolic assays and in vivo using a xenograft model. Cell cycle characteristics were determined by flow cytometry and immunoblotting. Immunostaining for TUNEL and γH2AX was used to measure DNA damage. Association of the FOXD1 pathway with cell cycle progression was investigated through correlation analysis using the TCGA database. RESULTS FOXD1 expression level in ccRCC correlated inversely with patient survival. Knockout of FOXD1 in 786-O cells altered expression of FOXD1 targets, particularly genes involved in metabolism (MICU1) and cell cycle progression. Investigation of metabolic state revealed significant alterations in mitochondrial metabolism and glycolysis, but no net change in energy production. In vitro growth rate assays showed a significant reduction in growth of 786-OFOXD1null. In vivo, xenografted 786-OFOXD1null showed reduced capacity for tumor formation and reduced tumor size. Cell cycle analysis showed that 786-OFOXD1null had an extended G2/M phase. Investigation of mitosis revealed a deficiency in phosphorylation of histone H3 in 786-OFOXD1null, and increased DNA damage. Genes correlate with FOXD1 in the TCGA dataset associate with several aspects of mitosis, including histone H3 phosphorylation. CONCLUSIONS We show that FOXD1 regulates the cell cycle in ccRCC cells by control of histone H3 phosphorylation, and that FOXD1 expression governs tumor formation and tumor growth. Transcriptome analysis supports this role for FOXD1 in ccRCC patient tumors and provides an explanation for the inverse correlation between tumor expression of FOXD1 and patient survival. Our findings reveal an important role for FOXD1 in maintaining chromatin stability and promoting cell cycle progression and provide a new tool with which to study the biology of FOXD1 in ccRCC.
Collapse
Affiliation(s)
- Kyle H Bond
- The Rogosin Institute, 310 East 67th Street, New York, NY, 10065, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, 168 College Ave, Orono, 04469, ME, USA
| | - Jennifer L Fetting
- Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04074, USA
- Current affiliation: ICON Plc, 2100 Pembrook Parkway, North Wales, 19446, PA, USA
| | - Christine W Lary
- Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04074, USA
| | - Ivette F Emery
- Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04074, USA
| | - Leif Oxburgh
- The Rogosin Institute, 310 East 67th Street, New York, NY, 10065, USA.
| |
Collapse
|
10
|
Meng L, Chen D, Meng G, Lu L, Han C. Dysregulation of the Sirt5/IDH2 axis contributes to sunitinib resistance in human renal cancer cells. FEBS Open Bio 2021; 11:921-931. [PMID: 33455080 PMCID: PMC7931237 DOI: 10.1002/2211-5463.13090] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 01/14/2021] [Indexed: 01/01/2023] Open
Abstract
Sunitinib (Sun), a tyrosine kinase inhibitor of vascular endothelial growth factor receptor, is the standard first‐line treatment against advanced clear cell renal cell carcinoma (RCC), but resistance to therapy is inevitable. Reactive oxygen species production is associated with sensitivity to chemotherapy, but the underlying mechanisms are not completely understood. Here, we investigated the mechanisms contributing to Sun resistance using the RCC cell lines ACHN and 786‐O. We report that Sun‐resistant cells exhibited reduced apoptosis, increased cell viability, increased reactive oxygen species production and disrupted mitochondrial function. Furthermore, chronic Sun treatment resulted in an up‐regulation of Sirt5/isocitrate dehydrogenase 2 (IDH2) expression levels. Knockdown of Sirt5/IDH2 impaired mitochondrial function and partially attenuated Sun resistance. Finally, up‐regulation of Sirt5 enhanced the expression of IDH2 via modulation of succinylation at K413 and promoted protein stability. In conclusion, dysregulation of Sirt5/IDH2 partially contributes to Sun resistance in RCC cells by affecting antioxidant capacity.
Collapse
Affiliation(s)
- Liang Meng
- Department of Computer Tomography, Cangzhou Central Hospital, China
| | - Deqiang Chen
- Department of Computer Tomography, Cangzhou Central Hospital, China
| | - Gaopei Meng
- Department of Computer Tomography, Cangzhou Central Hospital, China
| | - Li Lu
- Department of Computer Tomography, Cangzhou Central Hospital, China
| | - Chenggang Han
- Department of Computer Tomography, Cangzhou Central Hospital, China
| |
Collapse
|
11
|
Tsai LL, Bhatt RS, Strob MF, Jegede OA, Sun MRM, Alsop DC, Catalano P, McDermott D, Robson PM, Atkins MB, Pedrosa I. Arterial Spin Labeled Perfusion MRI for the Evaluation of Response to Tyrosine Kinase Inhibition Therapy in Metastatic Renal Cell Carcinoma. Radiology 2020; 298:332-340. [PMID: 33258745 DOI: 10.1148/radiol.2020201763] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Background Tumor perfusion may inform therapeutic response and resistance in metastatic renal cell carcinoma (RCC) treated with antiangiogenic therapy. Purpose To determine if arterial spin labeled (ASL) MRI perfusion changes are associated with tumor response and disease progression in metastatic RCC treated with vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors (TKIs). Materials and Methods In this prospective study (ClinicalTrials.gov identifier: NCT00749320), metastatic RCC perfusion was measured with ASL MRI before and during sunitinib or pazopanib therapy between October 2008 and March 2014. Objective response rate (ORR) and progression-free survival (PFS) were calculated. Perfusion was compared between responders and nonresponders at baseline, at week 2, after cycle 2 (12 weeks), after cycle 4 (24 weeks), and at disease progression and compared with the ORR by using the Wilcoxon rank sum test and with PFS by using the log-rank test. Results Seventeen participants received sunitinib (mean age, 59 years ± 7.0 [standard deviation]; 11 men); 11 participants received pazopanib (mean age, 63 years ± 6.6; eight men). Responders had higher baseline tumor perfusion than nonresponders (mean, 404 mL/100 g/min ± 213 vs 199 mL/100 g/min ± 136; P = .02). Perfusion decreased from baseline to week 2 (-53 mL/100 g/min ± 31; P < .001), after cycle 2 (-65 mL/100 g/min ± 25; P < .001), and after cycle 4 (-79 mL/100 g/min ± 15; P = .008). Interval reduction in perfusion at those three time points was not associated with ORR (P = .63, .29, and .27, respectively) or PFS (P = .28, .27, and .32). Perfusion increased from cycle 4 to disease progression (51% ± 11; P < .001). Conclusion Arterial spin labeled perfusion MRI may assist in identifying responders to vascular endothelial growth factor receptor tyrosine kinase inhibitors and may help detect early evidence of disease progression in patients with metastatic renal cell carcinoma. © RSNA, 2020 Online supplemental material is available for this article. See also the editorial by Goh and De Vita in this issue.
Collapse
Affiliation(s)
- Leo L Tsai
- From the Department of Radiology (L.L.T., M.F.S., D.C.A.) and Division of Hematology/Oncology (R.S.B., D.M.), Beth Israel Deaconess Medical Center, Boston, Mass; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Mass (O.A.J., P.C.); Department of Radiology, Lowell General Hospital, Lowell, Mass (M.R.M.S.); Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (P.M.R.); Division of Hematology/Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC (M.B.A.); and Department of Radiology, University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75390 (I.P.)
| | - Rupal S Bhatt
- From the Department of Radiology (L.L.T., M.F.S., D.C.A.) and Division of Hematology/Oncology (R.S.B., D.M.), Beth Israel Deaconess Medical Center, Boston, Mass; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Mass (O.A.J., P.C.); Department of Radiology, Lowell General Hospital, Lowell, Mass (M.R.M.S.); Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (P.M.R.); Division of Hematology/Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC (M.B.A.); and Department of Radiology, University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75390 (I.P.)
| | - Meaghan F Strob
- From the Department of Radiology (L.L.T., M.F.S., D.C.A.) and Division of Hematology/Oncology (R.S.B., D.M.), Beth Israel Deaconess Medical Center, Boston, Mass; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Mass (O.A.J., P.C.); Department of Radiology, Lowell General Hospital, Lowell, Mass (M.R.M.S.); Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (P.M.R.); Division of Hematology/Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC (M.B.A.); and Department of Radiology, University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75390 (I.P.)
| | - Opeyemi A Jegede
- From the Department of Radiology (L.L.T., M.F.S., D.C.A.) and Division of Hematology/Oncology (R.S.B., D.M.), Beth Israel Deaconess Medical Center, Boston, Mass; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Mass (O.A.J., P.C.); Department of Radiology, Lowell General Hospital, Lowell, Mass (M.R.M.S.); Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (P.M.R.); Division of Hematology/Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC (M.B.A.); and Department of Radiology, University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75390 (I.P.)
| | - Maryellen R M Sun
- From the Department of Radiology (L.L.T., M.F.S., D.C.A.) and Division of Hematology/Oncology (R.S.B., D.M.), Beth Israel Deaconess Medical Center, Boston, Mass; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Mass (O.A.J., P.C.); Department of Radiology, Lowell General Hospital, Lowell, Mass (M.R.M.S.); Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (P.M.R.); Division of Hematology/Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC (M.B.A.); and Department of Radiology, University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75390 (I.P.)
| | - David C Alsop
- From the Department of Radiology (L.L.T., M.F.S., D.C.A.) and Division of Hematology/Oncology (R.S.B., D.M.), Beth Israel Deaconess Medical Center, Boston, Mass; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Mass (O.A.J., P.C.); Department of Radiology, Lowell General Hospital, Lowell, Mass (M.R.M.S.); Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (P.M.R.); Division of Hematology/Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC (M.B.A.); and Department of Radiology, University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75390 (I.P.)
| | - Paul Catalano
- From the Department of Radiology (L.L.T., M.F.S., D.C.A.) and Division of Hematology/Oncology (R.S.B., D.M.), Beth Israel Deaconess Medical Center, Boston, Mass; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Mass (O.A.J., P.C.); Department of Radiology, Lowell General Hospital, Lowell, Mass (M.R.M.S.); Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (P.M.R.); Division of Hematology/Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC (M.B.A.); and Department of Radiology, University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75390 (I.P.)
| | - David McDermott
- From the Department of Radiology (L.L.T., M.F.S., D.C.A.) and Division of Hematology/Oncology (R.S.B., D.M.), Beth Israel Deaconess Medical Center, Boston, Mass; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Mass (O.A.J., P.C.); Department of Radiology, Lowell General Hospital, Lowell, Mass (M.R.M.S.); Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (P.M.R.); Division of Hematology/Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC (M.B.A.); and Department of Radiology, University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75390 (I.P.)
| | - Philip M Robson
- From the Department of Radiology (L.L.T., M.F.S., D.C.A.) and Division of Hematology/Oncology (R.S.B., D.M.), Beth Israel Deaconess Medical Center, Boston, Mass; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Mass (O.A.J., P.C.); Department of Radiology, Lowell General Hospital, Lowell, Mass (M.R.M.S.); Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (P.M.R.); Division of Hematology/Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC (M.B.A.); and Department of Radiology, University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75390 (I.P.)
| | - Michael B Atkins
- From the Department of Radiology (L.L.T., M.F.S., D.C.A.) and Division of Hematology/Oncology (R.S.B., D.M.), Beth Israel Deaconess Medical Center, Boston, Mass; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Mass (O.A.J., P.C.); Department of Radiology, Lowell General Hospital, Lowell, Mass (M.R.M.S.); Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (P.M.R.); Division of Hematology/Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC (M.B.A.); and Department of Radiology, University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75390 (I.P.)
| | - Ivan Pedrosa
- From the Department of Radiology (L.L.T., M.F.S., D.C.A.) and Division of Hematology/Oncology (R.S.B., D.M.), Beth Israel Deaconess Medical Center, Boston, Mass; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Mass (O.A.J., P.C.); Department of Radiology, Lowell General Hospital, Lowell, Mass (M.R.M.S.); Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (P.M.R.); Division of Hematology/Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC (M.B.A.); and Department of Radiology, University of Texas Southwestern Medical School, 5323 Harry Hines Blvd, Dallas, TX 75390 (I.P.)
| |
Collapse
|
12
|
The Role of Chemokine Receptor CXCR3 and Its Ligands in Renal Cell Carcinoma. Int J Mol Sci 2020; 21:ijms21228582. [PMID: 33202536 PMCID: PMC7696621 DOI: 10.3390/ijms21228582] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 11/08/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023] Open
Abstract
The major invasive subtype of kidney cancer is renal cell carcinoma (RCC). The essential components of cancer development are chronic inflammation and neoangiogenesis. It has been suggested that the chemokine ligand 9, -10, –11 (CXCL9–11) and chemokine receptor 3 (CXCR3) chemokines receptor expressed on monocytes, T and NK cells may be involved in the inhibition of angiogenesis. However, to date, little is known about the potential clinical significance of these chemokines and their receptor in renal cell carcinoma. Therefore, in this review, we described the role of CXCR3 and its ligands in pathogenesis of RCC. We performed an extensive search of the current literature in our investigation, using the MEDLINE/PubMed database. The changes of chemokines and their specific receptor in renal cell carcinoma were observed. Published studies revealed an increased expression of CXCR3 and elevated concentration of its ligands in RCC. The association between treatment of RCC and CXCL9–11/CXCR3 concentration and expression was also observed. Moreover, CXCR3 and its ligands levels were related to patient’s prognosis, risk of metastasis and tumor growth. This review describes the potential role of CXCR3 and its ligands in pathogenesis of RCC, as well as their potential immune-therapeutic significance. However, future studies should aim to confirm the clinical and prognostic role of CXCL9–11/CXCR3 in renal cell carcinoma.
Collapse
|
13
|
Xu W, Puligandla M, Manola J, Bullock AJ, Tamasauskas D, McDermott DF, Atkins MB, Haas NB, Flaherty K, Uzzo RG, Dutcher JP, DiPaola RS, Bhatt RS. Angiogenic Factor and Cytokine Analysis among Patients Treated with Adjuvant VEGFR TKIs in Resected Renal Cell Carcinoma. Clin Cancer Res 2019; 25:6098-6106. [PMID: 31471309 DOI: 10.1158/1078-0432.ccr-19-0818] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/06/2019] [Accepted: 07/09/2019] [Indexed: 12/30/2022]
Abstract
PURPOSE The use of VEGFR TKIs for the adjuvant treatment of renal cell carcinoma (RCC) remains controversial. We investigated the effects of adjuvant VEGFR TKIs on circulating cytokines in the ECOG-ACRIN 2805 (ASSURE) trial. EXPERIMENTAL DESIGN Patients with resected high-risk RCC were randomized to sunitinib, sorafenib, or placebo. Plasma from 413 patients was analyzed from post-nephrectomy baseline, 4 weeks, and 6 weeks after treatment initiation. Mixed effects and Cox proportional hazards models were used to test for changes in circulating cytokines and associations between disease-free survival (DFS) and cytokine levels. RESULTS VEGF and PlGF increased after 4 weeks on sunitinib or sorafenib (P < 0.0001 for both) and returned to baseline at 6 weeks on sunitinib (corresponding to the break in the sunitinib schedule) but not sorafenib (which was administered continuously). sFLT-1 decreased after 4 weeks on sunitinib and 6 weeks on sorafenib (P < 0.0001). sVEGFR-2 decreased after both 4 and 6 weeks of treatment on sunitinib or sorafenib (P < 0.0001). Patients receiving placebo had no significant changes in cytokine levels. CXCL10 was elevated at 4 and 6 weeks on sunitinib and sorafenib but not on placebo. Higher baseline CXCL10 was associated with worse DFS (HR 1.41 per log increase in CXCL10, Bonferroni-adjusted P = 0.003). This remained significant after adjustment for T-stage, Fuhrman grade, and ECOG performance status. CONCLUSIONS Among patients treated with adjuvant VEGFR TKIs for RCC, drug-host interactions mediate changes in circulating cytokines. Elevated baseline CXCL10 was associated with worse DFS. Studies to understand functional consequences of these changes are under way.
Collapse
Affiliation(s)
- Wenxin Xu
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Maneka Puligandla
- Dana-Farber Cancer Institute, ECOG-ACRIN Biostatistics Center, Boston, Massachusetts
| | - Judith Manola
- Dana-Farber Cancer Institute, ECOG-ACRIN Biostatistics Center, Boston, Massachusetts
| | | | | | | | - Michael B Atkins
- MedStar Georgetown University Hospital, Washington, District of Columbia
| | - Naomi B Haas
- Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | | | | | - Rupal S Bhatt
- Beth Israel Deaconess Medical Center, Boston, Massachusetts.
| |
Collapse
|
14
|
Yu S, Dai J, Ma M, Xu T, Kong Y, Cui C, Chi Z, Si L, Tang H, Yang L, Sheng X, Guo J. RBCK1 promotes p53 degradation via ubiquitination in renal cell carcinoma. Cell Death Dis 2019; 10:254. [PMID: 30874541 PMCID: PMC6420644 DOI: 10.1038/s41419-019-1488-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/22/2019] [Accepted: 02/25/2019] [Indexed: 01/05/2023]
Abstract
Renal cell carcinoma (RCC) accounts for approximately 3% of adult malignancies, and the incidence of RCC continues to rise worldwide. Although RCC can be treated with surgery at an early stages, the five-year survival rates have been observed to decline dramatically in patients with advanced disease. Most patients with RCC treated with cytotoxic or targeted drugs will develop resistance at some point during therapy. Thus, it is necessary to identify novel therapeutic targets for RCC. Here, we found that RANBP2-type and C3HC4-type zinc finger-containing 1 (RBCK1) expression was upregulated in human RCC samples. Analysis of multiple public databases revealed the correlation between RBCK1 expression and poor prognosis in RCC patients. Subsequently, we performed RBCK1 depletion experiments in RCC cells that severely affected the in vivo and in vitro proliferation of renal cancer cells. The effects of RBCK1 on cell proliferation could be rescued with p53 expression knockdown in two cell lines expressing wild-type p53. Further experiments demonstrated that RBCK1 could facilitate p53 poly-ubiquitination and degradation by direct interaction with p53. Together, our results show that RBCK1 may serve as a promising target for RCC therapy by restoring p53 functions.
Collapse
Affiliation(s)
- Sifan Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jie Dai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Meng Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Tianxiao Xu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yan Kong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Chuanliang Cui
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhihong Chi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Lu Si
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Huan Tang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Lu Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xinan Sheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China.
| | - Jun Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China.
| |
Collapse
|
15
|
Study of Cathepsin B inhibition in VEGFR TKI treated human renal cell carcinoma xenografts. Oncogenesis 2019; 8:15. [PMID: 30796200 PMCID: PMC6386754 DOI: 10.1038/s41389-019-0121-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 01/14/2019] [Indexed: 12/19/2022] Open
Abstract
Several therapeutic options are available for metastatic RCC, but responses are almost never complete, and resistance to therapy develops in the vast majority of patients. Consequently, novel treatments are needed to combat resistance to current therapies and to improve patient outcomes. We have applied integrated transcriptome and proteome analyses to identify cathepsin B (CTSB), a cysteine proteinase of the papain family, as one of the most highly upregulated gene products in established human RCC xenograft models of resistance to vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors (TKI). We used established RCC models to test the significance of CTSB in the progression of renal cancer. Our evaluation of CTSB showed that stable CTSB knockdown suppressed RCC growth in vitro and in vivo. Stable over-overexpression of wild-type CTSB (CTSBwt/hi), but not of an CTSB active site mutant (CTSBN298A), rescued cell growth in CTSB knockdown cells and abolished the efficacy of VEGFR TKI treatment. Genome-wide transcriptome profiling of CTSB knockdown cells demonstrated significant effects on multiple metabolic and stem cell-related pathways, with ALDHA1A (ALDH1) as one of the most significantly downregulated genes. Importantly, survival analysis across 16 major TCGA cancers revealed that CTSB overexpression is associated with low rates of three and five year patient survival rates (P = 2.5e-08, HR = 1.4). These data strongly support a contribution of CTSB activity to RCC cell growth and tumorigenicity. They further highlight the promise of CTSB inhibition in development of novel combination therapies designed to improve efficacy of current TKI treatments of metastatic RCC.
Collapse
|
16
|
Fischl AS, Wang X, Falcon BL, Almonte-Baldonado R, Bodenmiller D, Evans G, Stewart J, Wilson T, Hipskind P, Manro J, Uhlik MT, Chintharlapalli S, Gerald D, Alsop DC, Benjamin LE, Bhatt RS. Inhibition of Sphingosine Phosphate Receptor 1 Signaling Enhances the Efficacy of VEGF Receptor Inhibition. Mol Cancer Ther 2019; 18:856-867. [PMID: 30787172 DOI: 10.1158/1535-7163.mct-18-0548] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 10/04/2018] [Accepted: 02/04/2019] [Indexed: 01/15/2023]
Abstract
Inhibition of VEGFR signaling is an effective treatment for renal cell carcinoma, but resistance continues to be a major problem. Recently, the sphingosine phosphate (S1P) signaling pathway has been implicated in tumor growth, angiogenesis, and resistance to antiangiogenic therapy. S1P is a bioactive lipid that serves an essential role in developmental and pathologic angiogenesis via activation of the S1P receptor 1 (S1P1). S1P1 signaling counteracts VEGF signaling and is required for vascular stabilization. We used in vivo and in vitro angiogenesis models including a postnatal retinal angiogenesis model and a renal cell carcinoma murine tumor model to test whether simultaneous inhibition of S1P1 and VEGF leads to improved angiogenic inhibition. Here, we show that inhibition of S1P signaling reduces the endothelial cell barrier and leads to excessive angiogenic sprouting. Simultaneous inhibition of S1P and VEGF signaling further disrupts the tumor vascular beds, decreases tumor volume, and increases tumor cell death compared with monotherapies. These studies suggest that inhibition of angiogenesis at two stages of the multistep process may maximize the effects of antiangiogenic therapy. Together, these data suggest that combination of S1P1 and VEGFR-targeted therapy may be a useful therapeutic strategy for the treatment of renal cell carcinoma and other tumor types.
Collapse
MESH Headings
- Angiogenesis Inhibitors/pharmacology
- Animals
- Antibodies, Monoclonal/pharmacology
- Carcinoma, Renal Cell/blood supply
- Carcinoma, Renal Cell/drug therapy
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/pathology
- Cell Line, Tumor
- Drug Therapy, Combination
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Female
- Humans
- Kidney Neoplasms/blood supply
- Kidney Neoplasms/drug therapy
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/pathology
- Lysophospholipids/antagonists & inhibitors
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Nude
- Neovascularization, Pathologic/drug therapy
- Sphingosine/analogs & derivatives
- Sphingosine/antagonists & inhibitors
- Sphingosine-1-Phosphate Receptors/antagonists & inhibitors
- Sunitinib/pharmacology
- Treatment Outcome
- Tumor Burden/drug effects
- Vascular Endothelial Growth Factor A/pharmacology
- Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
| | - Xiaoen Wang
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | | | | | | | | | | | | | | | | | | | | | | | - David C Alsop
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | | | - Rupal S Bhatt
- Division of Hematology and Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
17
|
Characterisation of the Morphological, Functional and Molecular Changes in Sunitinib-Resistant Renal Cell Carcinoma Cells. J Kidney Cancer VHL 2018; 5:1-9. [PMID: 30109169 PMCID: PMC6088203 DOI: 10.15586/jkcvhl.2018.106] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Accepted: 07/16/2018] [Indexed: 12/12/2022] Open
Abstract
Sunitinib resistance is a major clinical problem hampering the treatment of renal cell carcinoma (RCC). Studies on the comprehensive characterisation of morphological, functional and molecular changes in sunitinib-resistant RCC cells are lacking. The aim of the current study was to develop sunitinib resistance in four human RCC cell lines (786-0, Caki-1, Caki-2 and SN12K1), and to characterise the changed cell biology with sunitinib resistance. RCC cells were made resistant by continuous, chronic exposure to 10 μM of sunitinib over a period of 12 months. Cell proliferation, morphology, transmigration, and gene expression for interleukin-6 (IL-6), interleukin-8 (IL-8), vascular endothelial growth factor (VEGF), Bcl-2 and Bax were studied. There was no significant difference in growth rate or transmigration between the parental and resistant cells. Sunitinib-resistant cells were significantly hypertrophic compared with parental cells as evidenced by increases in the surface areas of the whole cells and the nuclei. IL-6 was significantly increased in all resistant cells. IL-8 was increased in sunitinib-resistant Caki-2 and SN12K1 cells and decreased in 786-0 without any significant changes in Caki-1. VEGF was increased in resistant Caki-2 and SN12K1 cells but not in 786-0 and Caki-1. The Bcl2/Bax ratio was increased in Caki-1, Caki-2 and SN12K1 cells but decreased in 786-0 cells. The increased IL-6 may contribute to sunitinib resistance either via VEGF-mediated angiogenesis or through shifting of the Bcl2/Bax balance in favour of anti-apoptosis.
Collapse
|
18
|
Makhov P, Naito S, Haifler M, Kutikov A, Boumber Y, Uzzo RG, Kolenko VM. The convergent roles of NF-κB and ER stress in sunitinib-mediated expression of pro-tumorigenic cytokines and refractory phenotype in renal cell carcinoma. Cell Death Dis 2018; 9:374. [PMID: 29515108 PMCID: PMC5841329 DOI: 10.1038/s41419-018-0388-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 01/30/2018] [Accepted: 02/01/2018] [Indexed: 01/09/2023]
Abstract
Renal cell carcinoma (RCC) is the most common form of kidney cancer. While cure remains exceptionally infrequent in RCC patients with systemic or recurrent disease, current targeted molecular strategies, including multi-targeted tyrosine kinase inhibitors (TKIs), notably changed the treatment paradigm of advanced renal cancer. Yet, complete and durable responses have been noted in only a few cases. Our studies reveal that sunitinib triggers two resistance-promoting signaling pathways in RCC cells, which emanate from the endoplasmic reticulum (ER) stress response: a PERK-driven ER stress response that induces expression of the pro-tumorigenic cytokines IL-6, IL-8, and TNF-α, and a TRAF2-mediated NF-κB survival program that protects tumor cells against cell death. PERK blockade completely prevents sunitinib-induced expression of IL-6, IL-8 and TNF-α, whereas NF-κB inhibition reinstates sensitivity of RCC cells to sunitinib both in vitro and in vivo. Taken together, our findings indicate that ER stress response may contribute to sunitinib resistance in RCC patients.
Collapse
Affiliation(s)
- Peter Makhov
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA.
| | - Sei Naito
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Miki Haifler
- Division of Urologic Oncology, Department of Surgery, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Alexander Kutikov
- Division of Urologic Oncology, Department of Surgery, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Yanis Boumber
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Robert G Uzzo
- Division of Urologic Oncology, Department of Surgery, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Vladimir M Kolenko
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| |
Collapse
|
19
|
Li X, Shao C, Shi Y, Han W. Lessons learned from the blockade of immune checkpoints in cancer immunotherapy. J Hematol Oncol 2018; 11:31. [PMID: 29482595 PMCID: PMC6389077 DOI: 10.1186/s13045-018-0578-4] [Citation(s) in RCA: 246] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 02/16/2018] [Indexed: 12/16/2022] Open
Abstract
The advent of immunotherapy, especially checkpoint inhibitor-based immunotherapy, has provided novel and powerful weapons against cancer. Because only a subset of cancer patients exhibit durable responses, further exploration of the mechanisms underlying the resistance to immunotherapy in the bulk of cancer patients is merited. Such efforts may help to identify which patients could benefit from immune checkpoint blockade. Given the existence of a great number of pathways by which cancer can escape immune surveillance, and the complexity of tumor-immune system interaction, development of various combination therapies, including those that combine with conventional therapies, would be necessary. In this review, we summarize the current understanding of the mechanisms by which resistance to checkpoint blockade immunotherapy occurs, and outline how actionable combination strategies may be derived to improve clinical outcomes for patients.
Collapse
Affiliation(s)
- Xiaolei Li
- The First Affiliated Hospital of Soochow University and Jiangsu Engineering Research Center for Tumor Immunotherapy, Institutes for Translational Medicine and Suzhou Key Laboratory of Tumor Microenvironment and Pathology, Soochow University, Suzhou, Jiangsu, 215123, China.,Department of Molecular Biology, Immunology and Bio-therapeutic, Institute of Basic Medicine, Chinese PLA General Hospital, Beijing, 100853, China
| | - Changshun Shao
- The First Affiliated Hospital of Soochow University and Jiangsu Engineering Research Center for Tumor Immunotherapy, Institutes for Translational Medicine and Suzhou Key Laboratory of Tumor Microenvironment and Pathology, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yufang Shi
- The First Affiliated Hospital of Soochow University and Jiangsu Engineering Research Center for Tumor Immunotherapy, Institutes for Translational Medicine and Suzhou Key Laboratory of Tumor Microenvironment and Pathology, Soochow University, Suzhou, Jiangsu, 215123, China.
| | - Weidong Han
- Department of Molecular Biology, Immunology and Bio-therapeutic, Institute of Basic Medicine, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
20
|
Wang X, Solban N, Khanna P, Callea M, Song J, Alsop DC, Pearsall RS, Atkins MB, Mier JW, Signoretti S, Alimzhanov M, Kumar R, Bhasin MK, Bhatt RS. Inhibition of ALK1 signaling with dalantercept combined with VEGFR TKI leads to tumor stasis in renal cell carcinoma. Oncotarget 2018; 7:41857-41869. [PMID: 27248821 PMCID: PMC5173101 DOI: 10.18632/oncotarget.9621] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 05/05/2016] [Indexed: 12/21/2022] Open
Abstract
Treatment of metastatic renal cell carcinoma (mRCC) with agents that block signaling through vascular endothelial growth factor receptor 2 (VEGFR2) induces disease regression or stabilization in some patients; however, these responses tend to be short-lived. Therefore, development of combination therapies that can extend the efficacy of VEGFR antagonists in mRCC remains a priority. We studied murine xenograft models of RCC that become refractory to treatment with the VEGFR tyrosine kinase inhibitor (TKI) sunitinib. Dalantercept is a novel antagonist of Activin receptor-like kinase 1 (ALK1)/Bone morphogenetic protein (BMP) 9 signaling. Dalantercept inhibited growth in the murine A498 xenograft model which correlated with hyperdilation of the tumor vasculature and an increase in tumor hypoxia. When combined with sunitinib, dalantercept induced tumor necrosis and prevented tumor regrowth and revascularization typically seen with sunitinib monotherapy in two RCC models. Combination therapy led to significant downregulation of angiogenic genes as well as downregulation of endothelial specific gene expression particularly of the Notch signaling pathway. We demonstrate that simultaneous targeting of molecules that control distinct phases of angiogenesis, such as ALK1 and VEGFR, is a valid strategy for treatment of mRCC. At the molecular level, combination therapy leads to downregulation of Notch signaling.
Collapse
Affiliation(s)
- Xiaoen Wang
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | - Prateek Khanna
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Marcella Callea
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Jiaxi Song
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - David C Alsop
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | - Michael B Atkins
- Departments of Oncology and Medicine, Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | - James W Mier
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Ravi Kumar
- Acceleron Pharma, Inc., Cambridge, MA, USA
| | - Manoj K Bhasin
- Division of Interdisciplinary Medicine & Biotechnology, and Genomics, Proteomics, Bioinformatics and Systems Biology Center, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Rupal S Bhatt
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
21
|
Mastri M, Rosario S, Tracz A, Frink RE, Brekken RA, Ebos JML. The Challenges of Modeling Drug Resistance to Antiangiogenic Therapy. Curr Drug Targets 2017; 17:1747-1754. [PMID: 26648063 DOI: 10.2174/1389450117666151209123544] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 11/11/2015] [Accepted: 11/11/2015] [Indexed: 02/08/2023]
Abstract
Drug resistance remains an ongoing challenge for the majority of patients treated with inhibitors of the vascular endothelial growth factor (VEGF) pathway, a key regulator of tumor angiogenesis. Preclinical models have played a significant role in identifying multiple complex mechanisms of antiangiogenic treatment failure. Yet questions remain about the optimal methodology to study resistance that may assist in making clinically relevant choices about alternative or combination treatment strategies. The origins of antiangiogenic treatment failure may stem from the tumor vasculature, the tumor itself, or both together, and preclinical methods that define resistance are diverse and rarely compared. We performed a literature search of the preclinical methodologies used to examine resistance to VEGF pathway inhibitors and identified 109 papers from more than 400 that use treatment failure as the starting point for mechanistic study. We found that definitions of resistance are broad and inconsistent, involve only a small number of reagents, and derive mostly from in vitro and in vivo methodologies that often do not represent clinically relevant disease stages or progression. Together, this literature analysis highlights the challenges of studying inhibitors of the tumor microenvironment in the preclinical setting and the need for improved methodology to assist in qualifying (and quantifying) treatment failure to identify mechanisms that will help predict alternative strategies in patients.
Collapse
Affiliation(s)
| | | | | | | | | | - John M L Ebos
- Department of Cancer Genetics and Medicine, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY 142631, USA
| |
Collapse
|
22
|
Martínez-Sáez O, Gajate Borau P, Alonso-Gordoa T, Molina-Cerrillo J, Grande E. Targeting HIF-2 α in clear cell renal cell carcinoma: A promising therapeutic strategy. Crit Rev Oncol Hematol 2017; 111:117-123. [PMID: 28259286 DOI: 10.1016/j.critrevonc.2017.01.013] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 01/22/2017] [Indexed: 12/24/2022] Open
Abstract
The loss of the Von Hippel-Lindau tumor suppressor (VHL) is a key oncogenic event in the vast majority of patients with clear cell renal cell carcinoma (ccRCC). With the loss of the VHL protein (pVHL) function, the hypoxia inducible factor α (HIF-α) accumulates inside the tumor cell and dimerizes with HIF-β. The HIF-α/HIF-β complex transcriptionally activates hundreds of genes promoting the adaptation to hypoxia that is implicated in tumor development. There is growing evidence showing that HIF-2α subunit has a central role in ccRCC over HIF-1α. Thus, efforts have been made to specifically target this pathway. PT2385 and PT2399 are first-in-class, orally available, small molecule inhibitors of HIF-2 that selectively disrupt the heterodimerization of HIF-2α with HIF-1β. Preclinical and clinical data indicate that these new molecules are effective in blocking cancer cell growth, proliferation, and tumor angiogenesis characteristic in ccRCC. Treatment with HIF-2α specific antagonists, either alone or in combination with immunotherapy or other antiangiogenic agents have the potential to transform the therapeutic landscape in this tumor in the future. Herein, we summarize the molecular background behind the use of HIF-2α inhibitors in ccRCC and give an overview of the development of new agents in this setting.
Collapse
Affiliation(s)
- Olga Martínez-Sáez
- Medical Oncology Department, Ramon y Cajal University Hospital, Ctra, Colmenar Viejo km9100, 28029, Madrid, Spain.
| | - Pablo Gajate Borau
- Medical Oncology Department, Ramon y Cajal University Hospital, Ctra, Colmenar Viejo km9100, 28029, Madrid, Spain
| | - Teresa Alonso-Gordoa
- Medical Oncology Department, Ramon y Cajal University Hospital, Ctra, Colmenar Viejo km9100, 28029, Madrid, Spain
| | - Javier Molina-Cerrillo
- Medical Oncology Department, Ramon y Cajal University Hospital, Ctra, Colmenar Viejo km9100, 28029, Madrid, Spain
| | - Enrique Grande
- Medical Oncology Department, Ramon y Cajal University Hospital, Ctra, Colmenar Viejo km9100, 28029, Madrid, Spain
| |
Collapse
|
23
|
Sweis RF, Medved M, Towey S, Karczmar GS, Oto A, Szmulewitz RZ, O'Donnell PH, Fishkin P, Karrison T, Stadler WM. Dynamic Contrast-Enhanced Magnetic Resonance Imaging as a Pharmacodynamic Biomarker for Pazopanib in Metastatic Renal Carcinoma. Clin Genitourin Cancer 2016; 15:207-212. [PMID: 27634566 DOI: 10.1016/j.clgc.2016.08.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 08/08/2016] [Indexed: 01/18/2023]
Abstract
INTRODUCTION/BACKGROUND Traditional imaging assessment criteria might not correlate well with clinical benefit from vascular endothelial growth factor pathway-directed therapy in metastatic renal cancer. Preclinical data suggest tumor growth is preceded by a rise in Ktrans level, a parameter derived from dynamic contrast-enhanced (DCE) magnetic resonance imaging (MRI) that reflects vascular permeability. We thus hypothesized that Ktrans might be a predictive biomarker for pazopanib. PATIENTS AND METHODS Patients with metastatic renal cancer were treated with pazopanib at 800 mg oral daily until disease progression. MRI of the abdomen and pelvis with a DCE-MRI sequence was obtained at baseline and every 8 weeks. RESULTS Seventy-three DCE-MRI scans were completed and 66 were technically assessable. Of the 17 patients with at least 1 DCE-MRI scan after the baseline scan, 16 (94%) had a decline in Ktrans level. Changes in Ktrans compared with baseline after 1, 8, 16, and 24 weeks were -49%, -65%, -63%, and -53%, respectively (P = .0052, repeated measures analysis of variance). The median Ktrans nadir occurred at 8 weeks. The median progression-free survival (PFS) was 32.1 weeks. PFS was longer in patients with higher baseline Ktrans values (P = .036, log rank). Baseline Ktrans did not reach significance in a Cox proportional hazard model including clinical prognostic index and previous treatments (P = .083). CONCLUSION We show that Ktrans is a pharmacodynamic biomarker for pazopanib therapy in metastatic renal cancer. Because of the small sample size, the predictive capacity of Ktrans recovery could not be assessed, but baseline Ktrans correlated with PFS.
Collapse
Affiliation(s)
- Randy F Sweis
- Department of Medicine, Section of Hematology/Oncology, Comprehensive Cancer Center, University of Chicago, Chicago, IL
| | - Milica Medved
- Department of Radiology, University of Chicago, Chicago, IL
| | - Shannon Towey
- Department of Radiology, University of Chicago, Chicago, IL
| | | | - Aytekin Oto
- Department of Radiology, University of Chicago, Chicago, IL
| | - Russell Z Szmulewitz
- Department of Medicine, Section of Hematology/Oncology, Comprehensive Cancer Center, University of Chicago, Chicago, IL
| | - Peter H O'Donnell
- Department of Medicine, Section of Hematology/Oncology, Comprehensive Cancer Center, University of Chicago, Chicago, IL
| | | | - Theodore Karrison
- Department of Medicine, Section of Hematology/Oncology, Comprehensive Cancer Center, University of Chicago, Chicago, IL
| | - Walter M Stadler
- Department of Medicine, Section of Hematology/Oncology, Comprehensive Cancer Center, University of Chicago, Chicago, IL.
| |
Collapse
|
24
|
Gobe GC, Ng KL, Small DM, Vesey DA, Johnson DW, Samaratunga H, Oliver K, Wood S, Barclay JL, Rajandram R, Li L, Morais C. Decreased apoptosis repressor with caspase recruitment domain confers resistance to sunitinib in renal cell carcinoma through alternate angiogenesis pathways. Biochem Biophys Res Commun 2016; 473:47-53. [PMID: 26995091 DOI: 10.1016/j.bbrc.2016.03.048] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 03/10/2016] [Indexed: 11/15/2022]
Abstract
Apoptosis repressor with caspase recruitment domain (ARC), an endogenous inhibitor of apoptosis, is upregulated in a number of human cancers, thereby conferring drug resistance and giving a rationale for the inhibition of ARC to overcome drug resistance. Our hypothesis was that ARC would be similarly upregulated and targetable for therapy in renal cell carcinoma (RCC). Expression of ARC was assessed in 85 human RCC samples and paired non-neoplastic kidney by qPCR and immunohistochemistry, as well as in four RCC cell lines by qPCR, Western immunoblot and confocal microscopy. Contrary to expectations, ARC was significantly decreased in the majority of clear cell RCC and in three (ACHN, Caki-1 and 786-0) of the four RCC cell lines compared with the HK-2 non-cancerous human proximal tubular epithelial cell line. Inhibition of ARC with shRNA in the RCC cell line (SN12K1) that had shown increased ARC expression conferred resistance to Sunitinib, and upregulated interleukin-6 (IL-6) and vascular endothelial growth factor (VEGF). We therefore propose that decreased ARC, particularly in clear cell RCC, confers resistance to targeted therapy through restoration of tyrosine kinase-independent alternate angiogenesis pathways. Although the results are contrary to expectations from other cancer studies, they were confirmed here with multiple analytical methods. We believe the highly heterogeneous nature of cancers like RCC predicate that expression patterns of molecules must be interpreted in relation to respective matched non-neoplastic regions. In the current study, this procedure indicated that ARC is decreased in RCC.
Collapse
Affiliation(s)
- Glenda C Gobe
- Centre for Kidney Disease Research, School of Medicine, Translational Research Institute, The University of Queensland, Brisbane, Queensland, 4102, Australia
| | - Keng Lim Ng
- Centre for Kidney Disease Research, School of Medicine, Translational Research Institute, The University of Queensland, Brisbane, Queensland, 4102, Australia; Department of Urology, Princess Alexandra Hospital, Wollongabba Queensland, Australia
| | - David M Small
- Centre for Kidney Disease Research, School of Medicine, Translational Research Institute, The University of Queensland, Brisbane, Queensland, 4102, Australia
| | - David A Vesey
- Centre for Kidney Disease Research, School of Medicine, Translational Research Institute, The University of Queensland, Brisbane, Queensland, 4102, Australia; Department of Renal Medicine, The University of Queensland at Princess Alexandra Hospital, Brisbane, Queensland, 4102, Australia
| | - David W Johnson
- Centre for Kidney Disease Research, School of Medicine, Translational Research Institute, The University of Queensland, Brisbane, Queensland, 4102, Australia; Department of Renal Medicine, The University of Queensland at Princess Alexandra Hospital, Brisbane, Queensland, 4102, Australia
| | - Hemamali Samaratunga
- Centre for Kidney Disease Research, School of Medicine, Translational Research Institute, The University of Queensland, Brisbane, Queensland, 4102, Australia; Aquesta Pathology, Brisbane, Australia
| | - Kimberley Oliver
- Anatomical Pathology, Princess Alexandra Hospital, Wollongabba, Queensland, Australia
| | - Simon Wood
- Department of Urology, Princess Alexandra Hospital, Wollongabba Queensland, Australia
| | | | - Retnagowri Rajandram
- Centre for Kidney Disease Research, School of Medicine, Translational Research Institute, The University of Queensland, Brisbane, Queensland, 4102, Australia; Department of Surgery, Faculty of Medicine, University Malaya, Kuala Lumpur, Malaysia
| | - Li Li
- Laboratory of Translational Cancer Research, Ochsner Health System, New Orleans, LA, USA
| | - Christudas Morais
- Centre for Kidney Disease Research, School of Medicine, Translational Research Institute, The University of Queensland, Brisbane, Queensland, 4102, Australia.
| |
Collapse
|
25
|
Mahoney KM, Jacobus S, Bhatt RS, Song J, Carvo I, Cheng SC, Simpson M, Fay AP, Puzanov I, Michaelson MD, Atkins MB, McDermott DF, Signoretti S, Choueiri TK. Phase 2 Study of Bevacizumab and Temsirolimus After VEGFR TKI in Metastatic Renal Cell Carcinoma. Clin Genitourin Cancer 2016; 14:304-313.e6. [PMID: 27036973 DOI: 10.1016/j.clgc.2016.02.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 02/08/2016] [Accepted: 02/14/2016] [Indexed: 11/29/2022]
Abstract
BACKGROUND Inhibiting VEGF and mammalian target of rapamycin (mTOR) pathways are standard treatment approaches for patients with metastatic renal cell carcinoma (mRCC). Here we report the activity and safety of the VEGF ligand inhibitor bevacizumab and the mTOR inhibitor temsirolimus combination in patients with clear cell (CC) and non-clear cell (NCC) mRCC whose disease had failed to respond to prior VEGF blockade. PATIENTS AND METHODS In this phase 2 investigator-initiated multicenter study, patients received bevacizumab and temsirolimus. The primary end point was 4-month progression-free survival (PFS) rate. Secondary end points included overall response rate, median overall survival (OS), toxicity, and correlative studies of biomarkers downstream of mTOR. RESULTS Forty patients received at least 1 dose of therapy. Thirty-three (82.5%) had favorable/intermediate risk disease according to International Metastatic Renal Cell Carcinoma Database Consortium criteria, 13 (32.5%) with nccRCC histology. Nineteen (48.7%) had primary vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitor (TKI)-refractory disease. The 4-month PFS rate was 65%. Overall median PFS and OS were 5.6 and 12.2 months. Median PFS and OS were 6.5 and 9.6 months in patients with primary VEGFR TKI-refractory disease, and 5.6 months and 13.1 months in patients with nccRCC. Dose reductions were needed in 80% of patients. Most frequent toxicities included fatigue, hypertension, dyslipidemia, and proteinuria. Dose discontinuation due to adverse events occurred in 27.5% of patients. Baseline tumor immunohistochemistry for phospho-S6 protein was not associated with clinical benefit. CONCLUSION Combining bevacizumab and temsirolimus in patients previously treated with VEGFR TKI was possible but with dose reductions and treatment discontinuations. This combination resulted in modest activity, including in patients with primary VEGF-refractory disease and NCC histology.
Collapse
Affiliation(s)
- Kathleen M Mahoney
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Susanna Jacobus
- Department of Statistics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Rupal S Bhatt
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Jiaxi Song
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Ingrid Carvo
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Su-Chun Cheng
- Department of Statistics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Mekailah Simpson
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - André P Fay
- PUCRS School of Medicine, Porto Alegre, Brazil
| | - Igor Puzanov
- Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, TN
| | - M Dror Michaelson
- Department of Medical Oncology, Massachusetts General Hospital Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Michael B Atkins
- Department of Medical Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University School of Medicine, Washington, DC
| | - David F McDermott
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Sabina Signoretti
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Toni K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA.
| |
Collapse
|
26
|
Asić K. Dominant mechanisms of primary resistance differ from dominant mechanisms of secondary resistance to targeted therapies. Crit Rev Oncol Hematol 2016; 97:178-96. [DOI: 10.1016/j.critrevonc.2015.08.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 06/18/2015] [Accepted: 08/04/2015] [Indexed: 02/07/2023] Open
|
27
|
Kovács K, Decatur C, Toro M, Pham DG, Liu H, Jing Y, Murray TG, Lampidis TJ, Merchan JR. 2-Deoxy-Glucose Downregulates Endothelial AKT and ERK via Interference with N-Linked Glycosylation, Induction of Endoplasmic Reticulum Stress, and GSK3β Activation. Mol Cancer Ther 2015; 15:264-75. [PMID: 26637370 DOI: 10.1158/1535-7163.mct-14-0315] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 10/19/2015] [Indexed: 12/19/2022]
Abstract
Interference with endothelial cell metabolism is a promising, yet unexploited strategy for angiogenesis inhibition. We reported that the glucose analogue 2-deoxy-D-glucose (2-DG) inhibits angiogenesis at significantly lower concentrations than those required for tumor cytotoxicity. Here, we found that hypersensitivity to 2-DG in endothelial cells is not associated with enhanced drug uptake compared with tumor cells, but with time-dependent, endothelial-selective inhibition of AKT and ERK phosphorylation. Downregulation of these critical survival pathways is shown to be due to 2-DG's interference with N-linked glycosylation, leading to alterations in VEGFR2 (and downstream signaling) as well as induction of endoplasmic reticulum (ER) stress, GSK3β activation, and apoptosis. In vivo, periocular administration of 2-DG in LHBETATAG mice was associated with significant reduction of newly formed (CD105(+)) tumor capillaries, ER stress (GRP 78 expression), and endothelial apoptosis (TUNEL). These findings uniquely link N-linked glycosylation inhibition, ER stress, and ERK/AKT downregulation in endothelial cells, and provide a novel drug development strategy to overcome resistance mechanisms to currently available antiangiogenic agents.
Collapse
Affiliation(s)
- Krisztina Kovács
- Department of Medicine, Division of Hematology-Oncology, University of Miami Miller School of Medicine and Sylvester Comprehensive Cancer Center, Miami, Florida
| | - Christina Decatur
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine and Sylvester Comprehensive Cancer Center, Miami, Florida
| | - Marcela Toro
- Department of Medicine, Division of Hematology-Oncology, University of Miami Miller School of Medicine and Sylvester Comprehensive Cancer Center, Miami, Florida
| | - Dien G Pham
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine and Sylvester Comprehensive Cancer Center, Miami, Florida
| | - Huaping Liu
- Department of Cell Biology, University of Miami Miller School of Medicine and Sylvester Comprehensive Cancer Center, Miami, Florida
| | - Yuqi Jing
- Department of Medicine, Division of Hematology-Oncology, University of Miami Miller School of Medicine and Sylvester Comprehensive Cancer Center, Miami, Florida
| | - Timothy G Murray
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine and Sylvester Comprehensive Cancer Center, Miami, Florida
| | - Theodore J Lampidis
- Department of Cell Biology, University of Miami Miller School of Medicine and Sylvester Comprehensive Cancer Center, Miami, Florida
| | - Jaime R Merchan
- Department of Medicine, Division of Hematology-Oncology, University of Miami Miller School of Medicine and Sylvester Comprehensive Cancer Center, Miami, Florida.
| |
Collapse
|
28
|
Mahoney KM, Rennert PD, Freeman GJ. Combination cancer immunotherapy and new immunomodulatory targets. Nat Rev Drug Discov 2015; 14:561-84. [PMID: 26228759 DOI: 10.1038/nrd4591] [Citation(s) in RCA: 948] [Impact Index Per Article: 105.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Targeting immune checkpoints such as programmed cell death protein 1 (PD1), programmed cell death 1 ligand 1 (PDL1) and cytotoxic T lymphocyte antigen 4 (CTLA4) has achieved noteworthy benefit in multiple cancers by blocking immunoinhibitory signals and enabling patients to produce an effective antitumour response. Inhibitors of CTLA4, PD1 or PDL1 administered as single agents have resulted in durable tumour regression in some patients, and combinations of PD1 and CTLA4 inhibitors may enhance antitumour benefit. Numerous additional immunomodulatory pathways as well as inhibitory factors expressed or secreted by myeloid and stromal cells in the tumour microenvironment are potential targets for synergizing with immune checkpoint blockade. Given the breadth of potential targets in the immune system, critical questions to address include which combinations should move forward in development and which patients will benefit from these treatments. This Review discusses the leading drug targets that are expressed on tumour cells and in the tumour microenvironment that allow enhancement of the antitumour immune response.
Collapse
Affiliation(s)
- Kathleen M Mahoney
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215, USA. [2] Division of Haematology and Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA. [3]
| | - Paul D Rennert
- 1] SugarCone Biotech, Holliston, Massachusetts 01746, USA. [2] Videre Biotherapeutics, Watertown, Massachusetts 02472, USA. [3]
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215, USA
| |
Collapse
|
29
|
Shibasaki N, Yamasaki T, Kanno T, Arakaki R, Sakamoto H, Utsunomiya N, Inoue T, Tsuruyama T, Nakamura E, Ogawa O, Kamba T. Role of IL13RA2 in Sunitinib Resistance in Clear Cell Renal Cell Carcinoma. PLoS One 2015; 10:e0130980. [PMID: 26114873 PMCID: PMC4482605 DOI: 10.1371/journal.pone.0130980] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 05/26/2015] [Indexed: 12/31/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) and mammalian target of rapamycin are well-known therapeutic targets for renal cell carcinoma (RCC). Sunitinib is an agent that targets VEGF receptors and is considered to be a standard treatment for metastatic or unresectable clear cell RCC (ccRCC). However, ccRCC eventually develops resistance to sunitinib in most cases, and the mechanisms underlying this resistance are not fully elucidated. In the present study, we established unique primary xenograft models, KURC1 (Kyoto University Renal Cancer 1) and KURC2, from freshly isolated ccRCC specimens. The KURC1 xenograft initially responded to sunitinib treatment, however finally acquired resistance. KURC2 retained sensitivity to sunitinib for over 6 months. Comparing gene expression profiles between the two xenograft models with different sensitivity to sunitinib, we identified interleukin 13 receptor alpha 2 (IL13RA2) as a candidate molecule associated with the acquired sunitinib-resistance in ccRCC. And patients with high IL13RA2 expression in immunohistochemistry in primary ccRCC tumor tends to have sunitinib-resistant metastatic site. Next, we showed that sunitinib-sensitive 786-O cells acquired resistance in vivo when IL13RA2 was overexpressed. Conversely, shRNA-mediated knockdown of IL13RA2 successfully overcame the sunitinib-resistance in Caki-1 cells. Histopathological analyses revealed that IL13RA2 repressed sunitinib-induced apoptosis without increasing tumor vasculature in vivo. To our knowledge, this is a novel mechanism of developing resistance to sunitinib in a certain population of ccRCC, and these results indicate that IL13RA2 could be one of potential target to overcome sunitinib resistance.
Collapse
Affiliation(s)
- Noboru Shibasaki
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toshinari Yamasaki
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toru Kanno
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ryuichiro Arakaki
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiromasa Sakamoto
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Noriaki Utsunomiya
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takahiro Inoue
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tatsuaki Tsuruyama
- Department of Diagnostic Pathology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Eijiro Nakamura
- Laboratory for Malignancy Control Research, Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Osamu Ogawa
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomomi Kamba
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- * E-mail:
| |
Collapse
|
30
|
Zhang L, Wang X, Bullock AJ, Callea M, Shah H, Song J, Moreno K, Visentin B, Deutschman D, Alsop DC, Atkins MB, Mier JW, Signoretti S, Bhasin M, Sabbadini RA, Bhatt RS. Anti-S1P Antibody as a Novel Therapeutic Strategy for VEGFR TKI-Resistant Renal Cancer. Clin Cancer Res 2015; 21:1925-1934. [PMID: 25589614 DOI: 10.1158/1078-0432.ccr-14-2031] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 12/13/2014] [Indexed: 01/22/2023]
Abstract
PURPOSE VEGFR2 tyrosine kinase inhibition (TKI) is a valuable treatment approach for patients with metastatic renal cell carcinoma (RCC). However, resistance to treatment is inevitable. Identification of novel targets could lead to better treatment for patients with TKI-naïve or -resistant RCC. EXPERIMENTAL DESIGN In this study, we performed transcriptome analysis of VEGFR TKI-resistant tumors in a murine model and discovered that the SPHK-S1P pathway is upregulated at the time of resistance. We tested sphingosine-1-phosphate (S1P) pathway inhibition using an anti-S1P mAb (sphingomab), in two mouse xenograft models of RCC, and assessed tumor SPHK expression and S1P plasma levels in patients with metastatic RCC. RESULTS Resistant tumors expressed several hypoxia-regulated genes. The SPHK1 pathway was among the most highly upregulated pathways that accompanied resistance to VEGFR TKI therapy. SPHK1 was expressed in human RCC, and the product of SPHK1 activity, S1P, was elevated in patients with metastatic RCC, suggesting that human RCC behavior could, in part, be due to overproduction of S1P. Sphingomab neutralization of extracellular S1P slowed tumor growth in both mouse models. Mice bearing tumors that had developed resistance to sunitinib treatment also exhibited tumor growth suppression with sphingomab. Sphingomab treatment led to a reduction in tumor blood flow as measured by MRI. CONCLUSIONS Our findings suggest that S1P inhibition may be a novel therapeutic strategy in patients with treatment-naïve RCC and also in the setting of resistance to VEGFR TKI therapy.
Collapse
Affiliation(s)
- Liang Zhang
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, United States of America
| | - Xiaoen Wang
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, United States of America.,Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, United States of America
| | - Andrea J Bullock
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, United States of America
| | - Marcella Callea
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, Massachusetts, 02115, United States of America
| | - Harleen Shah
- Division of Interdisciplinary Medicine and Biotechnology, and Genomics and Proteomics Center, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, United States of America
| | - Jiaxi Song
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, Massachusetts, 02115, United States of America
| | - Kelli Moreno
- Lpath Inc., 4025 Sorrento Valley Blvd. San Diego, CA, 92121, United States of America
| | - Barbara Visentin
- Lpath Inc., 4025 Sorrento Valley Blvd. San Diego, CA, 92121, United States of America
| | - Douglas Deutschman
- Department of Biology, San Diego State University, 5500 Campanile Dr. San Diego, CA. 92182-4614, United States of America
| | - David C Alsop
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, United States of America
| | - Michael B Atkins
- Departments of Oncology and Medicine, Georgetown-Lombardi Comprehensive Cancer Center, 3970 Reservoir Road, NW, Washington, DC. United States of America
| | - James W Mier
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, United States of America
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, Massachusetts, 02115, United States of America
| | - Manoj Bhasin
- Division of Interdisciplinary Medicine and Biotechnology, and Genomics and Proteomics Center, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, United States of America
| | - Roger A Sabbadini
- Lpath Inc., 4025 Sorrento Valley Blvd. San Diego, CA, 92121, United States of America
| | - Rupal S Bhatt
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, United States of America
| |
Collapse
|
31
|
Ciamporcero E, Miles KM, Adelaiye R, Ramakrishnan S, Shen L, Ku S, Pizzimenti S, Sennino B, Barrera G, Pili R. Combination strategy targeting VEGF and HGF/c-met in human renal cell carcinoma models. Mol Cancer Ther 2014; 14:101-10. [PMID: 25381264 DOI: 10.1158/1535-7163.mct-14-0094] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Alternative pathways to the VEGF, such as hepatocyte growth factor or HGF/c-met, are emerging as key players in tumor angiogenesis and resistance to anti-VEGF therapies. The aim of this study was to assess the effects of a combination strategy targeting the VEGF and c-met pathways in clear cell renal cell carcinoma (ccRCC) models. Male SCID mice (8/group) were implanted with 786-O tumor pieces and treated with either a selective VEGF receptor tyrosine kinase inhibitor, axitinib (36 mg/kg, 2×/day); a c-met inhibitor, crizotinib (25 mg/kg, 1×/day); or combination. We further tested this drug combination in a human ccRCC patient-derived xenograft, RP-R-01, in both VEGF-targeted therapy-sensitive and -resistant models. To evaluate the resistant phenotype, we established an RP-R-01 sunitinib-resistant model by continuous sunitinib treatment (60 mg/kg, 1×/day) of RP-R-01-bearing mice. Treatment with single-agent crizotinib reduced tumor vascularization but failed to inhibit tumor growth in either model, despite also a significant increase of c-met expression and phosphorylation in the sunitinib-resistant tumors. In contrast, axitinib treatment was effective in inhibiting angiogenesis and tumor growth in both models, with its antitumor effect significantly increased by the combined treatment with crizotinib, independently from c-met expression. Combination treatment also induced prolonged survival and significant tumor growth inhibition in the 786-O human RCC model. Overall, our results support the rationale for the clinical testing of combined VEGF and HGF/c-met pathway blockade in the treatment of ccRCC, both in first- and second-line setting.
Collapse
Affiliation(s)
- Eric Ciamporcero
- Genitourinary Program, Roswell Park Cancer Institute, Buffalo, New York. Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | | | - Remi Adelaiye
- Genitourinary Program, Roswell Park Cancer Institute, Buffalo, New York. Department of Cancer Pathology and Prevention, Roswell Park Cancer Institute, Buffalo, New York
| | - Swathi Ramakrishnan
- Genitourinary Program, Roswell Park Cancer Institute, Buffalo, New York. Department of Cancer Pathology and Prevention, Roswell Park Cancer Institute, Buffalo, New York
| | - Li Shen
- Genitourinary Program, Roswell Park Cancer Institute, Buffalo, New York
| | - ShengYu Ku
- Genitourinary Program, Roswell Park Cancer Institute, Buffalo, New York. Department of Cancer Pathology and Prevention, Roswell Park Cancer Institute, Buffalo, New York
| | - Stefania Pizzimenti
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Barbara Sennino
- Department of Anatomy, Comprehensive Cancer Center, Cardiovascular Research Institute, University of California-San Francisco, San Francisco, California
| | - Giuseppina Barrera
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Roberto Pili
- Genitourinary Program, Roswell Park Cancer Institute, Buffalo, New York.
| |
Collapse
|
32
|
Sunitinib resistance in renal cell carcinoma. J Kidney Cancer VHL 2014; 1:1-11. [PMID: 28326244 PMCID: PMC5345511 DOI: 10.15586/jkcvhl.2014.7] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 04/10/2014] [Indexed: 12/26/2022] Open
Abstract
Of the many targeted therapies introduced since 2006, sunitinib has carved its way to become the most commonly used first-line therapy for the treatment of metastatic renal cell carcinoma (RCC). Despite significant improvements in progression-free survival, 30% of the patients are intrinsically resistant to sunitinib and the remaining 70% who respond initially will eventually become resistant in 6-15 months. While the molecular mechanisms of acquired resistance to sunitinib have been unravelling at a rapid rate, the mechanisms of intrinsic resistance remain elusive. Combination therapy, sunitinib rechallenge and sequential therapy have been investigated as means to overcome resistance to sunitinib. Of these, sequential therapy appears to be the most promising strategy. This mini review summarises our emerging understanding of the molecular mechanisms, and the strategies employed to overcome sunitinib resistance.
Collapse
|
33
|
Bielecka ZF, Czarnecka AM, Solarek W, Kornakiewicz A, Szczylik C. Mechanisms of Acquired Resistance to Tyrosine Kinase Inhibitors in Clear - Cell Renal Cell Carcinoma (ccRCC). ACTA ACUST UNITED AC 2014; 8:218-228. [PMID: 25152704 PMCID: PMC4141325 DOI: 10.2174/1574362409666140206223014] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 11/27/2013] [Accepted: 01/29/2014] [Indexed: 12/29/2022]
Abstract
Clear - cell renal cell carcinoma (ccRCC) is a histological subtype of renal cell carcinoma - the most prevalent adult kidney cancer. Causes of ccRCC are not completely understood and therefore number of available therapies is limited. As a consequence of tumor chemo- and radioresistance as well as restrictions in offered targeted therapies, overall response rate is still unsatisfactory. Moreover, a significant group of patients (circa 1/4) does not respond to the targeted first-line treatment, while in other cases, after an initial period of stable improvement, disease progression occurs. Owing to this, more data on resistance mechanisms are needed, especially those concerning widely used, relatively lately approved and more successful than previous therapies - tyrosine kinase inhibitors (TKIs). Up to date, five TKIs have been licensed for ccRCC treatment: sunitinib (SUTENT®, Pfizer Inc.), sorafenib (Nexavar®, Bayer HealthCare/Onyx Pharmaceuticals), pazopanib (Votrient®, GlaxoSmithKline), axitinib (Inlyta®, Pfitzer Inc.) and tivozanib (AV-951®, AVEO Pharmaceuticals). Researchers have specified different subsets of tyrosine kinase inhibitors potential resistance mechanisms in clear-cell renal cell carcinoma. In most papers published until now, drug resistance is divided into intrinsic and acquired, and typically multi-drug resistance (MDR) protein is described. Herein, the authors focus on molecular analysis concerning acquired, non-genetic resistance to TKIs, with insight into specific biological processes.
Collapse
Affiliation(s)
- Zofia F Bielecka
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine in Warsaw, Poland ; Postgraduate School of Molecular Medicine, Medical University of Warsaw, Poland
| | - Anna M Czarnecka
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine in Warsaw, Poland
| | - Wojciech Solarek
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine in Warsaw, Poland ; Postgraduate School of Molecular Medicine, Medical University of Warsaw, Poland
| | - Anna Kornakiewicz
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine in Warsaw, Poland ; I Faculty of Medicine, Medical University of Warsaw, Poland
| | - Cezary Szczylik
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine in Warsaw, Poland
| |
Collapse
|
34
|
Wang X, Bullock AJ, Zhang L, Wei L, Yu D, Mahagaokar K, Alsop DC, Mier JW, Atkins MB, Coxon A, Oliner J, Bhatt RS. The role of angiopoietins as potential therapeutic targets in renal cell carcinoma. Transl Oncol 2014; 7:188-95. [PMID: 24704536 PMCID: PMC4101387 DOI: 10.1016/j.tranon.2014.02.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Revised: 12/10/2013] [Accepted: 01/02/2014] [Indexed: 12/12/2022] Open
Abstract
Angiopoietin 2 (Ang2) is a secreted glycoprotein upregulated at sites of angiogenesis and has been implicated in cancer neovascularization. Recent studies have suggested efficacy of combined Ang and vascular endothelial growth factor receptor (VEGFR) inhibition for patients with metastatic renal cell carcinoma (mRCC). We measured Ang2 expression in human tissue and plasma, and tested the effect of dual Ang1/2 (trebananib; AMG386) or Ang2 alone (L1-7) inhibition with VEGFR inhibition on murine RCC growth and blood flow. Ang2 levels were higher in human tumors than normal tissues with RCC ranking highest for Ang2 expression across all tumor types tested. Plasma Ang2 was significantly higher in patients with mRCC compared to controls or patients with stage I disease. Plasma Ang2 decreased with sunitinib treatment and increased at time of disease progression. In the RCC mouse, dual Ang1/2 and Ang2 inhibition improved the activity of sunitinib. Combined Ang1/2 and VEGFR inhibition prevented the resumption of blood flow associated with sunitinib resistance. Thus, Ang2 inhibition, independent of Ang1 inhibition, improves the activity of sunitinib and plasma Ang2 increases in the setting of progression on sunitinib possibly contributing to resistance. Further, arterial spin-labeled perfusion magnetic resonance imaging might be a non-invasive marker of the antiangiogenic activity of Ang inhibitors.
Collapse
Affiliation(s)
- Xiaoen Wang
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Andrea J Bullock
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Liang Zhang
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Lin Wei
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Dongyin Yu
- Oncology Research, Amgen Inc, Thousand Oaks, CA
| | - Kedar Mahagaokar
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - David C Alsop
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - James W Mier
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Michael B Atkins
- Departments of Oncology and Medicine, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC
| | | | - Jon Oliner
- Oncology Research, Amgen Inc, Thousand Oaks, CA
| | - Rupal S Bhatt
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| |
Collapse
|
35
|
Chintalgattu V, Rees ML, Culver JC, Goel A, Jiffar T, Zhang J, Dunner K, Pati S, Bankson JA, Pasqualini R, Arap W, Bryan NS, Taegtmeyer H, Langley RR, Yao H, Kupferman ME, Entman ML, Dickinson ME, Khakoo AY. Coronary microvascular pericytes are the cellular target of sunitinib malate-induced cardiotoxicity. Sci Transl Med 2013; 5:187ra69. [PMID: 23720580 DOI: 10.1126/scitranslmed.3005066] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Sunitinib malate is a multitargeted receptor tyrosine kinase inhibitor used in the treatment of human malignancies. A substantial number of sunitinib-treated patients develop cardiac dysfunction, but the mechanism of sunitinib-induced cardiotoxicity is poorly understood. We show that mice treated with sunitinib develop cardiac and coronary microvascular dysfunction and exhibit an impaired cardiac response to stress. The physiological changes caused by treatment with sunitinib are accompanied by a substantial depletion of coronary microvascular pericytes. Pericytes are a cell type that is dependent on intact platelet-derived growth factor receptor (PDGFR) signaling but whose role in the heart is poorly defined. Sunitinib-induced pericyte depletion and coronary microvascular dysfunction are recapitulated by CP-673451, a structurally distinct PDGFR inhibitor, confirming the role of PDGFR in pericyte survival. Thalidomide, an anticancer agent that is known to exert beneficial effects on pericyte survival and function, prevents sunitinib-induced pericyte cell death in vitro and prevents sunitinib-induced cardiotoxicity in vivo in a mouse model. Our findings suggest that pericytes are the primary cellular target of sunitinib-induced cardiotoxicity and reveal the pericyte as a cell type of concern in the regulation of coronary microvascular function. Furthermore, our data provide preliminary evidence that thalidomide may prevent cardiotoxicity in sunitinib-treated cancer patients.
Collapse
|
36
|
Characterization of mechanism involved in acquired resistance to sorafenib in a mouse renal cell cancer RenCa model. Clin Transl Oncol 2013; 16:801-6. [DOI: 10.1007/s12094-013-1151-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 12/11/2013] [Indexed: 12/28/2022]
|
37
|
Sharpe K, Stewart GD, Mackay A, Van Neste C, Rofe C, Berney D, Kayani I, Bex A, Wan E, O'Mahony FC, O'Donnell M, Chowdhury S, Doshi R, Ho-Yen C, Gerlinger M, Baker D, Smith N, Davies B, Sahdev A, Boleti E, De Meyer T, Van Criekinge W, Beltran L, Lu YJ, Harrison DJ, Reynolds AR, Powles T. The effect of VEGF-targeted therapy on biomarker expression in sequential tissue from patients with metastatic clear cell renal cancer. Clin Cancer Res 2013; 19:6924-34. [PMID: 24130073 DOI: 10.1158/1078-0432.ccr-13-1631] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To investigate how biologically relevant markers change in response to antiangiogenic therapy in metastatic clear cell renal cancer (mRCC) and correlate these changes with outcome. EXPERIMENTAL DESIGN The study used sequential tumor tissue and functional imaging (taken at baseline and 12-16 weeks) obtained from three similar phase II studies. All three studies investigated the role of VEGF tyrosine kinase inhibitors (TKI) before planned nephrectomy in untreated mRCC (n = 85). The effect of targeted therapy on ten biomarkers was measured from sequential tissue. Comparative genomic hybridization (CGH) array and DNA methylation profiling (MethylCap-seq) was performed in matched frozen pairs. Biomarker expression was correlated with early progression (progression as best response) and delayed progression (between 12-16 weeks). RESULTS VEGF TKI treatment caused a significant reduction in vessel density (CD31), phospho-S6K expression, PDL-1 expression, and FOXP3 expression (P < 0.05 for each). It also caused a significant increase in cytoplasmic FGF-2, MET receptor expression in vessels, Fuhrman tumor grade, and Ki-67 (P < 0.05 for each). Higher levels of Ki-67 and CD31 were associated with delayed progression (P < 0.05). Multiple samples (n = 5) from the same tumor showed marked heterogeneity of tumor grade, which increased significantly with treatment. Array CGH showed extensive intrapatient variability, which did not occur in DNA methylation analysis. CONCLUSION TKI treatment is associated with dynamic changes in relevant biomarkers, despite significant heterogeneity in chromosomal and protein, but not epigenetic expression. Changes to Ki-67 expression and tumor grade indicate that treatment is associated with an increase in the aggressive phenotype of the tumor.
Collapse
Affiliation(s)
- Kevin Sharpe
- Authors' Affiliations: Barts Cancer Institute, Queen Mary University of London; Experimental Cancer Medicine Centre, University College; The Institute of Cancer Research; Guys and St Thomas' Hospital; The Royal Free Hospital London, London; Edinburgh Urological Cancer Group, University of Edinburgh, Edinburgh; Astra Zeneca, Manchester; School of Medicine, University of St Andrews, Fife, United Kingdom; National Cancer Institute, Amsterdam, the Netherlands; and University of Ghent, Ghent, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Stehle F, Schulz K, Seliger B. Towards defining biomarkers indicating resistances to targeted therapies. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1844:909-16. [PMID: 24269379 DOI: 10.1016/j.bbapap.2013.11.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 10/17/2013] [Accepted: 11/13/2013] [Indexed: 12/20/2022]
Abstract
An impressive, but often short objective response was obtained in many tumor patients treated with different targeted therapies, but most of the patients develop resistances against these drugs. So far, a number of distinct mechanisms leading to intrinsic as well as acquired resistances have been identified in tumors of distinct origin. These can arise from genetic alterations, like mutations, truncations, and amplifications or due to deregulated expression of various proteins and signal transduction pathways, but also from cellular heterogeneity within tumors after an initial response. Therefore, biomarkers are urgently needed for cancer prognosis and personalized cancer medicine. The application of "ome"-based technologies including cancer (epi)genomics, next generation sequencing, cDNA microarrays and proteomics might led to the predictive or prognostic stratification of patients to categorize resistance mechanisms and to postulate combinations of treatment strategies. This review discusses the implementation of proteome-based analysis to identify markers of pathway (in)activation in tumors and the resistance mechanisms, which represent major clinical problems as a tool to optimize individually tailored therapies based on targeted drugs. This article is part of a Special Issue entitled: Biomarkers: A Proteomic Challenge.
Collapse
Affiliation(s)
- Franziska Stehle
- Martin Luther University Halle-Wittenberg, Institute of Medical Immunology, Magdeburger Str. 2, D-06112 Halle, Saale, Germany
| | - Kristin Schulz
- Martin Luther University Halle-Wittenberg, Institute of Medical Immunology, Magdeburger Str. 2, D-06112 Halle, Saale, Germany
| | - Barbara Seliger
- Martin Luther University Halle-Wittenberg, Institute of Medical Immunology, Magdeburger Str. 2, D-06112 Halle, Saale, Germany.
| |
Collapse
|
39
|
Zhang T, Niu X, Liao L, Cho EA, Yang H. The contributions of HIF-target genes to tumor growth in RCC. PLoS One 2013; 8:e80544. [PMID: 24260413 PMCID: PMC3832366 DOI: 10.1371/journal.pone.0080544] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 10/14/2013] [Indexed: 12/12/2022] Open
Abstract
Somatic mutations or loss of expression of tumor suppressor VHL happen in the vast majority of clear cell Renal Cell Carcinoma, and it’s causal for kidney cancer development. Without VHL, constitutively active transcription factor HIF is strongly oncogenic and is essential for tumor growth. However, the contribution of individual HIF-responsive genes to tumor growth is not well understood. In this study we examined the contribution of important HIF-responsive genes such as VEGF, CCND1, ANGPTL4, EGLN3, ENO2, GLUT1 and IGFBP3 to tumor growth in a xenograft model using immune-compromised nude mice. We found that the suppression of VEGF or CCND1 impaired tumor growth, suggesting that they are tumor-promoting genes. We further discovered that the lack of ANGPTL4, EGLN3 or ENO2 expression did not change tumor growth. Surprisingly, depletion of GLUT1 or IGFBP3 significantly increased tumor growth, suggesting that they have tumor-inhibitory functions. Depletion of IGFBP3 did not lead to obvious activation of IGFIR. Unexpectedly, the depletion of IGFIR protein led to significant increase of IGFBP3 at both the protein and mRNA levels. Concomitantly, the tumor growth was greatly impaired, suggesting that IGFBP3 might suppress tumor growth in an IGFIR-independent manner. In summary, although the overall transcriptional activity of HIF is strongly tumor-promoting, the expression of each individual HIF-responsive gene could either enhance, reduce or do nothing to the kidney cancer tumor growth.
Collapse
Affiliation(s)
- Ting Zhang
- Tianjin Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Xiaohua Niu
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Lili Liao
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Eun-Ah Cho
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Haifeng Yang
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
40
|
García-Palmero I, López-Larrubia P, Cerdán S, Villalobo A. Nuclear magnetic resonance imaging of tumour growth and neovasculature performance in vivo reveals Grb7 as a novel antiangiogenic target. NMR IN BIOMEDICINE 2013; 26:1059-1069. [PMID: 23348935 DOI: 10.1002/nbm.2918] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 12/10/2012] [Accepted: 12/10/2012] [Indexed: 06/01/2023]
Abstract
Development of neovasculature is a necessary requirement for tumour growth and it provides additional opportunities for therapeutic intervention. However, current antiangiogenic therapies have limited efficacy, mostly because of the development of resistance. Hence, characterization of new antiangiogenic molecular targets is of considerable clinical interest. We report that a calmodulin-binding domain (CaM-BD) deletion mutant of the growth factor receptor bound protein 7 (Grb7) (denoted Grb7Δ) impairs tumour growth and associated angiogenesis in vivo. We implanted glioma C6 cells in rat brains transfected with an enhanced yellow fluorescent protein (EYFP) chimera of Grb7∆, its EYFP-Grb7 wild type counterpart, and EYFP alone. We systematically followed intracerebral growth of the tumours, their cellularity and the functional performance of tumour-associated microvasculature using magnetic resonance imaging, including anatomical T1W and T2W images and functional diffusion and perfusion parameters. Tumours grown from implanted C6 cells expressing EYFP-Grb7Δ developed slower, became smaller and presented lower apparent cellularity than those derived from cells expressing EYFP-Grb7 and EYFP. Vascular perfusion measurements within tumours derived from EYFP-Grb7∆-expressing cells showed significantly lower cerebral blood flow (CBF), cerebral blood volume (CBV) and mean transit time (MTT) values. These findings were independently validated by histological and immunohistochemical techniques. Taken together, these findings confirm that the CaM-BD of Grb7 plays an important role in tumour growth and associated angiogenesis in vivo, thus identifying this region of the protein as a novel target for antiangiogenic treatment.
Collapse
Affiliation(s)
- Irene García-Palmero
- Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Department of Cancer Biology, c/ Arturo Duperier 4, E-28029, Madrid, Spain
| | | | | | | |
Collapse
|
41
|
Antiangiogenic therapy promoted metastasis of hepatocellular carcinoma by suppressing host-derived interleukin-12b in mouse models. Angiogenesis 2013; 16:809-20. [PMID: 23716000 DOI: 10.1007/s10456-013-9357-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 05/22/2013] [Indexed: 02/07/2023]
Abstract
Antiangiogenic therapy, specially sorafenib, has become the standard of care for patients with advanced hepatocellular carcinoma (HCC), however, the improvement in survival time is not satisfactory. Previous studies have found that, in some circumstances, antiangiogenic therapy promoted tumor metastasis and the mechanistic studies were mainly focus on cancer-cell-autonomous manners. In two experimental metastasis models with tail-vein injection with hepatoma cells and an orthotopic HCC mouse model, we found that pretreatment with two vascular endothelial growth factor receptor (VEGFR) inhibitors, sunitinib and sorafenib, facilitated tumor cell survival in blood stream and promoted lung metastasis from tumors that were subsequently incubated after drug discontinuation, indicating that host response joined into the pro-metastatic effects. An antibody microarray identified that interleukin (IL)-12b was decreased in the peripheral blood of the mice treated with the two VEGFR inhibitors. IL-12b suppression in macrophages and dendritic cells from host organs was found to play a crucial role in treatment-induced metastasis. Supplement with recombinant mouse IL-12b or restoration of IL-12b expression in the host by zoledronic acid, which was previously reported to enhance IL-12 expression in vitro and in vivo, alleviated the metastasis-promoting effects of sunitinib and sorafenib. These studies suggest that host response to VEGFR inhibitors facilitates HCC metastasis and restoration of IL-12b expression could translate into clinical benefits.
Collapse
|
42
|
Ovatodiolide Targets β -Catenin Signaling in Suppressing Tumorigenesis and Overcoming Drug Resistance in Renal Cell Carcinoma. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:161628. [PMID: 23781255 PMCID: PMC3677612 DOI: 10.1155/2013/161628] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Accepted: 04/17/2013] [Indexed: 12/20/2022]
Abstract
Dysregulated β-catenin signaling is intricately involved in renal cell carcinoma (RCC) carcinogenesis and progression. Determining potential β-catenin signaling inhibitors would be helpful in ameliorating drug resistance in advanced or metastatic RCC. Screening for β-catenin signaling inhibitors involved in silico inquiry of the PubChem Bioactivity database followed by TCF/LEF reporter assay. The biological effects of ovatodiolide were evaluated in 4 RCC cell lines in vitro and 2 RCC cell lines in a mouse xenograft model. The synergistic effects of ovatodiolide and sorafenib or sunitinib were examined in 2 TKI-resistant RCC cell lines. Ovatodiolide, a pure compound of Anisomeles indica, inhibited β-catenin signaling and reduced RCC cell viability, survival, migration/invasion, and in vitro cell or in vivo mouse tumorigenicity. Cytotoxicity was significantly reduced in a normal kidney epithelial cell line with the treatment. Ovatodiolide reduced phosphorylated β-catenin (S552) that inhibited β-catenin nuclear translocation. Moreover, ovatodiolide decreased β-catenin stability and impaired the association of β-catenin and transcription factor 4. Ovatodiolide combined with sorafenib or sunitinib overcame drug resistance in TKI-resistant RCC cells. Ovatodiolide may be a potent β-catenin signaling inhibitor, with synergistic effects with sorafenib or sunitinib, and therefore, a useful candidate for improving RCC therapy.
Collapse
|
43
|
Wang X, Zhang L, O'Neill A, Bahamon B, Alsop DC, Mier JW, Goldberg SN, Signoretti S, Atkins MB, Wood CG, Bhatt RS. Cox-2 inhibition enhances the activity of sunitinib in human renal cell carcinoma xenografts. Br J Cancer 2013; 108:319-26. [PMID: 23322198 PMCID: PMC3566808 DOI: 10.1038/bjc.2012.591] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 12/07/2012] [Accepted: 12/07/2012] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Sunitinib (Su), a tyrosine kinase inhibitor of VEGFR, is effective at producing tumour response in clear cell renal cell carcinoma (cRCC), but resistance to therapy is inevitable. As COX-2 is a known mediator of tumour growth, we explored the potential benefit of COX-2 inhibition in combination with VEGFR inhibition in attempts at delaying tumour progression on Su. METHODS COX-2 expression was compared with areas of hypoxia in tumours that progressed on Su vs untreated tumours. Mice bearing human cRCC xenografts were treated with Su and the COX-2 inhibitor, celecoxib, and the effects on tumour growth were assessed. Sequential vs concurrent regimens were compared. RESULTS COX-2 expression was increased in cRCC xenografts in areas of tumour hypoxia. The combination of Su and celecoxib achieved longer times to tumour progression compared to treatment with either agent alone or to untreated control animals in four models. This effect was seen with concurrent but not with sequential therapy. CONCLUSION COX-2 inhibition can extend the effectiveness of VEGFR inhibition. This effect is dependent on the timing of therapy. Clinical trials combining Su and COX-2 inhibitors should be considered as a means delaying time to progression on sunitinib in patients with metastatic cRCC.
Collapse
Affiliation(s)
- X Wang
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | - L Zhang
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | - A O'Neill
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - B Bahamon
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - D C Alsop
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | - J W Mier
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | - S N Goldberg
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
- Department of Radiology, Hadassah Hebrew University Medical Center, PO Box 12000, Jerusalem 91120, Israel
| | - S Signoretti
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - M B Atkins
- Georgetown-Lombardi Comprehensive Cancer Center, 3970 Reservoir Road, NW, Washington, DC 20057, USA
| | - C G Wood
- Department of Urology, Division of Surgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe-Unit1373, Houston, TX 77030, USA
| | - R S Bhatt
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| |
Collapse
|
44
|
Sakai I, Miyake H, Fujisawa M. Acquired resistance to sunitinib in human renal cell carcinoma cells is mediated by constitutive activation of signal transduction pathways associated with tumour cell proliferation. BJU Int 2013; 112:E211-20. [PMID: 23305097 DOI: 10.1111/j.1464-410x.2012.11655.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
UNLABELLED WHAT'S KNOWN ON THE SUBJECT? AND WHAT DOES THE STUDY ADD?: Although there have been a few studies investigating the molecular mechanism mediating the acquisition of resistance to molecular-targeted agents, including sunitinib, by renal cell carcinoma (RCC) cells, this mechanism remains largely unclear. The maintenance of protein kinase activation during sunitinib treatment may be involved in the acquisition of a phenotype resistant to sunitinib in RCC, and additional treatment with agents targeting activated protein kinases could be a promising approach for overcoming resistance to sunitinib in RCC. OBJECTIVE To characterise the mechanism involved in the acquired resistance to sunitinib, a potential inhibitor of multiple receptor tyrosine kinases (RTKs), in renal cell carcinoma (RCC). MATERIALS AND METHODS A parental human RCC cell line, ACHN (ACHN/P), was continuously exposed to increasing doses of sunitinib, and a cell line resistant to sunitinib (ACHN/R), showing an ≈5-fold higher IC50 (concentration that reduces the effect by 50%) than that of ACHN/P, was developed. RESULTS ACHN/R appeared to acquire significant cross resistance to sorafenib; however, there were no significant differences in sensitivities to the Mammalian target of rapamycin inhibitors, temsirolimus and everolimus, between ACHN/P and ACHN/R. After sunitinib treatment, the expression levels of phosphorylated Akt and p44/42 mitogen-activated protein kinase in ACHN/P, but not those in ACHN/R, were significantly inhibited. RTK assay showed that treatment of ACHN/P with sunitinib resulted in the marked downregulation of several phosphorylated RTKs compared with that of ACHN/R. Additional treatment with a specific inhibitor of Akt significantly increased the sensitivity of ACHN/R to sunitinib, but not that of ACHN/P. There were no significant differences between in vivo growth patterns of ACHN/P and ACHN/R in mice before and after the administration of sunitinib; however, the proportion of cells positive for TUNEL (terminal deoxynucleotidyl transferase-mediated dUTP nick-end labelling) staining in ACHN/P tumour was significantly greater than that in ACHN/R tumour in mice treated with sunitinib. CONCLUSION The maintenance of protein kinase activation during sunitinib treatment may be involved in the acquisition of resistant phenotype to sunitinib in RCC cells.
Collapse
Affiliation(s)
- Iori Sakai
- Division of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | | | | |
Collapse
|
45
|
Abstract
INTRODUCTION The vascular endothelial growth factor (VEGF) pathway and the mammalian Target of Rapamycin (mTOR) represent the most frequently exploited targets in renal cell carcinoma (RCC). Temsirolimus is an inhibitor of mTOR, and is a unique ester derivative of sirolimus, a macrocyclic lactone, with improved pharmaceutical properties, including stability and solubility. Temsirolimus binds to the cytoplasmic protein FKBP-12, and the complex binds and inhibits mTOR. AREAS COVERED This review summarizes the clinical findings and safety of temsirolimus in RCC patients. EXPERT OPINION A Phase III clinical trial has demonstrated that temsirolimus has statistically significant advantages over treatment with IFN-α in RCC patients with poor prognosis, in terms of OS (overall survival), PFS (progression-free survival), and tumor response. Median OS was improved 49% compared to IFN-α, and median PFS was approximately doubled. It is now considered the standard for RCC patients with poor prognostic features. The possibility that this agent is useful in metastatic non-clear cell carcinoma patients has also been suggested by a subset analysis of the pivotal Phase III trial. Studies in untreated favorable and intermediate risk clear cell and refractory mRCC patients are required.
Collapse
|
46
|
Wang X, Zhang L, Goldberg SN, Bhasin M, Brown V, Alsop DC, Signoretti S, Mier JW, Atkins MB, Bhatt RS. High dose intermittent sorafenib shows improved efficacy over conventional continuous dose in renal cell carcinoma. J Transl Med 2011; 9:220. [PMID: 22188900 PMCID: PMC3258225 DOI: 10.1186/1479-5876-9-220] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Accepted: 12/21/2011] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Renal cell carcinoma (RCC) responds to agents that inhibit vascular endothelial growth factor (VEGF) pathway. Sorafenib, a multikinase inhibitor of VEGF receptor, is effective at producing tumor responses and delaying median progression free survival in patients with cytokine refractory RCC. However, resistance to therapy develops at a median of 5 months. In an effort to increase efficacy, we studied the effects of increased sorafenib dose and intermittent scheduling in a murine RCC xenograft model. METHODS Mice bearing xenografts derived from the 786-O RCC cell line were treated with sorafenib according to multiple doses and schedules: 1) Conventional dose (CD) continuous therapy; 2) high dose (HD) intermittent therapy, 3) CD intermittent therapy and 4) HD continuous therapy. Tumor diameter was measured daily. Microvessel density was assessed after 3 days to determine the early effects of therapy, and tumor perfusion was assessed serially by arterial spin labeled (ASL) MRI at day 0, 3, 7 and 10. RESULTS Tumors that were treated with HD sorafenib exhibited slowed tumor growth as compared to CD using either schedule. HD intermittent therapy was superior to CD continous therapy, even though the total dose of sorafenib was essentially equivalent, and not significantly different than HD continuous therapy. The tumors exposed to HD sorafenib had lower microvessel density than the untreated or the CD groups. ASL MRI showed that tumor perfusion was reduced to a greater extent with the HD sorafenib at day 3 and at all time points thereafter relative to CD therapy. Further the intermittent schedule appeared to maintain RCC sensitivity to sorafenib as determined by changes in tumor perfusion. CONCLUSIONS A modification of the sorafenib dosing schedule involving higher dose intermittent treatment appeared to improve its efficacy in this xenograft model relative to conventional dosing. MRI perfusion imaging and histologic analysis suggest that this benefit is related to enhanced and protracted antiangiogenic activity. Thus, better understanding of dosing and schedule issues may lead to improved therapeutic effectiveness of VEGF directed therapy in RCC and possibly other tumors.
Collapse
Affiliation(s)
- Xiaoen Wang
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Liang Zhang
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - S Nahum Goldberg
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Manoj Bhasin
- Division of Interdisciplinary Medicine and Biostatistics, Beth Israel Deaconess Medical Center, Harvard Medical School, Massachusetts, USA
| | - Victoria Brown
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - David C Alsop
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - James W Mier
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael B Atkins
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Rupal S Bhatt
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
47
|
Current World Literature. Curr Opin Support Palliat Care 2011; 5:297-305. [DOI: 10.1097/spc.0b013e32834a76ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
48
|
Epithelial-to-mesenchymal transition and acquired resistance to sunitinib in a patient with hepatocellular carcinoma. J Hepatol 2011; 54:1073-8. [PMID: 21145871 DOI: 10.1016/j.jhep.2010.11.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2010] [Revised: 11/03/2010] [Accepted: 11/22/2010] [Indexed: 01/13/2023]
Abstract
BACKGROUND & AIMS Based on the success of sorafenib, several anti-angiogenic therapies are currently evaluated in advanced hepatocellular carcinomas. Few biological data are currently available from patients that may help understanding mechanisms of acquired resistance to these drugs. Herein, we report translational data from a post-treatment surgical specimen in a patient who experienced acquired resistance to sunitinib. METHODS Clinical, radiological, and pathological data were collected before treatment, under treatment, and at the time of tumor progression. In addition, a biomolecular analysis was performed at the time of progression. RESULTS In this patient with non-alcoholic steatohepatitis, initial response to sunitinib was followed by tumor progression within 6 months of treatment, requesting salvage surgical resection. Surprisingly, pathological examination on post-treatment specimens revealed the presence of two juxtaposed tissue components containing either sarcomatoid-like mesenchymal cells or well- to moderately-differentiated hepatocellular carcinoma cells. Cancer cells retain a high α-fetoprotein expression in both components. However, while cells from carcinoma expressed E-cadherin but no vimentin, cancer cells from the mesenchymal component highly expressed vimentin and lost E-cadherin protein expression as measured by immunostaining. HMGA2 and Ki67 mRNA were also expressed at higher levels in mesenchymal than in carcinoma cells. CONCLUSION This case report suggests the occurrence of an epithelial-to-mesenchymal transition in discrete areas of hepatocellular carcinomas developing resistance to sunitinib.
Collapse
|
49
|
Zhang L, Bhasin M, Schor-Bardach R, Wang X, Collins MP, Panka D, Putheti P, Signoretti S, Alsop DC, Libermann T, Atkins MB, Mier JW, Goldberg SN, Bhatt RS. Resistance of renal cell carcinoma to sorafenib is mediated by potentially reversible gene expression. PLoS One 2011; 6:e19144. [PMID: 21559452 PMCID: PMC3084751 DOI: 10.1371/journal.pone.0019144] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Accepted: 03/27/2011] [Indexed: 12/29/2022] Open
Abstract
PURPOSE Resistance to antiangiogenic therapy is an important clinical problem. We examined whether resistance occurs at least in part via reversible, physiologic changes in the tumor, or results solely from stable genetic changes in resistant tumor cells. EXPERIMENTAL DESIGN Mice bearing two human RCC xenografts were treated with sorafenib until they acquired resistance. Resistant 786-O cells were harvested and reimplanted into naïve mice. Mice bearing resistant A498 cells were subjected to a 1 week treatment break. Sorafenib was then again administered to both sets of mice. Tumor growth patterns, gene expression, viability, blood vessel density, and perfusion were serially assessed in treated vs control mice. RESULTS Despite prior resistance, reimplanted 786-O tumors maintained their ability to stabilize on sorafenib in sequential reimplantation steps. A transcriptome profile of the tumors revealed that the gene expression profile of tumors upon reimplantation reapproximated that of the untreated tumors and was distinct from tumors exhibiting resistance to sorafenib. In A498 tumors, revascularization was noted with resistance and cessation of sorafenib therapy and tumor perfusion was reduced and tumor cell necrosis enhanced with re-exposure to sorafenib. CONCLUSIONS In two RCC cell lines, resistance to sorafenib appears to be reversible. These results support the hypothesis that resistance to VEGF pathway therapy is not solely the result of a permanent genetic change in the tumor or selection of resistant clones, but rather is due to a great extent to reversible changes that likely occur in the tumor and/or its microenvironment.
Collapse
Affiliation(s)
- Liang Zhang
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Manoj Bhasin
- Division of Interdisciplinary Medicine and Biotechnology, and Genomics and Proteomics Center, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Rachel Schor-Bardach
- Department of Radiology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
- Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Xiaoen Wang
- Department of Radiology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Michael P. Collins
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - David Panka
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Prabhakar Putheti
- Departments of Surgery and Medicine, The Transplant Institute at Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - David C. Alsop
- Department of Radiology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Towia Libermann
- Division of Interdisciplinary Medicine and Biotechnology, and Genomics and Proteomics Center, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Michael B. Atkins
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - James W. Mier
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - S. Nahum Goldberg
- Department of Radiology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
- Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Rupal S. Bhatt
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
- Kidney Cancer Program of the Dana Farber/Harvard Cancer Center, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|