1
|
Senrung A, Tripathi T, Aggarwal N, Janjua D, Chhokar A, Yadav J, Chaudhary A, Thakur K, Singh T, Bharti AC. Anti-angiogenic Potential of Trans-chalcone in an In Vivo Chick Chorioallantoic Membrane Model: An ATP Antagonist to VEGFR with Predicted Blood-brain Barrier Permeability. Cardiovasc Hematol Agents Med Chem 2024; 22:187-211. [PMID: 37936455 DOI: 10.2174/0118715257250417231019102501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 08/26/2023] [Accepted: 09/08/2023] [Indexed: 11/09/2023]
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is characterized by massive tumorinduced angiogenesis aiding tumorigenesis. Vascular endothelial growth factor A (VEGF-A) via VEGF receptor 2 (VEGFR-2) constitutes majorly to drive this process. Putting a halt to tumordriven angiogenesis is a major clinical challenge, and the blood-brain barrier (BBB) is the prime bottleneck in GBM treatment. Several phytochemicals show promising antiangiogenic activity across different models, but their ability to cross BBB remains unexplored. METHODS We screened over 99 phytochemicals having anti-angiogenic properties reported in the literature and evaluated them for their BBB permeability, molecular interaction with VEGFR-2 domains, ECD2-3 (extracellular domains 2-3) and TKD (tyrosine kinase domain) at VEGF-A and ATP binding site, cell membrane permeability, and hepatotoxicity using in silico tools. Furthermore, the anti-angiogenic activity of predicted lead Trans-Chalcone (TC) was evaluated in the chick chorioallantoic membrane. RESULTS Out of 99 phytochemicals, 35 showed an efficient ability to cross BBB with a probability score of > 0.8. Docking studies revealed 30 phytochemicals crossing benchmark binding affinity < -6.4 kcal/mol of TKD with the native ligand ATP alone. Out of 30 phytochemicals, 12 showed moderate to low hepatotoxicity, and 5 showed a violation of Lipinski's rule of five. Our in silico analysis predicted TC as a BBB permeable anti-angiogenic compound for use in GBM therapy. TC reduced vascularization in the CAM model, which was associated with the downregulation of VEGFR-2 transcript expression. CONCLUSION The present study showed TC to possess anti-angiogenic potential via the inhibition of VEGFR-2. In addition, the study predicted TC to cross BBB as well as a safe alternative for GBM therapy, which needs further investigation.
Collapse
Affiliation(s)
- Anna Senrung
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi (North Campus), Delhi, 110007, India
- Neuropharmacology & Drug Delivery Laboratory, Zoology Department, Daulat Ram College, University of Delhi, Delhi, 110007, India
| | - Tanya Tripathi
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi (North Campus), Delhi, 110007, India
| | - Nikita Aggarwal
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi (North Campus), Delhi, 110007, India
| | - Divya Janjua
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi (North Campus), Delhi, 110007, India
| | - Arun Chhokar
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi (North Campus), Delhi, 110007, India
- Department of Zoology, Deshbandhu College, University of Delhi, Delhi, 110019, India
| | - Joni Yadav
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi (North Campus), Delhi, 110007, India
| | - Apoorva Chaudhary
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi (North Campus), Delhi, 110007, India
| | - Kulbhushan Thakur
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi (North Campus), Delhi, 110007, India
| | - Tejveer Singh
- Department of Zoology, Hansraj College, University of Delhi, Delhi, India
| | - Alok Chandra Bharti
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi (North Campus), Delhi, 110007, India
| |
Collapse
|
2
|
Senrung A, Tripathi T, Yadav J, Janjua D, Chaudhary A, Chhokar A, Aggarwal N, Joshi U, Goswami N, Bharti AC. In vivo antiangiogenic effect of nimbolide, trans-chalcone and piperine for use against glioblastoma. BMC Cancer 2023; 23:1173. [PMID: 38036978 PMCID: PMC10691152 DOI: 10.1186/s12885-023-11625-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 11/09/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND Angiogenesis is an important hallmark of Glioblastoma (GBM) marked by elevated vascular endothelial growth factor-A (VEGF-A) and its receptor 2 (VEGFR-2). As previously reported nimbolide (NBL), trans-chalcone (TC) and piperine (PPR) possess promising antiangiogenic activity in several cancers however, their comparative efficacy and mechanism of antiangiogenic activity in GBM against VEGFR-2 has not been elucidated. METHODS 2D and 3D spheroids cultures of U87 (Uppsala 87 Malignant Glioma) were used for evaluation of non-cytotxoic dose for anti-angiogenic activity. The antiangiogenic effect was investigated by the GBM U87 cell line bearing chick CAM model. Excised U87 xenografts were histologically examined for blood vascular density by histochemistry. Reverse transcriptase polymerase chain reaction (RT-PCR) was used to detect the presence of avian and human VEGF-A and VEGFR-2 mRNA transcripts. RESULTS Using 2D and 3D spheroid models, the non-cytotoxic dose of NBL, TC and PPR was ≤ 11 µM. We found NBL, TC and PPR inhibit U87-induced neoangiogenesis in a dose-dependent manner in the CAM stand-alone model as well as in CAM U87 xenograft model. The results also indicate that these natural compounds inhibit the expression of notable angiogenic factors, VEGF-A and VEGFR-2. A positive correlation was found between blood vascular density and VEGF-A as well as VEGFR-2 transcripts. CONCLUSION Taken together, NBL, TC and PPR can suppress U87-induced neoangiogenesis via a reduction in VEGF-A and its receptor VEGFR-2 transcript expression at noncytotoxic concentrations. These phytochemicals showed their utility as adjuvants to GBM therapy, with Piperine demonstrating superior effectiveness among them all.
Collapse
Affiliation(s)
- Anna Senrung
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
- Neuropharmacology and Drug Delivery Laboratory, Daulat Ram College, University of Delhi, Delhi, India
| | - Tanya Tripathi
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Joni Yadav
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Divya Janjua
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Apoorva Chaudhary
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Arun Chhokar
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
- Deshbandhu College, University of Delhi, Delhi, India
| | - Nikita Aggarwal
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Udit Joshi
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Nidhi Goswami
- Neuropharmacology and Drug Delivery Laboratory, Daulat Ram College, University of Delhi, Delhi, India
| | - Alok Chandra Bharti
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India.
| |
Collapse
|
3
|
Khaki Bakhtiarvand V, Ramezani-Ali Akbari K, Amir Jalali S, Hojjat-Farsangi M, Jeddi-Tehrani M, Shokri F, Shabani M. Myeloid-derived suppressor cells (MDSCs) depletion by cabozantinib improves the efficacy of anti-HER2 antibody-based immunotherapy in a 4T1-HER2 murine breast cancer model. Int Immunopharmacol 2022; 113:109470. [DOI: 10.1016/j.intimp.2022.109470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 11/10/2022] [Accepted: 11/14/2022] [Indexed: 11/24/2022]
|
4
|
Santoni M, Iacovelli R, Colonna V, Klinz S, Mauri G, Nuti M. Antitumor effects of the multi-target tyrosine kinase inhibitor cabozantinib: a comprehensive review of the preclinical evidence. Expert Rev Anticancer Ther 2021; 21:1029-1054. [PMID: 34445927 DOI: 10.1080/14737140.2021.1919090] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Altered receptor tyrosine kinase (RTK) signaling contributes to tumorigenesis and suppression of immune-mediated destruction of cancer cells. Cabozantinib is an oral tyrosine kinase inhibitor that inhibits several RTKs involved in tumorigenesis, and is approved for the treatment of patients with progressive metastatic medullary thyroid cancer, advanced renal cell carcinoma, and hepatocellular carcinoma that has been previously treated with sorafenib. AREAS COVERED We present an up-to-date evaluation of preclinical evidence for RTK inhibition with cabozantinib, specifically VEGFR, MET, KIT, RET, AXL, FLT3, and associated antitumor effects. Preclinical investigations of cabozantinib in combination with other anticancer drugs are also reviewed. EXPERT OPINION Preclinical evidence shows that cabozantinib has antitumor activity against various cancer cells and exhibits synergy with other anticancer agents, including immune checkpoint inhibitors and hormone receptor or metabolic pathway inhibitors. Further optimization of cabozantinib treatment requires the identification of biomarkers of response and resistance, and exploration of complementary drug targets. Investigation of mechanisms of adaptive resistance, such as epithelial to mesenchymal transition (cancer intrinsic) and immunomodulation by the tumor microenvironment (cancer extrinsic), as well as identification of novel drug targets based on characterization of cancer stem cell metabolomic phenotypes, appear to be promising approaches.
Collapse
Affiliation(s)
- Matteo Santoni
- MD, U.O.C. Medical Oncology, Macerata Hospital, Macerata, Italy
| | - Roberto Iacovelli
- Medical Oncologist, Medical Oncology Unit, Fondazione Policlinico Agostino Gemelli IRCCS, Roma, Italy
| | - Valentina Colonna
- Global Medical Development Director, Global Oncology R&D, Ipsen S.p.A., Milano, Italy
| | - Stephan Klinz
- Senior Director, Translational Medicine & Biomarkers, Ipsen, United States, MA, USA
| | - Giorgio Mauri
- Medical Advisor Oncology, Ipsen S.p.A., Milano, Italy
| | - Marianna Nuti
- Professor, Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
5
|
Estaras M, Gonzalez A. Modulation of cell physiology under hypoxia in pancreatic cancer. World J Gastroenterol 2021; 27:4582-4602. [PMID: 34366624 PMCID: PMC8326256 DOI: 10.3748/wjg.v27.i28.4582] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/28/2021] [Accepted: 06/22/2021] [Indexed: 02/06/2023] Open
Abstract
In solid tumors, the development of vasculature is, to some extent, slower than the proliferation of the different types of cells that form the tissue, both cancer and stroma cells. As a consequence, the oxygen availability is compromised and the tissue evolves toward a condition of hypoxia. The presence of hypoxia is variable depending on where the cells are localized, being less extreme at the periphery of the tumor and more severe in areas located deep within the tumor mass. Surprisingly, the cells do not die. Intracellular pathways that are critical for cell fate such as endoplasmic reticulum stress, apoptosis, autophagy, and others are all involved in cellular responses to the low oxygen availability and are orchestrated by hypoxia-inducible factor. Oxidative stress and inflammation are critical conditions that develop under hypoxia. Together with changes in cellular bioenergetics, all contribute to cell survival. Moreover, cell-to-cell interaction is established within the tumor such that cancer cells and the microenvironment maintain a bidirectional communication. Additionally, the release of extracellular vesicles, or exosomes, represents short and long loops that can convey important information regarding invasion and metastasis. As a result, the tumor grows and its malignancy increases. Currently, one of the most lethal tumors is pancreatic cancer. This paper reviews the most recent advances in the knowledge of how cells grow in a pancreatic tumor by adapting to hypoxia. Unmasking the physiological processes that help the tumor increase its size and their regulation will be of major relevance for the treatment of this deadly tumor.
Collapse
Affiliation(s)
- Matias Estaras
- Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, Caceres 10003, Spain
| | - Antonio Gonzalez
- Department of Physiology, Cell Biology and Communication Research Group, University of Extremadura, Caceres 10003, Spain
| |
Collapse
|
6
|
Lue HW, Derrick DS, Rao S, Van Gaest A, Cheng L, Podolak J, Lawson S, Xue C, Garg D, White R, Ryan CW, Drake JM, Ritz A, Heiser LM, Thomas GV. Cabozantinib and dasatinib synergize to induce tumor regression in non-clear cell renal cell carcinoma. Cell Rep Med 2021; 2:100267. [PMID: 34095877 PMCID: PMC8149375 DOI: 10.1016/j.xcrm.2021.100267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 01/18/2021] [Accepted: 04/13/2021] [Indexed: 12/24/2022]
Abstract
The lack of effective treatment options for advanced non-clear cell renal cell carcinoma (NCCRCC) is a critical unmet clinical need. Applying a high-throughput drug screen to multiple human kidney cancer cells, we identify the combination of the VEGFR-MET inhibitor cabozantinib and the SRC inhibitor dasatinib acts synergistically in cells to markedly reduce cell viability. Importantly, the combination is well tolerated and causes tumor regression in vivo. Transcriptional and phosphoproteomic profiling reveals that the combination converges to downregulate the MAPK-ERK signaling pathway, a result not predicted by single-agent analysis alone. Correspondingly, the addition of a MEK inhibitor synergizes with either dasatinib or cabozantinib to increase its efficacy. This study, by using approved, clinically relevant drugs, provides the rationale for the design of effective combination treatments in NCCRCC that can be rapidly translated to the clinic.
Collapse
Affiliation(s)
- Hui-wen Lue
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Daniel S. Derrick
- Department of Biomedical Engineering, Oregon Health and Science University Center for Spatial Systems Biomedicine, Portland, OR, USA
| | - Soumya Rao
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Ahna Van Gaest
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Larry Cheng
- Graduate Program in Quantitative Biomedicine, Rutgers University, Piscataway, NJ, USA
| | - Jennifer Podolak
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Samantha Lawson
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Changhui Xue
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Devin Garg
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Ralph White
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Christopher W. Ryan
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
- Division of Hematology and Oncology, Oregon Health and Science University, Portland, OR, USA
| | - Justin M. Drake
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
- Department of Urology, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Anna Ritz
- Department of Biology, Reed College, Portland, OR, USA
| | - Laura M. Heiser
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
- Department of Biomedical Engineering, Oregon Health and Science University Center for Spatial Systems Biomedicine, Portland, OR, USA
| | - George V. Thomas
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
- Department of Pathology and Laboratory Medicine, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
7
|
Control of renal cell carcinoma brain metastases with cabozantinib following progression on immune checkpoint inhibitor therapy. CURRENT PROBLEMS IN CANCER: CASE REPORTS 2021. [DOI: 10.1016/j.cpccr.2021.100060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
8
|
Rathi N, McFarland TR, Nussenzveig R, Agarwal N, Swami U. Evolving Role of Immunotherapy in Metastatic Castration Refractory Prostate Cancer. Drugs 2020; 81:191-206. [PMID: 33369720 PMCID: PMC7932934 DOI: 10.1007/s40265-020-01456-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Immunotherapies have shown remarkable success in the treatment of multiple cancer types; however, despite encouraging preclinical activity, registration trials of immunotherapy in prostate cancer have largely been unsuccessful. Sipuleucel-T remains the only approved immunotherapy for the treatment of asymptomatic or minimally symptomatic metastatic castrate-resistant prostate cancer based on modest improvement in overall survival. This immune evasion in the case of prostate cancer has been attributed to tumor-intrinsic factors, an immunosuppressive tumor microenvironment, and host factors, which ultimately make it an inert 'cold' tumor. Recently, multiple approaches have been investigated to turn prostate cancer into a 'hot' tumor. Antibodies directed against programmed cell death protein 1 have a tumor agnostic approval for a small minority of patients with microsatellite instability-high or mismatch repair-deficient metastatic prostate cancer. Herein, we present an overview of the current immunotherapy landscape in metastatic castration-resistant prostate cancer with a focus on immune checkpoint inhibitors. We describe the results of clinical trials of immune checkpoint inhibitors in patients with metastatic castration-resistant prostate cancer; either as single agents or in combination with other checkpoint inhibitors, poly (ADP-ribose) polymerase (PARP) inhibitors, tyrosine kinase inhibitors, novel hormonal therapies, chemotherapies, and radioligands. Finally, we review upcoming immunotherapies, including novel monoclonal antibodies, chimeric-antigen receptor (CAR) T cells, Bi-Specific T cell Engagers (BiTEs), therapies targeting the adenosine pathway, and other miscellaneous agents.
Collapse
Affiliation(s)
- Nityam Rathi
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope Drive Suite 5726, Salt Lake City, UT, 84112, USA
| | - Taylor Ryan McFarland
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope Drive Suite 5726, Salt Lake City, UT, 84112, USA
| | - Roberto Nussenzveig
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope Drive Suite 5726, Salt Lake City, UT, 84112, USA
| | - Neeraj Agarwal
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope Drive Suite 5726, Salt Lake City, UT, 84112, USA
| | - Umang Swami
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope Drive Suite 5726, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
9
|
Tu J, Fang Y, Han D, Tan X, Jiang H, Gong X, Wang X, Hong W, Wei W. Activation of nuclear factor-κB in the angiogenesis of glioma: Insights into the associated molecular mechanisms and targeted therapies. Cell Prolif 2020; 54:e12929. [PMID: 33300633 PMCID: PMC7848966 DOI: 10.1111/cpr.12929] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/28/2020] [Accepted: 10/01/2020] [Indexed: 02/06/2023] Open
Abstract
Glioma is the most commonly observed primary intracranial tumour and is associated with massive angiogenesis. Glioma neovascularization provides nutrients for the growth and metabolism of tumour tissues, promotes tumour cell division and proliferation, and provides conditions ideal for the infiltration and migration of tumour cells to distant places. Growing evidence suggests that there is a correlation between the activation of nuclear factor (NF)‐κB and the angiogenesis of glioma. In this review article, we highlighted the functions of NF‐κB in the angiogenesis of glioma, showing that NF‐κB activation plays a pivotal role in the growth and progression of glioma angiogenesis and is a rational therapeutic target for antiangiogenic strategies aimed at glioma.
Collapse
Affiliation(s)
- Jiajie Tu
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Hefei, China
| | - Yilong Fang
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Hefei, China
| | - Dafei Han
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Hefei, China
| | - Xuewen Tan
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Hefei, China
| | - Haifeng Jiang
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Hefei, China
| | - Xun Gong
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Hefei, China
| | - Xinming Wang
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wenming Hong
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Hefei, China
| |
Collapse
|
10
|
Pezzella F, Ribatti D. Vascular co-option and vasculogenic mimicry mediate resistance to antiangiogenic strategies. Cancer Rep (Hoboken) 2020; 5:e1318. [PMID: 33295149 PMCID: PMC9780428 DOI: 10.1002/cnr2.1318] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 10/13/2020] [Accepted: 10/21/2020] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The concept that all the tumors need the formation of new vessels to grow inspired the hypothesis that inhibition of angiogenesis would have led to "cure" cancer. The expectancy that this type of therapy would have avoided the insurgence of resistance was based on the concept that targeting normal vessels, instead of the cancer cells which easily develop new mutations, would have allowed evasion of drug caused selection is, however, more complex as it was made apparent by the discovery of nonangiogenic tumors. At the same time an increasing number of trials with antiangiogenic drugs were coming out as not as successful as expected, mostly because of the appearance of unexpected resistance. RECENT FINDINGS Among the several different mechanisms of resistance to antiangiogenic treatment by now described, we review the evidences that vascular co-option and vasculogenic mimicry by nonangiogenic tumors are effectively two of such mechanisms. We focused on reviewing exclusively the study, both clinical and preclinical, that offer a demonstration that vascular co-option and vasculogenic mimicry are effectively two mechanisms of both intrinsic and acquired resistance. CONCLUSION The discovery that vascular co-opting and vasculogenic mimicry are two ways of escaping antiangiogenic treatment, prompts the need for a better understanding of this phenomenon in order to improve cancer treatment.
Collapse
Affiliation(s)
- Francesco Pezzella
- Nuffield Division of Laboratory Science, Radcliffe Department of MedicineJohn Radcliffe Hospital, University of OxfordOxfordUK
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory OrgansUniversity of Bari Medical SchoolBariItaly
| |
Collapse
|
11
|
Campanella R, Guarnaccia L, Caroli M, Zarino B, Carrabba G, La Verde N, Gaudino C, Rampini A, Luzzi S, Riboni L, Locatelli M, Navone SE, Marfia G. Personalized and translational approach for malignant brain tumors in the era of precision medicine: the strategic contribution of an experienced neurosurgery laboratory in a modern neurosurgery and neuro-oncology department. J Neurol Sci 2020; 417:117083. [PMID: 32784071 DOI: 10.1016/j.jns.2020.117083] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/16/2020] [Accepted: 08/04/2020] [Indexed: 12/20/2022]
Abstract
Personalized medicine (PM) aims to optimize patient management, taking into account the individual traits of each patient. The main purpose of PM is to obtain the best response, improving health care and lowering costs. Extending traditional approaches, PM introduces novel patient-specific paradigms from diagnosis to treatment, with greater precision. In neuro-oncology, the concept of PM is well established. Indeed, every neurosurgical intervention for brain tumors has always been highly personalized. In recent years, PM has been introduced in neuro-oncology also to design and prescribe specific therapies for the patient and the patient's tumor. The huge advances in basic and translational research in the fields of genetics, molecular and cellular biology, transcriptomics, proteomics, and metabolomics have led to the introduction of PM into clinical practice. The identification of a patient's individual variation map may allow to design selected therapeutic protocols that ensure successful outcomes and minimize harmful side effects. Thus, clinicians can switch from the "one-size-fits-all" approach to PM, ensuring better patient care and high safety margin. Here, we review emerging trends and the current literature about the development of PM in neuro-oncology, considering the positive impact of innovative advanced researches conducted by a neurosurgical laboratory.
Collapse
Affiliation(s)
- Rolando Campanella
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Laura Guarnaccia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Manuela Caroli
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Barbara Zarino
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giorgio Carrabba
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Chiara Gaudino
- Department of Neuroradiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Angela Rampini
- Neurosurgery Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Sabino Luzzi
- Neurosurgery Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy; Neurosurgery Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Laura Riboni
- Department of Medical Biotechnology and Translational Medicine, LITA-Segrate, University of Milan, Milan, Italy
| | - Marco Locatelli
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Aldo Ravelli" Research Center, Milan, Italy; Department of Medical-Surgical Physiopathology and Transplantation, University of Milan, Milan, Italy
| | - Stefania Elena Navone
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Aldo Ravelli" Research Center, Milan, Italy.
| | - Giovanni Marfia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Aldo Ravelli" Research Center, Milan, Italy; Clinical Pathology Unit, Istituto di Medicina Aerospaziale "A. Moosso", Aeronautica Militare, Milan, Italy
| |
Collapse
|
12
|
Montemagno C, Pagès G. Resistance to Anti-angiogenic Therapies: A Mechanism Depending on the Time of Exposure to the Drugs. Front Cell Dev Biol 2020; 8:584. [PMID: 32775327 PMCID: PMC7381352 DOI: 10.3389/fcell.2020.00584] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis, the formation of new blood vessels from preexisting one, represents a critical process for oxygen and nutrient supply to proliferating cells, therefore promoting tumor growth and metastasis. The Vascular Endothelial Growth Factor (VEGF) pathway is one of the key mediators of angiogenesis in cancer. Therefore, several therapies including monoclonal antibodies or tyrosine kinase inhibitors target this axis. Although preclinical studies demonstrated strong antitumor activity, clinical studies were disappointing. Antiangiogenic drugs, used to treat metastatic patients suffering of different types of cancers, prolonged survival to different extents but are not curative. In this review, we focused on different mechanisms involved in resistance to antiangiogenic therapies from early stage resistance involving mainly tumor cells to late stages related to the adaptation of the microenvironment.
Collapse
Affiliation(s)
- Christopher Montemagno
- Département de Biologie Médicale, Centre Scientifique de Monaco, Monaco, Monaco.,CNRS UMR 7284, Institute for Research on Cancer and Aging of Nice, Université Côte d'Azur, Nice, France.,INSERM U1081, Centre Antoine Lacassagne, Nice, France
| | - Gilles Pagès
- Département de Biologie Médicale, Centre Scientifique de Monaco, Monaco, Monaco.,CNRS UMR 7284, Institute for Research on Cancer and Aging of Nice, Université Côte d'Azur, Nice, France.,INSERM U1081, Centre Antoine Lacassagne, Nice, France
| |
Collapse
|
13
|
Saman H, Raza SS, Uddin S, Rasul K. Inducing Angiogenesis, a Key Step in Cancer Vascularization, and Treatment Approaches. Cancers (Basel) 2020; 12:cancers12051172. [PMID: 32384792 PMCID: PMC7281705 DOI: 10.3390/cancers12051172] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 04/10/2020] [Accepted: 04/17/2020] [Indexed: 12/27/2022] Open
Abstract
Angiogenesis is a term that describes the formation of new blood and lymphatic vessels from a pre-existing vasculature. This allows tumour cells to acquire sustenance in the form of nutrients and oxygen and the ability to evacuate metabolic waste. As one of the hallmarks of cancer, angiogenesis has been studied extensively in animal and human models to enable better understanding of cancer biology and the development of new anti-cancer treatments. Angiogenesis plays a crucial role in the process of tumour genesis, because solid tumour need a blood supply if they are to grow beyond a few millimeters in size. On the other hand, there is growing evidence that some solid tumour exploit existing normal blood supply and do not require a new vessel formation to grow and to undergo metastasis. This review of the literature will present the current understanding of this intricate process and the latest advances in the use of angiogenesis-targeting therapies in the fight against cancer.
Collapse
Affiliation(s)
- Harman Saman
- Barts Cancer Institute, Queen Mary University of London, London E1 4NS, UK
- Department of Medicine, Hazm Maubrairek Hospital, Ar-Rayyan PO Box 305, Qatar
- Correspondence: or ; Tel.: +97-466506781
| | - Syed Shadab Raza
- Department of Stem Cell Biology and Regenerative Medicine, ERA University, Lucknow 226003, India;
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar;
| | - Kakil Rasul
- National Cancer Care and Research, Hamad Medical Corporation, Doha 3050, Qatar;
| |
Collapse
|
14
|
Vessel co-option and resistance to anti-angiogenic therapy. Angiogenesis 2019; 23:55-74. [PMID: 31865479 DOI: 10.1007/s10456-019-09698-6] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/22/2019] [Indexed: 12/20/2022]
Abstract
Vessel co-option is a non-angiogenic mechanism of tumour vascularisation in which cancer cells utilise pre-existing blood vessels instead of inducing new blood vessel formation. Vessel co-option has been observed across a range of different tumour types, in both primary cancers and metastatic disease. Importantly, vessel co-option is now implicated as a major mechanism that mediates resistance to conventional anti-angiogenic drugs and this may help to explain the limited efficacy of this therapeutic approach in certain clinical settings. This includes the use of anti-angiogenic drugs to treat advanced-stage/metastatic disease, treatment in the adjuvant setting and the treatment of primary disease. In this article, we review the available evidence linking vessel co-option with resistance to anti-angiogenic therapy in numerous tumour types, including breast, colorectal, lung and pancreatic cancer, glioblastoma, melanoma, hepatocellular carcinoma, and renal cell carcinoma. The finding that vessel co-option is a significant mechanism of resistance to anti-angiogenic therapy may have important implications for the future of anti-cancer therapy, including (a) predicting response to anti-angiogenic drugs, (b) the need to develop therapies that target both angiogenesis and vessel co-option in tumours, and (c) predicting the response to other therapeutic modalities, including immunotherapy.
Collapse
|
15
|
Kuczynski EA, Vermeulen PB, Pezzella F, Kerbel RS, Reynolds AR. Vessel co-option in cancer. Nat Rev Clin Oncol 2019; 16:469-493. [PMID: 30816337 DOI: 10.1038/s41571-019-0181-9] [Citation(s) in RCA: 262] [Impact Index Per Article: 52.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
All solid tumours require a vascular supply in order to progress. Although the ability to induce angiogenesis (new blood vessel growth) has long been regarded as essential to this purpose, thus far, anti-angiogenic therapies have shown only modest efficacy in patients. Importantly, overshadowed by the literature on tumour angiogenesis is a long-standing, but continually emerging, body of research indicating that tumours can grow instead by hijacking pre-existing blood vessels of the surrounding nonmalignant tissue. This process, termed vessel co-option, is a frequently overlooked mechanism of tumour vascularization that can influence disease progression, metastasis and response to treatment. In this Review, we describe the evidence that tumours located at numerous anatomical sites can exploit vessel co-option. We also discuss the proposed molecular mechanisms involved and the multifaceted implications of vessel co-option for patient outcomes.
Collapse
Affiliation(s)
- Elizabeth A Kuczynski
- Bioscience, Oncology, IMED Biotech Unit, AstraZeneca, Cambridge, UK. .,Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Canada.
| | - Peter B Vermeulen
- HistoGeneX, Antwerp, Belgium.,Translational Cancer Research Unit, GZA Hospitals St Augustinus, University of Antwerp, Wilrijk-Antwerp, Belgium.,Tumour Biology Team, Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Francesco Pezzella
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Robert S Kerbel
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Andrew R Reynolds
- Tumour Biology Team, Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK. .,Oncology Translational Medicine Unit, IMED Biotech Unit, AstraZeneca, Cambridge, UK.
| |
Collapse
|
16
|
Lenting K, van den Heuvel CNAM, van Ewijk A, ElMelik D, de Boer R, Tindall E, Wei G, Kusters B, te Dorsthorst M, ter Laan M, Huynen MA, Leenders WP. Mapping actionable pathways and mutations in brain tumours using targeted RNA next generation sequencing. Acta Neuropathol Commun 2019; 7:185. [PMID: 31747973 PMCID: PMC6865071 DOI: 10.1186/s40478-019-0826-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 10/02/2019] [Indexed: 01/28/2023] Open
Abstract
Many biology-based precision drugs are available that neutralize aberrant molecular pathways in cancer. Molecular heterogeneity and the lack of reliable companion diagnostic biomarkers for many drugs makes targeted treatment of cancer inaccurate for many individuals. Identifying actionable hyperactive biological pathways in individual cancers may improve this situation. To achieve this we applied a novel targeted RNA next generation sequencing (t/RNA-NGS) technique to surgically obtained glioma tissues. The test combines mutation detection with analysis of biological pathway activities that are involved in tumour behavior in many cancer types (e.g. tyrosine kinase signaling, angiogenesis signaling, immune response, metabolism), via quantitative measurement of transcript levels and splice variants of hundreds of genes. We here present proof of concept that the technique, which uses molecular inversion probes, generates a histology-independent molecular diagnosis and identifies classifiers that are strongly associated with conventional histopathology diagnoses and even with patient prognosis. The test not only confirmed known glioma-associated molecular aberrations but also identified aberrant expression levels of actionable genes and mutations that have so far been considered not to be associated with glioma, opening up the possibility of drug repurposing for individual patients. Its cost-effectiveness makes t/RNA-NGS to an attractive instrument to aid oncologists in therapy decision making.
Collapse
|
17
|
Pathological features of vessel co-option versus sprouting angiogenesis. Angiogenesis 2019; 23:43-54. [DOI: 10.1007/s10456-019-09690-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 10/05/2019] [Indexed: 12/19/2022]
|
18
|
Ellingson BM, Aftab DT, Schwab GM, Hessel C, Harris RJ, Woodworth DC, Leu K, Chakhoyan A, Raymond C, Drappatz J, de Groot J, Prados MD, Reardon DA, Schiff D, Chamberlain M, Mikkelsen T, Desjardins A, Holland J, Ping J, Weitzman R, Wen PY, Cloughesy TF. Volumetric response quantified using T1 subtraction predicts long-term survival benefit from cabozantinib monotherapy in recurrent glioblastoma. Neuro Oncol 2019; 20:1411-1418. [PMID: 29660005 DOI: 10.1093/neuonc/noy054] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background To overcome challenges with traditional response assessment in anti-angiogenic agents, the current study uses T1 subtraction maps to quantify volumetric radiographic response in monotherapy with cabozantinib, an orally bioavailable tyrosine kinase inhibitor with activity against vascular endothelial growth factor receptor 2 (VEGFR2), hepatocyte growth factor receptor (MET), and AXL, in an open-label, phase II trial in patients with recurrent glioblastoma (GBM) (NCT00704288). Methods A total of 108 patients with adequate imaging data and confirmed recurrent GBM were included in this retrospective study from a phase II multicenter trial of cabozantinib monotherapy (XL184-201) at either 100 mg (N = 87) or 140 mg (N = 21) per day. Contrast enhanced T1-weighted digital subtraction maps were used to define volume of contrast-enhancing tumor at baseline and subsequent follow-up time points. Volumetric radiographic response (>65% reduction in contrast-enhancing tumor volume from pretreatment baseline tumor volume sustained for more than 4 wk) was tested as an independent predictor of overall survival (OS). Results Volumetric response rate for all therapeutic doses was 38.9% (41.4% and 28.6% for 100 mg and 140 mg doses, respectively). A log-linear association between baseline tumor volume and OS (P = 0.0006) and a linear correlation between initial change in tumor volume and OS (P = 0.0256) were observed. A significant difference in OS was observed between responders (median OS = 20.6 mo) and nonresponders (median OS = 8.0 mo) (hazard ratio [HR] = 0.3050, P < 0.0001). Multivariable analyses showed that continuous measures of baseline tumor volume (HR = 1.0233, P < 0.0001) and volumetric response (HR = 0.2240, P < 0.0001) were independent predictors of OS. Conclusions T1 subtraction maps provide value in determining response in recurrent GBM treated with cabozantinib and correlated with survival benefit.
Collapse
Affiliation(s)
- Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | | | | | | | - Robert J Harris
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Departments of Radiological Sciences and Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Davis C Woodworth
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Departments of Radiological Sciences and Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Kevin Leu
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Departments of Radiological Sciences and Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Ararat Chakhoyan
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Departments of Radiological Sciences and Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Catalina Raymond
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Departments of Radiological Sciences and Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Jan Drappatz
- Department of Neurology and Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - John de Groot
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael D Prados
- Department of Neurosurgery, University of California San Francisco (UCSF), San Francisco, California
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - David Schiff
- Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia
| | - Marc Chamberlain
- Department of Neurology, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | - Annick Desjardins
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | | | - Jerry Ping
- Exelixis, South San Francisco, California
| | | | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Timothy F Cloughesy
- UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
19
|
Clavreul A, Roger E, Pourbaghi-Masouleh M, Lemaire L, Tétaud C, Menei P. Development and characterization of sorafenib-loaded lipid nanocapsules for the treatment of glioblastoma. Drug Deliv 2019; 25:1756-1765. [PMID: 30338715 PMCID: PMC6225440 DOI: 10.1080/10717544.2018.1507061] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Anticancer agents that target both tumor cells and angiogenesis are of potential interest for glioblastoma (GB) therapy. One such agent is sorafenib (SFN), a tyrosine kinase inhibitor. However, poor aqueous solubility and undesirable side effects limit its clinical application, including local treatment. We encapsulated SFN in lipid nanocapsules (LNCs) to overcome these drawbacks. LNCs are nanocarriers formulated according to a solvent-free process, using only components that have received regulatory approval. SFN-LNCs had a diameter of 54 ± 1 nm, high encapsulation efficiency (>90%), and a drug payload of 2.11 ± 0.03 mg/g of LNC dispersion. They inhibited in vitro angiogenesis and decreased human U87MG GB cell viability similarly to free SFN. In vivo studies showed that the intratumoral administration of SFN-LNCs or free SFN in nude mice bearing an orthotopic U87MG human GB xenograft decreased the proportion of proliferating cells in the tumor relative to control groups. SFN-LNCs were more effective than free SFN for inducing early tumor vascular normalization, characterized by increases in tumor blood flow and decreases in tumor vessel area. These results highlight the potential of LNCs as delivery systems for SFN. The vascular normalization induced by SFN-LNCs could be used to improve the efficacy of chemotherapy or radiotherapy for treating GB.
Collapse
Affiliation(s)
- Anne Clavreul
- a Département de Neurochirurgie , CHU , Angers , France.,b CRCINA, INSERM , Université de Nantes, Université d'Angers , Angers , France
| | - Emilie Roger
- c MINT, INSERM 1066, CNRS 6021 , Université d'Angers, UNIV Angers , Angers , France
| | - Milad Pourbaghi-Masouleh
- b CRCINA, INSERM , Université de Nantes, Université d'Angers , Angers , France.,d Division of Drug Delivery and Tissue Engineering, School of Pharmacy , University of Nottingham , Nottingham , UK
| | - Laurent Lemaire
- c MINT, INSERM 1066, CNRS 6021 , Université d'Angers, UNIV Angers , Angers , France.,e PRISM-IRM , UNIV Angers , Angers , France
| | - Clément Tétaud
- b CRCINA, INSERM , Université de Nantes, Université d'Angers , Angers , France
| | - Philippe Menei
- a Département de Neurochirurgie , CHU , Angers , France.,b CRCINA, INSERM , Université de Nantes, Université d'Angers , Angers , France
| |
Collapse
|
20
|
Cheng F, Guo D. MET in glioma: signaling pathways and targeted therapies. J Exp Clin Cancer Res 2019; 38:270. [PMID: 31221203 PMCID: PMC6585013 DOI: 10.1186/s13046-019-1269-x] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 06/04/2019] [Indexed: 12/14/2022] Open
Abstract
Gliomas represent the most common type of malignant brain tumor, among which, glioblastoma remains a clinical challenge with limited treatment options and dismal prognosis. It has been shown that the dysregulated receptor tyrosine kinase (RTK, including EGFR, MET, PDGFRα, ect.) signaling pathways have pivotal roles in the progression of gliomas, especially glioblastoma. Increasing evidence suggests that expression levels of the RTK MET and its specific stimulatory factors are significantly increased in glioblastomas compared to those in normal brain tissues, whereas some negative regulators are found to be downregulated. Mutations in MET, as well as the dysregulation of other regulators of cross-talk with MET signaling pathways, have also been identified. MET and its ligand hepatocyte growth factor (HGF) play a critical role in the proliferation, survival, migration, invasion, angiogenesis, stem cell characteristics, and therapeutic resistance and recurrence of glioblastomas. Therefore, combined targeted therapy for this pathway and associated molecules could be a novel and attractive strategy for the treatment of human glioblastoma. In this review, we highlight progress made in the understanding of MET signaling in glioma and advances in therapies targeting HGF/MET molecules for glioma patients in recent years, in addition to studies on the expression and mutation status of MET.
Collapse
Affiliation(s)
- Fangling Cheng
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Avenue, Wuhan, 430030 China
| | - Dongsheng Guo
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Avenue, Wuhan, 430030 China
| |
Collapse
|
21
|
van den Heuvel CNAM, van Ewijk A, Zeelen C, de Bitter T, Huynen M, Mulders P, Oosterwijk E, Leenders WPJ. Molecular Profiling of Druggable Targets in Clear Cell Renal Cell Carcinoma Through Targeted RNA Sequencing. Front Oncol 2019; 9:117. [PMID: 30881919 PMCID: PMC6407434 DOI: 10.3389/fonc.2019.00117] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 02/11/2019] [Indexed: 01/05/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) comprises more than 80% of all renal cancers and when metastasized leads to a 5-year survival rate of only 10%. The high rate of therapy failure and resistance development calls for reliable methods that provide information on the actionable biological pathways and predict optimal treatment protocols for individual patients. We here applied targeted RNA sequencing (t/RNA-NGS) using single molecule Molecular Inversion Probes on tumor nephrectomy samples of five ccRCC patients, comparing tumor with healthy kidney tissues. Transcriptome profiling focused on expression of genes with involvement in ccRCC biology that can be targeted with clinically available drugs. Results confirm high expression of vascular endothelial growth factor-A (VEGF-A) in tumor tissue relative to healthy-appearing kidney, in line with the angiogenic nature of ccRCC. PDGFRα and KIT, targets of the multi-kinase inhibitor sunitinib which is one of the current choices of first-line drug in metastasized ccRCC patients, were expressed at relatively low levels in tumor tissues, whereas significantly increased in normal kidney. Of all measured druggable tyrosine kinases, MET, AXL, or EGFR were expressed at higher levels in tumors than in normal kidney tissues, although intertumor differences were observed. Using cancer cell lines we show that t/RNA-NGS gene expression profiles can be used to predict in vitro sensitivity to targeted drugs. In conclusion, t/RNA-NGS analysis may provide insights into the (druggable) molecular make-up of individual renal cancers, and may guide personalized therapy of renal cell cancers.
Collapse
Affiliation(s)
| | - Anne van Ewijk
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
| | - Carolien Zeelen
- Department of Pathology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Tessa de Bitter
- Department of Pathology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Martijn Huynen
- Center for Molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
| | - Peter Mulders
- Department of Urology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Egbert Oosterwijk
- Department of Urology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - William P. J. Leenders
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
| |
Collapse
|
22
|
Cloughesy TF, Drappatz J, de Groot J, Prados MD, Reardon DA, Schiff D, Chamberlain M, Mikkelsen T, Desjardins A, Ping J, Holland J, Weitzman R, Wen PY. Phase II study of cabozantinib in patients with progressive glioblastoma: subset analysis of patients with prior antiangiogenic therapy. Neuro Oncol 2019; 20:259-267. [PMID: 29036345 PMCID: PMC5777491 DOI: 10.1093/neuonc/nox151] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background Cabozantinib is a potent, multitarget inhibitor of MET and vascular endothelial growth factor receptor 2 (VEGFR2). This open-label, phase II trial evaluated cabozantinib in patients with recurrent or progressive glioblastoma (GBM). Methods Patients were initially enrolled to a starting cabozantinib dose of 140 mg/day, but the starting dose was amended to 100 mg/day because of safety concerns. Treatment continued until disease progression or unacceptable toxicity. The primary endpoint was objective response rate, assessed by an independent radiology facility using modified Response Assessment in Neuro-Oncology criteria. Additional endpoints included duration of response, 6-month and median progression-free survival, overall survival, glucocorticoid use, and safety. Results Among 222 patients enrolled, 70 had received prior antiangiogenic therapy. Herein, we report results in this subset of 70 patients. The objective response rate was 4.3%, and the median duration of response was 4.2 months. The proportion of patients alive and progression free at 6 months was 8.5%. Median progression-free survival was 2.3 months, and median overall survival was 4.6 months. The most common adverse events reported in all patients, regardless of dose group, included fatigue (74.3%), diarrhea (47.1%), increased alanine aminotransferase (37.1%), headache (35.7%), hypertension (35.7%), and nausea (35.7%); overall, 34 (48.6%) patients experienced adverse events that resulted in dose reductions. Conclusions Cabozantinib treatment appeared to have modest clinical activity with a 4.3% response rate in patients who had received prior antiangiogenic therapy for GBM. Clinical Trials Registration Number NCT00704288 (https://www.clinicaltrials.gov/ct2/show/NCT00704288)
Collapse
Affiliation(s)
- Timothy F Cloughesy
- The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W., J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California San Francisco, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.P., J.H., R.W.)
| | - Jan Drappatz
- The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W., J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California San Francisco, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.P., J.H., R.W.)
| | - John de Groot
- The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W., J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California San Francisco, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.P., J.H., R.W.)
| | - Michael D Prados
- The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W., J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California San Francisco, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.P., J.H., R.W.)
| | - David A Reardon
- The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W., J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California San Francisco, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.P., J.H., R.W.)
| | - David Schiff
- The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W., J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California San Francisco, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.P., J.H., R.W.)
| | - Marc Chamberlain
- The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W., J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California San Francisco, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.P., J.H., R.W.)
| | - Tom Mikkelsen
- The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W., J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California San Francisco, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.P., J.H., R.W.)
| | - Annick Desjardins
- The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W., J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California San Francisco, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.P., J.H., R.W.)
| | - Jerry Ping
- The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W., J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California San Francisco, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.P., J.H., R.W.)
| | - Jaymes Holland
- The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W., J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California San Francisco, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.P., J.H., R.W.)
| | - Ron Weitzman
- The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W., J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California San Francisco, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.P., J.H., R.W.)
| | - Patrick Y Wen
- The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.); Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W., J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California San Francisco, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.P., J.H., R.W.)
| |
Collapse
|
23
|
Wen PY, Drappatz J, de Groot J, Prados MD, Reardon DA, Schiff D, Chamberlain M, Mikkelsen T, Desjardins A, Holland J, Ping J, Weitzman R, Cloughesy TF. Phase II study of cabozantinib in patients with progressive glioblastoma: subset analysis of patients naive to antiangiogenic therapy. Neuro Oncol 2019; 20:249-258. [PMID: 29016998 PMCID: PMC5777496 DOI: 10.1093/neuonc/nox154] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background Cabozantinib is a tyrosine kinase inhibitor with activity against vascular endothelial growth factor receptor 2 (VEGFR2) and MET that has demonstrated clinical activity in advanced solid tumors. This open-label, phase II trial evaluated cabozantinib in patients with recurrent or refractory glioblastoma (GBM). Methods Patients were initially enrolled at a starting dose of 140 mg/day, but the starting dose was amended to 100 mg/day because of toxicity. Treatment continued until disease progression or unacceptable toxicity. The primary endpoint was objective response rate assessed by an independent radiology facility using modified Response Assessment in Neuro-Oncology criteria. Additional endpoints included duration of response, 6-month and median progression-free survival, overall survival, and safety. Results Among 152 patients naive to prior antiangiogenic therapy, the objective response rate was 17.6% and 14.5% in the 140 mg/day and 100 mg/day groups, respectively, which did not meet the predefined statistical target for success. The proportions of patients alive and progression free at 6 months were 22.3% and 27.8%, respectively. Median progression-free survival was 3.7 months in both groups, and median overall survival was 7.7 months and 10.4 months, respectively. The incidence of grade 3/4 adverse events (AEs) was 79.4% and 84.7% in the 140 mg/day and 100 mg/day groups, respectively, and dose reductions due to AEs were experienced by 61.8% and 72.0%, respectively. Common grade 3/4 AEs included fatigue, diarrhea, and palmar-plantar erythrodysesthesia syndrome. Conclusions Cabozantinib showed evidence of clinical activity in patients with recurrent GBM naive to antiangiogenic therapy, although the predefined statistical target for success was not met. At the starting doses assessed, AEs were frequently managed with dose reductions. Clinical Trials Registration Number NCT00704288 (https://www.clinicaltrials.gov/ct2/show/NCT00704288).
Collapse
Affiliation(s)
- Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W, J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California, San Francisco, Helen Diller Cancer Center Building, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.H., J.P., R.W.); The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.)
| | - Jan Drappatz
- Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W, J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California, San Francisco, Helen Diller Cancer Center Building, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.H., J.P., R.W.); The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.)
| | - John de Groot
- Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W, J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California, San Francisco, Helen Diller Cancer Center Building, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.H., J.P., R.W.); The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.)
| | - Michael D Prados
- Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W, J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California, San Francisco, Helen Diller Cancer Center Building, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.H., J.P., R.W.); The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.)
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W, J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California, San Francisco, Helen Diller Cancer Center Building, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.H., J.P., R.W.); The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.)
| | - David Schiff
- Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W, J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California, San Francisco, Helen Diller Cancer Center Building, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.H., J.P., R.W.); The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.)
| | - Marc Chamberlain
- Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W, J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California, San Francisco, Helen Diller Cancer Center Building, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.H., J.P., R.W.); The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.)
| | - Tom Mikkelsen
- Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W, J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California, San Francisco, Helen Diller Cancer Center Building, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.H., J.P., R.W.); The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.)
| | - Annick Desjardins
- Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W, J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California, San Francisco, Helen Diller Cancer Center Building, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.H., J.P., R.W.); The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.)
| | - Jaymes Holland
- Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W, J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California, San Francisco, Helen Diller Cancer Center Building, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.H., J.P., R.W.); The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.)
| | - Jerry Ping
- Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W, J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California, San Francisco, Helen Diller Cancer Center Building, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.H., J.P., R.W.); The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.)
| | - Ron Weitzman
- Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W, J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California, San Francisco, Helen Diller Cancer Center Building, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.H., J.P., R.W.); The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.)
| | - Timothy F Cloughesy
- Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Boston, Massachusetts (P.Y.W, J.D.); The University of Texas MD Anderson Cancer Center, Houston, Texas (J.dG.); University of California, San Francisco, Helen Diller Cancer Center Building, San Francisco, California (M.D.P.); Duke University, Durham, North Carolina (D.A.R., A.D.); Neuro-Oncology Center, University of Virginia Health System, Charlottesville, Virginia (D.S.); University of Washington, Department of Neurology, Fred Hutchinson Cancer Research Center, Seattle, Washington (M.C.); Henry Ford Health System, Detroit, Michigan (T.M.); Exelixis, South San Francisco, California (J.H., J.P., R.W.); The Ronald Reagan UCLA Medical Center, Los Angeles, California (T.F.C.)
| |
Collapse
|
24
|
Lenting K, Khurshed M, Peeters TH, van den Heuvel CNAM, van Lith SAM, de Bitter T, Hendriks W, Span PN, Molenaar RJ, Botman D, Verrijp K, Heerschap A, Ter Laan M, Kusters B, van Ewijk A, Huynen MA, van Noorden CJF, Leenders WPJ. Isocitrate dehydrogenase 1-mutated human gliomas depend on lactate and glutamate to alleviate metabolic stress. FASEB J 2018; 33:557-571. [PMID: 30001166 DOI: 10.1096/fj.201800907rr] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Diffuse gliomas often carry point mutations in isocitrate dehydrogenase ( IDH1mut), resulting in metabolic stress. Although IDHmut gliomas are difficult to culture in vitro, they thrive in the brain via diffuse infiltration, suggesting brain-specific tumor-stroma interactions that can compensate for IDH-1 deficits. To elucidate the metabolic adjustments in clinical IDHmut gliomas that contribute to their malignancy, we applied a recently developed method of targeted quantitative RNA next-generation sequencing to 66 clinical gliomas and relevant orthotopic glioma xenografts, with and without the endogenous IDH-1R132H mutation. Datasets were analyzed in R using Manhattan plots to calculate distance between expression profiles, Ward's method to perform unsupervised agglomerative clustering, and the Mann Whitney U test and Fisher's exact tests for supervised group analyses. The significance of transcriptome data was investigated by protein analysis, in situ enzymatic activity mapping, and in vivo magnetic resonance spectroscopy of orthotopic IDH1mut- and IDHwt-glioma xenografts. Gene set enrichment analyses of clinical IDH1mut gliomas strongly suggest a role for catabolism of lactate and the neurotransmitter glutamate, whereas, in IDHwt gliomas, processing of glucose and glutamine are the predominant metabolic pathways. Further evidence of the differential metabolic activity in these cancers comes from in situ enzymatic mapping studies and preclinical in vivo magnetic resonance spectroscopy imaging. Our data support an evolutionary model in which IDHmut glioma cells exist in symbiosis with supportive neuronal cells and astrocytes as suppliers of glutamate and lactate, possibly explaining the diffuse nature of these cancers. The dependency on glutamate and lactate opens the way for novel approaches in the treatment of IDHmut gliomas.-Lenting, K., Khurshed, M., Peeters, T. H., van den Heuvel, C. N. A. M., van Lith, S. A. M., de Bitter, T., Hendriks, W., Span, P. N., Molenaar, R. J., Botman, D., Verrijp, K., Heerschap, A., ter Laan, M., Kusters, B., van Ewijk, A., Huynen, M. A., van Noorden, C. J. F., Leenders, W. P. J. Isocitrate dehydrogenase 1-mutated human gliomas depend on lactate and glutamate to alleviate metabolic stress.
Collapse
Affiliation(s)
- Krissie Lenting
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands.,Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mohammed Khurshed
- Department of Medical Biology, Cancer Center Amsterdam, Academic Medical Centre, Amsterdam, The Netherlands
| | - Tom H Peeters
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Corina N A M van den Heuvel
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands.,Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sanne A M van Lith
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tessa de Bitter
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Wiljan Hendriks
- Department of Cell Biology, Radboud Institute of Molecular Life Sciences, Nijmegen, The Netherlands
| | - Paul N Span
- Radiotherapy and Oncoimmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Remco J Molenaar
- Department of Medical Biology, Cancer Center Amsterdam, Academic Medical Centre, Amsterdam, The Netherlands
| | - Dennis Botman
- Department of Medical Biology, Cancer Center Amsterdam, Academic Medical Centre, Amsterdam, The Netherlands
| | - Kiek Verrijp
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Arend Heerschap
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mark Ter Laan
- Department of Neurosurgery, Radboud University Medical Center, Nijmegen, The Netherlands; and
| | - Benno Kusters
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anne van Ewijk
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Martijn A Huynen
- Center for Molecular and Biomolecular Informatics, Radboud Institute of Molecular Life Sciences, Nijmegen, The Netherlands
| | - Cornelis J F van Noorden
- Department of Medical Biology, Cancer Center Amsterdam, Academic Medical Centre, Amsterdam, The Netherlands
| | - William P J Leenders
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands.,Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
25
|
Donnem T, Reynolds AR, Kuczynski EA, Gatter K, Vermeulen PB, Kerbel RS, Harris AL, Pezzella F. Non-angiogenic tumours and their influence on cancer biology. Nat Rev Cancer 2018; 18:323-336. [PMID: 29520090 DOI: 10.1038/nrc.2018.14] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Solid tumours need a blood supply, and a large body of evidence has previously suggested that they can grow only if they induce the development of new blood vessels, a process known as tumour angiogenesis. On the basis of this hypothesis, it was proposed that anti-angiogenic drugs should be able to suppress the growth of all solid tumours. However, clinical experience with anti-angiogenic agents has shown that this is not always the case. Reports of tumours growing without the formation of new vessels can be found in the literature dating back to the 1800s, yet no formal recognition, description and demonstration of their special biological status was made until recently. In 1996, we formally recognized and described non-angiogenic tumours in lungs where the only blood vessels present were those originating from normal lung tissue. This is far from an isolated scenario, as non-angiogenic tumour growth has now been observed in tumours of many different organs in both humans and preclinical animal models. In this Opinion article, we summarize how these tumours were discovered and discuss what we know so far about their biology and the potential implications of this knowledge for cancer treatment.
Collapse
Affiliation(s)
- Tom Donnem
- Department of Oncology, University Hospital of North Norway, Tromso, Norway
- Institute of Clinical Medicine, The Arctic University of Norway, Tromso, Norway
| | - Andrew R Reynolds
- Tumour Biology Team, Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
- Oncology Translational Medicine Unit, Oncology, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Elizabeth A Kuczynski
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
- Bioscience, Oncology, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Kevin Gatter
- Nuffield Department of Clinical Laboratory Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Peter B Vermeulen
- Tumour Biology Team, Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
- Translational Cancer Research Unit, GZA, Hospitals St Augustinus, University of Antwerp, Wilrijk-Antwerp, Belgium
- HistoGeneX, Antwerp, Belgium
| | - Robert S Kerbel
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Adrian L Harris
- Molecular Oncology Laboratories, Oxford University Department of Oncology, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK
| | - Francesco Pezzella
- Nuffield Department of Clinical Laboratory Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| |
Collapse
|
26
|
Schmidinger M, Danesi R. Management of Adverse Events Associated with Cabozantinib Therapy in Renal Cell Carcinoma. Oncologist 2018; 23:306-315. [PMID: 29146618 PMCID: PMC5905684 DOI: 10.1634/theoncologist.2017-0335] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 10/19/2017] [Indexed: 12/20/2022] Open
Abstract
Cabozantinib was recently approved for the treatment of advanced renal cell carcinoma (RCC) after treatment with vascular endothelial growth factor (VEGF)-targeted therapy. Cabozantinib is a multikinase inhibitor targeting VEGF receptor (VEGFR) 2, mesenchymal-epithelial transition receptor, and "anexelekto" receptor tyrosine kinase. A 60-mg daily dose led to improved overall survival and progression-free survival (PFS) versus everolimus in advanced RCC patients as a second- or later-line treatment in the METEOR trial. Improved PFS with cabozantinib versus sunitinib has also been demonstrated in the first-line setting in CABOSUN. However, cabozantinib, like other VEGFR inhibitors, is associated with toxicity that may affect the patient's quality of life. The most frequent adverse events (AEs) are diarrhea, fatigue, hypertension, hand-foot syndrome, weight loss, nausea, and stomatitis. This article summarizes the safety profile of cabozantinib in RCC patients and offers guidance for the management of these AEs. We discuss the underlying mechanisms of these AEs and, based on our experiences with cabozantinib and other multikinase inhibitors, we present approaches to manage toxicity. Prophylactic and therapeutic solutions are available to help with the management of toxicity associated with cabozantinib, and adequate interventions can ensure optimum adherence and maximize patient outcomes. IMPLICATIONS FOR PRACTICE Cabozantinib leads to improved survival outcomes in renal cell carcinoma patients compared with everolimus. However, management of the adverse event profile is crucial to achieve optimum adherence and outcomes with the use of cabozantinib. This review aims to provide appropriate guidance that will minimize the impact of adverse events and help to maximize the utility of this agent in patients with advanced renal cell carcinoma.
Collapse
Affiliation(s)
- Manuela Schmidinger
- Clinical Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Romano Danesi
- Clinical Pharmacology and Pharmacogenetic Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
27
|
van Lith SA, van den Brand D, Wallbrecher R, Wübbeke L, van Duijnhoven SM, Mäkinen PI, Hoogstad-van Evert JS, Massuger L, Ylä-Herttuala S, Brock R, Leenders WP. The effect of subcellular localization on the efficiency of EGFR-targeted VHH photosensitizer conjugates. Eur J Pharm Biopharm 2018; 124:63-72. [DOI: 10.1016/j.ejpb.2017.12.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 12/11/2017] [Accepted: 12/19/2017] [Indexed: 12/19/2022]
|
28
|
van Lith SAM, Roodink I, Verhoeff JJC, Mäkinen PI, Lappalainen JP, Ylä-Herttuala S, Raats J, van Wijk E, Roepman R, Letteboer SJ, Verrijp K, Leenders WPJ. In vivo phage display screening for tumor vascular targets in glioblastoma identifies a llama nanobody against dynactin-1-p150Glued. Oncotarget 2018; 7:71594-71607. [PMID: 27689404 PMCID: PMC5342104 DOI: 10.18632/oncotarget.12261] [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: 06/30/2016] [Accepted: 09/19/2016] [Indexed: 12/23/2022] Open
Abstract
Diffuse gliomas are primary brain cancers that are characterised by infiltrative growth. Whereas high-grade glioma characteristically presents with perinecrotic neovascularisation, large tumor areas thrive on pre-existent vasculature as well. Clinical studies have revealed that pharmacological inhibition of the angiogenic process does not improve survival of glioblastoma patients. Direct targeting of tumor vessels may however still be an interesting therapeutic approach as it allows pinching off the blood supply to tumor cells. Such tumor vessel targeting requires the identification of tumor-specific vascular targeting agents (TVTAs). Here we describe a novel TVTA, C-C7, which we identified via in vivo biopanning of a llama nanobody phage display library in an orthotopic mouse model of diffuse glioma. We show that C-C7 recognizes a subpopulation of tumor blood vessels in glioma xenografts and clinical glioma samples. Additionally, C-C7 recognizes macrophages and activated endothelial cells in atherosclerotic lesions. By using C-C7 as bait in yeast-2-hybrid (Y2H) screens we identified dynactin-1-p150Glued as its binding partner. The interaction was confirmed by co-immunostainings with C-C7 and a commercial anti-dynactin-1-p150Glued antibody, and via co-immunoprecipitation/western blot studies. Normal brain vessels do not express dynactin-1-p150Glued and its expression is reduced under anti-VEGF therapy, suggesting that dynactin-1-p150Glued is a marker for activated endothelial cells. In conclusion, we show that in vivo phage display combined with Y2H screenings provides a powerful approach to identify tumor-targeting nanobodies and their binding partners. Using this combination of methods we identify dynactin-1-p150Glued as a novel targetable protein on activated endothelial cells and macrophages.
Collapse
Affiliation(s)
| | - Ilse Roodink
- Department of Pathology, RadboudUMC, 6500 HB, Nijmegen, The Netherlands.,Modiquest BV, LSP, Molenstraat 110, 5342 CC, Oss, The Netherlands
| | | | - Petri I Mäkinen
- Department of Biotechnology and Molecular Medicine, University of Eastern Finland, FI-70211, Kuopio, Finland
| | - Jari P Lappalainen
- Department of Biotechnology and Molecular Medicine, University of Eastern Finland, FI-70211, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- Department of Biotechnology and Molecular Medicine, University of Eastern Finland, FI-70211, Kuopio, Finland.,Science Service Center and Gene Therapy Unit, Kuopio University Hospital, 70210 Kuopio, Finland
| | - Jos Raats
- Modiquest BV, LSP, Molenstraat 110, 5342 CC, Oss, The Netherlands
| | - Erwin van Wijk
- Department of Otorhinolaryngology, RadboudUMC, 6500 HB, Nijmegen, The Netherlands
| | - Ronald Roepman
- Department of Genetics, RadboudUMC, 6500 HB, Nijmegen,The Netherlands
| | - Stef J Letteboer
- Department of Genetics, RadboudUMC, 6500 HB, Nijmegen,The Netherlands
| | - Kiek Verrijp
- Department of Pathology, RadboudUMC, 6500 HB, Nijmegen, The Netherlands
| | | |
Collapse
|
29
|
Bell EH, Pugh SL, McElroy JP, Gilbert MR, Mehta M, Klimowicz AC, Magliocco A, Bredel M, Robe P, Grosu AL, Stupp R, Curran W, Becker AP, Salavaggione AL, Barnholtz-Sloan JS, Aldape K, Blumenthal DT, Brown PD, Glass J, Souhami L, Lee RJ, Brachman D, Flickinger J, Won M, Chakravarti A. Molecular-Based Recursive Partitioning Analysis Model for Glioblastoma in the Temozolomide Era: A Correlative Analysis Based on NRG Oncology RTOG 0525. JAMA Oncol 2017; 3:784-792. [PMID: 28097324 DOI: 10.1001/jamaoncol.2016.6020] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Importance There is a need for a more refined, molecularly based classification model for glioblastoma (GBM) in the temozolomide era. Objective To refine the existing clinically based recursive partitioning analysis (RPA) model by incorporating molecular variables. Design, Setting, and Participants NRG Oncology RTOG 0525 specimens (n = 452) were analyzed for protein biomarkers representing key pathways in GBM by a quantitative molecular microscopy-based approach with semiquantitative immunohistochemical validation. Prognostic significance of each protein was examined by single-marker and multimarker Cox regression analyses. To reclassify the prognostic risk groups, significant protein biomarkers on single-marker analysis were incorporated into an RPA model consisting of the same clinical variables (age, Karnofsky Performance Status, extent of resection, and neurologic function) as the existing RTOG RPA. The new RPA model (NRG-GBM-RPA) was confirmed using traditional immunohistochemistry in an independent data set (n = 176). Main Outcomes and Measures Overall survival (OS). Results In 452 specimens, MGMT (hazard ratio [HR], 1.81; 95% CI, 1.37-2.39; P < .001), survivin (HR, 1.36; 95% CI, 1.04-1.76; P = .02), c-Met (HR, 1.53; 95% CI, 1.06-2.23; P = .02), pmTOR (HR, 0.76; 95% CI, 0.60-0.97; P = .03), and Ki-67 (HR, 1.40; 95% CI, 1.10-1.78; P = .007) protein levels were found to be significant on single-marker multivariate analysis of OS. To refine the existing RPA, significant protein biomarkers together with clinical variables (age, Karnofsky Performance Status, extent of resection, and neurological function) were incorporated into a new model. Of 166 patients used for the new NRG-GBM-RPA model, 97 (58.4%) were male (mean [SD] age, 55.7 [12.0] years). Higher MGMT protein level was significantly associated with decreased MGMT promoter methylation and vice versa (1425.1 for methylated vs 1828.0 for unmethylated; P < .001). Furthermore, MGMT protein expression (HR, 1.84; 95% CI, 1.38-2.43; P < .001) had greater prognostic value for OS compared with MGMT promoter methylation (HR, 1.77; 95% CI, 1.28-2.44; P < .001). The refined NRG-GBM-RPA consisting of MGMT protein, c-Met protein, and age revealed greater separation of OS prognostic classes compared with the existing clinically based RPA model and MGMT promoter methylation in NRG Oncology RTOG 0525. The prognostic significance of the NRG-GBM-RPA was subsequently confirmed in an independent data set (n = 176). Conclusions and Relevance This new NRG-GBM-RPA model improves outcome stratification over both the current RTOG RPA model and MGMT promoter methylation, respectively, for patients with GBM treated with radiation and temozolomide and was biologically validated in an independent data set. The revised RPA has the potential to contribute to improving the accurate assessment of prognostic groups in patients with GBM treated with radiation and temozolomide and to influence clinical decision making. Trial Registration clinicaltrials.gov Identifier: NCT00304031.
Collapse
Affiliation(s)
- Erica Hlavin Bell
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital, Columbus, Ohio
| | - Stephanie L Pugh
- NRG Oncology Statistics and Data Management Center, Philadelphia, Pennsylvania
| | - Joseph P McElroy
- The Ohio State University Center for Biostatistics, Columbus, Ohio
| | | | - Minesh Mehta
- University of Maryland Medical Systems, Baltimore, Maryland (during trial)6now with Miami Cancer Institute, Coral Gables, Florida
| | | | | | | | | | | | - Roger Stupp
- University Hospital Zurich, Zürich, Switzerland
| | | | - Aline P Becker
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital, Columbus, Ohio
| | - Andrea L Salavaggione
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital, Columbus, Ohio
| | - Jill S Barnholtz-Sloan
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | | | | | | | - Jon Glass
- Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Luis Souhami
- McGill University Health Centre, Montreal, Québec, Canada
| | | | | | | | - Minhee Won
- NRG Oncology Statistics and Data Management Center, Philadelphia, Pennsylvania
| | - Arnab Chakravarti
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital, Columbus, Ohio
| |
Collapse
|
30
|
van den Heuvel CNAM, Navis AC, de Bitter T, Amiri H, Verrijp K, Heerschap A, Rex K, Dussault I, Caenepeel S, Coxon A, Span PN, Wesseling P, Hendriks W, Leenders WPJ. Selective MET Kinase Inhibition in MET-Dependent Glioma Models Alters Gene Expression and Induces Tumor Plasticity. Mol Cancer Res 2017; 15:1587-1597. [PMID: 28751462 DOI: 10.1158/1541-7786.mcr-17-0177] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 06/15/2017] [Accepted: 07/24/2017] [Indexed: 11/16/2022]
Abstract
The receptor tyrosine kinase (RTK) MET represents a promising tumor target in a subset of glioblastomas. Most RTK inhibitors available in the clinic today, including those inhibiting MET, affect multiple targets simultaneously. Previously, it was demonstrated that treatment with cabozantinib (MET/VEGFR2/RET inhibitor) prolonged survival of mice carrying orthotopic patient-derived xenografts (PDX) of the MET-addicted glioblastoma model E98, yet did not prevent development of recurrent and cabozantinib-resistant tumors. To exclude VEGFR2 inhibition-inflicted blood-brain barrier normalization and diminished tumor distribution of the drug, we have now investigated the effects of the novel MET-selective inhibitor Compound A in the orthotopic E98 xenograft model. In vitro, Compound A proved a highly potent inhibitor of proliferation of MET-addicted cell lines. In line with its target selectivity, Compound A did not restore the leaky blood-brain barrier and was more effective than cabozantinib in inhibiting MET phosphorylation in vivo Compound A treatment significantly prolonged survival of mice carrying E98 tumor xenografts, but did not prevent eventual progression. Contrasting in vitro results, the Compound A-treated xenografts displayed high levels of AKT phosphorylation despite the absence of phosphorylated MET. Profiling by RNA sequencing showed that in vivo transcriptomes differed significantly from those in control xenografts.Implications: Collectively, these findings demonstrate the plasticity of paracrine growth factor receptor signaling in vivo and urge for prudency with in vitro drug-testing strategies to validate monotherapies. Mol Cancer Res; 15(11); 1587-97. ©2017 AACR.
Collapse
Affiliation(s)
| | - Anna C Navis
- Department of Pathology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Tessa de Bitter
- Department of Pathology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Houshang Amiri
- Department of Radiology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Kiek Verrijp
- Department of Pathology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Arend Heerschap
- Department of Radiology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Karen Rex
- Department of Oncology Research, Amgen Inc., Thousand Oaks, California
| | - Isabelle Dussault
- Department of Oncology Research, Amgen Inc., Thousand Oaks, California
| | - Sean Caenepeel
- Department of Oncology Research, Amgen Inc., Thousand Oaks, California
| | - Angela Coxon
- Department of Oncology Research, Amgen Inc., Thousand Oaks, California
| | - Paul N Span
- Department of Radiation Oncology, Radboud University Medical Centre, Radiotherapy and Oncoimmunology Laboratory, Nijmegen, the Netherlands
| | - Pieter Wesseling
- Department of Pathology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Wiljan Hendriks
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Nijmegen, the Netherlands
| | - William P J Leenders
- Department of Pathology, Radboud University Medical Centre, Nijmegen, the Netherlands.
| |
Collapse
|
31
|
Safaie Qamsari E, Safaei Ghaderi S, Zarei B, Dorostkar R, Bagheri S, Jadidi-Niaragh F, Somi MH, Yousefi M. The c-Met receptor: Implication for targeted therapies in colorectal cancer. Tumour Biol 2017; 39:1010428317699118. [DOI: 10.1177/1010428317699118] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
c-Met (mesenchymal–epithelial transition factor) is a tyrosine kinase receptor activated by hepatocyte growth factor and regulates multiple biological processes, such as cell scattering, survival, and proliferation. Aberrant c-Met signaling has been implicated in a variety of cancer types, including colorectal cancer. c-Met is genetically altered through various mechanisms that is associated with colorectal cancer progression and metastasis. Especially, in colorectal cancer, preclinical evidence for the aberrant activation of the c-Met signaling exists. Accordingly, molecular targeting of c-Met receptor could be a promising strategy, in the treatment of colorectal cancer patients. Recently, it was also shown that crosstalk between c-Met and other cell surface receptors attributes to tumorigenesis and development of therapeutic resistance. Characterization of the molecular mechanisms through which c-Met crosstalks with other receptors in favor of tumor formation and progression remains to explore. This review will describe the mechanisms of aberrant c-Met signaling in colorectal cancer and discuss on additional roles for c-Met receptor through crosstalk with other tyrosine kinase receptors and cell surface proteins in colorectal cancer. Novel therapeutic approaches for c-Met pathway targeting will also be discussed.
Collapse
Affiliation(s)
- Elmira Safaie Qamsari
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Safaei Ghaderi
- Department of Biotechnology, Faculty of Advanced Science & Technology, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran
- Hybridoma Laboratory, Immunology Department, Pasteur Institute of Iran, Tehran, Iran
| | - Bahareh Zarei
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Ruhollah Dorostkar
- Applied Virology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Salman Bagheri
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Jadidi-Niaragh
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Somi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
32
|
Mahase S, Rattenni RN, Wesseling P, Leenders W, Baldotto C, Jain R, Zagzag D. Hypoxia-Mediated Mechanisms Associated with Antiangiogenic Treatment Resistance in Glioblastomas. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:940-953. [PMID: 28284719 PMCID: PMC5417003 DOI: 10.1016/j.ajpath.2017.01.010] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 12/31/2016] [Accepted: 01/05/2017] [Indexed: 12/28/2022]
Abstract
Glioblastomas (GBMs) are malignant tumors characterized by their vascularity and invasive capabilities. Antiangiogenic therapy (AAT) is a treatment option that targets GBM-associated vasculature to mitigate the growth of GBMs. However, AAT demonstrates transient effects because many patients eventually develop resistance to this treatment. Several recent studies attempt to explain the molecular and biochemical basis of resistance to AAT in GBM patients. Experimental investigations suggest that the induction of extensive intratumoral hypoxia plays a key role in GBM escape from AAT. In this review, we examine AAT resistance in GBMs, with an emphasis on six potential hypoxia-mediated mechanisms: enhanced invasion and migration, including increased expression of matrix metalloproteinases and activation of the c-MET tyrosine kinase pathway; shifts in cellular metabolism, including up-regulation of hypoxia inducible factor-1α's downstream processes and the Warburg effect; induction of autophagy; augmentation of GBM stem cell self-renewal; possible implications of GBM-endothelial cell transdifferentiation; and vasoformative responses, including vasculogenesis, alternative angiogenic pathways, and vascular mimicry. Juxtaposing recent studies on well-established resistance pathways with that of emerging mechanisms highlights the overall complexity of GBM treatment resistance while also providing direction for further investigation.
Collapse
Affiliation(s)
- Sean Mahase
- Microvascular and Molecular Neuro-Oncology Laboratory, New York University School of Medicine, New York, New York
| | - Rachel N Rattenni
- Microvascular and Molecular Neuro-Oncology Laboratory, New York University School of Medicine, New York, New York
| | - Pieter Wesseling
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands; Department of Pathology, Princess Máxima Center for Pediatric Oncology and University Medical Center, Utrecht, the Netherlands
| | - William Leenders
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Clarissa Baldotto
- Medical Oncology, Instituto Nacionale de Cancer, Rio de Janeiro, Brazil
| | - Rajan Jain
- Department of Radiology, New York University School of Medicine, New York, New York; Department of Neurosurgery, New York University School of Medicine, New York, New York
| | - David Zagzag
- Microvascular and Molecular Neuro-Oncology Laboratory, New York University School of Medicine, New York, New York; Department of Neurosurgery, New York University School of Medicine, New York, New York; Division of Neuropathology, Department of Pathology, New York University School of Medicine, New York, New York; Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, New York.
| |
Collapse
|
33
|
Lenting K, Verhaak R, Ter Laan M, Wesseling P, Leenders W. Glioma: experimental models and reality. Acta Neuropathol 2017; 133:263-282. [PMID: 28074274 PMCID: PMC5250671 DOI: 10.1007/s00401-017-1671-4] [Citation(s) in RCA: 191] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 01/06/2017] [Accepted: 01/06/2017] [Indexed: 12/12/2022]
Abstract
In theory, in vitro and in vivo models for human gliomas have great potential to not only enhance our understanding of glioma biology, but also to facilitate the development of novel treatment strategies for these tumors. For reliable prediction and validation of the effects of different therapeutic modalities, however, glioma models need to comply with specific and more strict demands than other models of cancer, and these demands are directly related to the combination of genetic aberrations and the specific brain micro-environment gliomas grow in. This review starts with a brief introduction on the pathological and molecular characteristics of gliomas, followed by an overview of the models that have been used in the last decades in glioma research. Next, we will discuss how these models may play a role in better understanding glioma development and especially in how they can aid in the design and optimization of novel therapies. The strengths and weaknesses of the different models will be discussed in light of genotypic, phenotypic and metabolic characteristics of human gliomas. The last part of this review provides some examples of how therapy experiments using glioma models can lead to deceptive results when such characteristics are not properly taken into account.
Collapse
Affiliation(s)
- Krissie Lenting
- Department of Pathology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Roel Verhaak
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Mark Ter Laan
- Department of Neurosurgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Pieter Wesseling
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
- Department of Pathology, Princess Máxima Center for Pediatric Oncology and University Medical Center Utrecht, Utrecht, The Netherlands
| | - William Leenders
- Department of Pathology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
34
|
Daudigeos-Dubus E, Le Dret L, Bawa O, Opolon P, Vievard A, Villa I, Bosq J, Vassal G, Geoerger B. Dual inhibition using cabozantinib overcomes HGF/MET signaling mediated resistance to pan-VEGFR inhibition in orthotopic and metastatic neuroblastoma tumors. Int J Oncol 2016; 50:203-211. [PMID: 27922668 DOI: 10.3892/ijo.2016.3792] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 10/06/2016] [Indexed: 11/06/2022] Open
Abstract
MET is expressed on neuroblastoma cells and may trigger tumor growth, neoangiogenesis and metastasis. MET upregulation further represents an escape mechanism to various anticancer treatments including VEGF signaling inhibitors. We developed in vitro a resistance model to pan-VEGFR inhibition and explored the simultaneous inhibition of VEGFR and MET in neuroblastoma models in vitro and in vivo using cabozantinib, an inhibitor of the tyrosine kinases including VEGFR2, MET, AXL and RET. Resistance in IGR-N91-Luc neuroblastoma cells under continuous in vitro exposure pressure to VEGFR1-3 inhibition using axitinib was associated with HGF and p-ERK overexpression. Cabozantinib exhibited anti-proliferative effects in neuroblastoma cells and reduced cell migration in vitro as measured by phase-contrast with IncuCyte system. In vivo, an enhanced number of animals with IGR-N91-Luc metastases was noted following axitinib treatment as compared to control animals. Orally administered cabozantinib per gavage at 30 and 60 mg/kg/day significantly inhibited tumor growth of orthotopic adrenal IGR-N91-Luc and metastatic IMR-32-Luc xenografts. Antitumor activity was associated with decreased vascularization, inhibition of p-SRC and induction of apoptotic cell death. Activation of the HGF-mediated MET pathway is involved in escape to selective VEGFR inhibition in neuroblastoma suggesting combined inhibition of MET and VEGFR signaling to reduce secondary resistance and enhanced invasiveness.
Collapse
Affiliation(s)
- Estelle Daudigeos-Dubus
- Vectorology and Anticancer Therapies, UMR 8203, CNRS, Univ. Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Ludivine Le Dret
- Vectorology and Anticancer Therapies, UMR 8203, CNRS, Univ. Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Olivia Bawa
- Preclinical Evaluation Platform, Gustave Roussy, Villejuif, France
| | - Paule Opolon
- Preclinical Evaluation Platform, Gustave Roussy, Villejuif, France
| | | | - Irène Villa
- Pathology Laboratory, Gustave Roussy, Villejuif, France
| | - Jacques Bosq
- Pathology Laboratory, Gustave Roussy, Villejuif, France
| | - Gilles Vassal
- Vectorology and Anticancer Therapies, UMR 8203, CNRS, Univ. Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Birgit Geoerger
- Vectorology and Anticancer Therapies, UMR 8203, CNRS, Univ. Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
35
|
Gebreyohannes YK, Schöffski P, Van Looy T, Wellens J, Vreys L, Cornillie J, Vanleeuw U, Aftab DT, Debiec-Rychter M, Sciot R, Wozniak A. Cabozantinib Is Active against Human Gastrointestinal Stromal Tumor Xenografts Carrying Different KIT Mutations. Mol Cancer Ther 2016; 15:2845-2852. [DOI: 10.1158/1535-7163.mct-16-0224] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 08/11/2016] [Accepted: 08/17/2016] [Indexed: 01/30/2023]
|
36
|
Yu SS, Quinn DI, Dorff TB. Clinical use of cabozantinib in the treatment of advanced kidney cancer: efficacy, safety, and patient selection. Onco Targets Ther 2016; 9:5825-5837. [PMID: 27713636 PMCID: PMC5045229 DOI: 10.2147/ott.s97397] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Clear cell (cc) renal cell carcinoma (RCC) is the most common type of cancer found in the kidney accounting for ~90% of all kidney cancers. In 2012, there were ~337,000 new cases of RCC diagnosed worldwide with an estimated 143,000 deaths, with the highest incidence and mortality in Western countries. Despite improvements in cancer control achieved with VEGF- and mTOR-targeted therapy for RCC, progression remains virtually universal and additional therapies are needed. The pivotal results of the METEOR trial led to cabozantinib's designation as a breakthrough drug by the US Food and Drug Administration and its approval for treatment of advanced RCC in 2016. Subsequent data from the CABOSUN trial, where caboxantinib is compared with sunitinib, will provide information on the relative activity of cabozantinib as first-line therapy for ccRCC. We review the development of cabozantinib in advanced RCC and its role in the treatment landscape for advanced RCC.
Collapse
Affiliation(s)
- Steven S Yu
- Division of Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - David I Quinn
- Division of Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Tanya B Dorff
- Division of Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
37
|
Náger M, Santacana M, Bhardwaj D, Valls J, Ferrer I, Nogués P, Cantí C, Herreros J. Nuclear phosphorylated Y142 β-catenin accumulates in astrocytomas and glioblastomas and regulates cell invasion. Cell Cycle 2016; 14:3644-55. [PMID: 26654598 DOI: 10.1080/15384101.2015.1104443] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a fast growing brain tumor characterized by extensive infiltration into the surrounding tissue and one of the most aggressive cancers. GBM is the most common glioma (originating from glial-derived cells) that either evolves from a low grade astrocytoma or appears de novo. Wnt/β-catenin and Hepatocyte Growth Factor (HGF)/c-Met signaling are hyperactive in human gliomas, where they regulate cell proliferation, migration and stem cell behavior. We previously demonstrated that β-catenin is phosphorylated at Y142 by recombinant c-Met kinase and downstream of HGF signaling in neurons. Here we studied phosphoY142 (PY142) β-catenin and dephospho S/T β-catenin (a classical Wnt transducer) in glioma biopsies, GBM cell lines and biopsy-derived glioma cell cultures. We found that PY142 β-catenin mainly localizes in the nucleus and signals through transcriptional activation in GBM cells. Tissue microarray analysis confirmed strong nuclear PY142 β-catenin immunostaining in astrocytoma and GBM biopsies. By contrast, active β-catenin showed nuclear localization only in GBM samples. Western blot analysis of tumor biopsies further indicated that PY142 and active β-catenin accumulate independently, correlating with the expression of Snail/Slug (an epithelial-mesenchymal transition marker) and Cyclin-D1 (a regulator of cell cycle progression), respectively, in high grade astrocytomas and GBMs. Moreover, GBM cells stimulated with HGF showed increasing levels of PY142 β-catenin and Snail/Slug. Importantly, the expression of mutant Y142F β-catenin decreased cell detachment and invasion induced by HGF in GBM cell lines and biopsy-derived cell cultures. Our results identify PY142 β-catenin as a nuclear β-catenin signaling form that downregulates adhesion and promotes GBM cell invasion.
Collapse
Affiliation(s)
- Mireia Náger
- a Departments of Basic Medical Sciences & Experimental Medicine ; University of Lleida & IRBLleida ; Lleida , Spain
| | - Maria Santacana
- b Immunohistochemical and Biostatistics and Epidemiology Units; IRBLleida ; Lleida , Spain
| | - Deepshikha Bhardwaj
- a Departments of Basic Medical Sciences & Experimental Medicine ; University of Lleida & IRBLleida ; Lleida , Spain
| | - Joan Valls
- b Immunohistochemical and Biostatistics and Epidemiology Units; IRBLleida ; Lleida , Spain
| | - Isidre Ferrer
- c Institute of Neuropathology; Hospital de Bellvitge-IDIBELL ; Barcelona , Spain
| | - Pere Nogués
- d Neurosurgery Unit; Hospital Arnau de Vilanova ; Lleida , Spain
| | - Carles Cantí
- a Departments of Basic Medical Sciences & Experimental Medicine ; University of Lleida & IRBLleida ; Lleida , Spain
| | - Judit Herreros
- a Departments of Basic Medical Sciences & Experimental Medicine ; University of Lleida & IRBLleida ; Lleida , Spain
| |
Collapse
|
38
|
Stella GM, Gentile A, Baderacchi A, Meloni F, Milan M, Benvenuti S. Ockham's razor for the MET-driven invasive growth linking idiopathic pulmonary fibrosis and cancer. J Transl Med 2016; 14:256. [PMID: 27590450 PMCID: PMC5010719 DOI: 10.1186/s12967-016-1008-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 08/16/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) identifies a specific lung disorder characterized by chronic, progressive fibrosing interstitial pneumonia of unknown etiology, which lacks effective treatment. According to the current pathogenic perspective, the aberrant proliferative events in IPF resemble those occurring during malignant transformation. MAIN BODY Receptor tyrosine kinases (RTK) are known to be key players in cancer onset and progression. It has been demonstrated that RTK expression is sometimes also altered and even druggable in IPF. One example of an RTK-the MET proto-oncogene-is a key regulator of invasive growth. This physiological genetic program supports embryonic development and post-natal organ regeneration, as well as cooperating in the evolution of cancer metastasis when aberrantly activated. Growing evidence sustains that MET activation may collaborate in maintaining tissue plasticity and the regenerative potential that characterizes IPF. CONCLUSION The present work aims to elucidate-by applying the logic of simplicity-the bio-molecular mechanisms involved in MET activation in IPF. This clarification is crucial to accurately design MET blockade strategies within a fully personalized approach to IPF.
Collapse
Affiliation(s)
- Giulia M. Stella
- Pneumology Unit, Cardiothoracic and Vascular Department, IRCCS Policlinico San Matteo Foundation and University of Pavia Medical School, Piazzale Golgi 19, 27100 Pavia, Italy
- Investigational Clinical Oncology (INCO), IRCCS Candiolo Cancer Institute-FPO, Candiolo, 20060 Turin, Italy
| | - Alessandra Gentile
- Experimental Clinical Molecular Oncology (ECMO), IRCCS Candiolo Cancer Institute-FPO, Candiolo, 20060 Turin, Italy
| | - Alice Baderacchi
- Investigational Clinical Oncology (INCO), IRCCS Candiolo Cancer Institute-FPO, Candiolo, 20060 Turin, Italy
| | - Federica Meloni
- Pneumology Unit, Cardiothoracic and Vascular Department, IRCCS Policlinico San Matteo Foundation and University of Pavia Medical School, Piazzale Golgi 19, 27100 Pavia, Italy
| | - Melissa Milan
- Experimental Clinical Molecular Oncology (ECMO), IRCCS Candiolo Cancer Institute-FPO, Candiolo, 20060 Turin, Italy
| | - Silvia Benvenuti
- Experimental Clinical Molecular Oncology (ECMO), IRCCS Candiolo Cancer Institute-FPO, Candiolo, 20060 Turin, Italy
| |
Collapse
|
39
|
Bourgonje AM, Verrijp K, Schepens JTG, Navis AC, Piepers JAF, Palmen CBC, van den Eijnden M, Hooft van Huijsduijnen R, Wesseling P, Leenders WPJ, Hendriks WJAJ. Comprehensive protein tyrosine phosphatase mRNA profiling identifies new regulators in the progression of glioma. Acta Neuropathol Commun 2016; 4:96. [PMID: 27586084 PMCID: PMC5009684 DOI: 10.1186/s40478-016-0372-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 08/19/2016] [Indexed: 12/20/2022] Open
Abstract
The infiltrative behavior of diffuse gliomas severely reduces therapeutic potential of surgical resection and radiotherapy, and urges for the identification of new drug-targets affecting glioma growth and migration. To address the potential role of protein tyrosine phosphatases (PTPs), we performed mRNA expression profiling for 91 of the 109 known human PTP genes on a series of clinical diffuse glioma samples of different grades and compared our findings with in silico knowledge from REMBRANDT and TCGA databases. Overall PTP family expression levels appeared independent of characteristic genetic aberrations associated with lower grade or high grade gliomas. Notably, seven PTP genes (DUSP26, MTMR4, PTEN, PTPRM, PTPRN2, PTPRT and PTPRZ1) were differentially expressed between grade II-III gliomas and (grade IV) glioblastomas. For DUSP26, PTEN, PTPRM and PTPRT, lower expression levels correlated with poor prognosis, and overexpression of DUSP26 or PTPRT in E98 glioblastoma cells reduced tumorigenicity. Our study represents the first in-depth analysis of PTP family expression in diffuse glioma subtypes and warrants further investigations into PTP-dependent signaling events as new entry points for improved therapy.
Collapse
|
40
|
Labak CM, Wang PY, Arora R, Guda MR, Asuthkar S, Tsung AJ, Velpula KK. Glucose transport: meeting the metabolic demands of cancer, and applications in glioblastoma treatment. Am J Cancer Res 2016; 6:1599-608. [PMID: 27648352 PMCID: PMC5004066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 07/25/2016] [Indexed: 06/06/2023] Open
Abstract
GLUT1, and to a lesser extent, GLUT3, appear to be interesting targets in the treatment of glioblastoma multiforme. The current review aims to give a brief history of the scientific community's understanding of these glucose transporters and to relate their importance to the metabolic changes that occur as a result of cancer. One of the primary changes that occurs in cancer, the Warburg Effect, is characterized by an extreme shift toward glycolysis from the usual reliance on oxidative phosphorylation and is currently being investigated to target the upstream and downstream factors responsible for Warburg-induced changes. Further, it aims to explain the differential expression of GLUT1 and GLUT3 in glioblastoma tissue, and how these modulations in expression can serve as targets to restore a more normal metabolism. Additionally, hypoxia-induced factor-1α's (HIF1α) role in a number of transcriptional changes typical to GBM will be discussed, including its role in GLUT upregulation. Finally, the four known subtypes of GBM [proneural, neural, mesenchymal, and classical] will be characterized in order to discuss how metabolic changes differ in each subtype. These changes have the potential to be selectively targeted in order to provide specificity to the clinical treatment options in GBM.
Collapse
Affiliation(s)
- Collin M Labak
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at PeoriaPeoria, IL, USA
| | - Paul Y Wang
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at PeoriaPeoria, IL, USA
| | - Rishab Arora
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at PeoriaPeoria, IL, USA
| | - Maheedhara R Guda
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at PeoriaPeoria, IL, USA
| | - Swapna Asuthkar
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at PeoriaPeoria, IL, USA
| | - Andrew J Tsung
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at PeoriaPeoria, IL, USA
- Department of Neurosurgery, University of Illinois College of Medicine at PeoriaPeoria, IL, USA
- Department of Illinois Neurological InstitutePeoria, IL, USA
| | - Kiran K Velpula
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at PeoriaPeoria, IL, USA
- Department of Neurosurgery, University of Illinois College of Medicine at PeoriaPeoria, IL, USA
- Department of Microbiology, Yogi Vemana UniversityKadapa, India
| |
Collapse
|
41
|
van Lith SAM, Navis AC, Lenting K, Verrijp K, Schepens JTG, Hendriks WJAJ, Schubert NA, Venselaar H, Wevers RA, van Rooij A, Wesseling P, Molenaar RJ, van Noorden CJF, Pusch S, Tops B, Leenders WPJ. Identification of a novel inactivating mutation in Isocitrate Dehydrogenase 1 (IDH1-R314C) in a high grade astrocytoma. Sci Rep 2016; 6:30486. [PMID: 27460417 PMCID: PMC4962051 DOI: 10.1038/srep30486] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 07/06/2016] [Indexed: 12/16/2022] Open
Abstract
The majority of low-grade and secondary high-grade gliomas carry heterozygous hotspot mutations in cytosolic isocitrate dehydrogenase 1 (IDH1) or the mitochondrial variant IDH2. These mutations mostly involve Arg132 in IDH1, and Arg172 or Arg140 in IDH2. Whereas IDHs convert isocitrate to alpha-ketoglutarate (α-KG) with simultaneous reduction of NADP+ to NADPH, these IDH mutants reduce α-KG to D-2-hydroxyglutarate (D-2-HG) while oxidizing NADPH. D-2-HG is a proposed oncometabolite, acting via competitive inhibition of α-KG-dependent enzymes that are involved in metabolism and epigenetic regulation. However, much less is known about the implications of the metabolic stress, imposed by decreased α-KG and NADPH production, for tumor biology. We here present a novel heterozygous IDH1 mutation, IDH1R314C, which was identified by targeted next generation sequencing of a high grade glioma from which a mouse xenograft model and a cell line were generated. IDH1R314C lacks isocitrate-to-α-KG conversion activity due to reduced affinity for NADP+, and differs from the IDH1R132 mutants in that it does not produce D-2-HG. Because IDH1R314C is defective in producing α-KG and NADPH, without concomitant production of the D-2-HG, it represents a valuable tool to study the effects of IDH1-dysfunction on cellular metabolism in the absence of this oncometabolite.
Collapse
Affiliation(s)
| | - Anna C Navis
- Department of Pathology, Radboudumc, Nijmegen, The Netherlands
| | - Krissie Lenting
- Department of Pathology, Radboudumc, Nijmegen, The Netherlands
| | - Kiek Verrijp
- Department of Pathology, Radboudumc, Nijmegen, The Netherlands
| | - Jan T G Schepens
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, The Netherlands
| | - Wiljan J A J Hendriks
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, The Netherlands
| | - Nil A Schubert
- Department of Pathology, Radboudumc, Nijmegen, The Netherlands
| | - Hanka Venselaar
- Centre for Molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, The Netherlands
| | - Ron A Wevers
- Translational Metabolic Laboratory, Department Laboratory Medicine, Radboudumc, Nijmegen, The Netherlands
| | - Arno van Rooij
- Translational Metabolic Laboratory, Department Laboratory Medicine, Radboudumc, Nijmegen, The Netherlands
| | - Pieter Wesseling
- Department of Pathology, Radboudumc, Nijmegen, The Netherlands.,Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - Remco J Molenaar
- Department of Cell Biology and Histology, Academic Medical Center, Amsterdam, The Netherlands
| | | | - Stefan Pusch
- Clinical Cooperation Unit Neuropathology, German Cancer Center (DKFZ), Heidelberg, Germany
| | - Bastiaan Tops
- Department of Pathology, Radboudumc, Nijmegen, The Netherlands
| | | |
Collapse
|
42
|
Ahmed M, Kumar G, Moussa M, Wang Y, Rozenblum N, Galun E, Goldberg SN. Hepatic Radiofrequency Ablation-induced Stimulation of Distant Tumor Growth Is Suppressed by c-Met Inhibition. Radiology 2016; 279:103-17. [PMID: 26418615 PMCID: PMC4819900 DOI: 10.1148/radiol.2015150080] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE To elucidate how hepatic radiofrequency (RF) ablation affects distant extrahepatic tumor growth by means of two key molecular pathways. MATERIALS AND METHODS Rats were used in this institutional animal care and use committee-approved study. First, the effect of hepatic RF ablation on distant subcutaneous in situ R3230 and MATBIII breast tumors was evaluated. Animals were randomly assigned to standardized RF ablation, sham procedure, or no treatment. Tumor growth rate was measured for 3½ to 7 days. Then, tissue was harvested for Ki-67 proliferative indexes and CD34 microvascular density. Second, hepatic RF ablation was performed for hepatocyte growth factor (HGF), vascular endothelial growth factor (VEGF), and c-Met receptor expression measurement in periablational rim, serum, and distant tumor 24 hours to 7 days after ablation. Third, hepatic RF ablation was combined with either a c-Met inhibitor (PHA-665752) or VEGF receptor inhibitor (semaxanib) and compared with sham or drug alone arms to assess distant tumor growth and growth factor levels. Finally, hepatic RF ablation was performed in rats with c-Met-negative R3230 tumors for comparison with the native c-Met-positive line. Tumor size and immunohistochemical quantification at day 0 and at sacrifice were compared with analysis of variance and the two-tailed Student t test. Tumor growth curves before and after treatment were analyzed with linear regression analysis to determine mean slopes of pre- and posttreatment growth curves on a per-tumor basis and were compared with analysis of variance and paired two-tailed t tests. RESULTS After RF ablation of normal liver, distant R3230 tumors were substantially larger at 7 days compared with tumors treated with the sham procedure and untreated tumors, with higher growth rates and tumor cell proliferation. Similar findings were observed in MATBIII tumors. Hepatic RF ablation predominantly increased periablational and serum HGF and downstream distant tumor VEGF levels. Compared with RF ablation alone, RF ablation combined with adjuvant PHA-665752 or semaxanib reduced distant tumor growth, proliferation, and microvascular density. For c-Met-negative tumors, hepatic RF ablation did not increase distant tumor growth, proliferation, or microvascular density compared with sham treatment. CONCLUSION RF ablation of normal liver can stimulate distant subcutaneous tumor growth mediated by HGF/c-Met pathway and VEGF activation. This effect was not observed in c-Met-negative tumors and can be blocked with adjuvant c-Met and VEGF inhibitors.
Collapse
Affiliation(s)
- Muneeb Ahmed
- From the Laboratory for Minimally Invasive Tumor Therapies, Department of Radiology, Beth Israel Deaconess Medical Center/Harvard Medical School, 1 Deaconess Rd, WCC 308-B, Boston, MA 02215 (M.A., G.K., M.M., Y.W., S.N.G.); and Goldyne Savad Institute of Gene Therapy (N.R., E.G.) and Division of Image-guided Therapy and Interventional Oncology, Department of Radiology (S.N.G.), Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Gaurav Kumar
- From the Laboratory for Minimally Invasive Tumor Therapies, Department of Radiology, Beth Israel Deaconess Medical Center/Harvard Medical School, 1 Deaconess Rd, WCC 308-B, Boston, MA 02215 (M.A., G.K., M.M., Y.W., S.N.G.); and Goldyne Savad Institute of Gene Therapy (N.R., E.G.) and Division of Image-guided Therapy and Interventional Oncology, Department of Radiology (S.N.G.), Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Marwan Moussa
- From the Laboratory for Minimally Invasive Tumor Therapies, Department of Radiology, Beth Israel Deaconess Medical Center/Harvard Medical School, 1 Deaconess Rd, WCC 308-B, Boston, MA 02215 (M.A., G.K., M.M., Y.W., S.N.G.); and Goldyne Savad Institute of Gene Therapy (N.R., E.G.) and Division of Image-guided Therapy and Interventional Oncology, Department of Radiology (S.N.G.), Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Yuanguo Wang
- From the Laboratory for Minimally Invasive Tumor Therapies, Department of Radiology, Beth Israel Deaconess Medical Center/Harvard Medical School, 1 Deaconess Rd, WCC 308-B, Boston, MA 02215 (M.A., G.K., M.M., Y.W., S.N.G.); and Goldyne Savad Institute of Gene Therapy (N.R., E.G.) and Division of Image-guided Therapy and Interventional Oncology, Department of Radiology (S.N.G.), Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Nir Rozenblum
- From the Laboratory for Minimally Invasive Tumor Therapies, Department of Radiology, Beth Israel Deaconess Medical Center/Harvard Medical School, 1 Deaconess Rd, WCC 308-B, Boston, MA 02215 (M.A., G.K., M.M., Y.W., S.N.G.); and Goldyne Savad Institute of Gene Therapy (N.R., E.G.) and Division of Image-guided Therapy and Interventional Oncology, Department of Radiology (S.N.G.), Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Eithan Galun
- From the Laboratory for Minimally Invasive Tumor Therapies, Department of Radiology, Beth Israel Deaconess Medical Center/Harvard Medical School, 1 Deaconess Rd, WCC 308-B, Boston, MA 02215 (M.A., G.K., M.M., Y.W., S.N.G.); and Goldyne Savad Institute of Gene Therapy (N.R., E.G.) and Division of Image-guided Therapy and Interventional Oncology, Department of Radiology (S.N.G.), Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - S. Nahum Goldberg
- From the Laboratory for Minimally Invasive Tumor Therapies, Department of Radiology, Beth Israel Deaconess Medical Center/Harvard Medical School, 1 Deaconess Rd, WCC 308-B, Boston, MA 02215 (M.A., G.K., M.M., Y.W., S.N.G.); and Goldyne Savad Institute of Gene Therapy (N.R., E.G.) and Division of Image-guided Therapy and Interventional Oncology, Department of Radiology (S.N.G.), Hadassah Hebrew University Hospital, Jerusalem, Israel
| |
Collapse
|
43
|
Zorzan M, Giordan E, Redaelli M, Caretta A, Mucignat-Caretta C. Molecular targets in glioblastoma. Future Oncol 2016; 11:1407-20. [PMID: 25952786 DOI: 10.2217/fon.15.22] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma is the most lethal brain tumor. The poor prognosis results from lack of defined tumor margins, critical location of the tumor mass and presence of chemo- and radio-resistant tumor stem cells. The current treatment for glioblastoma consists of neurosurgery, followed by radiotherapy and temozolomide chemotherapy. A better understanding of the role of molecular and genetic heterogeneity in glioblastoma pathogenesis allowed the design of novel targeted therapies. New targets include different key-role signaling molecules and specifically altered pathways. The new approaches include interference through small molecules or monoclonal antibodies and RNA-based strategies mediated by siRNA, antisense oligonucleotides and ribozymes. Most of these treatments are still being tested yet they stay as solid promises for a clinically relevant success.
Collapse
Affiliation(s)
- Maira Zorzan
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | | | | | | | | |
Collapse
|
44
|
Leibowitz-Amit R, Pintilie M, Khoja L, Azad AA, Berger R, Laird AD, Aftab DT, Chi KN, Joshua AM. Changes in plasma biomarkers following treatment with cabozantinib in metastatic castration-resistant prostate cancer: a post hoc analysis of an extension cohort of a phase II trial. J Transl Med 2016; 14:12. [PMID: 26762579 PMCID: PMC4712499 DOI: 10.1186/s12967-015-0747-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 12/04/2015] [Indexed: 11/10/2022] Open
Abstract
Background
Cabozantinib is an orally available inhibitor of tyrosine kinases including VEGFR2 and c-MET. We performed a post hoc analysis to find associations between select plasma biomarkers and treatment response in patients (pts) with metastatic castration resistant prostate cancer (mCRPC) who received cabozantinib 100 mg daily as part of a phase 2 non-randomized expansion cohort (NCT00940225). Methods
Plasma samples were collected at baseline, 6 weeks and at time of maximal response from 81 mCRPC pts with bone metastases, of which 33 also had measurable soft-tissue disease. Levels of 27 biomarkers were measured in duplicate using enzyme-linked immunosorbent assay. Spearman correlation coefficients were calculated for the association between biomarker levels or their change on treatment and either bone scan response (BSR) or soft tissue response according to RECIST. Results A BSR and RECIST response were seen in 66/81 pts (81 %) and 6/33 pts (18 %) respectively. No significant associations were found between any biomarker at any time point and either type of response. Plasma concentrations of VEGFA, FLT3L, c-MET, AXL, Gas6A, bone-specific alkaline phosphatase, interleukin-8 and the hypoxia markers CA9 and clusterin significantly increased during treatment with cabozantinib irrespective of response. The plasma concentrations of VEGFR2, Trap5b, Angiopoietin-2, TIMP-2 and TIE-2 significantly decreased during treatment with caboznatinib. Conclusions Our data did not reveal plasma biomarkers associated with response to cabozantinib. The observed alterations in several biomarkers during treatment with cabozantinib may provide insights on the effects of cabozantinib on tumor cells and on tumor micro-environment and may help point to potential co-targeting approaches.
Collapse
Affiliation(s)
| | - Melania Pintilie
- Division of Biostatistics, Princess Margaret Cancer Center, University Health Network, Toronto, Canada.
| | - Leila Khoja
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Center, University Health Network, 610 University Ave, Toronto, ON, M5G 2M9, Canada.
| | - Arun A Azad
- Department of Medical Oncology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
| | - Raanan Berger
- Department of Oncology, Sheba Medical Center, Tel Hashomer, Israel.
| | | | | | - Kim N Chi
- Department of Medical Oncology, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
| | - Anthony M Joshua
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Center, University Health Network, 610 University Ave, Toronto, ON, M5G 2M9, Canada.
| |
Collapse
|
45
|
Wang H, Xu T, Jiang Y, Xu H, Yan Y, Fu D, Chen J. The challenges and the promise of molecular targeted therapy in malignant gliomas. Neoplasia 2015; 17:239-55. [PMID: 25810009 PMCID: PMC4372648 DOI: 10.1016/j.neo.2015.02.002] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 02/06/2015] [Indexed: 11/18/2022] Open
Abstract
Malignant gliomas are the most common malignant primary brain tumors and one of the most challenging forms of cancers to treat. Despite advances in conventional treatment, the outcome for patients remains almost universally fatal. This poor prognosis is due to therapeutic resistance and tumor recurrence after surgical removal. However, over the past decade, molecular targeted therapy has held the promise of transforming the care of malignant glioma patients. Significant progress in understanding the molecular pathology of gliomagenesis and maintenance of the malignant phenotypes will open opportunities to rationally develop new molecular targeted therapy options. Recently, therapeutic strategies have focused on targeting pro-growth signaling mediated by receptor tyrosine kinase/RAS/phosphatidylinositol 3-kinase pathway, proangiogenic pathways, and several other vital intracellular signaling networks, such as proteasome and histone deacetylase. However, several factors such as cross-talk between the altered pathways, intratumoral molecular heterogeneity, and therapeutic resistance of glioma stem cells (GSCs) have limited the activity of single agents. Efforts are ongoing to study in depth the complex molecular biology of glioma, develop novel regimens targeting GSCs, and identify biomarkers to stratify patients with the individualized molecular targeted therapy. Here, we review the molecular alterations relevant to the pathology of malignant glioma, review current advances in clinical targeted trials, and discuss the challenges, controversies, and future directions of molecular targeted therapy.
Collapse
Affiliation(s)
- Hongxiang Wang
- Department of Neurosurgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Tao Xu
- Department of Neurosurgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Ying Jiang
- Department of Neurosurgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Hanchong Xu
- Department of Neurosurgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Yong Yan
- Department of Neurosurgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Da Fu
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Juxiang Chen
- Department of Neurosurgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China.
| |
Collapse
|
46
|
Oosterwijk-Wakka JC, de Weijert MCA, Franssen GM, Leenders WPJ, van der Laak JAWM, Boerman OC, Mulders PFA, Oosterwijk E. Successful combination of sunitinib and girentuximab in two renal cell carcinoma animal models: a rationale for combination treatment of patients with advanced RCC. Neoplasia 2015; 17:215-24. [PMID: 25748241 PMCID: PMC4351300 DOI: 10.1016/j.neo.2014.12.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 12/19/2014] [Accepted: 12/23/2014] [Indexed: 11/18/2022] Open
Abstract
Anti-angiogenic treatment with tyrosine kinase inhibitors (TKI) has lead to an impressive increase in progression-free survival for patients with metastatic RCC (mRCC), but mRCC remains largely incurable. We combined sunitinib, targeting the endothelial cells with Girentuximab (monoclonal antibody cG250, recognizing carbonic anhydrase IX (CAIX) targeting the tumor cells to study the effect of sunitinib on the biodistribution of Girentuximab because combination of modalities targeting tumor vasculature and tumor cells might result in improved effect. Nude mice with human RCC xenografts (NU12, SK-RC-52) were treated orally with 0.8 mg/day sunitinib, or vehicle for 7 to 14 days. Three days before start or cessation of treatment mice were injected i.v. with 0.4 MBq/5 μg (111)In-Girentuximab followed by biodistribution studies. Immunohistochemical analyses were performed to study the tumor vasculature and CAIX expression and to confirm Girentuximab uptake. NU12 appeared to represent a sunitinib sensitive tumor: sunitinib treatment resulted in extensive necrosis and decreased microvessel density (MVD). Accumulation of Girentuximab was significantly decreased when sunitinib treatment preceded the antibody injection but remained unchanged when sunitinib followed Girentuximab injection. Cessation of therapy led to a rapid neovascularization, reminiscent of a tumor flare. SK-RC-52 appeared to represent a sunitinib-resistant tumor: (central) tumor necrosis was minimal and MVD was not affected. Sunitinib treatment resulted in increased Girentuximab uptake, regardless of the sequence of treatment. These data indicate that sunitinib can be combined with Girentuximab. Since these two modalities have different modes of action, this combination might lead to enhanced therapeutic efficacy.
Collapse
Affiliation(s)
- Jeannette C Oosterwijk-Wakka
- Radboud University Medical Center, Department of Urology, 267 Experimental Urology, PO Box 9101, 6500 HB Nijmegen, The Netherlands.
| | - Mirjam C A de Weijert
- Radboud University Medical Center, Department of Urology, 267 Experimental Urology, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Gerben M Franssen
- Radboud University Medical Center, Department of Nuclear Medicine, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - William P J Leenders
- Radboud University Medical Center, Department of Pathology, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Jeroen A W M van der Laak
- Radboud University Medical Center, Department of Pathology, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Otto C Boerman
- Radboud University Medical Center, Department of Nuclear Medicine, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Peter F A Mulders
- Radboud University Medical Center, Department of Urology, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Egbert Oosterwijk
- Radboud University Medical Center, Department of Urology, 267 Experimental Urology, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| |
Collapse
|
47
|
Wang X, Huang Y, Christie A, Bowden M, Lee GSM, Kantoff PW, Sweeney CJ. Cabozantinib Inhibits Abiraterone's Upregulation of IGFIR Phosphorylation and Enhances Its Anti-Prostate Cancer Activity. Clin Cancer Res 2015; 21:5578-87. [PMID: 26289068 DOI: 10.1158/1078-0432.ccr-15-0824] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 08/10/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Abiraterone improves the overall survival of men with metastatic castration-resistant prostate cancer. However, de novo or adaptive resistance to abiraterone limits its activity. Rational combinations of drugs with different mechanisms of action that overcome resistance mechanisms may improve the efficacy of therapy. To that end, we studied the molecular and phenotypic effects of the combination of cabozantinib plus abiraterone. EXPERIMENTAL DESIGN Three prostate cancer cell lines were used to interrogate the in vitro molecular and antiproliferative effects of the single agents and combination of cabozantinib and abiraterone. The in vivo impact of the combination was assessed using the LAPC4-CR xenograft mouse model. RESULTS In vitro proliferation studies demonstrated single-agent doses between 2 μmol/L and 10 μmol/L for abiraterone and cabozantinib inhibit prostate cancer cell proliferation in a dose-dependent manner, and the anticancer activity of abiraterone is enhanced when combined with cabozantinib. In vivo LAPC4-CR xenograft mouse studies also showed that cabozantinib can improve the antitumor activity of abiraterone. Cabozantinib, a multiple receptor tyrosine kinase inhibitor, enhances the ability of abiraterone to inhibit AR activity in a cell line-dependent manner. In addition, our cell line studies demonstrate abiraterone-stimulated insulin-like growth factor I receptor (IGFIR) phosphorylation with downstream activation of MEK1/2 and ERK1/2, and that this potential adaptive resistance mechanism was inhibited by cabozantinib. CONCLUSIONS Cabozantinib can enhance the efficacy of abiraterone by blocking multiple compensatory survival mechanisms, including IGFIR activation, and supports the assessment of the combination in a clinical trial.
Collapse
Affiliation(s)
- Xiaodong Wang
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Ying Huang
- Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Amanda Christie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Michaela Bowden
- Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Gwo-Shu Mary Lee
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Philip W Kantoff
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| | - Christopher J Sweeney
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
48
|
Sun Y, Sun L, An Y, Shen X. Cabozantinib, a Novel c-Met Inhibitor, Inhibits Colorectal Cancer Development in a Xenograft Model. Med Sci Monit 2015; 21:2316-21. [PMID: 26255947 PMCID: PMC4536870 DOI: 10.12659/msm.893590] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Background Angiogenesis plays a critical role during tumor development. c-Met has recently been implicated in the angiogenesis of various tumors, leaving its role in colorectal cancer (CRC) unknown. In this study, we aimed to evaluate the effect of a novel c-Met inhibitor, cabozantinib, on the tumor growth and angiogenesis in a CRC mouse model. Material/Methods A mouse CRC xenograft model was used to evaluate the effect of cabozantinib on vivo growth of tumors and angiogenesis. The expression of angiogenesis-related factors was evaluated by immunohistochemistry (IHC) and Western blotting. Levels of serum cytokines were detected by ELISA. Results Cabozantinib effectively reduced tumor size and angiogenesis, and suppressed the expression of vascular endothelial growth factor (VEGF) in tumor tissues, possibly via the inhibition of Sonic Hedgehog (SHH) pathway. Conclusions The blockade of c-Met inhibits the tumor growth and angiogenesis via modulating SHH pathway, suggesting a potential strategy in the treatment of CRC.
Collapse
Affiliation(s)
- Yinggang Sun
- Department of General Surgery, Jinan Military General Hospital, Jinan, Shandong, China (mainland)
| | - Liyong Sun
- Department of General Surgery, Jinan Military General Hospital, Jinan, Shandong, China (mainland)
| | - Yanxin An
- Department of General Surgery, Jinan Military General Hospital, Jinan, Shandong, China (mainland)
| | - Xin Shen
- Department of General Surgery, Jinan Military General Hospital, Jinan, Shandong, China (mainland)
| |
Collapse
|
49
|
Towner RA, Ihnat M, Saunders D, Bastian A, Smith N, Pavana RK, Gangjee A. A new anti-glioma therapy, AG119: pre-clinical assessment in a mouse GL261 glioma model. BMC Cancer 2015; 15:522. [PMID: 26177924 PMCID: PMC4504175 DOI: 10.1186/s12885-015-1538-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 07/13/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND High grade gliomas (HGGs; grades III and IV) are the most common primary brain tumors in adults, and their malignant nature ranks them fourth in incidence of cancer death. Standard treatment for glioblastomas (GBM), involving surgical resection followed by radiation and chemotherapy with temozolomide (TMZ) and the anti-angiogenic therapy bevacizumab, have not substantially improved overall survival. New therapeutic agents are desperately needed for this devastating disease. Here we study the potential therapeutic agent AG119 in a pre-clinical model for gliomas. AG119 possesses both anti-angiogenic (RTK inhibition) and antimicrotubule cytotoxic activity in a single molecule. METHODS GL261 glioma-bearing mice were either treated with AG119, anti-VEGF (vascular endothelial growth factor) antibody, anti c-Met antibody or TMZ, and compared to untreated tumor-bearing mice. Animal survival was assessed, and tumor volumes and vascular alterations were monitored with morphological magnetic resonance imaging (MRI) and perfusion-weighted imaging, respectively. RESULTS Percent survival of GL261 HGG-bearing mice treated with AG119 was significantly higher (p < 0.001) compared to untreated tumors. Tumor volumes (21-31 days following intracerebral implantation of GL261 cells) were found to be significantly lower for AG119 (p < 0.001), anti-VEGF (p < 0.05) and anti-c-Met (p < 0.001) antibody treatments, and TMZ-treated (p < 0.05) mice, compared to untreated controls. Perfusion data indicated that both AG119 and TMZ were able to reduce the effect of decreasing perfusion rates significantly (p < 0.05 for both), when compared to untreated tumors. It was also found that IC50 values for AG119 were much lower than those for TMZ in T98G and U251 cells. CONCLUSIONS These data support further exploration of the anticancer activity AG119 in HGG, as this compound was able to increase animal survival and decrease tumor volumes in a mouse GL261 glioma model, and that AG119 is also not subject to methyl guanine transferase (MGMT) mediated resistance, as is the case with TMZ, indicating that AG119 may be potentially useful in treating resistant gliomas.
Collapse
Affiliation(s)
- Rheal A Towner
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA. .,Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73117, USA.
| | - Michael Ihnat
- Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73117, USA.
| | - Debra Saunders
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA.
| | - Anja Bastian
- Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73117, USA. .,Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73117, USA.
| | - Nataliya Smith
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA.
| | - Roheeth Kumar Pavana
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, 15282, USA.
| | - Aleem Gangjee
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, 15282, USA.
| |
Collapse
|
50
|
Womeldorff M, Gillespie D, Jensen RL. Hypoxia-inducible factor-1 and associated upstream and downstream proteins in the pathophysiology and management of glioblastoma. Neurosurg Focus 2015; 37:E8. [PMID: 25581937 DOI: 10.3171/2014.9.focus14496] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive brain tumor with an exceptionally poor patient outcome despite aggressive therapy including surgery, radiation, and chemotherapy. This aggressive phenotype may be associated with intratumoral hypoxia, which probably plays a key role in GBM tumor growth, development, and angiogenesis. A key regulator of cellular response to hypoxia is the protein hypoxia-inducible factor–1 (HIF-1). An examination of upstream hypoxic and nonhypoxic regulation of HIF-1 as well as a review of the downstream HIF-1– regulated proteins may provide further insight into the role of this transcription factor in GBM pathophysiology. Recent insights into upstream regulators that intimately interact with HIF-1 could provide potential therapeutic targets for treatment of this tumor. The same is potentially true for HIF-1–mediated pathways of glycolysis-, angiogenesis-, and invasion-promoting proteins. Thus, an understanding of the relationship between HIF-1, its upstream protein regulators, and its downstream transcribed genes in GBM pathogenesis could provide future treatment options for the care of patients with these tumors.
Collapse
|