201
|
Yun JA, Kim J, Baek YY, Park W, Park M, Kim S, Kim T, Choi S, Jeoung D, Lee H, Won MH, Kim JY, Ha KS, Kwon YG, Kim YM. N-Terminal Modification of the Tetrapeptide Arg-Leu-Tyr-Glu, a Vascular Endothelial Growth Factor Receptor-2 (VEGFR-2) Antagonist, Improves Antitumor Activity by Increasing its Stability against Serum Peptidases. Mol Pharmacol 2019; 96:692-701. [PMID: 31594790 DOI: 10.1124/mol.119.117234] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 10/03/2019] [Indexed: 02/14/2025] Open
Abstract
The tetrapeptide Arg-Leu-Tyr-Glu (RLYE), a vascular endothelial growth factor (VEGF) receptor-2 antagonist, has been used previously either alone or in combination with chemotherapeutic drugs for treating colorectal cancer in a mouse model. We analyzed the half-life of the peptide and found that because of degradation by aminopeptidases B and N, it had a short half-life of 1.2 hours in the serum. Therefore, to increase the stability and potency of the peptide, we designed the modified peptide, N-terminally acetylated RLYE (Ac-RLYE), which had a strongly stabilized half-life of 8.8 hours in serum compared with the original parent peptide. The IC50 value of Ac-RLYE for VEGF-A-induced endothelial cell migration decreased to approximately 37.1 pM from 89.1 pM for the parent peptide. Using a mouse xenograft tumor model, we demonstrated that Ac-RLYE was more potent than RLYE in inhibiting tumor angiogenesis and growth, improving vascular integrity and normalization through enhanced endothelial cell junctions and pericyte coverage of the tumor vasculature, and impeding the infiltration of macrophages into tumor and their polarization to the M2 phenotype. Furthermore, combined treatment of Ac-RLYE and irinotecan exhibited synergistic effects on M1-like macrophage activation and apoptosis and growth inhibition of tumor cells. These findings provide evidence that the N-terminal acetylation augments the therapeutic effect of RLYE in solid tumors via inhibition of tumor angiogenesis, improvement of tumor vessel integrity and normalization, and enhancement of the livery and efficacy of the coadministered chemotherapeutic drugs. SIGNIFICANCE STATEMENT: The results of this study demonstrate that the N-terminal acetylation of the tetrapeptide RLYE (Ac-RLYE), a novel vascular endothelial growth factor receptor-2 (VEGFR-2) inhibitor, significantly improves its serum stability, antiangiogenic activity, and vascular normalizing potency, resulting in enhanced therapeutic effect on solid tumors. Furthermore, the combined treatment of Ac-RLYE with the chemotherapeutic drug, irinotecan, synergistically enhanced its antitumor efficacy by improving the perfusion and delivery of the drug into the tumors and stimulating the conversion of the tumor-associated macrophages to an immunostimulatory M1-like antitumor phenotype.
Collapse
Affiliation(s)
- Jung-A Yun
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| | - Joohwan Kim
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| | - Yi-Yong Baek
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| | - Wonjin Park
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| | - Minsik Park
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| | - Suji Kim
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| | - Taesam Kim
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| | - Seunghwan Choi
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| | - Dooil Jeoung
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| | - Hansoo Lee
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| | - Moo-Ho Won
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| | - Ji-Yoon Kim
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| | - Kwon-Soo Ha
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| | - Young-Guen Kwon
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| | - Young-Myeong Kim
- Departments of Molecular and Cellular Biochemistry (J.-A.Y., J.K., Y.-Y.B., W.P., M.P., S.K., T.K., S.C., K.-S.H., Y.-M.K.) and Neurobiology, School of Medicine (M.-H.W.), Departments of Biochemistry, College of Natural Sciences (D.J.) and Life Sciences, College of Natural Sciences (H.L.), and Kangwon Institute of Inclusive Technology (J.K., Y.-M.K.), Kangwon National University, Chuncheon, Gangwon-do, and Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul (J.-Y.K.), and Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul (Y.-G.K.), Korea
| |
Collapse
|
202
|
Cho HD, Kim JH, Park JK, Hong SM, Kim DH, Seo KI. Kochia scoparia seed extract suppresses VEGF-induced angiogenesis via modulating VEGF receptor 2 and PI3K/AKT/mTOR pathways. PHARMACEUTICAL BIOLOGY 2019; 57:684-693. [PMID: 31608754 PMCID: PMC6807910 DOI: 10.1080/13880209.2019.1672753] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 09/20/2019] [Indexed: 06/10/2023]
Abstract
Context: Kochia scoparia (L.) Schrad (Amaranthaceae), known as a traditional medicine in China, Japan and Korea, is reported to have various biological activities. However, K. scoparia seed extract (KSE) functional roles on angiogenesis and prostate cancer inhibition have not been elucidated. Objective: This study elucidates the effects of KSE on vascular endothelial growth factor (VEGF)-induced angiogenesis in human umbilical vein endothelial cells (HUVECs) and inhibition of proliferation in prostate cancer cells. Materials and methods: HUVECs were treated with 10-20 µg/mL of KSE and 20-50 ng/mL of VEGF for 12-72 h. Anti-angiogenesis properties of KSE were determined by wound healing, trans-well, tube formation, rat aortic ring assay and western blotting. Prostate cancer and normal cells were incubated with 10-250 µg/mL of KSE for 24 h, and cell viability was measured by SRB assay. Phenolic compounds in KSE were analyzed using a HPLC-PDA system. Results: IC50 for cell viability of HUVECs, LNCaP, PC-3, RC-58T and RWPE-1 by KSE were 30.64, 89.25, 123.41, 141.62 and >250 µg/mL, respectively. Treatment with KSE (20 µg/mL) significantly suppressed VEGF-induced migration, invasion and capillary-like structure formation of HUVECs and microvessel sprouting from rat aortic rings. In addition, KSE down-regulated PI3K/AKT/mTOR levels and phosphorylation of VEGF receptor 2 in HUVECs. 3-OH-tyrosol (1.63 mg/g) and morin hydrate (0.17 mg/g) were identified in KSE. Conclusions: KSE inhibits angiogenesis in HUVECs as well as proliferation in human prostate cancer cells, suggesting KSE may be useful herbal medicine for preventing progression of prostate cancer and angiogenesis.
Collapse
Affiliation(s)
- Hyun-Dong Cho
- Department of Food Science and Biotechnology,
Kyungpook National University, Daegu, Republic of
Korea
| | - Jeong-Ho Kim
- Department of Food Science and Biotechnology,
Kyungpook National University, Daegu, Republic of
Korea
| | - Jun-Kyu Park
- Department of Biotechnology, Dong-A
University, Busan, Republic of Korea
| | - Seong-Min Hong
- College of Pharmacy and Gachon Institute of
Pharmaceutical Science, Gachon University, Incheon, Republic of
Korea
| | - Du-Hyun Kim
- Department of Life Resources Industry, Dong-A
University, Busan, Republic of Korea
| | - Kwon-Il Seo
- Department of Biotechnology, Dong-A
University, Busan, Republic of Korea
| |
Collapse
|
203
|
Lam SY, Lee CS, Sharma S, Cheng K. Bevacizumab-induced dysphonia: A case report with brief review of literature. J Oncol Pharm Pract 2019; 26:1032-1036. [PMID: 31771464 DOI: 10.1177/1078155219889388] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
INTRODUCTION Anti-angiogenic treatment in adjunct with chemotherapy is widely used for the treatment of various cancers. These agents inhibit vascular endothelial growth factor (VEGF) signaling thereby inhibiting tumor proliferation and invasion. Dysphonia, or voice changes, has been documented, but is an underreported side effect of anti-angiogenic agents. We report a case of intermittent dysphonia in a patient with metastatic, platinum-refractory ovarian cancer treated with bevacizumab. CASE REPORT A 48-year-old female with high grade mixed type ovarian adenocarcinoma and concurrent left sided breast cancer was transitioned to palliative therapy with gemcitabine-bevacizumab for her ovarian cancer. At a follow-up visit after three cycles of the new therapy, the patient complained of intermittent changes in her voice, describing periods of hoarseness or softness in her voice after the chemotherapy-sometimes to the point that her voice was inaudible. Management and outcome: A new pelvic thrombus was discovered upon assessment of the patient's disease. Bevacizumab was held and she was referred to ear, nose, and throat evaluation for dysphonia. Laryngoscopic examination showed normal vocal cord, with normal movements and no lesion or necrosis. During subsequent follow-up, the patient reported improvement in her voice with no additional dysphonia. DISCUSSION Vocal adverse effects of anti-VEGF agents have been documented in landmark trials and case reports; however, clinicians are often unaware of this rare side effect. Although VEGF-induced dysphonia may be rare and may not impede the patient's quality of life in some cases, it is critical to acknowledge and not underestimate this adverse effect.
Collapse
Affiliation(s)
- So Yi Lam
- St. John's University, New York, NY, USA
| | - Chung-Shien Lee
- Department of Clinical Health Professions, St. John's University College of Pharmacy and Health Sciences, New York, NY, USA
| | | | - Kit Cheng
- Monter Cancer Center of Northwell Health, New York, NY, USA
| |
Collapse
|
204
|
Lugano R, Ramachandran M, Dimberg A. Tumor angiogenesis: causes, consequences, challenges and opportunities. Cell Mol Life Sci 2019; 77:1745-1770. [PMID: 31690961 PMCID: PMC7190605 DOI: 10.1007/s00018-019-03351-7] [Citation(s) in RCA: 1033] [Impact Index Per Article: 172.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/10/2019] [Accepted: 10/21/2019] [Indexed: 02/07/2023]
Abstract
Tumor vascularization occurs through several distinct biological processes, which not only vary between tumor type and anatomic location, but also occur simultaneously within the same cancer tissue. These processes are orchestrated by a range of secreted factors and signaling pathways and can involve participation of non-endothelial cells, such as progenitors or cancer stem cells. Anti-angiogenic therapies using either antibodies or tyrosine kinase inhibitors have been approved to treat several types of cancer. However, the benefit of treatment has so far been modest, some patients not responding at all and others acquiring resistance. It is becoming increasingly clear that blocking tumors from accessing the circulation is not an easy task to accomplish. Tumor vessel functionality and gene expression often differ vastly when comparing different cancer subtypes, and vessel phenotype can be markedly heterogeneous within a single tumor. Here, we summarize the current understanding of cellular and molecular mechanisms involved in tumor angiogenesis and discuss challenges and opportunities associated with vascular targeting.
Collapse
Affiliation(s)
- Roberta Lugano
- The Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 75185, Uppsala, Sweden
| | - Mohanraj Ramachandran
- The Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 75185, Uppsala, Sweden
| | - Anna Dimberg
- The Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 75185, Uppsala, Sweden.
| |
Collapse
|
205
|
Targeted cancer therapies: Oral health care implications. J Am Dent Assoc 2019; 149:100-111. [PMID: 29389333 DOI: 10.1016/j.adaj.2017.08.046] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 08/23/2017] [Accepted: 08/28/2017] [Indexed: 12/30/2022]
Abstract
BACKGROUND Targeted treatments have been incorporated into oncology protocols, often with more traditional therapies, and are not totally free of adverse reactions, some of which affect the orofacial region. METHODS The authors searched PubMed, the Cochrane Library, and the US Food and Drug Administration Approved Drug Products database to identify reported adverse effects of targeted agents in the orofacial region as well as other implications in oral health care. Their principal focus was the relatively newer category of molecularly targeted drugs which are called small molecules (SMs). RESULTS The authors identified several categories of SMs and biological agents (for example, monoclonal antibodies) with adverse effects in the orofacial region. The oral and perioral regions are also fields for which there are therapeutic applications for targeted therapies, particularly to treat malignant neoplasms such as head and neck cancers. CONCLUSIONS SMs are the most rapidly growing group of targeted cancer treatments. Patients receiving SMs and other targeted antineoplastic agents may require oral medicine advice and special-care dentistry. PRACTICAL IMPLICATIONS In this narrative review, the authors focus mainly on the orofacial adverse effects of targeted cancer therapies and outline many of the agents that are in use so the dentally focused reader can familiarize themselves with these adverse effects and agents.
Collapse
|
206
|
Necrotizing enterocolitis after intravitreal bevacizumab in an infant with Incontinentia Pigmenti - a case report. BMC Pediatr 2019; 19:353. [PMID: 31615465 PMCID: PMC6792241 DOI: 10.1186/s12887-019-1732-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 09/20/2019] [Indexed: 11/17/2022] Open
Abstract
Background Incontinentia Pigmenti is a rare disease affecting multiple organs. Fifty of patients show affection of the eye with retinopathy and possible amaurosis being the worst outcome. Treatment has commonly been panretinal laser coagulation but intravitreal application of bevacizumab as VEGF-inhibitor has shown to effectively suppress retinal neovascularization. Case presentation A six-week-old female infant with Incontinentia Pigmenti developed a foudroyant necrotizing enterocolitis shortly after intravitreal injection of bevazicumab due to a retinopathy with impending tractional detachment of the left eye. Since the onset of abdominal symptoms occurred immediately after the intravitreal application, a link between the two events seemed likely. Sequential analyses of the VEGF serum concentrations showed a massive suppression of endogenous VEGF with only a very slow recovery over weeks. Such a severe systemic adverse event has not been reported after intravitreal treatment with bevacizumab in an infant. Conclusion This case report shows a relevant systemic uptake of bevacizumab after intravitreal application as suppressed VEGF levels show. There seems to be a connection between suppressed VEGF levels and the onset of necrotizing enterocolitis. Therefore, treatment with bevacizumab should be carefully considered and further research is needed to assess this drug’s safety profile.
Collapse
|
207
|
Applications of microneedles in delivering drugs for various ocular diseases. Life Sci 2019; 237:116907. [PMID: 31606378 DOI: 10.1016/j.lfs.2019.116907] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 09/21/2019] [Accepted: 09/23/2019] [Indexed: 01/20/2023]
Abstract
Treatment of majority of eye diseases involve the use of eye drops or eye ointments, which have major drawbacks of needing frequent administration, lower bioavailability and inability to cross the various eye barriers. This necessitates the use of novel delivery systems. Microneedles (MNs) as an alternate novel delivery system facilitate drug delivery to various ocular diseases with promising approaches in healthcare. Advances in pharmaceutical technology have made MNs provide localized, effective, less invasive and targeted drug delivery in the eye. The purpose of this review is to provide an insight to efficacious therapeutic applications the MNs can bring in various ocular diseases. Out of which, glaucoma, age-related macular degeneration, uveitis, retinal vascular occlusion and retinitis pigmentosa are majorly discussed. Among the various types of MNs; solid coated, hollow and dissolving polymeric MNs are specifically focused for their applications in ocular diseases. In addition, MNs shows improvement in the visual acuity and decreases the progression of the different ocular diseases.
Collapse
|
208
|
Ravindran D, Cartland SP, Bursill CA, Kavurma MM. Broad-spectrum chemokine inhibition blocks inflammation-induced angiogenesis, but preserves ischemia-driven angiogenesis. FASEB J 2019; 33:13423-13434. [PMID: 31574232 DOI: 10.1096/fj.201900232rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
M3 is a broad-spectrum chemokine-binding protein that inactivates inflammatory chemokines, including CCL2, CCL5, and CX3CL1. The aim of this study was to compare whether M3 could inhibit angiogenesis driven by inflammation or ischemia. Here, apolipoprotein E-/- mice were injected with adenoviral M3 (AdM3) or control adenoviral green fluorescent protein (AdGFP) 3 d prior to stimulating angiogenesis using 2 established models that distinctly represent inflammatory or ischemia-driven angiogenesis, namely the periarterial femoral cuff and hind limb ischemia. AdM3 reduced intimal thickening, adventitial capillary density, and macrophage accumulation in femoral arteries 21 d after periarterial femoral cuff placement compared with AdGFP-treated mice (P < 0.05). AdM3 also reduced mRNA expression of proangiogenic VEGF, inflammatory markers IL-6 and IL-1β, and vascular smooth muscle cell (VSMC)-activated synthetic markers Krüppel-like family of transcription factor 4 (KLF4) and platelet-derived growth factor receptor β (PDGFRβ) in the inflammatory cuff model. In contrast, capillary density, VSMC content, blood flow perfusion, and VEGF gene expression were unaltered between groups in skeletal muscle following hind limb ischemia. In vitro, AdM3 significantly reduced human microvascular endothelial cell 1 proliferation, migration, and tubule formation by ∼17, 71.3, and 8.7% (P < 0.05) in macrophage-conditioned medium associating with reduced VEGF and hypoxia-inducible factor 1α mRNA but not in hypoxia (1% O2). Compared with AdGFP, AdM3 also inhibited VSMC proliferation and migration and reduced mRNA expression of KLF4 and PDGFRβ under inflammatory conditions. In contrast, AdM3 had no effect on VSMC processes in response to hypoxia in vitro. Our findings show that broad-spectrum inhibition of inflammatory chemokines by M3 inhibits inflammatory-driven but not ischemia-driven angiogenesis, presenting a novel strategy for the treatment of diseases associated with inflammatory-driven angiogenesis.-Ravindran, D., Cartland, S. P., Bursill, C. A., Kavurma, M. M. Broad-spectrum chemokine inhibition blocks inflammation-induced angiogenesis, but preserves ischemia-driven angiogenesis.
Collapse
Affiliation(s)
- Dhanya Ravindran
- The Heart Research Institute, Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Siân P Cartland
- The Heart Research Institute, Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Christina A Bursill
- Heart Health, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Mary M Kavurma
- The Heart Research Institute, Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
209
|
Fugazzola L, Elisei R, Fuhrer D, Jarzab B, Leboulleux S, Newbold K, Smit J. 2019 European Thyroid Association Guidelines for the Treatment and Follow-Up of Advanced Radioiodine-Refractory Thyroid Cancer. Eur Thyroid J 2019; 8:227-245. [PMID: 31768334 PMCID: PMC6873012 DOI: 10.1159/000502229] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/19/2019] [Indexed: 01/03/2023] Open
Abstract
The vast majority of thyroid cancers of follicular origin (TC) have a very favourable outcome, but 5-10% of cases will develop metastatic disease. Around 60-70% of this subset, hence less than 5% of all patients with TC, will become radioiodine refractory (RAI-R), with a significant negative impact on prognosis and a mean life expectancy of 3-5 years. Since no European expert consensus or guidance for this challenging condition is currently available, a task force of TC experts was nominated by the European Thyroid Association (ETA) to prepare this document based on the principles of clinical evidence. The task force started to work in September 2018 and after several revision rounds, prepared a list of recommendations to support the treatment and follow-up of patients with advanced TC. Criteria for advanced RAI-R TC were proposed, and the most appropriate diagnostic tools and the local, systemic and palliative treatments are described. Systemic therapy with multikinase inhibitors is fully discussed, including recommendations on how to start it and at which dosage, on the duration of treatment, and on the management of side effects. The appropriate relationship between the specialist and the patient/family as well as ethical issues are covered. Based on the available studies and on personal experience, the experts provided 39 recommendations aimed to improve the management of advanced RAI-R TCs. Above all of them is the indication to treat and follow these patients in a specialized setting which allows the interaction between several specialists in a multidisciplinary team.
Collapse
Affiliation(s)
- Laura Fugazzola
- Division of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Rossella Elisei
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Dagmar Fuhrer
- Department of Endocrinology, Diabetes and Metabolism, Endocrine Tumour Center at West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Duisburg, Germany
| | - Barbara Jarzab
- Department of Nuclear Medicine and Endocrine Oncology, Maria Sklodowska-Curie Institute, Oncology Center, Gliwice Branch, Gliwice, Poland
| | - Sophie Leboulleux
- Department of Nuclear Medicine and Endocrine Oncology, Gustave Roussy and Université Paris Saclay, Villejuif, France
| | - Kate Newbold
- Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, United Kingdom
| | - Jan Smit
- Division of Endocrinology, Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
210
|
Toenjes ST, Garcia V, Maddox SM, Dawson GA, Ortiz MA, Piedrafita FJ, Gustafson JL. Leveraging Atropisomerism to Obtain a Selective Inhibitor of RET Kinase with Secondary Activities toward EGFR Mutants. ACS Chem Biol 2019; 14:1930-1939. [PMID: 31424197 DOI: 10.1021/acschembio.9b00407] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Unstable atropisomerism is innate in many common scaffolds in drug discovery, commonly existing as freely rotating aryl-aryl bonds. Such compounds can access the majority of dihedral conformations around the bond axis; however, most small molecules bind their target within a narrow range of these available conformations. The remaining accessible conformations can interact with other proteins leading to compound promiscuity. Herein, we leverage atropisomerism to restrict the accessible low-energy dihedral conformations available to a promiscuous kinase inhibitor and achieve highly selective and potent inhibitors of the oncogenic target rearranged during transfection (RET) kinase. We then evaluate our lead inhibitor against kinases that were predicted to bind compounds in a similar conformational window to RET, discovering a potent inhibitor of drug-resistant epidermal growth factor receptor (EGFR) mutants including L858R/T790M/C797S EGFR. Leveraging atropisomerism to restrict accessible conformational space should be a generally applicable strategy due to the prevalence of unstable atropisomerism in drug discovery.
Collapse
|
211
|
Darweesh RS, Ayoub NM, Nazzal S. Gold nanoparticles and angiogenesis: molecular mechanisms and biomedical applications. Int J Nanomedicine 2019; 14:7643-7663. [PMID: 31571869 PMCID: PMC6756918 DOI: 10.2147/ijn.s223941] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 08/18/2019] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis is the formation of new blood vessels from pre-existing vessels. It is a highly regulated process as determined by the interplay between pro-angiogenic and anti-angiogenic factors. Under certain conditions the balance between angiogenesis stimulators and inhibitors is altered, which results in a shift from physiological to pathological angiogenesis. Therefore, the goal of therapeutic targeting of angiogenic process is to normalize vasculature in target tissues by enhancing angiogenesis in disease conditions of reduced vascularity and blood flow, such as tissue ischemia, or alternatively to inhibit excessive and abnormal angiogenesis in disorders like cancer. Gold nanoparticles (AuNPs) are special particles that are generated by nanotechnology and composed of an inorganic core containing gold which is encircled by an organic monolayer. The ability of AuNPs to alter vasculature has captured recent attention in medical literature as potential therapeutic agents for the management of pathologic angiogenesis. This review provides an overview of the effects of AuNPs on angiogenesis and the molecular mechanisms and biomedical applications associated with their effects. In addition, the main synthesis methods, physical properties, uptake mechanisms, and toxicity of AuNPs are briefly summarized.
Collapse
Affiliation(s)
- Ruba S Darweesh
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid22110, Jordan
| | - Nehad M Ayoub
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid22110, Jordan
| | - Sami Nazzal
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Dallas, TX75235-6411, USA
| |
Collapse
|
212
|
Sadeghi A, Behdani M, Muyldermans S, Habibi-Anbouhi M, Kazemi-Lomedasht F. Development of a mono-specific anti-VEGF bivalent nanobody with extended plasma half-life for treatment of pathologic neovascularization. Drug Test Anal 2019; 12:92-100. [PMID: 31476257 DOI: 10.1002/dta.2693] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 08/21/2019] [Accepted: 08/29/2019] [Indexed: 12/11/2022]
Abstract
Vascular endothelial growth factor (VEGF) plays a crucial role in angiogenesis within solid cancers. Thus, targeting VEGF might be part of a feasible therapy for treating pathological neovascularization, and nanobodies - derived from heavy chain-only antibodies occurring within Camelidae - are a novel class of nanometer-sized antibodies possessing unique properties that could be developed into a promising therapeutic. However, nanobodies have a very short half-life in vivo due to their small size. Development of a bivalent nanobody is one way to remediate the half-life problem of nanobodies. Two identical anti-VEGF nanobodies were connected using the hinge region of llama IgG2c. The recombinant plasmid (pHEN6c-bivalent nanobody) was transformed into E.coli WK6 cells and expression of the bivalent nanobody construct was induced with 1mM Isopropyl β-D-1-thiogalactopyranoside (IPTG). Recombinant bivalent nanobody was purified using nickel affinity chromatography and its activity on human endothelial cells was assessed using 3-(4,5-Dimethylthiazol-2-yr)-2,5-diphenyltetrazolium bromide (MTT), tube formation, and cell migration assays. The pharmacokinetic study was performed after intravenous (i.v.) injection of recombinant bivalent nanobody into six-week-old C57BL/6 mice. Recombinant bivalent nanobody performed significantly better than monovalent nanobody in inhibiting proliferation, tube formation, and migration of human endothelial cells. Pharmacokinetic results showed a 1.8-fold longer half-life of bivalent nanobody in comparison with the monovalent nanobody. These results underscore the potential of recombinant anti-VEGF bivalent nanobody as a promising tool for development of a novel therapeutic with an extended plasma half-life for VEGF-related diseases.
Collapse
Affiliation(s)
- Amir Sadeghi
- Biotechnology Research Center, Venom and Biotherapeutics Molecules Laboratory, Pasteur Institute of Iran, Tehran, Iran
| | - Mahdi Behdani
- Biotechnology Research Center, Venom and Biotherapeutics Molecules Laboratory, Pasteur Institute of Iran, Tehran, Iran
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Fatemeh Kazemi-Lomedasht
- Biotechnology Research Center, Venom and Biotherapeutics Molecules Laboratory, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
213
|
|
214
|
Melo ÉGA, Silveira PAL, Mello CA. Transient vocal fold lesion and hoarseness associated with the use of ramucirumab: Case report. Eur Ann Otorhinolaryngol Head Neck Dis 2019; 136:317-319. [PMID: 30876850 DOI: 10.1016/j.anorl.2018.08.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 08/17/2018] [Accepted: 08/27/2018] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Gastric cancer (GC) is among the leading neoplasms with highest morbidity and mortality in the world. Tumor angiogenesis represents one of important targets of antineoplastic treatment, as it has an important role in cell development and growth. CASE REPORT Ramucirumab, an IgG1 monoclonal antibody (MoAb), inhibits the angiogenesis pathway by blocking the VEGFR-2 activation. The most common adverse events associated with angiogenic inhibitors are cardiovascular and healing disorders, such as systemic arterial hypertension (HTN), thromboembolism, bleeding, wound healing delay and fistulas. We report a case of vocal fold lesion associated with ramucirumab in a patient with metastatic gastric cancer. The patient presented with transient hoarseness that was related to the exposure to ramucirumab. DISCUSSION Laryngeal toxicity due to anti-angiogenic agents is rare. Despite the fact that this adverse event is not a life-threatening complication, it can significantly impair quality of life and should be promptly recognized.
Collapse
Affiliation(s)
- É G A Melo
- Department of Medical Oncology, AC Camargo Cancer Center, São Paulo, SP, Brazil.
| | - P A L Silveira
- Department of Otorrinolaryngology, AC Camargo Cancer Center, São Paulo, SP, Brazil
| | - C A Mello
- Department of Medical Oncology, AC Camargo Cancer Center, São Paulo, SP, Brazil
| |
Collapse
|
215
|
Lupo G, Cambria MT, Olivieri M, Rocco C, Caporarello N, Longo A, Zanghì G, Salmeri M, Foti MC, Anfuso CD. Anti-angiogenic effect of quercetin and its 8-methyl pentamethyl ether derivative in human microvascular endothelial cells. J Cell Mol Med 2019; 23:6565-6577. [PMID: 31369203 PMCID: PMC6787496 DOI: 10.1111/jcmm.14455] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 05/03/2019] [Accepted: 05/17/2019] [Indexed: 12/27/2022] Open
Abstract
Angiogenesis is involved in many pathological states such as progression of tumours, retinopathy of prematurity and diabetic retinopathy. The latter is a more complex diabetic complication in which neurodegeneration plays a significant role and a leading cause of blindness. The vascular endothelial growth factor (VEGF) is a powerful pro‐angiogenic factor that acts through three tyrosine kinase receptors (VEGFR‐1, VEGFR‐2 and VEGFR‐3). In this work we studied the anti‐angiogenic effect of quercetin (Q) and some of its derivates in human microvascular endothelial cells, as a blood retinal barrier model, after stimulation with VEGF‐A. We found that a permethylated form of Q, namely 8MQPM, more than the simple Q, is a potent inhibitor of angiogenesis both in vitro and ex vivo. Our results showed that these compounds inhibited cell viability and migration and disrupted the formation of microvessels in rabbit aortic ring. The addition of Q and more significantly 8MQPM caused recoveries or completely re‐establish the transendothelial electrical resistance (TEER) to the control values and suppressed the activation of VEGFR2 downstream signalling molecules such as AKT, extracellular signal‐regulated kinase, and c‐Jun N‐terminal kinase. Taken together, these data suggest that 8MQPM might have an important role in the contrast of angiogenesis‐related diseases.
Collapse
Affiliation(s)
- Gabriella Lupo
- Section of Medical Biochemistry, Department of Biomedical and Biotechnological Sciences (Biometec), School of Medicine, University of Catania, Catania, Italy
| | - Maria Teresa Cambria
- Section of Medical Biochemistry, Department of Biomedical and Biotechnological Sciences (Biometec), School of Medicine, University of Catania, Catania, Italy
| | - Melania Olivieri
- Section of Medical Biochemistry, Department of Biomedical and Biotechnological Sciences (Biometec), School of Medicine, University of Catania, Catania, Italy
| | - Concetta Rocco
- Institute of Biomolecular Chemistry of CNR, Catania, Italy
| | - Nunzia Caporarello
- Section of Medical Biochemistry, Department of Biomedical and Biotechnological Sciences (Biometec), School of Medicine, University of Catania, Catania, Italy
| | - Anna Longo
- Section of Medical Biochemistry, Department of Biomedical and Biotechnological Sciences (Biometec), School of Medicine, University of Catania, Catania, Italy
| | - Guido Zanghì
- Department of Surgery (CHIR), School of Medicine, University of Catania, Catania, Italy
| | - Mario Salmeri
- Section of Medical Biochemistry, Department of Biomedical and Biotechnological Sciences (Biometec), School of Medicine, University of Catania, Catania, Italy
| | - Mario C Foti
- Institute of Biomolecular Chemistry of CNR, Catania, Italy
| | - Carmelina Daniela Anfuso
- Section of Medical Biochemistry, Department of Biomedical and Biotechnological Sciences (Biometec), School of Medicine, University of Catania, Catania, Italy
| |
Collapse
|
216
|
Gara E, Csikó KG, Ruzsa Z, Földes G, Merkely B. Anti-cancer drugs-induced arterial injury: risk stratification, prevention, and treatment. Med Oncol 2019; 36:72. [PMID: 31292791 DOI: 10.1007/s12032-019-1295-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 06/30/2019] [Indexed: 12/21/2022]
Abstract
Vascular side effects of standard chemotherapeutic drugs and novel anti-tumor agents complicate treatment cycles, increase non-cancer-related mortality rates, and decrease the quality of life in cancer survivors. Arterial thromboembolic events (ATEE) are associated with most anti-cancer medications. Previous articles have reported a variety of vascular events including ST-segment elevation myocardial infarction as one of the most severe acute arterial attacks. Cardiologists should play an early role in identifying those at high risk for vascular complications and tailor anti-thrombotic therapies in keeping with thromboembolic and bleeding risks. Early preventive steps and individualized chemotherapy may decrease anti-tumor treatment-related vascular events. Here, we aim to provide an extensive review of anti-tumor drug-induced vascular injury (DIVI), pathomechanisms, and risk stratification underlining arterial events. We give a summary of clinical manifestations, treatment options, and possible preventive measures of DIVI. Additionally, the treatment of modifiable risk factors and tailored choice of chemotherapy must be considered in all oncology patients to prevent DIVI. We propose a complex tool for ATEE risk stratification which is warranted for early prediction leading to less frequent complications in cancer patients.
Collapse
Affiliation(s)
- Edit Gara
- Heart and Vascular Centre, Semmelweis University, 68 Városmajor str., Budapest, 1122, Hungary.
| | - Kristóf György Csikó
- Heart and Vascular Centre, Semmelweis University, 68 Városmajor str., Budapest, 1122, Hungary
| | - Zoltán Ruzsa
- Heart and Vascular Centre, Semmelweis University, 68 Városmajor str., Budapest, 1122, Hungary
| | - Gábor Földes
- Heart and Vascular Centre, Semmelweis University, 68 Városmajor str., Budapest, 1122, Hungary
| | - Béla Merkely
- Heart and Vascular Centre, Semmelweis University, 68 Városmajor str., Budapest, 1122, Hungary
| |
Collapse
|
217
|
Zhu J, Zhang J, Wang Y, Chen J, Li X, Liu X, Kong E, Su SB, Zhang Z. The Effect of Interleukin 38 on Inflammation-induced Corneal Neovascularization. Curr Mol Med 2019; 19:589-596. [PMID: 31244436 DOI: 10.2174/1566524019666190627122655] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 05/27/2019] [Accepted: 05/30/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Angiogenesis is tightly linked to inflammation. Cytokines of interleukin 1 (IL-1) family are key mediators in modulating inflammatory responses. METHODS In this study, we examined the role of IL-38, a member of the IL-1 family, in mediating inflammation-induced angiogenesis. RESULTS The results showed that the angiogenesis was attenuated by topical administration of IL-38 to the injured corneas in a mouse model of alkali-induced corneal neovascularization (CNV). Further study showed that the expression of inflammatory cytokines TNF-α, IL-6, IL-8 and IL-1β was decreased in the IL-38-treated corneas. Moreover, the angiogenic activities including the proliferation, migration and tube formation of human retinal endothelial cells were reduced by IL-38 treatment in vitro. CONCLUSION The data indicate that IL-38 modulates inflammation-induced angiogenesis.
Collapse
Affiliation(s)
- Jiangli Zhu
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453000, China
| | - Jing Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Yan Wang
- Guangdong Science and Technology Library (Guangdong Institute of Scientific and Technical Information and Development Strategy), China
| | - Jianping Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Xiaopeng Li
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453000, China
| | - Xiangling Liu
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453000, China
| | - Eryan Kong
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, Henan, 453000, China
| | - Shao B Su
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453000, China.,State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Zhongjian Zhang
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, Henan, 453000, China
| |
Collapse
|
218
|
Sánchez Ramírez J, Bequet-Romero M, Morera Díaz Y, Hernández-Bernal F, Ayala Avila M. Does VEGF-targeted active immunotherapy induce complete abrogation of platelet VEGF levels? BMC Res Notes 2019; 12:323. [PMID: 31182141 PMCID: PMC6558718 DOI: 10.1186/s13104-019-4368-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 06/05/2019] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVES Vascular endothelial growth factor (VEGF) is involved in physiological angiogenesis, but also is considered one of the key factors that promotes tumor angiogenesis. CIGB-247 is a VEGF-based vaccine that has been evaluated in phase I clinical trial patients with advanced solid tumors. This specific active immunotherapy is able to reduce platelet VEGF levels; however it is unknown whether this effect leads to a decrease in VEGF below the levels that can be observed in healthy individuals. The objective of the present study is to investigate platelet VEGF levels in cancer patients vaccinated with CIGB-247, and then compare these values with those obtained in healthy individuals. To achieve this, platelet VEGF levels of 62 cancer patients and 93 healthy individuals were compared. Cancer patients were those individuals recruited in CENTAURO and CENTAURO-2 clinical trials. RESULTS Before vaccination, platelets of cancer patients carried more VEGF than the levels seen in platelet of healthy individuals. However, after vaccination, cancer patients had platelet VEGF values within the range established by healthy individuals, indicating that the antibody response elicited by CIGB-247 is not able to induce a complete suppression of VEGF. Vaccination with CIGB-247 helps to normalize VEGF levels within platelets.
Collapse
Affiliation(s)
- Javier Sánchez Ramírez
- Department of Pharmaceuticals, Center for Genetic Engineering and Biotechnology, Ave 31 e/158 y 190, Playa, P.O. Box 6162, Havana, Cuba.
| | - Mónica Bequet-Romero
- Department of Pharmaceuticals, Center for Genetic Engineering and Biotechnology, Ave 31 e/158 y 190, Playa, P.O. Box 6162, Havana, Cuba
| | - Yanelys Morera Díaz
- Department of Pharmaceuticals, Center for Genetic Engineering and Biotechnology, Ave 31 e/158 y 190, Playa, P.O. Box 6162, Havana, Cuba
| | - Francisco Hernández-Bernal
- Department of Clinical Research, Center for Genetic Engineering and Biotechnology, Ave 31 e/158 y 190, Playa, P.O. Box 6162, Havana, Cuba
| | - Marta Ayala Avila
- Department of Pharmaceuticals, Center for Genetic Engineering and Biotechnology, Ave 31 e/158 y 190, Playa, P.O. Box 6162, Havana, Cuba
| |
Collapse
|
219
|
Fields GB. Mechanisms of Action of Novel Drugs Targeting Angiogenesis-Promoting Matrix Metalloproteinases. Front Immunol 2019; 10:1278. [PMID: 31214203 PMCID: PMC6558196 DOI: 10.3389/fimmu.2019.01278] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 05/20/2019] [Indexed: 12/16/2022] Open
Abstract
Angiogenesis is facilitated by the proteolytic activities of members of the matrix metalloproteinase (MMP) family. More specifically, MMP-9 and MT1-MMP directly regulate angiogenesis, while several studies indicate a role for MMP-2 as well. The correlation of MMP activity to tumor angiogenesis has instigated numerous drug development programs. However, broad-based and Zn2+-chelating MMP inhibitors have fared poorly in the clinic. Selective MMP inhibition by antibodies, biologicals, and small molecules has utilized unique modes of action, such as (a) binding to protease secondary binding sites (exosites), (b) allosterically blocking the protease active site, or (c) preventing proMMP activation. Clinical trials have been undertaken with several of these inhibitors, while others are in advanced pre-clinical stages. The mechanistically non-traditional MMP inhibitors offer treatment strategies for tumor angiogenesis that avoid the off-target toxicities and lack of specificity that plagued Zn2+-chelating inhibitors.
Collapse
Affiliation(s)
- Gregg B Fields
- Department of Chemistry and Biochemistry, Florida Atlantic University, Jupiter, FL, United States.,Department of Chemistry, The Scripps Research Institute/Scripps Florida, Jupiter, FL, United States
| |
Collapse
|
220
|
Lane N, Nyman J, Uppuganti S, Chaudhari A, Aguirre J, Shidara K, Liu X, Yao W, Kimmel D. Inhibition of vascular endothelial growth factor in young adult mice causes low bone blood flow and bone strength with no effect on bone mass in trabecular regions. Bone Rep 2019; 10:100210. [PMID: 31193542 PMCID: PMC6535464 DOI: 10.1016/j.bonr.2019.100210] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 05/03/2019] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE To determine the effect of an antibody to vascular endothelial growth factor (VEGF) on bone blood flow, bone strength, and bone mass in the young adult mouse. METHODS Ten-week-old male BALB/cJ mice were body weight-randomized into either a rodent anti-VEGF monoclonal antibody (anti-VEGF, B20-4.1.1; 5 mg/kg 2×/wk.; n = 12) group or a vehicle (VEH; n = 12) group. After 42 days, mice were evaluated for bone blood flow at the distal femur by 18F-NaF-PET/CT and then necropsied. Samples from trabecular and cortical bone regions were evaluated for bone strength by mechanical testing, bone mass by peripheral quantitative computed tomography (pQCT), and micoarchitecture (MicroCT). Hydration of the whole femur was studied by proton nuclear magnetic resonance relaxometry (1H NMR). RESULTS Distal femur blood flow was 43% lower in anti-VEGF mice than in VEH mice (p = 0.009). Ultimate load in the lumbar vertebral body was 25% lower in anti-VEGF than in VEH mice (p = 0.013). Bone mineral density (BMD) in the trabecular region of the proximal humeral metaphysis by pQCT, and bone volume fraction and volumetric BMD by MicroCT were the same in the two groups. Volume fraction of bound water (BW) of the whole femur was 14% lower in anti-VEGF than in VEH mice (p = 0.003). Finally, BW, but not cortical tissue mineral density, helped section modulus explain the variance in the ultimate moment experienced by the femur in three-point bending. CONCLUSION Anti-VEGF caused low bone blood flow and bone strength in trabecular bone regions without influencing BMD and microarchitecture. Low bone strength was also associated with low bone hydration. These data suggest that bone blood flow is a novel bone property that affects bone quality.
Collapse
Affiliation(s)
- N.E. Lane
- Center for Musculoskeletal Health, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - J.S. Nyman
- Department of Orthopaedic Surgery and Rehabilitation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - S. Uppuganti
- Department of Orthopaedic Surgery and Rehabilitation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - A.J. Chaudhari
- Center for Molecular and Genomic Imaging, Department of Radiology, University of California at Davis, Davis, CA 95616, USA
| | - J.I. Aguirre
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA
| | - K. Shidara
- Center for Musculoskeletal Health, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - X.P. Liu
- Center for Musculoskeletal Health, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - W. Yao
- Center for Musculoskeletal Health, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - D.B. Kimmel
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
221
|
Melegh Z, Oltean S. Targeting Angiogenesis in Prostate Cancer. Int J Mol Sci 2019; 20:E2676. [PMID: 31151317 PMCID: PMC6600172 DOI: 10.3390/ijms20112676] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 05/24/2019] [Accepted: 05/29/2019] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer is the most commonly diagnosed cancer among men in the Western world. Although localized disease can be effectively treated with established surgical and radiopharmaceutical treatments options, the prognosis of castration-resistant advanced prostate cancer is still disappointing. The objective of this study was to review the role of angiogenesis in prostate cancer and to investigate the effectiveness of anti-angiogenic therapies. A literature search of clinical trials testing the efficacy of anti-angiogenic therapy in prostate cancer was performed using Pubmed. Surrogate markers of angiogenic activity (microvessel density and vascular endothelial growth factor A (VEGF-A) expression) were found to be associated with tumor grade, metastasis, and prognosis. Six randomizedstudies were included in this review: two phase II trials on localized and hormone-sensitive disease (n = 60 and 99 patients) and four phase III trials on castration-resistant refractory disease (n = 873 to 1224 patients). Although the phase II trials showed improved relapse-free survival and stabilisation of the disease, the phase III trials found increased toxicity and no significant improvement in overall survival. Although angiogenesis appears to have an important role in prostate cancer, the results of anti-angiogenic therapy in castration-resistant refractory disease have hitherto been disappointing. There are various possible explanations for this lack of efficacy in castration-resistant refractory disease: redundancy of angiogenic pathways, molecular heterogeneity of the disease, loss of tumor suppressor protein phosphatase and tensin homolog (PTEN) expression as well as various VEGF-A splicing isoforms with pro- and anti-angiogenic activity. A better understanding of the molecular mechanisms of angiogenesis may help to develop effective anti-angiogenic therapy in prostate cancer.
Collapse
Affiliation(s)
- Zsombor Melegh
- Department of Cellular Pathology, Southmead Hospital, Bristol BS10 5NB, UK.
| | - Sebastian Oltean
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Exeter EX12LU, UK.
| |
Collapse
|
222
|
Palmela C, Gouveia C, Fidalgo C, Ferreira AO. Rare case of a giant duodenal ulcer penetrating the pancreas during antiangiogenic treatment. BMJ Case Rep 2019; 12:12/5/e228612. [PMID: 31061194 DOI: 10.1136/bcr-2018-228612] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
We report a case of a patient with renal cell carcinoma on pazopanib, who presented with severe upper gastrointestinal bleeding. Endoscopy showed a giant bulbar ulcer with a visible vessel of 4 mm. Due to unavailability of surgical rescue backup, large calibre vessel treatment was delayed. Endoscopy was repeated after 48 hours and showed a reduction in the vessel diameter. Endoscopic adrenalin injection and electrocoagulation were performed. However, the vessel increased in size and became pulsatile. The patient was operated, confirming a giant bulbar ulcer penetrating the pancreas with active bleeding from the gastroduodenal artery. Pazopanib therapy was suspended, and the patient is asymptomatic. Antiangiogenic treatment has been associated with gastrointestinal bleeding, perforation and fistulisation. Although we cannot confirm the causal association between the penetrating ulcer and pazopanib, the absence of Helicobacter pylori infection or non-steroidal anti-inflammatory drugs, and the reported cases of gastrointestinal bleeding during these therapies favour a possible association.
Collapse
Affiliation(s)
- Carolina Palmela
- Gastroenterology Department, Hospital Beatriz Angelo, Loures, Portugal
| | - Catarina Gouveia
- Gastroenterology Department, Hospital Beatriz Angelo, Loures, Portugal
| | - Catarina Fidalgo
- Gastroenterology Department, Hospital Beatriz Angelo, Loures, Portugal
| | | |
Collapse
|
223
|
Makker V, Rasco D, Vogelzang NJ, Brose MS, Cohn AL, Mier J, Di Simone C, Hyman DM, Stepan DE, Dutcus CE, Schmidt EV, Guo M, Sachdev P, Shumaker R, Aghajanian C, Taylor M. Lenvatinib plus pembrolizumab in patients with advanced endometrial cancer: an interim analysis of a multicentre, open-label, single-arm, phase 2 trial. Lancet Oncol 2019; 20:711-718. [PMID: 30922731 PMCID: PMC11686814 DOI: 10.1016/s1470-2045(19)30020-8] [Citation(s) in RCA: 353] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/09/2019] [Accepted: 01/10/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Lenvatinib is a multikinase inhibitor of VEGFR1, VEGFR2, and VEGFR3, and other receptor tyrosine kinases. Pembrolizumab, an antibody targeting PD-1, has moderate efficacy in biomarker-unselected endometrial cancer. We aimed to assess the combination of lenvatinib plus pembrolizumab in patients with advanced endometrial carcinoma, after establishing the maximum tolerated dose in a phase 1b study. METHODS In this open-label, single-arm, phase 2 study done at 11 centres in the USA, eligible patients were aged 18 years or older and had metastatic endometrial cancer (unselected for microsatellite instability or PD-L1), had an Eastern Cooperative Oncology Group performance status of 0 or 1, had received no more than two previous systemic therapies, had measurable disease according to the immune-related Response Evaluation Criteria In Solid Tumors (irRECIST), and had a life expectancy of 12 weeks or longer. Patients received 20 mg oral lenvatinib daily plus 200 mg intravenous pembrolizumab every 3 weeks. Treatment continued until disease progression, development of unacceptable toxic effects, or withdrawal of consent. The primary endpoint of this interim analysis was the proportion of patients with an objective response at week 24 as assessed by investigators according to irRECIST in the per-protocol population. This trial is registered with ClinicalTrials.gov, number NCT02501096. FINDINGS Between Sept 10, 2015, and July 24, 2017, 54 patients were enrolled, 53 of whom were included in the analysis. At the cutoff date for anti-tumour activity data (Dec 15, 2017), median study follow-up was 13·3 months (IQR 6·7-20·1). 21 (39·6% [95% CI 26·5-54·0]) patients had an objective response at week 24. Serious treatment-related adverse events occurred in 16 (30%) patients, and one treatment-related death was reported (intracranial haemorrhage). The most frequently reported any-grade treatment-related adverse events were hypertension (31 [58%]), fatigue (29 [55%]), diarrhoea (27 [51%]), and hypothyroidism (25 [47%]). The most common grade 3 treatment-related adverse events were hypertension (18 [34%]) and diarrhoea (four [8%]). No grade 4 treatment-related adverse events were reported. Five (9%) patients discontinued study treatment because of treatment-related adverse events. INTERPRETATION Lenvatinib plus pembrolizumab showed anti-tumour activity in patients with advanced recurrent endometrial cancer with a safety profile that was similar to those previously reported for lenvatinib and pembrolizumab monotherapies, apart from an increased frequency of hypothyroidism. Lenvatinib plus pembrolizumab could represent a new potential treatment option for this patient population, and is being investigated in a randomised phase 3 study. FUNDING Eisai and Merck.
Collapse
Affiliation(s)
- Vicky Makker
- Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | | | - Nicholas J Vogelzang
- Comprehensive Cancer Centers of Nevada and US Oncology Research, Las Vegas, NV, USA
| | - Marcia S Brose
- Perelman Center for Advanced Medicine, Philadelphia, PA, USA
| | - Allen L Cohn
- Rocky Mountain Cancer Center and US Oncology Research, Denver, CO, USA
| | - James Mier
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | - David M Hyman
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | | | | | | | | | | | - Matthew Taylor
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
224
|
Debeuckelaere C, Murgioni S, Lonardi S, Girardi N, Alberti G, Fano C, Gallimberti S, Magro C, Ahcene-Djaballah S, Daniel F, Fassan M, Prenen H, Loupakis F. Ramucirumab: the long and winding road toward being an option for mCRC treatment. Expert Opin Biol Ther 2019; 19:399-409. [PMID: 30917706 DOI: 10.1080/14712598.2019.1600505] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/25/2019] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Colorectal cancer (CRC) is one of the main causes of cancer-related morbidity and mortality worldwide. Mortality is most often attributable to metastatic disease. Despite the progress achieved so far, life expectancy continues to be limited in most patients. Ramucirumab, a most recent antiangiogenic drug, is vying in the race to metastatic CRC (mCRC) treatment since its approval by the Food and Drug Administration (FDA), based on the results of the RAISE study. AREAS COVERED This article reviews the role of ramucirumab in mCRC, including clinical indication, safety issues, and future perspectives. EXPERT OPINION The use of Ramucirumab in clinical practice is still limited, probably due to economic burden and the lack of specific biomarkers. Future efforts will be addressed to improve our knowledge in the use of this drug and better guide us in patients' care.
Collapse
Affiliation(s)
| | - Sabina Murgioni
- b Department of Oncology, Unit of Oncology 1 , Veneto Institute of Oncology, Scientific Institute for Research and Healthcare (IRCCS) , Padua , Italy
| | - Sara Lonardi
- b Department of Oncology, Unit of Oncology 1 , Veneto Institute of Oncology, Scientific Institute for Research and Healthcare (IRCCS) , Padua , Italy
| | - Noemi Girardi
- c Department of Surgery, Oncology and Gastroenterology , University of Padua , Padua , Italy
| | - Giulia Alberti
- b Department of Oncology, Unit of Oncology 1 , Veneto Institute of Oncology, Scientific Institute for Research and Healthcare (IRCCS) , Padua , Italy
- c Department of Surgery, Oncology and Gastroenterology , University of Padua , Padua , Italy
| | - Carolina Fano
- d Research Nurses Coordinating Center, Unit of Oncology 1 , Veneto Institute of Oncology, Scientific Institute for Research and Healthcare (IRCCS) , Padua , Italy
| | - Sara Gallimberti
- d Research Nurses Coordinating Center, Unit of Oncology 1 , Veneto Institute of Oncology, Scientific Institute for Research and Healthcare (IRCCS) , Padua , Italy
| | - Cristina Magro
- d Research Nurses Coordinating Center, Unit of Oncology 1 , Veneto Institute of Oncology, Scientific Institute for Research and Healthcare (IRCCS) , Padua , Italy
| | - Selma Ahcene-Djaballah
- b Department of Oncology, Unit of Oncology 1 , Veneto Institute of Oncology, Scientific Institute for Research and Healthcare (IRCCS) , Padua , Italy
| | - Francesca Daniel
- b Department of Oncology, Unit of Oncology 1 , Veneto Institute of Oncology, Scientific Institute for Research and Healthcare (IRCCS) , Padua , Italy
- e Clinical Oncology, Department of Morphology, Surgery and Experimental Medicine , S. Anna University Hospital , Ferrara , Italy
| | - Matteo Fassan
- f Department of Medicine, Pathology and Cytopathology Unit , Padua University Hospital , Padua , Italy
| | - Hans Prenen
- a Department of Oncology , University Hospital Antwerp , Edegem , Belgium
- g Center for Oncological Research , Antwerp University , Edegem , Belgium
| | - Fotios Loupakis
- b Department of Oncology, Unit of Oncology 1 , Veneto Institute of Oncology, Scientific Institute for Research and Healthcare (IRCCS) , Padua , Italy
| |
Collapse
|
225
|
Seebacher NA, Stacy AE, Porter GM, Merlot AM. Clinical development of targeted and immune based anti-cancer therapies. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:156. [PMID: 30975211 PMCID: PMC6460662 DOI: 10.1186/s13046-019-1094-2] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 02/07/2019] [Indexed: 02/08/2023]
Abstract
Cancer is currently the second leading cause of death globally and is expected to be responsible for approximately 9.6 million deaths in 2018. With an unprecedented understanding of the molecular pathways that drive the development and progression of human cancers, novel targeted therapies have become an exciting new development for anti-cancer medicine. These targeted therapies, also known as biologic therapies, have become a major modality of medical treatment, by acting to block the growth of cancer cells by specifically targeting molecules required for cell growth and tumorigenesis. Due to their specificity, these new therapies are expected to have better efficacy and limited adverse side effects when compared with other treatment options, including hormonal and cytotoxic therapies. In this review, we explore the clinical development, successes and challenges facing targeted anti-cancer therapies, including both small molecule inhibitors and antibody targeted therapies. Herein, we introduce targeted therapies to epidermal growth factor receptor (EGFR), vascular endothelial growth factor (VEGF), human epidermal growth factor receptor 2 (HER2), anaplastic lymphoma kinase (ALK), BRAF, and the inhibitors of the T-cell mediated immune response, cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein-1 (PD-1)/ PD-1 ligand (PD-1 L).
Collapse
Affiliation(s)
- N A Seebacher
- Faculty of Medicine, The University of Sydney, Camperdown, New South Wales, 2006, Australia
| | - A E Stacy
- Faculty of Medicine, The University of Notre Dame, Darlinghurst, New South Wales, 2010, Australia
| | - G M Porter
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Kensington, New South Wales, 2031, Australia
| | - A M Merlot
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Kensington, New South Wales, 2031, Australia. .,School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Kensington, New South Wales, 2031, Australia. .,UNSW Centre for Childhood Cancer Research, Faculty of Medicine, University of New South Wales, Kensington, New South Wales, 2031, Australia.
| |
Collapse
|
226
|
Flores AM, Ye J, Jarr KU, Hosseini-Nassab N, Smith BR, Leeper NJ. Nanoparticle Therapy for Vascular Diseases. Arterioscler Thromb Vasc Biol 2019; 39:635-646. [PMID: 30786744 PMCID: PMC6436996 DOI: 10.1161/atvbaha.118.311569] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Nanoparticles promise to advance strategies to treat vascular disease. Since being harnessed by the cancer field to deliver safer and more effective chemotherapeutics, nanoparticles have been translated into applications for cardiovascular disease. Systemic exposure and drug-drug interactions remain a concern for nearly all cardiovascular therapies, including statins, antithrombotic, and thrombolytic agents. Moreover, off-target effects and poor bioavailability have limited the development of completely new approaches to treat vascular disease. Through the rational design of nanoparticles, nano-based delivery systems enable more efficient delivery of a drug to its therapeutic target or even directly to the diseased site, overcoming biological barriers and enhancing a drug's therapeutic index. In addition, advances in molecular imaging have led to the development of theranostic nanoparticles that may simultaneously act as carriers of both therapeutic and imaging payloads. The following is a summary of nanoparticle therapy for atherosclerosis, thrombosis, and restenosis and an overview of recent major advances in the targeted treatment of vascular disease.
Collapse
Affiliation(s)
- Alyssa M. Flores
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine., Hanover, NH
- Geisel School of Medicine at Dartmouth, Hanover, NH
| | - Jianqin Ye
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine., Hanover, NH
| | - Kai-Uwe Jarr
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine., Hanover, NH
| | - Niloufar Hosseini-Nassab
- Department of Radiology, Stanford University School of Medicine, Michigan State University, East Lansing, MI, USA
| | - Bryan R. Smith
- Department of Biomedical Engineering and Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Nicholas J. Leeper
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine., Hanover, NH
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA
- Stanford Cardiovascular Institute, Stanford, CA 94305, USA
| |
Collapse
|
227
|
Structure-based identification of potent VEGFR-2 inhibitors from in vivo metabolites of a herbal ingredient. J Mol Model 2019; 25:98. [PMID: 30904971 DOI: 10.1007/s00894-019-3979-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 03/05/2019] [Indexed: 12/22/2022]
Abstract
Vascular endothelial growth factor receptor-2 (VEGFR-2) is one of the regulatory elements of angiogenesis that is expressed highly in various diseases and is also essential for solid tumor growth. The present study was aimed at identifying potent inhibitors of VEGFR-2 by considering herbal secondary metabolites; as natural molecules are less toxic than synthetic derivatives. A structure-based virtual screening protocol consisting of molecular docking, MM-GBSA and ADME/T analysis was initially used to screen a library of in vivo metabolites of the herbal ingredient. Using a fixed cutoff value, four potent virtual hits were identified from molecular docking, ADME/T and binding affinity calculations, which were considered further for molecular dynamics (MD) simulation to broadly describe the binding mechanisms to VEGFR-2. The results suggested that these molecules have high affinity for the catalytic region of VEGFR-2, and form strong hydrophobic and polar interactions with the amino acids involved in the binding site of ATP and linker regions of the catalytic site. Subsequently, the stability of the docked complexes and binding mechanisms were evaluated by MD simulations, and the energy of binding was calculated through MM-PBSA analysis. The results uncovered two virtual hits, designated ZINC14762520 and ZINC36470466, as VEGFR-2 inhibitors, and suggested that they bind to kinase domain in an ATP-competitive manner. These virtual hits will offer a suitable starting point for the further design of their various analogs, allowing a rational search for more effective inhibitors in the future. Graphical abstract.
Collapse
|
228
|
Yeung AWK, Abdel-Daim MM, Abushouk AI, Kadonosono K. A literature analysis on anti-vascular endothelial growth factor therapy (anti-VEGF) using a bibliometric approach. Naunyn Schmiedebergs Arch Pharmacol 2019; 392:393-403. [PMID: 30826857 DOI: 10.1007/s00210-019-01629-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 02/06/2019] [Indexed: 12/24/2022]
Abstract
We performed the current study to assess the citation performance of research works on anti-vascular endothelial growth factor (anti-VEGF) therapy. We searched Web of Science (WoS) to identify relevant publications and analyze them with reference to their publication year, journal title, citation count, WoS category, and article type. The bibliometric software (VOSviewer) was used for citation analyses of countries and journals and to generate a term map that visualizes the recurring terms appearing in the titles and abstracts of published articles. The literature search resulted in 7364 articles, with a mean citation count of 26.2. Over half of them (50.2%) were published during the past 5 years. Original articles constituted the majority (67.8%). The publications were mainly classified into WoS categories of ophthalmology (43.2%) and oncology (20.6%). The most prolific ophthalmology and cancer journals were Investigative Ophthalmology & Visual Science (7.3%) and Cancer Research (1.4%), respectively. The correlation between journal impact factor and citation count was weak to moderate (for journals with impact factor up to 5 and 10, respectively), and open-access articles had significantly more citations than non-open-access articles (p < 0.001). The frequently targeted tumors by anti-VEGF therapy included metastatic colorectal cancer (196; 49.2 citations per article (CPA)), breast cancers (167; 37.2 CPA), and renal cell carcinoma (122; 38.2 CPA). The frequently targeted eye pathologies were age-related macular degeneration (828; 18.2 CPA), diabetic macular edema (466; 10.8 CPA), and diabetic retinopathy (358; 31.9 CPA). These results indicate that anti-VEGF therapy has a wide range of applications and its publications are highly cited.
Collapse
Affiliation(s)
- Andy Wai Kan Yeung
- Oral and Maxillofacial Radiology, Applied Oral Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong S.A.R., China.
| | - Mohamed M Abdel-Daim
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt.
- Department of Ophthalmology and Micro-Technology, Yokohama City University, Yokohama, Japan.
| | | | - Kazuaki Kadonosono
- Department of Ophthalmology and Micro-Technology, Yokohama City University, Yokohama, Japan
| |
Collapse
|
229
|
Zou J, Zhu W, Meng H, Luo P, Zhang J. Efficacy and safety of selective internal radiotherapy versus sorafenib for intermediate-locally advanced hepatocellular carcinoma: a systematic review and meta-analysis. Expert Rev Gastroenterol Hepatol 2019; 13:271-279. [PMID: 30791765 DOI: 10.1080/17474124.2019.1570135] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Sorafenib (SOR) is recommended for locally advanced and metastatic hepatocellular carcinoma (HCC), but the tolerability of SOR is unsatisfactory. Selective internal radiotherapy (SIRT) has shown efficacy in intermediate-locally advanced HCC patients. This meta-analysis aimed to compare the efficacy and safety of SIRT and SOR in the treatment of intermediate-locally advanced HCC. METHODS We systematically searched the PubMed, Embase, Cochrane Library and Web of Science databases for eligible studies. The endpoints evaluated included the overall survival (OS), disease control rate (DCR), objective response rate (ORR) and grade≥3 adverse events (AEs). RESULTS Six studies were included in this analysis. The OS was similar between the two groups (HR 1.06, 95%CI 0.93-1.20; P = 0.40). There was no difference in the DCR between the two groups (RR 1.13, 95%CI 0.87-1.46; P = 0.35). However, the ORR in the SIRT group was significantly higher than that in the SOR group (RR 4.10, 95%CI 1.92-8.76; P = 0.0003). The incidence rate of grade≥3 AEs was higher in the SOR group. CONCLUSIONS In patients with intermediate-locally advanced HCC, SIRT and SOR result in similar survival rates. The improved toxicity profile of SIRT may help when choosing between the two treatments.
Collapse
Affiliation(s)
- Jingwen Zou
- a Oncology Center , Zhujiang Hospital of Southern Medical University , Guangzhou , Guangdong Province , China
| | - Weiliang Zhu
- a Oncology Center , Zhujiang Hospital of Southern Medical University , Guangzhou , Guangdong Province , China
| | - Hui Meng
- a Oncology Center , Zhujiang Hospital of Southern Medical University , Guangzhou , Guangdong Province , China
| | - Peng Luo
- a Oncology Center , Zhujiang Hospital of Southern Medical University , Guangzhou , Guangdong Province , China
| | - Jian Zhang
- a Oncology Center , Zhujiang Hospital of Southern Medical University , Guangzhou , Guangdong Province , China
| |
Collapse
|
230
|
Pitoia F, Schmidt A, Bueno F, Abelleira E, Jerkovich F. Rare complications of multikinase inhibitor treatment. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2019; 62:636-640. [PMID: 30624504 PMCID: PMC10118667 DOI: 10.20945/2359-3997000000090] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 10/30/2018] [Indexed: 11/23/2022]
Abstract
OBJECTIVE The advent of multikinase inhibitor (MKI) therapy has led to a radical change in the treatment of patients with advanced thyroid carcinoma. The aim of this manuscript is to communicate rare adverse events that occurred in less than 5% of patients in clinical trials in a subset of patients treated in our hospital. SUBJECTS AND METHODS Out of 760 patients with thyroid cancer followed up with in our Division of Endocrinology, 29 (3.8%) received treatment with MKIs. The median age at diagnosis of these patients was 53 years (range 20-70), and 75.9% of them were women. Sorafenib was prescribed as first-line treatment to 23 patients with differentiated thyroid cancer and as second-line treatment to one patient with advanced medullary thyroid cancer (MTC). Vandetanib was indicated as first-line treatment in 6 patients with MTC and lenvatinib as second-line treatment in two patients with progressive disease under sorafenib treatment. RESULTS During the follow-up of treatment (mean 13.7 ± 7 months, median 12 months, range 6-32), 5/29 (17.2%) patients presented rare adverse events. These rare adverse effects were: heart failure, thrombocytopenia, and squamous cell carcinoma during sorafenib therapy and squamous cell carcinoma and oophoritis with intestinal perforation during vandetanib treatment. CONCLUSIONS About 3 to 5 years after the approval of MKI therapy, we learned that MKIs usually lead to adverse effects in the majority of patients. Although most of them are manageable, we still need to be aware of potentially serious and rare or unreported adverse effects that can be life-threatening.
Collapse
Affiliation(s)
- Fabián Pitoia
- Division of Endocrinology, University of Buenos Aires, Buenos Aires, Argentina
| | - Angélica Schmidt
- Division of Endocrinology, University of Buenos Aires, Buenos Aires, Argentina
| | - Fernanda Bueno
- Division of Endocrinology, University of Buenos Aires, Buenos Aires, Argentina
| | - Erika Abelleira
- Division of Endocrinology, University of Buenos Aires, Buenos Aires, Argentina
| | - Fernando Jerkovich
- Division of Endocrinology, University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
231
|
Kugeratski FG, Atkinson SJ, Neilson LJ, Lilla S, Knight JRP, Serneels J, Juin A, Ismail S, Bryant DM, Markert EK, Machesky LM, Mazzone M, Sansom OJ, Zanivan S. Hypoxic cancer-associated fibroblasts increase NCBP2-AS2/HIAR to promote endothelial sprouting through enhanced VEGF signaling. Sci Signal 2019; 12:eaan8247. [PMID: 30723174 PMCID: PMC6794160 DOI: 10.1126/scisignal.aan8247] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Intratumoral hypoxia causes the formation of dysfunctional blood vessels, which contribute to tumor metastasis and reduce the efficacy of therapeutic treatments. Blood vessels are embedded in the tumor stroma of which cancer-associated fibroblasts (CAFs) constitute a prominent cellular component. We found that hypoxic human mammary CAFs promoted angiogenesis in CAF-endothelial cell cocultures in vitro. Mass spectrometry-based proteomic analysis of the CAF secretome unraveled that hypoxic CAFs contributed to blood vessel abnormalities by altering their secretion of various pro- and anti-angiogenic factors. Hypoxia induced pronounced remodeling of the CAF proteome, including proteins that have not been previously related to this process. Among those, the uncharacterized protein NCBP2-AS2 that we renamed HIAR (hypoxia-induced angiogenesis regulator) was the protein most increased in abundance in hypoxic CAFs. Silencing of HIAR abrogated the pro-angiogenic and pro-migratory function of hypoxic CAFs by decreasing secretion of the pro-angiogenic factor VEGFA and consequently reducing VEGF/VEGFR downstream signaling in the endothelial cells. Our study has identified a regulator of angiogenesis and provides a map of hypoxia-induced molecular alterations in mammary CAFs.
Collapse
Affiliation(s)
| | | | - Lisa J Neilson
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK
| | - Sergio Lilla
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK
| | | | - Jens Serneels
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology (CCB), VIB, 3000 Leuven, Belgium
| | - Amelie Juin
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK
| | - Shehab Ismail
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - David M Bryant
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Elke K Markert
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Laura M Machesky
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology (CCB), VIB, 3000 Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, KU Leuven, 3000 Leuven, Belgium
| | - Owen J Sansom
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Sara Zanivan
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK.
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| |
Collapse
|
232
|
van Cleemput J, Sonaglioni A, Wuyts WA, Bengus M, Stauffer JL, Harari S. Idiopathic Pulmonary Fibrosis for Cardiologists: Differential Diagnosis, Cardiovascular Comorbidities, and Patient Management. Adv Ther 2019; 36:298-317. [PMID: 30554332 PMCID: PMC6824347 DOI: 10.1007/s12325-018-0857-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Indexed: 02/06/2023]
Abstract
The presence of rare comorbidities in patients with cardiovascular disease (CVD) presents a diagnostic challenge to cardiologists. In evaluating these patients, cardiologists are faced with a unique opportunity to shorten diagnosis times and direct patients towards correct treatment pathways. Idiopathic pulmonary fibrosis (IPF), a type of interstitial lung disease (ILD), is an example of a rare disease where patients frequently demonstrate comorbid CVD. Both CVD and IPF most commonly affect a similar patient demographic: men over the age of 60 years with a history of smoking. Moreover, IPF and heart failure (HF) share a number of symptoms. As a result, patients with IPF can be misdiagnosed with HF and vice versa. This article aims to increase awareness of IPF among cardiologists, providing an overview for cardiologists on the differential diagnosis of IPF from HF, and describing the signs and symptoms that would warrant referral to a pulmonologist with expertise in ILD. Once patients with IPF have received a diagnosis, cardiologists can have an important role in managing patients who are candidates for a lung transplant or those who develop pulmonary hypertension (PH). Group 3 PH is one of the most common cardiovascular complications diagnosed in patients with IPF, its prevalence varying between reports but most often cited as between 30% and 50%. This review summarizes the current knowledge on Group 3 PH in IPF, discusses data from clinical trials assessing treatments for Group 1 PH in patients with IPF, and highlights that treatment guidelines recommend against these therapies in IPF. Finally, this article provides the cardiologist with an overview on the use of the two approved treatments for IPF, the antifibrotics pirfenidone and nintedanib, in patients with IPF and CVD comorbidities. Conversely, the impact of treatments for CVD comorbidities on patients with IPF is also discussed.Funding: F. Hoffmann-La Roche, Ltd.Plain Language Summary: Plain language summary available for this article.
Collapse
Affiliation(s)
| | - Andrea Sonaglioni
- U.O. di Cardiologia, Ospedale San Giuseppe - MultiMedica IRCCS, Milan, Italy
| | - Wim A Wuyts
- Department of Respiratory Medicine, Unit for Interstitial Lung Diseases, University Hospitals Leuven, Leuven, Belgium
| | | | | | - Sergio Harari
- U.O. di Pneumologia e Terapia Semi-Intensiva Respiratoria - Servizio di Fisiopatologia Respiratoria ed Emodinamica Polmonare, Ospedale San Giuseppe - MultiMedica IRCCS, Milan, Italy
| |
Collapse
|
233
|
Abd-El-Barr MM, Huang KT, Moses ZB, Iorgulescu JB, Chi JH. Recent advances in intradural spinal tumors. Neuro Oncol 2019; 20:729-742. [PMID: 29216380 DOI: 10.1093/neuonc/nox230] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Intradural spinal tumors are rare tumors of the central nervous system. Due to the eloquence of the spinal cord and its tracts, the compact architecture of the cord and nerves, and the infiltrative nature of some of these tumors, surgical resection is difficult to achieve without causing neurological deficits. Likewise, chemotherapy and radiotherapy are utilized more cautiously in the treatment of intradural spinal tumors than their cranial counterparts. Targeted therapies aimed at the genetic alterations and molecular biology tailored to these tumors would be helpful but are lacking.Here, we review the major types of intradural spinal tumors, with an emphasis on genetic alterations, molecular biology, and experimental therapies for these difficult to treat neoplasms.
Collapse
Affiliation(s)
- Muhammad M Abd-El-Barr
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Kevin T Huang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ziev B Moses
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - J Bryan Iorgulescu
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - John H Chi
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
234
|
Long-term therapy with bevacizumab in a young patient affected by NF-2: a case report and review of the literature. Anticancer Drugs 2019; 30:318-321. [PMID: 30640792 DOI: 10.1097/cad.0000000000000732] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Neurofibromatosis type 2 (NF-2) is an autosomal dominant inherited disease caused by heterozygous mutations in the NF-2 tumor suppressor gene. It is characterized by the development of multiple benign tumors in the central nervous system. A majority of these tumors can be treated with surgery or radiotherapy in the case of the symptomatic disease. Cytotoxic chemotherapy has no established role in the treatment of NF-2. Vascular endothelial growth factor (VEGF) is a critical mediator of tumor angiogenesis and vessel permeability. VEGF and its receptor VEGFR-1 have been detected in schwannomas, and increased levels of these factors correlate with increased rates of tumor growth. The use of bevacizumab has made many progresses in recent years in NF-2 patients. We report a case of a young patient treated with more than 100 administration of bevacizumab, with clinical and instrumental benefits.
Collapse
|
235
|
Murota H, Shinya T, Nishiuchi A, Sakanaka M, Toda KI, Ogata T, Hayama N, Kimachi T, Takahashi S. Inhibition of angiogenesis and tumor growth by a novel 1,4-naphthoquinone derivative. Drug Dev Res 2019; 80:395-402. [PMID: 30632632 DOI: 10.1002/ddr.21513] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 12/26/2018] [Indexed: 01/07/2023]
Abstract
Hit, Lead & Candidate Discovery Antiangiogenesis therapy is a promising way for treatment of solid cancers, and many angiogenesis inhibitors that target vascular endothelial growth factor (VEGF) or its receptors have been developed. We explored novel antiangiogenic compounds other than anti-VEGF drugs by screening our synthetic compound library and found that 6-thiophen-3-yl-2-methoxy-1,4-naphthoquinone (6-TMNQ) had potential as a novel angiogenesis inhibitor. This paper describes the effects of 6-TMNQ on angiogenesis and tumor growth in vitro and in vivo. 6-TMNQ inhibited serum-, VEGF-, and basic fibroblast growth factor (bFGF)-stimulated proliferation of endothelial cells in a concentration-dependent manner, but had no effect on the proliferation of fibroblasts. VEGF-induced activation of VEGF receptor-2 in endothelial cells was not affected by the compound. 6-TMNQ markedly abrogated both migration and tube formation of endothelial cells. Orally administered 6-TMNQ inhibited angiogenesis in response to VEGF or bFGF in mice in a dose-dependent manner. Furthermore, when tumor-bearing mice were treated with 6-TMNQ, increase in tumor size was significantly prevented due to inhibition of angiogenesis in the tumor tissues. These results demonstrate that 6-TMNQ is an orally available compound that selectively inhibits endothelial cell proliferation and migration, and abrogates angiogenesis, resulting in the prevention of tumor growth. The mechanism of 6-TMNQ action is different from that of conventional anti-VEGF drugs.
Collapse
Affiliation(s)
- Hiroko Murota
- Department of Immunobiology, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
| | - Tomohiro Shinya
- Department of Immunobiology, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
| | - Arisa Nishiuchi
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
| | - Mariko Sakanaka
- Department of Immunobiology, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
| | - Ken-Ichi Toda
- Department of Dermatology, Kitano Hospital, The Tazuke Kofukai Medical Institute, Osaka, Japan
| | - Tokutaro Ogata
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
| | - Noboru Hayama
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
| | - Tetsutaro Kimachi
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
| | - Satoru Takahashi
- Department of Immunobiology, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, Nishinomiya, Hyogo, Japan.,Institute for Biosciences, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
| |
Collapse
|
236
|
Cabanillas ME, Takahashi S. Managing the adverse events associated with lenvatinib therapy in radioiodine-refractory differentiated thyroid cancer. Semin Oncol 2018; 46:57-64. [PMID: 30685073 DOI: 10.1053/j.seminoncol.2018.11.004] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 11/21/2018] [Indexed: 02/06/2023]
Abstract
Lenvatinib is a multikinase inhibitor of vascular endothelial growth factor (VEGF) receptors 1-3, fibroblast growth factor receptors 1-4, RET, KIT, and platelet-derived growth factor receptor-α. Lenvatinib is approved as a monotherapy for the treatment of radioiodine-refractory differentiated thyroid cancer and in combination with everolimus for the second-line treatment of advanced renal cell carcinoma. Lenvatinib is also under investigation for the treatment of several malignancies including unresectable hepatocellular carcinoma. Although lenvatinib is associated with favorable efficacy, it is associated with adverse events (AEs) that the clinician will have to closely monitor for and proactively manage. Most of these AEs are known class effects of VEGF-targeted therapies, including hypertension, diarrhea, fatigue or asthenia, decreased appetite, and weight loss. This review summarizes the safety profile of lenvatinib and offers guidance for the management of both frequent and rare AEs. We discuss the potential mechanisms underlying these AEs and present practical recommendations for managing toxicities. The development of treatment plans that include prophylactic and therapeutic strategies for the management of lenvatinib-associated AEs has the potential to improve patient quality of life, optimize adherence, minimize the need for dose reductions, treatment interruptions, or discontinuations, and maximize patient outcomes.
Collapse
Affiliation(s)
- Maria E Cabanillas
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Shunji Takahashi
- Department of Medical Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
237
|
Ikeuchi T, de Vega S, Forcinito P, Doyle AD, Amaral J, Rodriguez IR, Arikawa-Hirasawa E, Yamada Y. Extracellular Protein Fibulin-7 and Its C-Terminal Fragment Have In Vivo Antiangiogenic Activity. Sci Rep 2018; 8:17654. [PMID: 30518776 PMCID: PMC6281620 DOI: 10.1038/s41598-018-36182-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 11/12/2018] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis is crucial for tissue development and homeostasis; however, excessive angiogenesis can lead to diseases, including arthritis and cancer metastasis. Some antiangiogenic drugs are available, but side effects remain problematic. Thus, alternative angiogenesis inhibition strategies are needed. Fibulin-7 (Fbln7) is a newly discovered member of the fibulin protein family, a group of cell-secreted glycoproteins, that functions as a cell adhesion molecule and interacts with other extracellular matrix (ECM) proteins as well as cell receptors. We previously showed that a recombinant C-terminal Fbln7 fragment (Fbln7-C) inhibits tube formation by human umbilical vein endothelial cells (HUVECs) in vitro. In the present study, we examined the in vivo antiangiogenic activity of recombinant full-length Fbln7 (Fbln7-FL) and Fbln7-C proteins using a rat corneal angiogenesis model. We found that both Fbln7-FL and Fbln7-C inhibited neovascularization. Fbln7-C bound to vascular endothelial growth factor receptor 2 (VEGFR2), inhibiting VEGFR2 and ERK phosphorylation and resulting in reduced HUVEC motility. HUVEC attachment to Fbln7-C occurred through an interaction with integrin α5β1 and regulated changes in cellular morphology. These results suggest that Fbln7-C action may target neovascularization by altering cell/ECM associations. Therefore, Fbln7-C could have potential as a therapeutic agent for diseases associated with angiogenesis.
Collapse
Affiliation(s)
- Tomoko Ikeuchi
- Molecular Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, 20892, USA.
| | - Susana de Vega
- Molecular Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, 20892, USA
- Research Department of Pathophysiology for Locomotive and Neoplastic Diseases, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Patricia Forcinito
- Molecular Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, 20892, USA
- Office of Portfolio Analysis, Office of the Director, Bethesda, Maryland, 20892, USA
| | - Andrew D Doyle
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Juan Amaral
- Mechanism of Retinal Diseases Section, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, Maryland, 20892, USA
- Division of Intermural Research, National Eye Institute, National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Ignacio R Rodriguez
- Mechanism of Retinal Diseases Section, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, Maryland, 20892, USA
- Sterculia Farms, 11601 SW Fox Brown Rd, Indiantown, Florida, 33496, USA
| | - Eri Arikawa-Hirasawa
- Research Institute for Diseases of Old Age, Juntendo University School of Medicine, Tokyo, 113-8421, Japan
| | - Yoshihiko Yamada
- Molecular Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, 20892, USA.
| |
Collapse
|
238
|
Cheng C, Nayernama A, Christopher Jones S, Casey D, Waldron PE. Wound healing complications with lenvatinib identified in a pharmacovigilance database. J Oncol Pharm Pract 2018; 25:1817-1822. [DOI: 10.1177/1078155218817109] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The U.S. Food and Drug Administration (FDA) has approved several vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors, including lenvatinib, for thyroid and renal malignancies. Inhibition of the VEGFR signaling pathway impairs angiogenesis and can disrupt wound healing. The objective of this work was to evaluate wound healing complications as a potential safety risk for patients treated with lenvatinib. We searched the FDA Adverse Event Reporting System (FAERS) database for postmarketing reports of wound healing complications with lenvatinib between 13 February 2015 (FDA approval date) and 15 February 2017. The search identified nine FAERS cases of lenvatinib-associated wound healing complications that were not previously reported in the medical literature. Seven cases involved postoperative wound healing complications, such as impaired healing or wound dehiscence. In our case series, the reported time to identification of delayed wound healing from tissue injury or surgery varied over a wide range (4–58 days). The time of initial lenvatinib exposure relative to the tissue injury was also highly varied in our series, which may have influenced the development and detection of impaired healing. FAERS case-level evidence suggests that lenvatinib may have contributed to wound healing complications based on temporality and biologic plausibility. Healthcare professionals should be aware of this safety risk to facilitate prompt recognition and risk mitigation.
Collapse
Affiliation(s)
- Connie Cheng
- Division of Pharmacovigilance, Office of Surveillance and Epidemiology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Afrouz Nayernama
- Division of Pharmacovigilance, Office of Surveillance and Epidemiology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - S Christopher Jones
- Division of Pharmacovigilance, Office of Surveillance and Epidemiology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Denise Casey
- Division of Oncology Products, Office of Hematology and Oncology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Peter E Waldron
- Division of Pharmacovigilance, Office of Surveillance and Epidemiology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
239
|
Corsini LR, Fanale D, Passiglia F, Incorvaia L, Gennusa V, Bazan V, Russo A. Monoclonal antibodies for the treatment of non-hematological tumors: a safety review. Expert Opin Drug Saf 2018; 17:1197-1209. [PMID: 30457416 DOI: 10.1080/14740338.2018.1550068] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: The introduction of monoclonal antibodies (moAbs) into clinical practice revolutionized the treatment strategies in several solid tumors. These agents differ from cytotoxic chemotherapy for their mechanism of action and toxicity. By targeting specific antigens present on healthy cells and modulating immune system activity, these biological drugs are able to generate a wide spectrum of peculiar adverse events that can negatively impact on patients' quality of life. Areas covered: In this review, the main side effects associated with the use of moAbs have been described to show their incidence and current management strategies, which may drive clinicians in their daily practice. Expert opinion: The majority of these drugs represents an example of successful innovation, since they are able to induce a significant improvement of patients' survival and quality of life without any increase in related side effects as compared to standard cancer treatments. For this reason, they have become new milestones in personalized therapy for different non-hematological malignancies. With the increasing use of moAbs in treatment regimens, it is strongly recommended that clinicians are knowledgeable about the side effects associated with these agents, their management and monitoring, to optimize the clinical treatment of cancer patients.
Collapse
Affiliation(s)
- Lidia Rita Corsini
- a Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology , University of Palermo , Palermo , Italy
| | - Daniele Fanale
- a Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology , University of Palermo , Palermo , Italy
| | - Francesco Passiglia
- a Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology , University of Palermo , Palermo , Italy
| | - Lorena Incorvaia
- a Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology , University of Palermo , Palermo , Italy
| | - Vincenzo Gennusa
- a Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology , University of Palermo , Palermo , Italy
| | - Viviana Bazan
- a Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology , University of Palermo , Palermo , Italy
| | - Antonio Russo
- a Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology , University of Palermo , Palermo , Italy
| |
Collapse
|
240
|
Bartmańska A, Tronina T, Popłoński J, Milczarek M, Filip-Psurska B, Wietrzyk J. Highly Cancer Selective Antiproliferative Activity of Natural Prenylated Flavonoids. Molecules 2018; 23:molecules23112922. [PMID: 30423918 PMCID: PMC6278664 DOI: 10.3390/molecules23112922] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 10/31/2018] [Accepted: 11/08/2018] [Indexed: 02/05/2023] Open
Abstract
Xanthohumol (XN) and four minor hops prenylflavonoids: α,β-dihydroxanthohumol (2HXN), isoxanthohumol (IXN), 8-prenylnaringenin (8PN), and 6-prenylnaringenin (6PN), were tested for antiproliferative activity towards human cancer and normal cell lines. Nonprenylated naringenin (NG) was used as a model compound. Xanthohumol, α,β-dihydroxanthohumol and 6-prenylnaringenin were the most active compounds. Xanthohumol exhibited higher antiproliferative activity than cisplatin (CP) against five cancer cell lines: ovarian resistant to cisplatin A2780cis, breast MDA-MB-231 and T-47D, prostate PC-3, and colon HT-29. Isoxanthohumol was more potent than cisplatin against breast cancer cell lines MDA-MB-231 and T-47D whereas 6-prenylnaringenin was stronger than cisplatin against breast cancer cell line T-47D. It was found that tested chalcones possessed highly selective antiproliferative activity towards all tested breast cancer lines compared to the normal breast MCF 10A cell line (the calculated selectivity index ranged from 5 to 10). Low antiproliferative activity of naringenin indicates the importance of the prenyl group with respect to antiproliferative activity.
Collapse
Affiliation(s)
- Agnieszka Bartmańska
- Department of Chemistry, Wrocław University of Environmental and Life Sciences, C.K. Norwida 25, 50-375 Wrocław, Poland.
| | - Tomasz Tronina
- Department of Chemistry, Wrocław University of Environmental and Life Sciences, C.K. Norwida 25, 50-375 Wrocław, Poland.
| | - Jarosław Popłoński
- Department of Chemistry, Wrocław University of Environmental and Life Sciences, C.K. Norwida 25, 50-375 Wrocław, Poland.
| | - Magdalena Milczarek
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114 Wrocław, Poland.
| | - Beata Filip-Psurska
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114 Wrocław, Poland.
| | - Joanna Wietrzyk
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114 Wrocław, Poland.
| |
Collapse
|
241
|
Benndorf RA. Renal Biomarker and Angiostatic Mediator? Cystatin C as a Negative Regulator of Vascular Endothelial Cell Homeostasis and Angiogenesis. J Am Heart Assoc 2018; 7:e010997. [PMID: 30571391 PMCID: PMC6404208 DOI: 10.1161/jaha.118.010997] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
See Article by Li et al.
Collapse
Affiliation(s)
- Ralf A Benndorf
- 1 Department of Clinical Pharmacy and Pharmacotherapy Martin-Luther-University Halle-Wittenberg Halle (Saale) Germany
| |
Collapse
|
242
|
Cho HD, Moon KD, Park KH, Lee YS, Seo KI. Effects of auriculasin on vascular endothelial growth factor (VEGF)-induced angiogenesis via regulation of VEGF receptor 2 signaling pathways in vitro and in vivo. Food Chem Toxicol 2018; 121:612-621. [PMID: 30236598 DOI: 10.1016/j.fct.2018.09.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 09/07/2018] [Accepted: 09/15/2018] [Indexed: 12/31/2022]
Abstract
Angiogenesis plays an important role in various pathological conditions such as cancer via excessive delivery of oxygen and nutrients. Recent studies have demonstrated that understanding the molecular basis of natural agents in angiogenesis is critical for the development of promising cancer therapeutics. In this study, auriculasin, an active component from Flemingia philippinensis, was found to exert strong anti-angiogenesis activity. Treatment with auriculasin suppressed proliferation of human umbilical vein endothelial cells (HUVECs) by modulating expression of Bcl-2, Bcl-XL, and vascular endothelial growth factor (VEGF). Further, auriculasin inhibited VEGF-induced chemotactic migration, invasion, and capillary-like structure formation of endothelial cells. In addition, auriculasin abrogated VEGF-induced vascular network formation around rat aortic rings as well as blocked accumulation of hemoglobin, endothelial cells and VEGF in the Matrigel plug of C57BL/6 mice. The inhibitory effect of auriculasin on angiogenesis was well correlated with inhibition of VEGF receptor 2 (VEGFR2) activation as well as phosphorylation of intracellular downstream protein kinases of VEGFR2 containing Akt, mammalian target of rapamycin (mTOR), phosphoinositide 3-kinase (PI3K), p-38, extracellular signal-related kinase (ERK), and Src. Taken together, this study reports that auriculasin potently inhibits angiogenesis by modulating VEGFR2-related signaling pathways, which further validates its great potential in clinical applications.
Collapse
Affiliation(s)
- Hyun-Dong Cho
- Department of Food Science and Technology, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Kwang-Deog Moon
- Department of Food Science and Technology, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Ki-Hun Park
- Division of Applied Life Science (BK21 plus), Institute of Agriculture and Life Science (IALS), Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Yong-Suk Lee
- Department of Biotechnology, Dong-A University, Busan, 49315, Republic of Korea
| | - Kwon-Il Seo
- Department of Biotechnology, Dong-A University, Busan, 49315, Republic of Korea.
| |
Collapse
|
243
|
Manolis AA, Manolis TA, Mikhailidis DP, Manolis AS. Cardiovascular safety of oncologic agents: A double-edged sword even in the era of targeted therapies - part 1. Expert Opin Drug Saf 2018; 17:875-892. [PMID: 30126304 DOI: 10.1080/14740338.2018.1513488] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Patients with cancer are subject to the cardiotoxic effects of cancer therapy and as more patients survive cancer due to improved treatment they are exposed to various forms of cardiovascular (CV) disease as they age, and vice-versa. Such an interplay of age with both malignancy and CV disease may contribute to increased morbidity and mortality. AREAS COVERED This two-part review considers the effects of cancer drug treatment on the CV system. In Part I, the various types of CV and cardiometabolic toxicity of anti-cancer drugs and the possible mechanisms involved are discussed. Also, among the specific oncologic agents, the CV effects of the classical agents and of the large molecule immunological agents (monoclonal antibodies, including immune checkpoint inhibitors) are detailed. EXPERT OPINION Oncologic agents produce a variety of CV adverse effects, including cardiomyopathy and heart failure, peri-myocarditis, coronary artery disease, peripheral vascular disease, hypertension (HTN), cardiac arrhythmias, valvular heart disease, and pulmonary HTN. Both the oncologist and the cardiologist need to be aware of such adverse effects and of the specific agents that produce them. They need to join forces to prevent, anticipate, recognize, and manage such complications.
Collapse
Affiliation(s)
| | | | - Dimitri P Mikhailidis
- c Department of Clinical Biochemistry , University College London Medical School , London , UK
| | - Antonis S Manolis
- d Third Department of Cardiology , Athens University School of Medicine , Athens , Greece
| |
Collapse
|
244
|
Justice CN, Derbala MH, Baich TM, Kempton AN, Guo AS, Ho TH, Smith SA. The Impact of Pazopanib on the Cardiovascular System. J Cardiovasc Pharmacol Ther 2018; 23:387-398. [PMID: 29706106 PMCID: PMC6257996 DOI: 10.1177/1074248418769612] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Pazopanib is an approved treatment for renal cell carcinoma and a second-line treatment for nonadipocytic soft-tissue sarcoma. However, its clinical efficacy is limited by its cardiovascular side effects. Pazopanib and other vascular endothelial growth factor receptor tyrosine kinase inhibitors have been associated with the development of hypertension, QT interval prolongation, and other cardiovascular events; however, these mechanisms are largely unknown. Gaining a deeper understanding of these mechanisms is essential for the development of appropriate surveillance strategies and possible diagnostic biomarkers to allow us to monitor patients and modulate therapy prior to significant cardiac insult. This approach will be vital in keeping patients on these life-saving therapies and may be applicable to other tyrosine kinase inhibitors as well. In this review, we provide a comprehensive overview of the preclinical and clinical side effects of pazopanib with a focus on the mechanisms responsible for its toxicity to the cardiovascular system.
Collapse
Affiliation(s)
- Cody N. Justice
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Mohamed H. Derbala
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Tesla M. Baich
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Amber N. Kempton
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Aaron S. Guo
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Thai H. Ho
- Mayo Clinic Scottsdale, Arizona, Phoenix, AZ, USA
| | - Sakima A. Smith
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
- Division of Cardiology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
245
|
Gadgeel SM, Stevenson JP, Langer CJ, Gandhi L, Borghaei H, Patnaik A, Villaruz LC, Gubens M, Hauke R, Yang JCH, Sequist LV, Bachman R, Saraf S, Raftopoulos H, Papadimitrakopoulou V. Pembrolizumab and platinum-based chemotherapy as first-line therapy for advanced non-small-cell lung cancer: Phase 1 cohorts from the KEYNOTE-021 study. Lung Cancer 2018; 125:273-281. [PMID: 30429032 DOI: 10.1016/j.lungcan.2018.08.019] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 08/16/2018] [Accepted: 08/23/2018] [Indexed: 12/26/2022]
Abstract
OBJECTIVES Platinum-based chemotherapy for advanced non-small-cell lung cancer (NSCLC) has modest benefit overall, but has the potential to amplify immune responses. In cohorts A-C of the multicohort phase 1/2 study KEYNOTE-021 (Clinicaltrials.gov, NCT02039674), we evaluated combinations of platinum-doublet chemotherapy with the anti-programmed death 1 monocloncal antibody pembrolizumab. MATERIALS AND METHODS Patients with previously untreated, advanced NSCLC without EGFR/ALK aberrations were randomized to pembrolizumab 2 or 10 mg/kg Q3W plus carboplatin area under the serum concentration-time curve (AUC) 6 mg/mL/min plus paclitaxel 200 mg/m2 (cohort A, any histology), carboplatin AUC 6 mg/mL/min plus paclitaxel 200 mg/m2 plus bevacizumab 15 mg/kg (cohort B, non-squamous), or carboplatin AUC 5 mg/mL/min plus pemetrexed 500 mg/m2 (cohort C, non-squamous) for 4 cycles followed by maintenance pembrolizumab (cohort A), pembrolizumab plus bevacizumab (cohort B), or pembrolizumab plus pemetrexed (cohort C). Response was assessed by blinded independent central review. RESULTS Overall, 74 patients were randomized; median follow-up was 21.4, 16.4, and 17.4 months in cohorts A, B, and C, respectively. No dose-limiting toxicities occurred in any cohort at either pembrolizumab dose. Most frequent treatment-related adverse events (AEs) were alopecia, fatigue, and nausea. Treatment-related grade 3/4 AEs occurred in 40%, 42%, and 46% of patients in cohorts A, B, and C, respectively; AEs with possible immune etiology occurred in 24%, 50%, and 38% of patients, respectively. Objective response rates were 48%, 56%, and 75% in cohorts A, B, and C, respectively. CONCLUSION Pembrolizumab in combination with carboplatin-paclitaxel and with pemetrexed-carboplatin yielded encouraging antitumor activity and toxicity consistent with known toxicities of platinum-based chemotherapy or pembrolizumab monotherapy.
Collapse
Affiliation(s)
| | | | - Corey J Langer
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Amita Patnaik
- South Texas Accelerated Research Therapeutics, San Antonio, TX, USA
| | - Liza C Villaruz
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Matthew Gubens
- University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| | | | - James Chih-Hsin Yang
- National Taiwan University Hospital and National Taiwan University Cancer Center, Taipei, Taiwan
| | - Lecia V Sequist
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, USA
| | | | | | | | | |
Collapse
|
246
|
Sui A, Zhong Y, Demetriades AM, Shen J, Su T, Yao Y, Gao Y, Zhu Y, Shen X, Xie B. ATN-161 as an Integrin α5β1 Antagonist Depresses Ocular Neovascularization by Promoting New Vascular Endothelial Cell Apoptosis. Med Sci Monit 2018; 24:5860-5873. [PMID: 30133427 PMCID: PMC6116638 DOI: 10.12659/msm.907446] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Accepted: 04/24/2018] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND ATN-161 (Ac-PHSCN-NH2), an antagonist of integrin α5β1, has shown an important influence in inhibiting tumor angiogenesis and metastasis of other tumor types. However, the mechanism of action of ATN-161 and whether it can inhibit ocular neovascularization (NV) are unclear. This study investigated the role of ATN-161 in regulating ocular angiogenesis in mouse models and explored the underlying signaling pathway. MATERIAL AND METHODS An oxygen-induced retinopathy (OIR) mouse model and a laser-induced choroidal neovascularization (CNV) mouse model were used to test integrin a5b1 expression and the effect of ATN-161 on ocular NV by immunofluorescence staining, Western blot analysis, and flat-mount analysis. The activation of nuclear factor-κB (NF-κB), matrix metalloproteinase-2/9 (MMP-2/9), and cell apoptosis were detected by immunofluorescence staining, Western blot, real-time RT-PCR, and terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL). The cell proliferation was detected by BrdU labeling. RESULTS In OIR and CNV mice, the protein expression level of integrin α5β1 increased compared with that in age-matched controls. The mice given ATN-161 had significantly reduced retinal neovascularization (RNV) and CNV. Blocking integrin a5b1 by ATN-161 strongly inhibited nuclear factor-κB (NF-κB) activation and matrix metalloproteinase-2/9 (MMP-2/9) expression and promoted cell apoptosis, but the effect of ATN-161 on proliferation in CNV mice was indirect and required the inhibition of neovascularization. Inhibiting NF-κB activation by ammonium pyrrolidinedithiocarbamate (PDTC) reduced RNV and promoted cell apoptosis in ocular NV. CONCLUSIONS Blocking integrin α5β1 by ATN-161 reduced ocular NV by inhibiting MMP-2/MMP-9 expression and promoting the cell apoptosis of ocular NV.
Collapse
Affiliation(s)
- Ailing Sui
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Yisheng Zhong
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Anna M. Demetriades
- Department of Ophthalmology, New York Presbyterian Hospital-Weill Cornell Medicine, New York, NY, U.S.A
| | - Jikui Shen
- Departments of Ophthalmology and Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD, U.S.A
| | - Ting Su
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Yiyun Yao
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Yushuo Gao
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Yanji Zhu
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Xi Shen
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Bing Xie
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| |
Collapse
|
247
|
Xiao B, Wang W, Zhang D. Risk of bleeding associated with antiangiogenic monoclonal antibodies bevacizumab and ramucirumab: a meta-analysis of 85 randomized controlled trials. Onco Targets Ther 2018; 11:5059-5074. [PMID: 30174444 PMCID: PMC6110629 DOI: 10.2147/ott.s166151] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aim Bevacizumab and ramucirumab are antiangiogenic monoclonal antibodies, which target vascular endothelial growth factor-A and vascular endothelial growth factor receptor-2, respectively, used in various cancers. Bleeding events have been described with these two agents. We conducted an up-to-date meta-analysis to determine the relative risk (RR) associated with the use of antiangiogenic monoclonal antibodies, bevacizumab and ramucirumab. Methods This meta-analysis of randomized controlled trials was performed after searching PubMed, American Society for Clinical Oncology Abstracts, European Society for Medical Oncology Abstracts, and the proceedings of major conferences for relevant clinical trials. RR and 95% CIs were calculated by random-effects or fixed-effects models for all-grade and high-grade bleeding events related to the angiogenesis inhibitors. Results Eighty-five randomized controlled trials were selected for the meta-analysis, covering 46,630 patients. The results showed that antiangiogenic monoclonal antibodies significantly increased the risk of all-grade (RR: 2.38, 95% CI: 2.09–2.71, p<0.00001) and high-grade (RR: 1.71, 95% CI: 1.48–1.97, p<0.00001) bleeding compared with control arms. In the subgroup analysis, bevacizumab significantly increased the risk of all-grade (RR: 2.73, 95% CI: 2.24–3.33, p<0.00001) and high-grade bleeding (RR: 1.98, 95% CI: 1.68–2.34, p<0.00001), but ramucirumab only increased the risk of all-grade bleeding (RR: 1.94, 95% CI: 1.76–2.13, p<0.00001) and no difference was observed for the risk of high-grade bleeding (RR: 1.04, 95% CI: 0.78–1.39, p=0.79) compared with the control group. For lung cancer patients, bevacizumab significantly increased the risk of all-grade (RR: 4.72, 95% CI: 1.99–11.19, p=0.0004) and high-grade pulmonary hemorrhage (RR: 3.97, 95% CI: 1.70–9.29, p=0.001), but no significant differences in the risk of all-grade (RR: 1.09, 95% CI: 0.76–1.57, p=0.64) and high-grade (RR: 1.22, 95% CI: 0.35–4.21, p=0.75) pulmonary hemorrhage were observed for ramucirumab. The increased risk of all-grade and high-grade bleeding was also observed in colorectal cancer or non-colorectal tumors and low-dose or high-dose angiogenesis inhibitors. Conclusion Antiangiogenic monoclonal antibodies are associated with a significant increase in the risk of all-grade and high-grade bleeding. Ramucirumab may be different from bevacizumab in terms of the risk of high-grade bleeding and the risk of all-grade and high-grade pulmonary hemorrhage in lung cancer patients.
Collapse
Affiliation(s)
- Bingkun Xiao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Weilan Wang
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China,
| | - Dezhi Zhang
- Department of Pharmacy, The 264th Hospital of PLA, Taiyuan, Shanxi, China
| |
Collapse
|
248
|
Rust R, Gantner C, Schwab ME. Pro- and antiangiogenic therapies: current status and clinical implications. FASEB J 2018; 33:34-48. [PMID: 30085886 DOI: 10.1096/fj.201800640rr] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Blood vessels nurture every part of the human body. Consequently, abnormalities in the vasculature are closely associated with a variety of diseases, including cerebral stroke, heart disease, retinopathy, and cancer. Pro- or antiangiogenic therapies can influence these diseases by regulating the growth of new blood vessels from a pre-existing vascular network or dampening excessive blood growth. However, clinical translation of these approaches is slow and challenging. In this review, we discuss recent preclinical approaches to regulate angiogenesis and their potential and risks in a clinical setting.-Rust, R., Gantner, C., Schwab, M. E. Pro- and antiangiogenic therapies: current status and clinical implications.
Collapse
Affiliation(s)
- Ruslan Rust
- Brain Research Institute, University of Zurich, Zurich, Switzerland.,Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland; and
| | - Christina Gantner
- Department of Biology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Martin E Schwab
- Brain Research Institute, University of Zurich, Zurich, Switzerland.,Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland; and
| |
Collapse
|
249
|
Zhao B, Zhang W, Yu D, Xu J, Wei Y. Erlotinib in combination with bevacizumab has potential benefit in non-small cell lung cancer: A systematic review and meta-analysis of randomized clinical trials. Lung Cancer 2018; 122:10-21. [DOI: 10.1016/j.lungcan.2018.05.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/06/2018] [Accepted: 05/17/2018] [Indexed: 12/09/2022]
|
250
|
Zuazo-Gaztelu I, Casanovas O. Unraveling the Role of Angiogenesis in Cancer Ecosystems. Front Oncol 2018; 8:248. [PMID: 30013950 PMCID: PMC6036108 DOI: 10.3389/fonc.2018.00248] [Citation(s) in RCA: 182] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 06/19/2018] [Indexed: 12/12/2022] Open
Abstract
Activation of the tumor and stromal cell-driven angiogenic program is one of the first requirements in the tumor ecosystem for growth and dissemination. The understanding of the dynamic angiogenic tumor ecosystem has rapidly evolved over the last decades. Beginning with the canonical sprouting angiogenesis, followed by vasculogenesis and intussusception, and finishing with vasculogenic mimicry, the need for different neovascularization mechanisms is further explored. In addition, an overview of the orchestration of angiogenesis within the tumor ecosystem cellular and molecular components is provided. Clinical evidence has demonstrated the effectiveness of traditional vessel-directed antiangiogenics, stressing on the important role of angiogenesis in tumor establishment, dissemination, and growth. Particular focus is placed on the interaction between tumor cells and their surrounding ecosystem, which is now regarded as a promising target for the development of new antiangiogenics.
Collapse
Affiliation(s)
- Iratxe Zuazo-Gaztelu
- Tumor Angiogenesis Group, ProCURE, Catalan Institute of Oncology - IDIBELL, Barcelona, Spain
| | - Oriol Casanovas
- Tumor Angiogenesis Group, ProCURE, Catalan Institute of Oncology - IDIBELL, Barcelona, Spain
| |
Collapse
|