1
|
Monaghan NP, Duckett KA, Nguyen SA, Newman JG, Albergotti WG, Kejner AE. Vascular events in patients with head and neck cancer: A systematic review and meta-analysis. Head Neck 2024; 46:1557-1572. [PMID: 38334324 DOI: 10.1002/hed.27675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/02/2024] [Accepted: 01/29/2024] [Indexed: 02/10/2024] Open
Abstract
OBJECTIVE To assess the incidence of vascular events in patients with head and neck cancer. REVIEW METHODS Primary studies identified through April 2023. Meta-analysis was performed. RESULTS There were 146 studies included in the systematic review. Rates of events were collected in the overall group, those with chemoprophylaxis, and those that underwent surgery, radiation, or chemotherapy. Of 1 184 160 patients, 4.3% had a vascular event. Radiation therapy had highest risk of overall events and stroke when compared to surgery and chemotherapy. Chemotherapy had a higher risk of stroke and overall events when compared to surgery. CONCLUSIONS Vascular events occur in 4%-5% of patients with head and neck cancer. Our data does not support the use of routine anticoagulation. Patients undergoing radiation therapy had the highest frequency of events.
Collapse
Affiliation(s)
- Neil P Monaghan
- Department of Otolaryngology, Medical University of South Carolina, Charleston, South Carolina, USA
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Kelsey A Duckett
- Department of Otolaryngology, Medical University of South Carolina, Charleston, South Carolina, USA
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Shaun A Nguyen
- Department of Otolaryngology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Jason G Newman
- Department of Otolaryngology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - W Greer Albergotti
- Department of Otolaryngology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Alexandra E Kejner
- Department of Otolaryngology, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
2
|
Hsieh CY, Lin CC, Chang WC. Taxanes in the Treatment of Head and Neck Squamous Cell Carcinoma. Biomedicines 2023; 11:2887. [PMID: 38001888 PMCID: PMC10669519 DOI: 10.3390/biomedicines11112887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/22/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023] Open
Abstract
Taxanes, particularly docetaxel (DTX), has been widely used for combination therapy of head and neck squamous cell carcinoma (HNSCC). For locally advanced unresectable HNSCC, DTX combined with cisplatin and 5-fluorouracil as a revolutionary treatment revealed an advantage in the improvement of patient outcome. In addition, DTX plus immune check inhibitors (ICIs) showed low toxicity and an increased response of patients with recurrent or metastatic HNSCC (R/M HNSCC). Accumulated data indicate that taxanes not only function as antimitotics but also impair diverse oncogenic signalings, including angiogenesis, inflammatory response, ROS production, and apoptosis induction. However, despite an initial response, the development of resistance remains a major obstacle to treatment response. Taxane resistance could result from intrinsic mechanisms, such as enhanced DNA/RNA damage repair, increased drug efflux, and apoptosis inhibition, and extrinsic effects, such as angiogenesis and interactions between tumor cells and immune cells. This review provides an overview of taxanes therapy applied in different stages of HNSCC and describe the mechanisms of taxane resistance in HNSCC. Through a detailed understanding, the mechanisms of resistance may help in developing the potential therapeutic methods and the effective combination strategies to overcome drug resistance.
Collapse
Affiliation(s)
- Ching-Yun Hsieh
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan;
| | - Ching-Chan Lin
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan;
| | - Wei-Chao Chang
- Center for Molecular Medicine, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan
| |
Collapse
|
3
|
Ansari MJ, Bokov D, Markov A, Jalil AT, Shalaby MN, Suksatan W, Chupradit S, AL-Ghamdi HS, Shomali N, Zamani A, Mohammadi A, Dadashpour M. Cancer combination therapies by angiogenesis inhibitors; a comprehensive review. Cell Commun Signal 2022; 20:49. [PMID: 35392964 PMCID: PMC8991477 DOI: 10.1186/s12964-022-00838-y] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 02/03/2022] [Indexed: 02/06/2023] Open
Abstract
Abnormal vasculature is one of the most conspicuous traits of tumor tissue, largely contributing to tumor immune evasion. The deregulation mainly arises from the potentiated pro-angiogenic factors secretion and can also target immune cells' biological events, such as migration and activation. Owing to this fact, angiogenesis blockade therapy was established to fight cancer by eliminating the nutrient and oxygen supply to the malignant cells by impairing the vascular network. Given the dominant role of vascular-endothelium growth factor (VEGF) in the angiogenesis process, the well-known anti-angiogenic agents mainly depend on the targeting of its actions. However, cancer cells mainly show resistance to anti-angiogenic agents by several mechanisms, and also potentiated local invasiveness and also distant metastasis have been observed following their administration. Herein, we will focus on clinical developments of angiogenesis blockade therapy, more particular, in combination with other conventional treatments, such as immunotherapy, chemoradiotherapy, targeted therapy, and also cancer vaccines. Video abstract.
Collapse
Affiliation(s)
- Mohammad Javed Ansari
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Kingdom of Saudi Arabia
| | - Dmitry Bokov
- Institute of Pharmacy, Sechenov First Moscow State Medical University, 8 Trubetskaya St., bldg. 2, Moscow, 119991 Russian Federation
- Laboratory of Food Chemistry, Federal Research Center of Nutrition, Biotechnology and Food Safety, 2/14 Ustyinsky pr., Moscow, 109240 Russian Federation
| | - Alexander Markov
- Tyumen State Medical University, Tyumen, Russian Federation
- Industrial University, Tyumen, Russian Federation
| | - Abduladheem Turki Jalil
- Faculty of Biology and Ecology, Yanka Kupala State University of Grodno, 230023 Grodno, Belarus
- College of Technical Engineering, The Islamic University, Najaf, Iraq
- Department of Dentistry, Kut University College, Kut, Wasit 52001 Iraq
| | - Mohammed Nader Shalaby
- Biological Sciences and Sports Health Department, Faculty of Physical Education, Suez Canal University, Ismailia, Egypt
| | - Wanich Suksatan
- Faculty of Nursing, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Supat Chupradit
- Department of Occupational Therapy, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200 Thailand
| | - Hasan S. AL-Ghamdi
- Internal Medicine Department, Division of Dermatology, Albaha University, Al Bahah, Kingdom of Saudi Arabia
| | - Navid Shomali
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Zamani
- Shiraz Transplant Center, Abu Ali Sina Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Mohammadi
- Department of Neurology, Imam Khomeini Hospital, Urmia University of Medical Sciences, Urmia, Iran
| | - Mehdi Dadashpour
- Department of Medical Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
4
|
Hyytiäinen A, Wahbi W, Väyrynen O, Saarilahti K, Karihtala P, Salo T, Al-Samadi A. Angiogenesis Inhibitors for Head and Neck Squamous Cell Carcinoma Treatment: Is There Still Hope? Front Oncol 2021; 11:683570. [PMID: 34195084 PMCID: PMC8236814 DOI: 10.3389/fonc.2021.683570] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 05/17/2021] [Indexed: 01/27/2023] Open
Abstract
Background Head and neck squamous cell carcinoma (HNSCC) carries poor survival outcomes despite recent progress in cancer treatment in general. Angiogenesis is crucial for tumour survival and progression. Therefore, several agents targeting the pathways that mediate angiogenesis have been developed. We conducted a systematic review to summarise the current clinical trial data examining angiogenesis inhibitors in HNSCC. Methods We carried out a literature search on three angiogenesis inhibitor categories—bevacizumab, tyrosine kinase inhibitors and endostatin—from Ovid MEDLINE, Cochrane Library, Scopus and ClinicalTrials.gov database. Results Here, we analysed 38 clinical trials, total of 1670 patients, investigating 12 angiogenesis inhibitors. All trials were in phase I or II, except one study in phase III on bevacizumab. Angiogenesis inhibitors were used as mono- and combination therapies together with radio-, chemo-, targeted- or immunotherapy. Among 12 angiogenesis inhibitors, bevacizumab was the most studied drug, included in 13 trials. Although bevacizumab appeared effective in various combinations, it associated with high toxicity levels. Endostatin and lenvatinib were well-tolerated and their anticancer effects appeared promising. Conclusions Most studies did not show benefit of angiogenesis inhibitors in HNSCC treatment. Additionally, angiogenesis inhibitors were associated with considerable toxicity. However, some results appear encouraging, suggesting that further investigations of angiogenesis inhibitors, particularly in combination therapies, for HNSCC patients are warranted. Systematic Review Registration PROSPERO (https://www.crd.york.ac.uk/prospero/), identifier CRD42020157144.
Collapse
Affiliation(s)
- Aini Hyytiäinen
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland.,Translational Immunology Programme, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Wafa Wahbi
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland.,Translational Immunology Programme, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Otto Väyrynen
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland
| | - Kauko Saarilahti
- Department of Oncology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Peeter Karihtala
- Department of Oncology, Helsinki University Hospital Comprehensive Cancer Centre and University of Helsinki, Helsinki, Finland
| | - Tuula Salo
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland.,Translational Immunology Programme, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Department of Pathology, University of Helsinki, Helsinki, Finland.,Cancer Research and Translational Medicine Research Unit, University of Oulu, Oulu, Finland.,Oulu Medical Research Centre, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Ahmed Al-Samadi
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland.,Translational Immunology Programme, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
5
|
VEGF-A in Cardiomyocytes and Heart Diseases. Int J Mol Sci 2020; 21:ijms21155294. [PMID: 32722551 PMCID: PMC7432634 DOI: 10.3390/ijms21155294] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 12/11/2022] Open
Abstract
The vascular endothelial growth factor (VEGF), a homodimeric vasoactive glycoprotein, is the key mediator of angiogenesis. Angiogenesis, the formation of new blood vessels, is responsible for a wide variety of physio/pathological processes, including cardiovascular diseases (CVD). Cardiomyocytes (CM), the main cell type present in the heart, are the source and target of VEGF-A and express its receptors, VEGFR1 and VEGFR2, on their cell surface. The relationship between VEGF-A and the heart is double-sided. On the one hand, VEGF-A activates CM, inducing morphogenesis, contractility and wound healing. On the other hand, VEGF-A is produced by CM during inflammation, mechanical stress and cytokine stimulation. Moreover, high concentrations of VEGF-A have been found in patients affected by different CVD, and are often correlated with an unfavorable prognosis and disease severity. In this review, we summarized the current knowledge about the expression and effects of VEGF-A on CM and the role of VEGF-A in CVD, which are the most important cause of disability and premature death worldwide. Based on clinical studies on angiogenesis therapy conducted to date, it is possible to think that the control of angiogenesis and VEGF-A can lead to better quality and span of life of patients with heart disease.
Collapse
|
6
|
Muratori L, La Salvia A, Sperone P, Di Maio M. Target therapies in recurrent or metastatic head and neck cancer: state of the art and novel perspectives. A systematic review. Crit Rev Oncol Hematol 2019; 139:41-52. [PMID: 31112881 DOI: 10.1016/j.critrevonc.2019.05.002] [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: 10/21/2018] [Revised: 12/03/2018] [Accepted: 05/01/2019] [Indexed: 12/21/2022] Open
Abstract
Recurrent or metastatic head and neck squamous-cell carcinomas (R/M HNSCC) are a group of cancers with a very poor prognosis. Many clinical trials testing novel target therapies in this setting are currently ongoing. We performed a systematic review focusing our attention on all clinical trials, ongoing or already published, concerning the use of novel drugs for treatment of R/M HNSCC. We found that the research of novel molecules effective in treatment of R/M HNSCC has been intense during last decade, and nowadays it is still very active. Unfortunately, the results in this setting have been, overall, disappointing: until now, only cetuximab and, recently, nivolumab and pembrolizumab received authorization for treatment of R/M HNSCC. Nevertheless, the promising results showed by some novel drugs may lead to continue the research in this field, with the aim of producing more evidence and finding new therapeutic indication.
Collapse
Affiliation(s)
- Leonardo Muratori
- Medical Oncology, Department of Oncology, University of Turin, Azienda Ospedaliera Universitaria San Luigi Gonzaga, Regione Gonzole 10, 10043 Orbassano, Turin, Italy.
| | - Anna La Salvia
- Medical Oncology, Department of Oncology, Hospital Universitario 12 de Octubre, Avenida Cordoba, s/n, 28041 Madrid, Spain.
| | - Paola Sperone
- Medical Oncology, Department of Oncology, University of Turin, Azienda Ospedaliera Universitaria San Luigi Gonzaga, Regione Gonzole 10, 10043 Orbassano, Turin, Italy.
| | - Massimo Di Maio
- Medical Oncology, Department of Oncology, University of Turin, Azienda Ospedaliera Ordine Mauriziano, Corso Re Umberto 109, 10128 Turin, Italy.
| |
Collapse
|
7
|
Touyz RM, Herrmann SMS, Herrmann J. Vascular toxicities with VEGF inhibitor therapies-focus on hypertension and arterial thrombotic events. ACTA ACUST UNITED AC 2018; 12:409-425. [PMID: 29703600 PMCID: PMC6168784 DOI: 10.1016/j.jash.2018.03.008] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 03/02/2018] [Accepted: 03/14/2018] [Indexed: 12/21/2022]
Abstract
The vascular endothelial growth factor (VEGF) signaling pathway (VSP) fulfills a cardinal role in endothelial cells and its inhibition has profound cardiovascular impact. This is true not only for the normal vasculature but also for the tumor vasculature when VSP inhibitors are used as anti-angiogenic therapies. Generalized endothelial dysfunction predisposes to vasoconstriction, atherosclerosis, platelet activation, and thrombosis (arterial more than venous). All of these have been reported with VSP inhibitors and collectively give rise to vascular toxicities, the most concerning of which are arterial thromboembolic events (ATE). VSP inhibitors include antibodies, acting extracelluarly on VEGF, such as bevacizumab and tyrosine kinases inhibitors, acting intracellularly on the kinase domain of VEGF receptors, such as sunintib and sorafenib. The addition of bevacizumab and VSP tyrosine kinase inhibitor therapy to the cancer treatment regimen is associated with a 1.5-2.5-fold and 2.3-4.6-fold increase risk of ATEs, respectively. Risk factors for ATEs while on VSP inhibitor therapy include age older than 65 years, previous thromboembolic events, history of atherosclerotic disease, and duration of VSP inhibitor therapy. In clinical practice, hypertension remains the most commonly noted vascular manifestation of VSP inhibition. Optimal blood pressure goals and preferred therapeutic strategies toward reaching these goals are not defined at present. This review summarizes current data on this topic and proposes a more intensive management approach to patients undergoing VSP inhibitor therapy including Systolic Blood PRessure Intervention Trial (SPRINT) blood pressure goals, pleiotropic vasoprotective agents such as angiotensin converting enzyme inhibitors, amlodipine, and carvedilol, high-dose statin therapy, and aspirin.
Collapse
Affiliation(s)
- Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Sandra M S Herrmann
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
8
|
Redaelli S, Plaza-Menacho I, Mologni L. Novel targeted therapeutics for MEN2. Endocr Relat Cancer 2018; 25:T53-T68. [PMID: 29348306 DOI: 10.1530/erc-17-0297] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 11/14/2017] [Indexed: 01/28/2023]
Abstract
The rearranged during transfection (RET) proto-oncogene was recognized as the multiple endocrine neoplasia type 2 (MEN2) causing gene in 1993. Since then, much effort has been put into a clear understanding of its oncogenic signaling, its biochemical function and ways to block its aberrant activation in MEN2 and related cancers. Several small molecules have been designed, developed or redirected as RET inhibitors for the treatment of MEN2 and sporadic MTC. However, current drugs are mostly active against several other kinases, as they were not originally developed for RET. This limits efficacy and poses safety issues. Therefore, there is still much to do to improve targeted MEN2 treatments. New, more potent and selective molecules, or combinatorial strategies may lead to more effective therapies in the near future. Here, we review the rationale for RET targeting in MEN2, the use of currently available drugs and novel preclinical and clinical RET inhibitor candidates.
Collapse
Affiliation(s)
- Sara Redaelli
- School of Medicine and SurgeryUniversity of Milano-Bicocca, Monza, Italy
| | | | - Luca Mologni
- School of Medicine and SurgeryUniversity of Milano-Bicocca, Monza, Italy
| |
Collapse
|
9
|
Le T, New J, Jones JW, Usman S, Yalamanchali S, Tawfik O, Hoover L, Bruegger DE, Thomas SM. Inhibition of fibroblast growth factor receptor with AZD4547 mitigates juvenile nasopharyngeal angiofibroma. Int Forum Allergy Rhinol 2017; 7:973-979. [PMID: 28707818 PMCID: PMC5827962 DOI: 10.1002/alr.21987] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 06/10/2017] [Accepted: 06/18/2017] [Indexed: 01/27/2023]
Abstract
BACKGROUND Juvenile nasopharyngeal angiofibroma (JNA) is a benign tumor that presents in adolescent males. Although surgical excision is the mainstay of treatment, recurrences complicate treatment. There is a need to develop less invasive approaches for management. JNA tumors are composed of fibroblasts and vascular endothelial cells. We identified fibroblast growth factor receptor (FGFR) and vascular endothelial growth factor (VEGF) expression in JNA-derived fibroblasts. FGFR influences fibroblast proliferation and VEGF is necessary for angiogenesis. We hypothesized that targeting FGFR would mitigate JNA fibroblast proliferation, invasion, and migration, and that targeting the VEGF receptor would attenuate endothelial tubule formation. METHODS After informed consent, fibroblasts from JNA explants of 3 patients were isolated. Fibroblasts were treated with FGFR inhibitor AZD4547, 0 to 25 μg/mL for 72 hours and proliferation was quantified using CyQuant assay. Migration and invasion of JNA were assessed using 24-hour transwell assays with subsequent fixation and quantification. Mitigation of FGFR and downstream signaling was evaluated by immunoblotting. Tubule formation was assessed in human umbilical vein endothelial cells (HUVECs) treated with vehicle control (dimethylsulfoxide [DMSO]) or semaxanib (SU5416) as well as in serum-free media (SFM) or JNA conditioned media (CM). Tubule length was compared between treatment groups. RESULTS Compared to control, AZD4547 inhibited JNA fibroblast proliferation, migration, and invasion through inhibition of FGFR and downstream signaling, specifically phosphorylation of - p44/42 mitogen activated protein kinase (p44/42 MAPK). JNA fibroblast CM significantly increased HUVEC tubule formation (p = 0.0039). CONCLUSION AZD4547 effectively mitigates FGFR signaling and decreases JNA fibroblast proliferation, migration, and invasion. SU5416 attenuated JNA fibroblast-induced tubule formation. AZD4547 may have therapeutic potential in the treatment of JNA.
Collapse
Affiliation(s)
- Tran Le
- Department of Otolaryngology, University of Kansas Medical Center and University of Kansas Cancer Center, Kansas City, KS
| | - Jacob New
- Department of Otolaryngology, University of Kansas Medical Center and University of Kansas Cancer Center, Kansas City, KS
| | - Joel W. Jones
- Department of Otolaryngology, University of Kansas Medical Center and University of Kansas Cancer Center, Kansas City, KS
| | - Shireen Usman
- Department of Otolaryngology, University of Kansas Medical Center and University of Kansas Cancer Center, Kansas City, KS
| | - Sreeya Yalamanchali
- Department of Otolaryngology, University of Kansas Medical Center and University of Kansas Cancer Center, Kansas City, KS
| | - Ossama Tawfik
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center and University of Kansas Cancer Center, Kansas City, KS
| | - Larry Hoover
- Department of Otolaryngology, University of Kansas Medical Center and University of Kansas Cancer Center, Kansas City, KS
| | - Dan E. Bruegger
- Department of Otolaryngology, University of Kansas Medical Center and University of Kansas Cancer Center, Kansas City, KS
| | - Sufi Mary Thomas
- Department of Otolaryngology, University of Kansas Medical Center and University of Kansas Cancer Center, Kansas City, KS
| |
Collapse
|
10
|
Specenier PM, Vermorken JB. Recurrent head and neck cancer: current treatment and future prospects. Expert Rev Anticancer Ther 2014; 8:375-91. [DOI: 10.1586/14737140.8.3.375] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
11
|
Kieran MW, Kalluri R, Cho YJ. The VEGF pathway in cancer and disease: responses, resistance, and the path forward. Cold Spring Harb Perspect Med 2012; 2:a006593. [PMID: 23209176 DOI: 10.1101/cshperspect.a006593] [Citation(s) in RCA: 152] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Antiangiogenesis was proposed as a novel target for the treatment of cancer 40 years ago. Since the original hypothesis put forward by Judah Folkman in 1971, factors that mediate angiogenesis, their cellular targets, many of the pathways they signal, and inhibitors of the cytokines and receptors have been identified. Vascular endothelial growth factor (VEGF) is the most prominent among the angiogenic cytokines and is believed to play a central role in the process of neovascularization, both in cancer as well as other inflammatory diseases. This article reviews the biology of VEGF and its receptors, the use of anti-VEGF approaches in clinical disease, the toxicity of these therapies, and the resistance mechanisms that have limited the activity of these agents when used as monotherapy.
Collapse
Affiliation(s)
- Mark W Kieran
- Department of Pediatric Medical Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA.
| | | | | |
Collapse
|
12
|
The mechanisms that regulate the localization and overexpression of VEGF receptor-2 are promising therapeutic targets in cancer biology. Anticancer Drugs 2012; 23:347-54. [PMID: 22261741 DOI: 10.1097/cad.0b013e32835004ac] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The vascular endothelial growth factor (VEGF) family has been proposed to be the most important signaling protein family in vessel formation and maturation. VEGF receptor-2 (VEGFR-2) plays an abundant role in the most common forms of cancer. The localization of VEGFR-2 expression is important in cancer pathogenesis; however, so far, little attention has been paid to this phenomenon. Induced cytoplasmic VEGFR-2 transition from the nucleus is associated with poor prognostic cancer stages. Current VEGFR-2-targeted therapy approaches are effective in inhibiting or arresting tumor growth. Moreover, VEGFR-2-targeted therapy was demonstrated to restore the abnormal vasculature in tumors, enhancing their susceptibility toward conventional therapy. Most effects can be found when VEGFR-2-targeted therapy inhibits not only the induced angiogenesis but also the cancer cells that sometimes overexpress VEGFR-2. Nevertheless, we still have little knowledge about the mechanisms that regulate VEGFR-2 expression and how its localization is exactly involved in cancer prognosis. Further research and evaluation of VEGFR-2 regulation and its nuclear transition is necessary to develop more accurate therapeutic strategies to improve the patients' quality of life and their survival.
Collapse
|
13
|
Keefe D, Bowen J, Gibson R, Tan T, Okera M, Stringer A. Noncardiac vascular toxicities of vascular endothelial growth factor inhibitors in advanced cancer: a review. Oncologist 2011; 16:432-44. [PMID: 21441297 PMCID: PMC3228115 DOI: 10.1634/theoncologist.2010-0271] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Accepted: 01/13/2011] [Indexed: 12/15/2022] Open
Abstract
The introduction of molecularly targeted anticancer therapies has brought the promise of longer survival times for select patients with cancers previously considered untreatable. However, it has also brought new toxicities that require understanding and management, sometimes for long periods of time. Vascular endothelial growth factor inhibitors are associated with a broad range of adverse effects, with vascular toxicity being particularly serious. This review focuses on the current understanding of the pathophysiology and mechanisms of macrovascular toxicities (hypertension, hemorrhage, and thromboembolism), their incidence and severity, the current clinical management, and implications in the advanced cancer setting. Movement of these agents into the early disease setting will alter the impact of these toxicities. Search Strategy and Selection Criteria. Information for this review was collected by searching PubMed/Medline and American Society of Clinical Oncology abstract databases. The medical subject heading terms used included toxicity, hypertension, thromboembolism, hemorrhage, intestinal perforation, risk factors, pharmacokinetics, and metabolism, combined with free text search terms including, but not limited to, VEGF inhibitor*, bevacizumab, sunitinib, and sorafenib. Articles published in English before March 2010 were included, in addition to information from case reports and pharmaceutical agent package inserts.
Collapse
Affiliation(s)
- Dorothy Keefe
- Discipline of Medicine, School of Medicine, School of Medical Sciences, Faculty of Health Sciences, University of Adelaide, Adelaide, South Australia.
| | | | | | | | | | | |
Collapse
|
14
|
Kim C, Lee JL, Choi YH, Kang BW, Ryu MH, Chang HM, Kim TW, Kang YK. Phase I dose-finding study of sorafenib in combination with capecitabine and cisplatin as a first-line treatment in patients with advanced gastric cancer. Invest New Drugs 2010; 30:306-15. [PMID: 20839031 DOI: 10.1007/s10637-010-9531-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Accepted: 08/24/2010] [Indexed: 02/06/2023]
Abstract
BACKGROUND To define maximum-tolerated dose (MTD), dose-limiting toxicities (DLTs), and preliminary efficacy of sorafenib plus capecitabine/cisplatin in advanced gastric cancer (AGC) patients. METHODS Four dose-level combinations were tested in a standard 3 + 3 dose escalation design. Level 1: sorafenib 400 mg/d, capecitabine 1,600 mg/m(2)/d, cisplatin 80 mg/m(2). Level 2: sorafenib 800 mg/d, capecitabine 1,600 mg/m(2)/d, cisplatin 80 mg/m(2). Level 3: sorafenib 800 mg/d, capecitabine 2,000 mg/m(2)/d, cisplatin 80 mg/m(2). Level 1A: sorafenib 800 mg/d, capecitabine 1,600 mg/m(2)/d, cisplatin 60 mg/m(2). RESULTS There were 1 DLT at Level 2, and 2 DLTs at Level 3 (Level 3 was MTD). Since the relative dose intensity (RDI) of sorafenib and capecitabine could not be maintained at Level 2, Level 1A was newly investigated. As no DLT was observed and RDI remained above 80%, Level 1A is the recommended dose for the next clinical trial. Objective response rate was 62.5% (10 of 16 patients, 95% CI; 38.8-86.2%). Median progression-free survival and overall survival were 10.0 months (95% CI; 7.4-13.8) and 14.7 months (95% CI; 12.0-20.0), respectively. CONCLUSIONS Sorafenib 400 mg bid daily, capecitabine 800 mg/m(2) bid (days 1-14), and cisplatin 60 mg/m(2) (day 1) is recommended for further development in AGC.
Collapse
Affiliation(s)
- Chul Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 86 Asanbyeongwon-gil, Songpa-gu, Seoul 138-736, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Regulation of invasive behavior by vascular endothelial growth factor is HEF1-dependent. Oncogene 2010; 29:4449-59. [PMID: 20498643 DOI: 10.1038/onc.2010.185] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We previously reported a vascular endothelial growth factor (VEGF) autocrine loop in head and neck squamous cell carcinoma (HNSCC) cell lines, supporting a role for VEGF in HNSCC tumorigenesis. Using a phosphotyrosine proteomics approach, we screened the HNSCC cell line, squamous cell carcinoma-9 for effectors of VEGFR2 signaling. A cluster of proteins involved in cell migration and invasion, including the p130Cas paralog, human enhancer of filamentation 1 (HEF1/Cas-L/Nedd9) was identified. HEF1 silencing and overexpression studies revealed a role for VEGF in regulating cell migration, invasion and matrix metalloproteinase (MMP) expression in a HEF1-dependent manner. Moreover, cells plated on extracellular matrix-coated coverslips showed enhanced invadopodia formation in response to VEGF that was HEF1-dependent. Immunolocalization revealed that HEF1 colocalized to invadopodia with MT1-MMP. Analysis of HNSCC tissue microarrays for HEF1 immunoreactivity revealed a 6.5-fold increase in the odds of having a metastasis with a high HEF1 score compared with a low HEF1 score. These findings suggest that HEF1 may be prognostic for advanced stage HNSCC. They also show for the first time that HEF1 is required for VEGF-mediated HNSCC cell migration and invasion, consistent with HEF1's recent identification as a metastatic regulator. These results support a strategy targeting VEGF:VEGFR2 in HNSCC therapeutics.
Collapse
|
16
|
Zangari M, Fink LM, Elice F, Zhan F, Adcock DM, Tricot GJ. Thrombotic events in patients with cancer receiving antiangiogenesis agents. J Clin Oncol 2009; 27:4865-73. [PMID: 19704059 DOI: 10.1200/jco.2009.22.3875] [Citation(s) in RCA: 156] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Tumor-associated neoangiogenesis has recently become a suitable target for antineoplastic drug development. In this overview, we discuss specific drug-associated hemostatic complications, the already known pathogenetic mechanisms involved, and the effect of varying antithrombotic strategies. Multiple agents with angiogenic inhibitory capacity (thalidomide, lenalidomide, bevacizumab, sunitinib, sorafenib, and sirolimus) have obtained US Food and Drug Administration approval, and many others have entered clinical trials. Arterial and venous thromboembolism and hemorrhage have emerged as significant toxicities associated with the use of angiogenesis inhibitors. We present a detailed analysis of the literature on thrombotic complication of antiangiogenic drugs. Close attention to hemostatic complications during antiangiogenic treatment is warranted. Further studies are required to better understand the pathophysiologic mechanisms involved and to define a safe prophylactic strategy.
Collapse
Affiliation(s)
- Maurizio Zangari
- University of Utah, Division of Hematology, Blood/Marrow Transplant and Myeloma Program, Salt Lake City, UT, USA.
| | | | | | | | | | | |
Collapse
|
17
|
Bozec A, Peyrade F, Fischel JL, Milano G. Emerging molecular targeted therapies in the treatment of head and neck cancer. Expert Opin Emerg Drugs 2009; 14:299-310. [DOI: 10.1517/14728210902997947] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
18
|
Mooney CJ, Nagaiah G, Fu P, Wasman JK, Cooney MM, Savvides PS, Bokar JA, Dowlati A, Wang D, Agarwala SS, Flick SM, Hartman PH, Ortiz JD, Lavertu PN, Remick SC. A phase II trial of fosbretabulin in advanced anaplastic thyroid carcinoma and correlation of baseline serum-soluble intracellular adhesion molecule-1 with outcome. Thyroid 2009; 19:233-40. [PMID: 19265494 PMCID: PMC2913806 DOI: 10.1089/thy.2008.0321] [Citation(s) in RCA: 148] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Fosbretabulin is a novel vascular-disrupting agent that has antitumor activity against anaplastic thyroid cancer (ATC) cell lines, xenografts, and demonstrable efficacy in a phase I trial. This phase II study determined the efficacy and safety of fosbretabulin in patients with advanced ATC and whether fosbretabulin altered the natural history of ATC by virtue of doubling the median survival. A secondary aim evaluated the prognostic value of serum soluble intracellular adhesion molecule-1 (sICAM). METHODS Twenty-six patients received fosbretabulin 45 mg/m(2) as a 10-minute intravenous infusion on days 1, 8, and 15 of a 28-day cycle. sICAM levels were obtained at baseline, over the first two cycles, and end of therapy. Treatment was continued until disease progression. RESULTS Fosbretabulin was well tolerated; grade 3 toxicity was observed in nine patients (35%), and grade 4 toxicity in one (4%). QTc prolongation delayed treatment in four causing one to stop treatment. Median survival was 4.7 months with 34% and 23% alive at 6 and 12 months, respectively. Median duration of stable disease in seven patients was 12.3 months (range, 4.4-37.9 months). Baseline serum sICAM levels were measured in 24 patients with a median 253.5 ng/mL. There was a significant difference in event-free survival among tertiles of baseline sICAM levels (p < 0.009). CONCLUSIONS There were no objective responses seen with single-agent fosbretabulin as administered in this trial, and we did not observe a doubling of survival as our primary endpoint. This is among the largest prospective trials ever conducted for ATC. Fosbretabulin has an acceptable safety profile in patients with advanced ATC, and one-third survived more than 6 months. Despite a small sample size, low baseline sICAM levels were predictive of event-free survival. Further prospective validation of sICAM as a therapeutic biomarker and exploring combination regimens with fosbretabulin are warranted.
Collapse
Affiliation(s)
- Colin J. Mooney
- Developmental Therapeutics Program, Case Comprehensive Cancer Center, Cleveland, Ohio
| | - Govardhanan Nagaiah
- Developmental Therapeutics Program, Case Comprehensive Cancer Center, Cleveland, Ohio
| | - Pingfu Fu
- Developmental Therapeutics Program, Case Comprehensive Cancer Center, Cleveland, Ohio
- Department of Biostatistics and Epidemiology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Jay K. Wasman
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Matthew M. Cooney
- Developmental Therapeutics Program, Case Comprehensive Cancer Center, Cleveland, Ohio
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Panos S. Savvides
- Developmental Therapeutics Program, Case Comprehensive Cancer Center, Cleveland, Ohio
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Joseph A. Bokar
- Developmental Therapeutics Program, Case Comprehensive Cancer Center, Cleveland, Ohio
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Afshin Dowlati
- Developmental Therapeutics Program, Case Comprehensive Cancer Center, Cleveland, Ohio
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Ding Wang
- Karmanos Cancer Institute (formerly Josephine Ford Cancer Center), Detroit, Michigan
| | - Sanjiv S. Agarwala
- St. Luke's Hospital and Health Network, Bethlehem, Pennsylvania (formerly University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania)
| | - Susan M. Flick
- Developmental Therapeutics Program, Case Comprehensive Cancer Center, Cleveland, Ohio
| | - Paul H. Hartman
- Translational Research Shared Resource, Case Comprehensive Cancer Center, Cleveland, Ohio
| | - Jose D. Ortiz
- Developmental Therapeutics Program, Case Comprehensive Cancer Center, Cleveland, Ohio
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Pierre N. Lavertu
- Developmental Therapeutics Program, Case Comprehensive Cancer Center, Cleveland, Ohio
- Otolaryngology and Head and Neck Surgery, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Scot C. Remick
- Developmental Therapeutics Program, Case Comprehensive Cancer Center, Cleveland, Ohio
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio
- Mary Babb Randolph Cancer Center, West Virginia University School of Medicine, Morgantown, West Virginia
| |
Collapse
|
19
|
Place des thérapeutiques moléculaires ciblées dans les carcinomes épidermoïdes des voies aérodigestives supérieures. ONCOLOGIE 2009. [DOI: 10.1007/s10269-008-1038-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
20
|
Kieran MW, Supko JG, Wallace D, Fruscio R, Poussaint TY, Phillips P, Pollack I, Packer R, Boyett JM, Blaney S, Prados M, Geyer R, Friedman H, Goldman S, Kun LE, MacDonald T. Phase I study of SU5416, a small molecule inhibitor of the vascular endothelial growth factor receptor (VEGFR) in refractory pediatric central nervous system tumors. Pediatr Blood Cancer 2009; 52:169-76. [PMID: 19065567 PMCID: PMC2775441 DOI: 10.1002/pbc.21873] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
SU5416 is a novel small molecule tyrosine kinase inhibitor of the VEGF receptors 1 and 2. A phase I dose escalation study stratified by concurrent use (stratum II) or absence (stratum I) of enzyme-inducing anticonvulsant drugs was undertaken to estimate the maximum-tolerated dose (MTD) and to describe the toxicity profile of SU5416 in pediatric patients with refractory brain tumors. Dose escalations were conducted independently for stratum I starting at 110 mg/m(2) while stratum II started at 48 mg/m(2). Thirty-three eligible patients were treated on stratum I (n = 23) and stratum II (n = 10). Tumor types included 23 glial tumors, 4 neural tumors, 4 ependymomas, and 2 choroid plexus carcinomas. The MTD in stratum I was initially estimated to be 110 mg/m(2). The protocol was amended to determine the MTD after excluding transient AST elevation. Re-estimation of the MTD began at the 145 mg/m(2) dose level but due to development of SU5416 being stopped by the sponsor, the trial was closed before completion. The most serious drug-related toxicities were grade 3 liver enzyme abnormalities, arthralgia, and hallucinations. The plasma pharmacokinetics of SU5416 was not significantly affected by the concurrent administration of enzyme-inducing anticonvulsant drugs. Mean values of the total body clearance, apparent volume of distribution, and terminal phase half-life of SU5416 for the 19 patients in stratum I were 26.1 +/- 12.5 l/hr/m(2), 41.9 +/- 21.4 l/m(2), and 1.11 +/- 0.41 hr, respectively. The plasma pharmacokinetics of SU5416 in children was similar to previously reported findings in adult cancer patients. Prolonged disease stabilization was observed in 4 of 16 stratum I patients.
Collapse
Affiliation(s)
- Mark W. Kieran
- Pediatric Neuro-Oncology Program, Dana-Farber Cancer Institute, Boston, MA, USA,To whom requests for reprints should be addressed: Mark W. Kieran, M.D., Pediatric Neuro-Oncology, Dana-Farber Cancer Institute, 44 Binney Street, Room SW331, Boston, MA 02115. Phone (617) 632-4386, Fax (617) 632-4897,
| | - Jeffrey G. Supko
- Division of Hematology/Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Dana Wallace
- Operations and Biostatistics Center, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Robert Fruscio
- Division of Hematology/Oncology, Massachusetts General Hospital, Boston, MA, USA
| | | | - Peter Phillips
- Division of Pediatric Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ian Pollack
- Department of Neurosurgery, Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Roger Packer
- Department of Neurology, Children’s National Medical Center, Washington, DC, USA
| | - James M. Boyett
- Operations and Biostatistics Center, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Susan Blaney
- Texas Children’s Hospital/Baylor College of Medicine, Houston, TX, USA
| | - Michael Prados
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Russ Geyer
- Department of Pediatrics, Children’s Hospital. & Regional Medical Center, Seattle, WA, USA
| | - Henry Friedman
- The Preston Robert Tisch Brain Tumor Center at Duke, Duke University Medical Center, Durham, NC, USA
| | | | - Larry E. Kun
- Department of Radiological Sciences, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Tobey MacDonald
- Division of Hematology/Oncology, Children’s National Medical Center, Washington, D.C, USA
| | | |
Collapse
|
21
|
Elice F, Jacoub J, Rickles FR, Falanga A, Rodeghiero F. Hemostatic complications of angiogenesis inhibitors in cancer patients. Am J Hematol 2008; 83:862-70. [PMID: 18819092 DOI: 10.1002/ajh.21277] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Tumor vasculature and tumor-associated neo-angiogenesis have recently become major targets for rational drug design of antineoplastic agents. Five such agents with angiogenesis inhibiting activity (thalidomide, lenalidomide, bevacizumab, sunitinib, sorafenib) have already obtained US Food and Drug Administration approval for clinical use and many others have entered clinical trials. Vascular complications, including venous or arterial thromboembolism and hemorrhage, have emerged as relevant toxicities in several clinical trials with angiogenesis inhibitors. Given the well-known interplay between the blood clotting system, angiogenesis, and tumor growth, a better understanding of the impact of these new drugs on overall hemostatic balance is required. In this brief overview, we discuss the incidence of hemostatic complications, the likely pathogenetic mechanisms involved, and the critical need to establish in randomized clinical trials the usefulness of thrombosis prophylaxis to prevent these complications. Careful documentation of hemostatic complications during treatment with each of the new antiangiogenic drugs is warranted. Further studies are urgently required to better define the causal association of these new agents with hemostatic complications and to establish the best prophylactic strategy.
Collapse
Affiliation(s)
- Francesca Elice
- Department of Cell Therapy and Hematology, San Bortolo Hospital, Vicenza, Italy
| | | | | | | | | |
Collapse
|
22
|
Carden CP, Larkin JMG, Rosenthal MA. What is the risk of intracranial bleeding during anti-VEGF therapy? Neuro Oncol 2008; 10:624-30. [PMID: 18539884 PMCID: PMC2666237 DOI: 10.1215/15228517-2008-010] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2007] [Accepted: 01/04/2008] [Indexed: 12/13/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) is a key mediator of physiological and pathological angiogenesis. All solid tumors are dependent on pathological angiogenesis, and anti-VEGF therapy has demonstrated clinical benefit in breast, colorectal, non-small-cell lung, and renal carcinomas. Central nervous system metastases are common in many of these tumor types. An increased risk of bleeding has been reported with anti-VEGF therapy, but the risk of intracranial bleeding is unknown with this type of therapy. We reviewed the available data to investigate the risk of intracranial bleeding with anti-VEGF therapy in the presence and absence of CNS metastases. The PubMed and Medline databases and the Proceedings of the American Society of Clinical Oncology (ASCO) annual meetings were searched for articles, abstracts, and presentations of clinical trials. We identified 57 trials examining the safety and efficacy of anti-VEGF therapy in a total of 10,598 patients. Four trials examined the use of anti-VEGF therapy in treating patients with brain metastases. The presence of CNS metastases was a stated exclusion criterion in 76% of trials. The rate of intracranial bleeding was negligible. We conclude that there is no trial evidence that anti-VEGF therapy confers an increased risk of intracranial bleeding, even in the presence of CNS metastases. Future trials of anti-VEGF therapy should not exclude patients with controlled CNS metastases at enrollment.
Collapse
Affiliation(s)
- Craig P Carden
- Drug Development Unit, Royal Marsden Hospital, Sutton, London, UK.
| | | | | |
Collapse
|
23
|
Bozec A, Thariat J, Bensadoun R, Milano G. La chimioradiothérapie des carcinomes épidermoïdes des voies aérodigestives supérieures : point sur les thérapeutiques ciblées. Cancer Radiother 2008; 12:14-24. [DOI: 10.1016/j.canrad.2007.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2007] [Revised: 11/22/2007] [Accepted: 11/23/2007] [Indexed: 10/22/2022]
|
24
|
Overmoyer B, Fu P, Hoppel C, Radivoyevitch T, Shenk R, Persons M, Silverman P, Robertson K, Ziats NP, Wasman JK, Abdul-Karim FW, Jesberger JA, Duerk J, Hartman P, Hanks S, Lewin J, Dowlati A, McCrae K, Ivy P, Remick SC. Inflammatory Breast Cancer as a Model Disease to Study Tumor Angiogenesis: Results of a Phase IB Trial of Combination SU5416 and Doxorubicin. Clin Cancer Res 2007; 13:5862-8. [PMID: 17908980 DOI: 10.1158/1078-0432.ccr-07-0688] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE We used inflammatory breast cancer (IBC) as a model disease to investigate biological changes associated with an antiangiogenesis agent, SU5416, combined with doxorubicin. EXPERIMENTAL DESIGN Patients with stage IIIB or IV IBC were treated neoadjuvantly with the combination of SU5416 and doxorubicin for induction therapy. The dose of SU5416 (administered on days 1 and 4, every 3 weeks) and doxorubicin (administered on day 1 every 3 weeks) were escalated in cohorts of three patients starting at 110 and 60 mg/m2, respectively, for a total of five cycles leading up to mastectomy. Patients underwent serial assessment (pharmacokinetic sampling, biopsy of breast, tumor blood flow dynamic contrast-enhanced magnetic resonance imaging, plasma angiogenesis, and endothelial cell damage markers) prior to treatment, at the end of cycles no. 2 and no. 5, and after mastectomy. RESULTS Eighteen patients were enrolled; neutropenia was dose-limiting, and overall median survival was not reached (50 months of study follow-up). Four patients (22%) experienced congestive heart failure, which resolved and were likely attributable to a smaller volume of distribution and higher Cmax of doxorubicin in combination with SU5416. We did observe a significant decline in tumor blood flow using Kep calculated by Brix (pretreatment versus post-cycle no. 5; P = 0.033), trend for a decline in tumor microvessel density after treatment, and low baseline levels of soluble intracellular adhesion molecule were associated with improved event-free survival. CONCLUSIONS This study showed evidence of an unfavorable cardiac interaction between SU5416 and doxorubicin, which prohibits further investigation of this combination. However, this study supports the importance of using IBC as a model for investigating angiogenesis inhibitors.
Collapse
Affiliation(s)
- Beth Overmoyer
- Connecticut Oncology & Hematology Associates/US Oncology, Torrington, Connecticut 06790, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Peng JH, Zhang CE, Wei W, Hong XP, Pan XP, Wang JZ. Dehydroevodiamine attenuates tau hyperphosphorylation and spatial memory deficit induced by activation of glycogen synthase kinase-3 in rats. Neuropharmacology 2007; 52:1521-7. [PMID: 17434540 DOI: 10.1016/j.neuropharm.2007.02.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2006] [Revised: 02/08/2007] [Accepted: 02/19/2007] [Indexed: 10/23/2022]
Abstract
Tau hyperphosphorylation and memory deficit are characteristic alterations of Alzheimer's disease (AD), and glycogen synthase kinase-3 (GSK-3) plays a crucial role in these AD-like changes. We have reported that activation of GSK-3 through ventricular injection of wortmannin and GF-109203X (WT/GFX, 100 microM each) induces tau hyperphosphorylation and memory impairment of rats [Liu, S.J. et al., 2003. Overactivation of glycogen synthase kinase-3 by inhibition of phosphoinositol-3 kinase and protein kinase C leads to hyperphosphorylation of tau and impairment of spatial memory. J. Neurochem. 87, 1333-1344]. By using this model, we explored in the present study the effects of dehydroevodiamine (DHED), a quinazoline alkaloid isolated from Evodia rutaecarpa Bentham, on the memory retention, tau phosphorylation and the activity of GSK-3. We found that pre-administration of DHED through vena caudalis for 1 week efficiently improved the WT/GFX-induced spatial memory retention impairment of the rats; it also antagonized tau hyperphosphorylation at multiple AD sites and arrested the overactivation of GSK-3 induced by WT/GFX. Our study gave the first in vivo evidence that DHED could suppress the overactivation of GSK-3 and improve tau hyperphosphorylation and spatial memory deficit of the rats, suggesting that this chemical may be served as a candidate for arresting AD-like pathological and behavioral alterations.
Collapse
Affiliation(s)
- Jun-Hua Peng
- Pathophysiology Department, Hubei Provincial Key Laboratory of Neurological Disease, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | | | | | | | | | | |
Collapse
|
26
|
|
27
|
Noy A, von Roenn J, Politsmakher A, Miles S, Bernd C. Fatal hepatorenal failure and thrombocytopenia with SU5416, a vascular endothelial growth factor Flk-1 receptor inhibitor, in AIDS-Kaposi's sarcoma. AIDS 2007; 21:113-5. [PMID: 17148979 DOI: 10.1097/01.aids.0000254688.06831.28] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
28
|
Eskens FALM, Verweij J. The clinical toxicity profile of vascular endothelial growth factor (VEGF) and vascular endothelial growth factor receptor (VEGFR) targeting angiogenesis inhibitors; a review. Eur J Cancer 2006; 42:3127-39. [PMID: 17098419 DOI: 10.1016/j.ejca.2006.09.015] [Citation(s) in RCA: 260] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2006] [Accepted: 09/27/2006] [Indexed: 01/09/2023]
Abstract
Clinical experience with vascular endothelial growth factor (VEGF) and vascular endothelial growth factor receptor (VEGFR) targeting angiogenesis inhibitors is rapidly increasing, and some compounds have already been approved for regular anticancer treatment. Apart from their activity, much attention has been focussed on the clinical toxicity profile of these compounds. This review describes the most frequently occurring side-effects of both antibodies and tyrosine kinase inhibitors and discusses some of the underlying mechanisms. Some practical guidelines for treatment of the side-effects are given.
Collapse
Affiliation(s)
- Ferry A L M Eskens
- Erasmus University Medical Center, Department of Medical Oncology PO Box 2040, 3000 CA Rotterdam, Netherlands.
| | | |
Collapse
|
29
|
Egloff AM, Grandis J. Epidermal growth factor receptor – targeted molecular therapeutics for head and neck squamous cell carcinoma. Expert Opin Ther Targets 2006; 10:639-47. [PMID: 16981821 DOI: 10.1517/14728222.10.5.639] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Several molecular-targeted therapeutics have been tested in clinical trials for the treatment of head and neck squamous cell carcinoma (HNSCC). Of these, therapeutics targeting the epidermal growth factor receptor (EGFR) have been studied most extensively and some agents have demonstrated measurable clinical effectiveness. However, molecular studies designed to define HNSCC patient subcohorts of likely responders to EGFR-targeted therapy have not identified molecular signatures that correlate with clinical response. Here, the authors summarise the relevant clinical findings and highlight reported molecular correlative studies for EGFR-targeted therapeutics for HNSCC. The authors focus especially on molecular markers evaluated for association with clinical response and include data from EGFR-targeted clinical studies in other cancer sites that they anticipate will be of interest to the head and neck cancer research and treatment communities.
Collapse
|
30
|
Fury MG, Zahalsky A, Wong R, Venkatraman E, Lis E, Hann L, Aliff T, Gerald W, Fleisher M, Pfister DG. A Phase II study of SU5416 in patients with advanced or recurrent head and neck cancers. Invest New Drugs 2006; 25:165-72. [PMID: 16983506 DOI: 10.1007/s10637-006-9011-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2006] [Accepted: 08/28/2006] [Indexed: 11/30/2022]
Abstract
BACKGROUND SU5416 (semaxanib) is a synthetic small molecule inhibitor of the tyrosine kinase domain of vascular endothelial growth factor receptor 2 (VEGFR2). This Phase II study was conducted to determine the safety and efficacy of SU5416 in patients with recurrent or metastatic head and neck cancers. PATIENTS AND METHODS This was an open label, single arm, Phase 2 study for patients who had received no more than 2 cytotoxic regimens. Thirty-five patients received intravenous SU5416 (145 mg/m(2)) twice per week by intravenous catheter. Radiologic imaging for response assessment was planned at the conclusion of each 8 week cycle. Serum VEGF levels and power Doppler ultrasound tumor imaging were explored as potential surrogate markers for SU5416 activity. RESULTS Thirty-two patients had received prior radiotherapy, including 18 patients who received prior concurrent chemoradiotherapy. Twelve patients had received prior chemotherapy in the recurrent disease setting. Thirty-one patients were evaluable for response. There was one partial response and one minor response. The median number of 8-week cycles received was 1 (range 1-4). Median overall survival was 6.25 months. The most common > or = grade 3 toxicity was headache (31%). There was one fatal carotid artery hemorrhage. Fatigue, nausea, and vomiting were common grade 1-2 adverse events. Power Doppler ultrasound demonstrated decreased tumor vascularity in 5 of 7 patients. CONCLUSIONS Treatment with SU5416 in patients with head and neck cancers is feasible, but objective responses are rare. Studies evaluating more potent anti-angiogenic agents in this disease are of interest.
Collapse
Affiliation(s)
- Matthew G Fury
- Head and Neck Medical Oncology Service, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Gietema JA, Hoekstra R, de Vos FYFL, Uges DRA, van der Gaast A, Groen HJM, Loos WJ, Knight RA, Carr RA, Humerickhouse RA, Eskens FALM. A phase I study assessing the safety and pharmacokinetics of the thrombospondin-1-mimetic angiogenesis inhibitor ABT-510 with gemcitabine and cisplatin in patients with solid tumors. Ann Oncol 2006; 17:1320-7. [PMID: 16728485 DOI: 10.1093/annonc/mdl102] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The aim of the study was to determine the safety profile, pharmacokinetics and potential drug interactions of the angiogenesis inhibitor ABT-510 combined with gemcitabine-cisplatin chemotherapy in patients with solid tumors. PATIENTS AND METHODS Patients with advanced solid tumors received gemcitabine 1250 mg/m2 intravenously (i.v.) on days 1 and 8 and cisplatin 80 mg/m2 on day 1 of a 3-week cycle in combination with ABT-510. ABT-510 was administered subcutaneously twice daily at doses of 50 mg or 100 mg. Plasma samples for pharmacokinetics were obtained on days 1 (gemcitabine, cisplatin as single agents), 15 (ABT-510 as single agent) and 22 (gemcitabine, cisplatin and ABT-510 as combination). RESULTS Thirteen patients received ABT-510 as either 50 mg b.i.d. (seven patients) or 100 mg b.i.d. (six patients) in combination with gemcitabine-cisplatin. The most common reported adverse events reflected the known toxicity profile induced by gemcitabine-cisplatin without ABT-510. One episode of hemoptysis occurred in a patient with non-small-cell lung cancer (NSCLC) after 13 days of treatment. No clinically significant pharmacokinetic interactions between ABT-510, gemcitabine and platinum were observed. Three partial responses were observed in 12 evaluable patients (one head and neck cancer, one melanoma and one NSCLC). CONCLUSIONS Combining ABT-510 at doses of 50 mg and 100 mg with gemcitabine-cisplatin is feasible. Pharmacokinetic interactions were not observed and adding ABT-510 does not appear to increase toxicity.
Collapse
Affiliation(s)
- J A Gietema
- Department of Medical Oncology, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Caponigro F, Milano A, Basile M, Ionna F, Iaffaioli RV. Recent advances in head and neck cancer therapy: the role of new cytotoxic and molecular-targeted agents. Curr Opin Oncol 2006; 18:247-52. [PMID: 16552236 DOI: 10.1097/01.cco.0000219253.53091.fb] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW To provide an update on novel compounds in head and neck cancer (HNC) therapy, with emphasis on biologic agents. RECENT FINDINGS Cisplatin-5-fluorouracil (5-FU) is the standard chemotherapeutic approach in HNC. Strategies to improve its activity include the substitution of 5-FU with oral fluoropyrimidines; the substitution of cisplatin with different analogs or formulations; and the use of additional or alternative compounds. Epidermal growth factor receptor (EGFR) is the most appealing target for novel therapies in HNC. Cetuximab, a chimeric anti-EGFR monoclonal antibody, has undergone evaluation in platinum-refractory recurrent or metastatic HNC with a satisfactory and consistent response rate (10-13%) across three different Phase II studies in association with platinum or as single agent. A recent Phase III placebo-controlled trial has shown better response rate for patients treated with cetuximab and cisplatin, with respect to those treated with cisplatin alone. EGFR tyrosine kinase inhibitors (TKIs) are under investigation in HNC, and efforts are made to understand which molecular features are associated with objective responses. One appealing way to use EGFR TKIs is in combination with other biologic compounds, such as anti-angiogenic agents. SUMMARY New molecular-targeted therapies are inducing consistent, small improvements in HNC management. The major challenge regards how to better combine them with the final aim of obtaining long-term stabilization of advanced disease.
Collapse
Affiliation(s)
- Francesco Caponigro
- Division of Medical Oncology B, National Tumor Institute of Naples, Fondazione G. Pascale, Naples, Italy.
| | | | | | | | | |
Collapse
|
33
|
Citrin D, Ménard C, Camphausen K. Combining radiotherapy and angiogenesis inhibitors: clinical trial design. Int J Radiat Oncol Biol Phys 2006; 64:15-25. [PMID: 16377411 DOI: 10.1016/j.ijrobp.2005.03.065] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2005] [Revised: 03/30/2005] [Accepted: 03/31/2005] [Indexed: 01/12/2023]
Abstract
Radiotherapy (RT) plays a vital role in the multimodality treatment of cancer. Recent advances in RT have primarily involved improvements in dose delivery. Future improvements in tumor control and disease outcomes will likely involve the combination of RT with targeted therapies. Preclinical evaluations of angiogenesis inhibitors in combination with RT have yielded promising results with increased tumor "cure." It remains to be seen whether these improvements in tumor control in the laboratory will translate into improved outcomes in the clinic. Multiple differences between these agents and cytotoxic chemotherapy must be taken into account when designing clinical trials evaluating their effectiveness in combination with RT. We discuss important considerations for designing clinical trials of angiogenesis inhibitors with RT.
Collapse
Affiliation(s)
- Deborah Citrin
- Radiation Oncology Branch, NCI, Bethesda, MD 20892, USA.
| | | | | |
Collapse
|
34
|
Rosenthal DI, Fuller CD, Machtay M, Algazy KM, Meyer DM, Kaiser LR, Yardley DA, Loiacano ME, Carbone DP. Phase I Study of Paclitaxel Given by Seven-Week Continuous Infusion Concurrent with Radiation Therapy for Locally Advanced Non–Small Cell Lung Cancer. J Thorac Oncol 2006. [DOI: 10.1016/s1556-0864(15)31511-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
35
|
Phase I Study of Paclitaxel Given by Seven-Week Continuous Infusion Concurrent with Radiation Therapy for Locally Advanced Non???Small Cell Lung Cancer. J Thorac Oncol 2006. [DOI: 10.1097/01243894-200601000-00009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
36
|
O'Donnell A, Padhani A, Hayes C, Kakkar AJ, Leach M, Trigo JM, Scurr M, Raynaud F, Phillips S, Aherne W, Hardcastle A, Workman P, Hannah A, Judson I. A Phase I study of the angiogenesis inhibitor SU5416 (semaxanib) in solid tumours, incorporating dynamic contrast MR pharmacodynamic end points. Br J Cancer 2005; 93:876-83. [PMID: 16222321 PMCID: PMC2361651 DOI: 10.1038/sj.bjc.6602797] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
SU5416 (Z-3-[(2,4-dimethylpyrrol-5-yl)methylidenyl]-2-indolinone; semaxanib) is a small molecule inhibitor of the vascular endothelial growth factor receptor (VEGFR2). A Phase I dose escalation study was performed. Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) was used as a pharmacodynamic assessment tool. In all, 27 patients were recruited. SU5416 was administered twice weekly by fixed rate intravenous infusion. Patients were treated in sequential cohorts of three patients at 48, 65, 85 110 and 145 mg m-2. A further dose level of 190 mg m-2 after a 2-week lead in period at a lower dose was completed; thereafter, the cohort at 145 mg m-2 was expanded. SU5416 showed linear pharmacokinetics to 145 mg m-2 with a large volume of distribution and rapid clearance. A significant degree of interpatient variability was seen. SU5416 was well tolerated, by definition a maximum-tolerated dose was not defined. No reproducible changes were seen in DCE-MRI end points. Serial assessments of VEGF in a cohort of patients treated at 145 mg m-2 did not show a statistically significant treatment-related change. Parallel assessments of the impact of SU5416 on coagulation profiles in six patients showed a transient effect within the fibrinolytic pathway. Clinical experience showed that patients who had breaks of therapy longer than a week could not have treatment reinitiated at a dose of 190 mg m-2 without unacceptable toxicity. The 145 mg m-2 dose level is thus the recommended dose for future study.
Collapse
Affiliation(s)
- A O'Donnell
- Institute of Cancer Research, and Royal Marsden Hospital, Sutton SM2 5PT, UK. anne.o'
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
The field of cancer research has seen a marked shift in the past decade towards the exploration and development of non-conventional antitumour agents. One of the most widely studied approaches to therapy during this period has been that of antiangiogenesis. The published clinical trials and subsequent FDA approval (in February 2004) of the anti-vascular endothelial growth factor (VEGF) monoclonal antibody bevacizumab (Avastin, Genentech) for the treatment of colorectal cancer marked a milestone for antiangiogenesis therapy. Currently, preclinical and clinical research involving therapeutic targeting of VEGF and other mediators of angiogenesis continues in multiple tumour types. In addition to colorectal cancer, angiogenesis inhibitors are being investigated in the treatment of renal cell carcinoma, head and neck carcinoma, lung cancer, breast cancer, prostate cancer, and a variety of haematological malignancies. This article will discuss the background of antiangiogenesis research, preclinical and clinical data relating to the use of bevacizumab in the treatment of colorectal cancer, other completed clinical trials involving antiangiogenesis agents, and the potential future utility of these agents in the treatment of malignancy.
Collapse
Affiliation(s)
- Jay Rhee
- University of Texas MD Anderson Cancer Center, Department of GI Medical Oncology, 1515 Holcombe Boulevard, Unit 426 Houston, TX 77030, USA
| | | |
Collapse
|