1
|
Andersen MS, Kofoed MS, Paludan-Müller AS, Pedersen CB, Mathiesen T, Mawrin C, Wirenfeldt M, Kristensen BW, Olsen BB, Halle B, Poulsen FR. Meningioma animal models: a systematic review and meta-analysis. J Transl Med 2023; 21:764. [PMID: 37898750 PMCID: PMC10612271 DOI: 10.1186/s12967-023-04620-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/11/2023] [Indexed: 10/30/2023] Open
Abstract
BACKGROUND Animal models are widely used to study pathological processes and drug (side) effects in a controlled environment. There is a wide variety of methods available for establishing animal models depending on the research question. Commonly used methods in tumor research include xenografting cells (established/commercially available or primary patient-derived) or whole tumor pieces either orthotopically or heterotopically and the more recent genetically engineered models-each type with their own advantages and disadvantages. The current systematic review aimed to investigate the meningioma model types used, perform a meta-analysis on tumor take rate (TTR), and perform critical appraisal of the included studies. The study also aimed to assess reproducibility, reliability, means of validation and verification of models, alongside pros and cons and uses of the model types. METHODS We searched Medline, Embase, and Web of Science for all in vivo meningioma models. The primary outcome was tumor take rate. Meta-analysis was performed on tumor take rate followed by subgroup analyses on the number of cells and duration of incubation. The validity of the tumor models was assessed qualitatively. We performed critical appraisal of the methodological quality and quality of reporting for all included studies. RESULTS We included 114 unique records (78 using established cell line models (ECLM), 21 using primary patient-derived tumor models (PTM), 10 using genetically engineered models (GEM), and 11 using uncategorized models). TTRs for ECLM were 94% (95% CI 92-96) for orthotopic and 95% (93-96) for heterotopic. PTM showed lower TTRs [orthotopic 53% (33-72) and heterotopic 82% (73-89)] and finally GEM revealed a TTR of 34% (26-43). CONCLUSION This systematic review shows high consistent TTRs in established cell line models and varying TTRs in primary patient-derived models and genetically engineered models. However, we identified several issues regarding the quality of reporting and the methodological approach that reduce the validity, transparency, and reproducibility of studies and suggest a high risk of publication bias. Finally, each tumor model type has specific roles in research based on their advantages (and disadvantages). SYSTEMATIC REVIEW REGISTRATION PROSPERO-ID CRD42022308833.
Collapse
Affiliation(s)
- Mikkel Schou Andersen
- Department of Neurosurgery, Odense University Hospital, Odense, Denmark.
- BRIDGE (Brain Research - Inter Disciplinary Guided Excellence), University of Southern Denmark, Odense, Denmark.
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark.
| | - Mikkel Seremet Kofoed
- Department of Neurosurgery, Odense University Hospital, Odense, Denmark
- BRIDGE (Brain Research - Inter Disciplinary Guided Excellence), University of Southern Denmark, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Asger Sand Paludan-Müller
- Nordic Cochrane Centre, Rigshospitalet, Copenhagen University, Copenhagen, Denmark
- Centre for Evidence-Based Medicine Odense (CEBMO) and NHTA: Market Access & Health Economics Consultancy, Copenhagen, Denmark
| | - Christian Bonde Pedersen
- Department of Neurosurgery, Odense University Hospital, Odense, Denmark
- BRIDGE (Brain Research - Inter Disciplinary Guided Excellence), University of Southern Denmark, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Tiit Mathiesen
- Department of Neurosurgery, Rigshospitalet, Copenhagen University, Copenhagen, Denmark
| | - Christian Mawrin
- Department of Neuropathology, Otto-Von-Guericke University, Magdeburg, Germany
| | - Martin Wirenfeldt
- Department of Pathology and Molecular Biology, Hospital South West Jutland, Esbjerg, Denmark
- Department of Regional Health Research, University of Southern, Odense, Denmark
| | | | - Birgitte Brinkmann Olsen
- Clinical Physiology and Nuclear Medicine, Odense University Hospital, Odense, Denmark
- Department of Surgical Pathology, Zealand University Hospital, Roskilde, Denmark
| | - Bo Halle
- Department of Neurosurgery, Odense University Hospital, Odense, Denmark
- BRIDGE (Brain Research - Inter Disciplinary Guided Excellence), University of Southern Denmark, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Frantz Rom Poulsen
- Department of Neurosurgery, Odense University Hospital, Odense, Denmark
- BRIDGE (Brain Research - Inter Disciplinary Guided Excellence), University of Southern Denmark, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
2
|
Khan M, Hanna C, Findlay M, Lucke-Wold B, Karsy M, Jensen RL. Modeling Meningiomas: Optimizing Treatment Approach. Neurosurg Clin N Am 2023; 34:479-492. [PMID: 37210136 DOI: 10.1016/j.nec.2023.02.014] [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] [Indexed: 05/22/2023]
Abstract
Preclinical meningioma models offer a setting to test molecular mechanisms of tumor development and targeted treatment options but historically have been challenging to generate. Few spontaneous tumor models in rodents have been established, but cell culture and in vivo rodent models have emerged along with artificial intelligence, radiomics, and neural networks to differentiate the clinical heterogeneity of meningiomas. We reviewed 127 studies using PRISMA guideline methodology, including laboratory and animal studies, that addressed preclinical modeling. Our evaluation identified that meningioma preclinical models provide valuable molecular insight into disease progression and effective chemotherapeutic and radiation approaches for specific tumor types.
Collapse
Affiliation(s)
- Majid Khan
- Reno School of Medicine, University of Nevada, Reno, NV, USA
| | - Chadwin Hanna
- Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Matthew Findlay
- School of Medicine, University of Utah, Salt Lake City, UT, USA
| | | | - Michael Karsy
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah, 175 North Medical Drive East, Salt Lake City, UT 84132, USA.
| | - Randy L Jensen
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah, 175 North Medical Drive East, Salt Lake City, UT 84132, USA
| |
Collapse
|
3
|
Wach J, Naegeli J, Vychopen M, Seidel C, Barrantes-Freer A, Grunert R, Güresir E, Arlt F. Impact of Shape Irregularity in Medial Sphenoid Wing Meningiomas on Postoperative Cranial Nerve Functioning, Proliferation, and Progression-Free Survival. Cancers (Basel) 2023; 15:3096. [PMID: 37370707 DOI: 10.3390/cancers15123096] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/24/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Medial sphenoid wing meningiomas (MSWM) are surgically challenging skull base tumors. Irregular tumor shapes are thought to be linked to histopathology. The present study aims to investigate the impact of tumor shape on postoperative functioning, progression-free survival, and neuropathology. This monocentric study included 74 patients who underwent surgery for primary sporadic MSWM (WHO grades 1 and 2) between 2010 and 2021. Furthermore, a systematic review of the literature regarding meningioma shape and the MIB-1 index was performed. Irregular MSWM shapes were identified in 31 patients (41.9%). Multivariable analysis revealed that irregular shape was associated with postoperative cranial nerve deficits (OR: 5.75, 95% CI: 1.15-28.63, p = 0.033). In multivariable Cox regression analysis, irregular MSWM shape was independently associated with tumor progression (HR:8.0, 95% CI: 1.04-62.10, p = 0.046). Multivariable regression analysis showed that irregular shape is independently associated with an increased MIB-1 index (OR: 7.59, 95% CI: 2.04-28.25, p = 0.003). A systematic review of the literature and pooled data analysis, including the present study, showed that irregularly shaped meningiomas had an increase of 1.98 (95% CI: 1.38-2.59, p < 0.001) in the MIB-1 index. Irregular MSWM shape is independently associated with an increased risk of postoperative cranial nerve deficits and a shortened time to tumor progression. Irregular MSWM shapes might be caused by highly proliferative tumors.
Collapse
Affiliation(s)
- Johannes Wach
- Department of Neurosurgery, University Hospital Leipzig, University of Leipzig, 04103 Leipzig, Germany
| | - Johannes Naegeli
- Department of Neurosurgery, University Hospital Leipzig, University of Leipzig, 04103 Leipzig, Germany
| | - Martin Vychopen
- Department of Neurosurgery, University Hospital Leipzig, University of Leipzig, 04103 Leipzig, Germany
| | - Clemens Seidel
- Department of Radiation Oncology, University Hospital Leipzig, University of Leipzig, 04103 Leipzig, Germany
| | - Alonso Barrantes-Freer
- Department of Neuropathology, University Hospital Leipzig, University of Leipzig, 04103 Leipzig, Germany
| | - Ronny Grunert
- Department of Neurosurgery, University Hospital Leipzig, University of Leipzig, 04103 Leipzig, Germany
- Fraunhofer Institute for Machine Tools and Forming Technology, Theodor-Koerner-Allee 6, 02763 Zittau, Germany
| | - Erdem Güresir
- Department of Neurosurgery, University Hospital Leipzig, University of Leipzig, 04103 Leipzig, Germany
| | - Felix Arlt
- Department of Neurosurgery, University Hospital Leipzig, University of Leipzig, 04103 Leipzig, Germany
| |
Collapse
|
4
|
Wach J, Güresir Á, Vatter H, Herrlinger U, Becker A, Toma M, Hölzel M, Güresir E. Low-Dose Acetylsalicylic Acid Treatment in Non-Skull-Base Meningiomas: Impact on Tumor Proliferation and Seizure Burden. Cancers (Basel) 2022; 14:cancers14174285. [PMID: 36077817 PMCID: PMC9454729 DOI: 10.3390/cancers14174285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/26/2022] [Accepted: 08/31/2022] [Indexed: 11/16/2022] Open
Abstract
MIB-1 index is an important predictor of meningioma progression and was found to be correlated with COX-2 expression. However, the impact of low-dose acetylsalicylic acid (ASA) on MIB-1 index and clinical symptoms is unclear. Between 2009 and 2022, 710 patients with clinical data, tumor-imaging data, inflammatory laboratory (plasma fibrinogen, serum C-reactive protein) data, and neuropathological reports underwent surgery for primary cranial WHO grade 1 and 2 meningioma. ASA intake was found to be significantly associated with a low MIB-1 labeling index in female patients ≥ 60 years. Multivariable analysis demonstrated that female patients ≥ 60 years with a non-skull-base meningioma taking ASA had a significantly lower MIB-1 index (OR: 2.6, 95%: 1.0–6.6, p = 0.04). Furthermore, the intake of ASA was independently associated with a reduced burden of symptomatic epilepsy at presentation in non-skull-base meningiomas in both genders (OR: 3.8, 95%CI: 1.3–10.6, p = 0.03). ASA intake might have an anti-proliferative effect in the subgroup of elderly female patients with non-skull-base meningiomas. Furthermore, anti-inflammatory therapy seems to reduce the burden of symptomatic epilepsy in non-skull-base meningiomas. Further research is needed to investigate the role of anti-inflammatory therapy in non-skull-base meningiomas.
Collapse
Affiliation(s)
- Johannes Wach
- Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany
- Correspondence: ; Tel.: +49-228-287-16521
| | - Ági Güresir
- Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Hartmut Vatter
- Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Ulrich Herrlinger
- Division of Clinical Neurooncology, Department of Neurology and Centre of Integrated Oncology, University Hospital Bonn, 53127 Bonn, Germany
| | - Albert Becker
- Department of Neuropathology, University Hospital Bonn, 53127 Bonn, Germany
| | - Marieta Toma
- Institute of Pathology, University Hospital Bonn, 53127 Bonn, Germany
| | - Michael Hölzel
- Institute of Experimental Oncology, University Hospital Bonn, 53127 Bonn, Germany
| | - Erdem Güresir
- Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany
| |
Collapse
|
5
|
Boetto J, Peyre M, Kalamarides M. Mouse Models in Meningioma Research: A Systematic Review. Cancers (Basel) 2021; 13:cancers13153712. [PMID: 34359639 PMCID: PMC8345085 DOI: 10.3390/cancers13153712] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/10/2021] [Accepted: 07/21/2021] [Indexed: 12/21/2022] Open
Abstract
Meningiomas are the most frequent primitive central nervous system tumors found in adults. Mouse models of cancer have been instrumental in understanding disease mechanisms and establishing preclinical drug testing. Various mouse models of meningioma have been developed over time, evolving in light of new discoveries in our comprehension of meningioma biology and with improvements in genetic engineering techniques. We reviewed all mouse models of meningioma described in the literature, including xenograft models (orthotopic or heterotopic) with human cell lines or patient derived tumors, and genetically engineered mouse models (GEMMs). Xenograft models provided useful tools for preclinical testing of a huge range of innovative drugs and therapeutic options, which are summarized in this review. GEMMs offer the possibility of mimicking human meningiomas at the histological, anatomical, and genetic level and have been invaluable in enabling tumorigenesis mechanisms, including initiation and progression, to be dissected. Currently, researchers have a range of different mouse models that can be used depending on the scientific question to be answered.
Collapse
Affiliation(s)
- Julien Boetto
- Department of Neurosurgery, Gui de Chauliac Hospital, Montpellier Universitary Hospital Center, 80 Avenue Augustin Fliche, 34090 Montpellier, France;
- Institut du Cerveau et de la Moelle Épinière, INSERM U1127 CNRS UMR 7225, F-75013 Paris, France;
| | - Matthieu Peyre
- Institut du Cerveau et de la Moelle Épinière, INSERM U1127 CNRS UMR 7225, F-75013 Paris, France;
- Department of Neurosurgery, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris, France
- Sorbonne Université, Université Pierre et Marie Curie Paris 06, F-75013 Paris, France
| | - Michel Kalamarides
- Institut du Cerveau et de la Moelle Épinière, INSERM U1127 CNRS UMR 7225, F-75013 Paris, France;
- Department of Neurosurgery, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris, France
- Sorbonne Université, Université Pierre et Marie Curie Paris 06, F-75013 Paris, France
- Correspondence:
| |
Collapse
|
6
|
Suppiah S, Nassiri F, Bi WL, Dunn IF, Hanemann CO, Horbinski CM, Hashizume R, James CD, Mawrin C, Noushmehr H, Perry A, Sahm F, Sloan A, Von Deimling A, Wen PY, Aldape K, Zadeh G. Molecular and translational advances in meningiomas. Neuro Oncol 2020; 21:i4-i17. [PMID: 30649490 DOI: 10.1093/neuonc/noy178] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Meningiomas are the most common primary intracranial neoplasm. The current World Health Organization (WHO) classification categorizes meningiomas based on histopathological features, but emerging molecular data demonstrate the importance of genomic and epigenomic factors in the clinical behavior of these tumors. Treatment options for symptomatic meningiomas are limited to surgical resection where possible and adjuvant radiation therapy for tumors with concerning histopathological features or recurrent disease. At present, alternative adjuvant treatment options are not available in part due to limited historical biological analysis and clinical trial investigation on meningiomas. With advances in molecular and genomic techniques in the last decade, we have witnessed a surge of interest in understanding the genomic and epigenomic landscape of meningiomas. The field is now at the stage to adopt this molecular knowledge to refine meningioma classification and introduce molecular algorithms that can guide prediction and therapeutics for this tumor type. Animal models that recapitulate meningiomas faithfully are in critical need to test new therapeutics to facilitate rapid-cycle translation to clinical trials. Here we review the most up-to-date knowledge of molecular alterations that provide insight into meningioma behavior and are ready for application to clinical trial investigation, and highlight the landscape of available preclinical models in meningiomas.
Collapse
Affiliation(s)
- Suganth Suppiah
- Division of Neurosurgery, University Health Network, University of Toronto, Ontario, Canada.,MacFeeters-Hamilton Center for Neuro-Oncology, Princess Margaret Cancer Center, Toronto, Ontario, Canada
| | - Farshad Nassiri
- Division of Neurosurgery, University Health Network, University of Toronto, Ontario, Canada.,MacFeeters-Hamilton Center for Neuro-Oncology, Princess Margaret Cancer Center, Toronto, Ontario, Canada
| | - Wenya Linda Bi
- Centre for Skull Base and Pituitary Surgery, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ian F Dunn
- Centre for Skull Base and Pituitary Surgery, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Clemens Oliver Hanemann
- Institute of Translational and Stratified Medicine, Peninsula Schools of Medicine and Dentistry, Plymouth University, Plymouth, United Kingdom
| | - Craig M Horbinski
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Rintaro Hashizume
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Charles David James
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Christian Mawrin
- Institute of Neuropathology, Otto-von-Guericke University, Magdeburg, Germany
| | - Houtan Noushmehr
- Department of Neurosurgery, Henry Ford Health System, Detroit, Michigan, USA
| | - Arie Perry
- Department of Pathology, University of California San Francisco, San Francisco, California, USA
| | - Felix Sahm
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Andrew Sloan
- Department of Neurological Surgery, University Hospital-Case Medical Center, Cleveland, Ohio, USA
| | - Andreas Von Deimling
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Kenneth Aldape
- Department of Laboratory Pathology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA.,MacFeeters-Hamilton Center for Neuro-Oncology, Princess Margaret Cancer Center, Toronto, Ontario, Canada
| | - Gelareh Zadeh
- Division of Neurosurgery, University Health Network, University of Toronto, Ontario, Canada.,MacFeeters-Hamilton Center for Neuro-Oncology, Princess Margaret Cancer Center, Toronto, Ontario, Canada
| | | |
Collapse
|
7
|
Burnett BA, Womeldorff MR, Jensen R. Meningioma: Signaling pathways and tumor growth. HANDBOOK OF CLINICAL NEUROLOGY 2020; 169:137-150. [PMID: 32553285 DOI: 10.1016/b978-0-12-804280-9.00009-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Meningiomas are the most common primary intracranial brain tumor in adult humans; however, our understanding of meningioma tumorigenesis is relatively limited in comparison with the body of research available for other intracranial tumors such as gliomas. Here we briefly describe the current understanding of aberrant signaling pathways and tumor growth mechanisms responsible for meningioma differentiation, cellular growth, development, inhibition, and death. Numerous cellular functions impacted by these signaling pathways are critical for angiogenesis, proliferation, and apoptosis. Ultimately, a further understanding of the signaling pathways involved in meningioma tumorigenesis will lead to better treatment modalities in the future.
Collapse
Affiliation(s)
- Brian Andrew Burnett
- Department of Neurosurgery, University of Utah, Salt Lake City, UT, United States
| | | | - Randy Jensen
- Department of Neurosurgery, University of Utah, Salt Lake City, UT, United States.
| |
Collapse
|
8
|
Choudhury A, Raleigh DR. Preclinical models of meningioma: Cell culture and animal systems. HANDBOOK OF CLINICAL NEUROLOGY 2020; 169:131-136. [PMID: 32553284 DOI: 10.1016/b978-0-12-804280-9.00008-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Meningioma is the most common primary intracranial tumor; yet there are no effective systemic or molecular therapies for meningioma patients. One of the primary barriers to understanding meningioma biology and identifying novel therapeutic targets is the lack of tractable preclinical models. While numerous model systems have been created for meningioma, many have fundamental drawbacks. This chapter details the strengths and limitations of existing meningioma models and suggests possible future model systems. Cell culture meningioma models consist of human meningioma cell lines derived from tumor resection specimens, but unfortunately, in vitro systems do not capture the histologic architecture, the tumor microenvironment, or the heterogeneity of meningiomas. Mouse meningioma systems range from genetically engineered mouse models (GEMMs) to patient-derived xenografts (PDXs) and overcome some of the limitations of cultured meningioma cells. However, many in vivo systems have poor reproducibility or fail to recapitulate important aspects of meningioma biology, such as tumor latency. Despite these drawbacks, new discoveries in meningioma biology and advances in the technologies used to develop model systems provide hope that more representative models of meningioma will be developed in the near future.
Collapse
Affiliation(s)
- Abrar Choudhury
- Brain Tumor Center, University of California San Francisco, San Francisco, CA, United States
| | - David R Raleigh
- Brain Tumor Center, University of California San Francisco, San Francisco, CA, United States.
| |
Collapse
|
9
|
Abstract
More than 70,000 primary central nervous system tumors are diagnosed in the United States each year. Approximately 36% of these are meningiomas, making it the most common primary brain tumor. Because meningioma risk increases dramatically with age, the healthcare burden of meningioma in the developed world will continue to rise as demographics shift toward an older population. In addition to demographic factors associated with increased meningioma risk (i.e., older age, female sex, African American ethnicity), increased body mass index is a strong risk factor. A history of atopic allergies, eczema, and increased serum IgE are all consistently associated with reduced meningioma risk, suggesting a potential role for immunosurveillance. Although ionizing radiation is a strong meningioma risk factor, it accounts for very few cases at the population level. Recent studies suggest that diagnostic radiation (e.g., dental X-rays) increases meningioma risk. Because radiation dosages associated with medical imaging have decreased dramatically, the public health impact of this exposure is likely in decline. Genome-wide association studies have identified common inherited variants in the gene MLLT10 and RIC8A as low-penetrance meningioma risk alleles. To provide further insight into the etiology of meningioma, future studies will need to simultaneously examine genetic and environmental risk factors, while also stratifying analyses by subject sex.
Collapse
Affiliation(s)
- Kyle M Walsh
- Department of Neurosurgery, Duke University, Durham, NC, United States.
| |
Collapse
|
10
|
Mortensen R, Clemmensen HS, Woodworth JS, Therkelsen ML, Mustafa T, Tonby K, Jenum S, Agger EM, Dyrhol-Riise AM, Andersen P. Cyclooxygenase inhibitors impair CD4 T cell immunity and exacerbate Mycobacterium tuberculosis infection in aerosol-challenged mice. Commun Biol 2019; 2:288. [PMID: 31396568 PMCID: PMC6683187 DOI: 10.1038/s42003-019-0530-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 07/02/2019] [Indexed: 01/06/2023] Open
Abstract
Tuberculosis, caused by infection with Mycobacterium tuberculosis (Mtb), kills over 1.6 million people each year despite availability of antibiotics. The increase in drug resistant Mtb strains is a major public health emergency and host-directed therapy as adjunct to antibiotic treatment has gained increased interest. Cyclooxygenase inhibitors (COXi) are frequently used drugs to alleviate tuberculosis related symptoms. Mouse studies of acute intravenous Mtb infection have suggested a potential benefit of COXi for host-directed therapy. Here we show that COXi treatment (ibuprofen and celecoxib) is detrimental to Mtb control in different mouse models of respiratory infection. This effect links to impairments of the Type-1 helper (Th1) T-cell response as CD4 T-cells in COXi-treated animals have significantly decreased Th1 differentiation, reduced IFNγ expression and decreased protective capacity upon adoptive transfer. If confirmed in clinical trials, these findings could have major impact on global health and question the use of COXi for host-directed therapy.
Collapse
Affiliation(s)
- Rasmus Mortensen
- Department of Infectious Disease Immunology, Statens Serum Institut, 2300 Copenhagen S, Denmark
| | | | - Joshua S. Woodworth
- Department of Infectious Disease Immunology, Statens Serum Institut, 2300 Copenhagen S, Denmark
| | - Marie Louise Therkelsen
- Department of Infectious Disease Immunology, Statens Serum Institut, 2300 Copenhagen S, Denmark
| | - Tehmina Mustafa
- Centre for International Health, Department of Global Public Health and Primary Care, University of Bergen & Department of Thoracic Medicine, Haukeland University Hospital, 5021 Bergen, Norway
| | - Kristian Tonby
- Department of Infectious Diseases, Oslo University Hospital, 0424 Oslo, Norway
| | - Synne Jenum
- Department of Infectious Diseases, Oslo University Hospital, 0424 Oslo, Norway
| | - Else Marie Agger
- Department of Infectious Disease Immunology, Statens Serum Institut, 2300 Copenhagen S, Denmark
| | - Anne Ma Dyrhol-Riise
- Department of Infectious Diseases, Oslo University Hospital, 0424 Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, 0424 Oslo, Norway
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institut, 2300 Copenhagen S, Denmark
- Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen N, Denmark
| |
Collapse
|
11
|
La Cava F, Fringuello Mingo A, Irrera P, Di Vito A, Cordaro A, Brioschi C, Colombo Serra S, Cabella C, Terreno E, Miragoli L. Orthotopic induction of CH157MN convexity and skull base meningiomas into nude mice using stereotactic surgery and MRI characterization. Animal Model Exp Med 2019; 2:58-63. [PMID: 31016288 PMCID: PMC6431243 DOI: 10.1002/ame2.12050] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 12/06/2018] [Accepted: 12/07/2018] [Indexed: 12/02/2022] Open
Abstract
Meningioma in vivo research is hampered by the difficulty of establishing an easy and reproducible orthotopic model able to mimic the characteristics of a human meningioma. Moreover, leptomeningeal dissemination and high mortality are often associated with such orthotopical models, making them useless for clinical translation studies. An optimized method for inducing meningiomas in nude mice at two different sites is described in this paper and the high reproducibility and low mortality of the models are demonstrated. Skull base meningiomas were induced in the auditory meatus and convexity meningiomas were induced on the brain surface of 23 and 24 nude mice, respectively. Both models led to the development of a mass easily observable by imaging methods. Dynamic contrast enhanced MRI was used as a tool to monitor and characterize the pathology onset and progression. At the end of the study, histology was performed to confirm the neoplastic origin of the diseased mass.
Collapse
Affiliation(s)
- Francesca La Cava
- Department of Molecular Biotechnologies and Health SciencesCenter of Excellence for Preclinical Imaging (CEIP)University of TorinoColleretto GiacosaTOItaly
| | | | - Pietro Irrera
- Department of Molecular Biotechnologies and Health SciencesCenter of Excellence for Preclinical Imaging (CEIP)University of TorinoColleretto GiacosaTOItaly
| | - Aldo Di Vito
- Bracco Research CentreBracco Imaging SpAColleretto GiacosaTOItaly
| | - Alessia Cordaro
- Bracco Research CentreBracco Imaging SpAColleretto GiacosaTOItaly
| | - Chiara Brioschi
- Bracco Research CentreBracco Imaging SpAColleretto GiacosaTOItaly
| | | | - Claudia Cabella
- Bracco Research CentreBracco Imaging SpAColleretto GiacosaTOItaly
| | - Enzo Terreno
- Department of Molecular Biotechnologies and Health SciencesCenter of Excellence for Preclinical Imaging (CEIP)University of TorinoColleretto GiacosaTOItaly
| | - Luigi Miragoli
- Bracco Research CentreBracco Imaging SpAColleretto GiacosaTOItaly
| |
Collapse
|
12
|
Hailat MM, Ebrahim HY, Mohyeldin MM, Goda AA, Siddique AB, El Sayed KA. The tobacco cembranoid (1S,2E,4S,7E,11E)-2,7,11-cembratriene-4,6-diol as a novel angiogenesis inhibitory lead for the control of breast malignancies. Bioorg Med Chem 2017; 25:3911-3921. [PMID: 28583806 DOI: 10.1016/j.bmc.2017.05.028] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 05/09/2017] [Accepted: 05/12/2017] [Indexed: 12/01/2022]
Abstract
(1S,2E,4S,6R,7E,11E)-2,7,11-cembratriene-4,6-diol (1) and its 4-epi-analog (2) are diterpene precursors of the key flavor components in most Nicotiana (tobacco) species that purposely degraded during commercial tobacco fermentation. Angiogenesis, recruitment of new blood vessels, is important for tumor growth, survival and metastasis that can be targeted to control cancer. This study shows evidences and potential of the cembranoid 1 as a potent angiogenesis modulator through targeting VEGFR2. In silico study suggested favorable docking scores and binding affinity of 1 at the ATP binding pocket of VEGFR2. The binding mode of 1 was parallel to the standard FDA-approved antiangiogenic drug sunitinib (4). In vitro, cembranoid 1 significantly reduced the activated VEGFR2 levels in multiple breast cancer cell lines. Intraperitoneal 40mg/kg, 3X/week treatment of 1 significantly reduced the MDA-MB-231 cells breast tumor size in mice. Immunohistochemistry and Western blotting analysis of the treated mice tumors showed significant downregulation of the vasculogenesis marker CD31 and suppressed activated VEGFR2-paxillin-FAK pathway. Matrigel study in Swiss albino mice showed similar trend. The tobacco cembranoid 1 is a potential antiangiogenic lead useful for future use to control breast malignancies.
Collapse
Affiliation(s)
- Mohammad M Hailat
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Hassan Y Ebrahim
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Mohamed M Mohyeldin
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Amira A Goda
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Abu Bakar Siddique
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Khalid A El Sayed
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA.
| |
Collapse
|
13
|
Antitumor activity of gemcitabine against high-grade meningioma in vitro and in vivo. Oncotarget 2017; 8:90996-91008. [PMID: 29207619 PMCID: PMC5710900 DOI: 10.18632/oncotarget.18827] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 06/10/2017] [Indexed: 12/11/2022] Open
Abstract
Currently, there is no established therapeutic option for high-grade meningioma recurring after surgery and radiotherapy, and few chemotherapeutic agents are in development for the treatment of high-grade meningioma. Here in this study, we screened a panel of chemotherapeutic agents for their possible antitumor activity in high-grade meningioma and discovered that high-grade meningioma cells show a preferential sensitivity to antimetabolites, in particular, to gemcitabine. In vitro, gemcitabine inhibited the growth of high-grade meningioma cells effectively by inducing S-phase arrest and apoptotic cell death. In vivo, systemic gemcitabine chemotherapy suppressed not only tumor initiation but also inhibited the growth and achieved a long-term control of established tumors in xenograft models of high-grade meningioma. Histological analysis indicated that systemic gemcitabine blocks cell cycle progression and promotes apoptotic cell death in tumor cells in vivo. Together, our data demonstrate that gemcitabine exerts potent antitumor activity against high-grade meningioma through cytostatic and cytotoxic mechanisms. We therefore propose gemcitabine is a promising chemotherapeutic agent that warrants further investigation as a treatment option for high-grade meningioma.
Collapse
|
14
|
Bundscherer A, Malsy M, Bitzinger D, Graf BM. [Interaction of anesthetics and analgesics with tumor cells]. Anaesthesist 2014; 63:313-25. [PMID: 24584840 DOI: 10.1007/s00101-014-2310-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The results of preclinical and clinical studies indicate that the perioperative period is a vulnerable period for cancer progression and metastasis. The risk of cancer cell dissemination is enhanced by the combination of surgical manipulation and perioperative immunosuppression. Whether the oncological outcome of cancer patients can be influenced by the choice of anesthetic techniques is still a matter of debate. This review summarizes the molecular characteristics of cancer and interaction of anesthetic and analgesic drugs with cancer cells.
Collapse
Affiliation(s)
- A Bundscherer
- Klinik für Anästhesiologie, Universitätsklinikum Regensburg, Franz Josef Strauß Allee 11, 93053, Regensburg, Deutschland,
| | | | | | | |
Collapse
|
15
|
Yang S, Gao Q, Jiang W. Relationship between tumour angiogenesis and expression of cyclo-oxygenase-2 and vascular endothelial growth factor-A in human renal cell carcinoma. J Int Med Res 2014; 43:110-7. [PMID: 25488950 DOI: 10.1177/0300060514545799] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE *These authors contributed equally to this work. To study the relationship between tumour angiogenesis and expression of cyclo-oxygenase (COX)-2 and vascular endothelial growth factor (VEGF)-A in human renal cell carcinoma. METHODS Archival samples of primary human renal cell carcinoma tissue and surrounding normal renal tissue (control samples) obtained from patients diagnosed with renal cell carcinoma were analysed for COX-2 and VEGF-A expression by immunohistochemistry using specific monoclonal antibodies. Tumour microvasculature was examined using factor VIII-related antigen antibody staining. RESULTS A total of 33 renal cell carcinoma and 12 control renal tissue specimens were included. COX-2 and VEGF-A genes were overexpressed in tumour specimens compared with normal epithelia. A significant correlation was found between COX-2 and VEGF-A expression. Microvessel density was found to be increased in tumour tissues that expressed COX-2 and VEGF-A. CONCLUSION Microvessel density was increased in tumour tissues that expressed COX-2 and VEGF-A, suggesting that COX-2 and VEGF-A are related to tumour angiogenesis in human renal cell carcinoma.
Collapse
Affiliation(s)
- Sheng Yang
- Department of Internal Medicine Oncology, The Union Hospital of Fujian Medical University, Fuzhou, China Fujian Provincial Key Laboratory of Tumour Translational Medicine, Fuzhou, China
| | - Qin Gao
- Teaching and Research Department of Thoracic Surgery, Union Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Wei Jiang
- Department of Urology, The Union Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
16
|
Mahzouni P, Sarmadi T. An observational study on the expression of cyclooxygenase-2 in meningioma. Adv Biomed Res 2014; 3:211. [PMID: 25371868 PMCID: PMC4219207 DOI: 10.4103/2277-9175.143256] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 05/18/2014] [Indexed: 11/15/2022] Open
Abstract
Background: The cyclooxygenase-2 (COX-2) enzyme is overexpressed in different types of tumors and is known to be associated with malignant behavior of tumors. We determined the association of COX-2 expression and different grades of human meningioma. Materials and Methods: This retrospective study was conducted on specimens obtained from adult patients with meningioma. Meningioma was classified according to the WHO 2007 classification protocol (I, II, and III). COX-2 expression intensity was scored based on the percentage of immunopositive cells as 0: 0-10%; +1: >10% and a part of the cell membrane; +2: >10% and complete cell membrane; and +3: >30% and complete cell membrane. Scores of +2 or +3 were considered as COX-2 positive. Results: Ninety meningioma cases (mean age = 53.0 ± 13.2 years, 71.1% female) were studied. COX-2 was positive in 25% (17/68), 68.4% (13/19), and 100% (3/3) of cases with tumor grade I, II, and III, respectively (P < 0.001). There was a significant correlation between tumor grade and COX-2 expression score (Spearman's correlation coefficient = 0.422, P < 0.001). Conclusions: There is a strong association between COX-2 expression and tumoral grade in meningioma with more aggressive tumors expressing COX-2 with more intensity. Prospective studies examining the association of COX-2 expression with tumor recurrence and interventional studies examining the role of COX-2 inhibitors anticancer therapy of meningioma are warranted.
Collapse
Affiliation(s)
- Parvin Mahzouni
- Department of Pathology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Tannaz Sarmadi
- Department of Pathology, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
17
|
Rosas C, Sinning M, Ferreira A, Fuenzalida M, Lemus D. Celecoxib decreases growth and angiogenesis and promotes apoptosis in a tumor cell line resistant to chemotherapy. Biol Res 2014; 47:27. [PMID: 25027008 PMCID: PMC4101715 DOI: 10.1186/0717-6287-47-27] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 05/07/2014] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND During the last few years it has been shown in several laboratories that Celecoxib (Cx), a non-steroidal anti-inflammatory agent (NSAID) normally used for pain and arthritis, mediates antitumor and antiangiogenic effects. However, the effects of this drug on a tumor cell line resistant to chemotherapeutical drugs used in cancer have not been described. RESULTS Cx reduces angiogenesis in the chick embryonic chorioallantoic membrane assay (CAM), inhibits the growth and microvascular density of the murine TA3-MTXR tumor, reduces microvascular density of tumor metastases, promotes apoptosis and reduces vascular endothelial growth factor (VEGF) production and cell proliferation in the tumor. CONCLUSION The antiangiogenic and antitumor Cx effects correlate with its activity on other tumor cell lines, suggesting that Prostaglandins (PGs) and VEGF production are involved. These results open the possibility of using Celecoxib combined with other experimental therapies, ideally aiming to get synergic effects.
Collapse
|
18
|
Kang HC, Kim IH, Park CI, Park SH. Immunohistochemical analysis of cyclooxygenase-2 and brain fatty acid binding protein expression in grades I-II meningiomas: correlation with tumor grade and clinical outcome after radiotherapy. Neuropathology 2014; 34:446-54. [PMID: 24779988 DOI: 10.1111/neup.12128] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 04/04/2014] [Accepted: 04/06/2014] [Indexed: 11/27/2022]
Abstract
This study was done to evaluate the association of cyclooxygenase 2 (COX-2) and brain fatty acid binding protein (BFABP) with tumor grade and outcome of grades I-II meningiomas treated with radiotherapy. From 1996 to 2008, 40 patients with intracranial grades I-II meningiomas were treated with radiotherapy. Immunohistochemical staining for COX-2 and BFABP were performed on formalin-fixed paraffin-embedded tissues. COX-2 expression was significantly associated with BFABP status and both COX-2 (P < 0.01) and BFABP (P = 0.01) expression were stronger in the grade II meningiomas than in grade I tumors. Among the clinicopathologic factors, age and COX-2 status were prognostic in progression-free survival. Patients with moderate or strong COX-2 expression had worse outcome than those with negative or weak COX-2 expression (P = 0.03) after controlling for potential confounders. Our results suggest that the molecular biomarker COX-2 has prognostic significance in intracranial grades I-II meningiomas following radiotherapy.
Collapse
Affiliation(s)
- Hyun-Cheol Kang
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, Korea; Department of Radiation Oncology, Dongnam Institute of Radiological and Medical Sciences, Busan, Korea
| | | | | | | |
Collapse
|
19
|
Sui W, Zhang Y, Wang Z, Wang Z, Jia Q, Wu L, Zhang W. Antitumor effect of a selective COX-2 inhibitor, celecoxib, may be attributed to angiogenesis inhibition through modulating the PTEN/PI3K/Akt/HIF-1 pathway in an H₂₂ murine hepatocarcinoma model. Oncol Rep 2014; 31:2252-60. [PMID: 24647425 DOI: 10.3892/or.2014.3093] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Accepted: 02/04/2014] [Indexed: 11/06/2022] Open
Abstract
Celecoxib, a selective cyclooxygenase-2 (COX-2) inhibitor, has recently been shown to affect the development of different types of cancer. The present study utilized a murine H22 hepatocarcinoma model to investigate the molecular mechanisms involved in celecoxib-induced inhibition of tumor angiogenesis. Tumor-bearing mice were randomly divided into five groups: i) control; ii) low-dose celecoxib (50 mg/kg); iii) high-dose celecoxib (200 mg/kg); iv) 5-fluorouracil (5-FU), (20 mg/kg) and v) combination of 5-FU and celecoxib (50 mg/kg). The antitumor effect of celecoxib was determined by measuring tumor volume. Tumor angiogenesis was evaluated by microvessel density (MVD). Tumor histology and immunostaining for CD34 in endothelial cells were performed to detect MVD. The expression levels of phosphatase and tensin homologue deleted from chromosome 10 (PTEN), phosphatidylinositol 3-kinase (PI3K), phospho‑Akt (P-Akt), COX-2, hypoxia-inducible factor-1α (HIF-1α) and vascular endothelial growth factor-A (VEGF-A) were detected by ELISA, immunohistochemistry and western blotting, respectively. We discovered substantial growth delay in murine H22 hepatoma as a result of celecoxib treatment. The inhibition rate of tumor growth induced by high-dose and low-dose celecoxib was 49.3 and 37.0%, respectively (P<0.05). The expression of PI3K, P-Akt, COX-2, HIF-1α, VEGF-A and PTEN in tumor tissues treated with celecoxib was demonstrated by immunohistochemistry, and the MVD was decreased in a dose-dependent manner (P<0.05). Reduced PI3K and P-Akt was particularly apparent in the high-dose celecoxib group (P<0.05). ELISA and western blotting data showed that the expression of PI3K, P-Akt, COX-2, HIF-1α and VEGF-A were reduced and PTEN was increased after treatment with celecoxib. In conclusion, the impact of celecoxib-induced tumor growth delay of murine H22 hepatocarcinoma may correlate with the inhibition of angiogenesis by reducing PI3K, P-Akt, COX-2, HIF-1α and VEGF-A expression and increasing PTEN expression in tumor tissue.
Collapse
Affiliation(s)
- Wenwen Sui
- Key Laboratory for Modern Medicine and Technology of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong, P.R. China
| | - Yueying Zhang
- Key Laboratory for Modern Medicine and Technology of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong, P.R. China
| | - Zhaopeng Wang
- Key Laboratory for Modern Medicine and Technology of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong, P.R. China
| | - Zhaoxia Wang
- Key Laboratory for Modern Medicine and Technology of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong, P.R. China
| | - Qing Jia
- Key Laboratory for Modern Medicine and Technology of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong, P.R. China
| | - Licun Wu
- Latner Thoracic Surgery Research Laboratories and Division of Thoracic Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Weidong Zhang
- Key Laboratory for Modern Medicine and Technology of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong, P.R. China
| |
Collapse
|
20
|
Non-steroidal anti-inflammatory drug use and brain tumour risk: a case-control study within the Clinical Practice Research Datalink. Cancer Causes Control 2013; 24:2027-34. [PMID: 23990380 DOI: 10.1007/s10552-013-0279-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 08/17/2013] [Indexed: 12/23/2022]
Abstract
PURPOSE The aetiology of primary brain tumours is largely unknown; the role of non-steroidal anti-inflammatory drugs (NSAIDs) or aspirin use and glioma risk has been inconclusive, but few population-based studies with reliable prescribing data have been conducted, and the association with meningioma risk has yet to be assessed. METHODS The UK Clinical Practice Research Datalink was used to assess the association between aspirin and non-aspirin NSAID use and primary brain tumour risk using a nested case-control study design. Conditional logistic regression analysis was performed on 5,052 brain tumour patients aged 16 years and over, diagnosed between 1987 and 2009 and 42,678 controls matched on year of birth, gender and general practice, adjusting for history of allergy and hormone replacement therapy use in the glioma and meningioma models, respectively. RESULTS In conditional logistic regression analysis, excluding drug use in the year preceding the index date, there was no association with non-aspirin NSAID use (OR 0.96, 95 % CI 0.81-1.13) or glioma risk comparing the highest category of daily defined dose to non-users; however, non-aspirin NSAID use was positively associated with meningioma risk (OR 1.35, 95 % CI 1.06-1.71). No association was seen with high- or low-dose aspirin use irrespective of histology. CONCLUSIONS This large nested case-control study finds no association between aspirin or non-aspirin NSAID use and risk of glioma but a slight increased risk with non-aspirin NSAIDs and meningioma.
Collapse
|
21
|
Lee SH, Lee YS, Hong YG, Kang CS. Significance of COX-2 and VEGF expression in histopathologic grading and invasiveness of meningiomas. APMIS 2013; 122:16-24. [DOI: 10.1111/apm.12079] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 02/22/2013] [Indexed: 01/28/2023]
Affiliation(s)
- Sung Hak Lee
- Department of Hospital Pathology; The Catholic University of Korea; College of Medicine; Seoul Korea
| | - Youn Soo Lee
- Department of Hospital Pathology; The Catholic University of Korea; College of Medicine; Seoul Korea
| | - Yong Gil Hong
- Department of Neurosurgery; The Catholic University of Korea; College of Medicine; Seoul Korea
| | - Chang Suk Kang
- Department of Hospital Pathology; The Catholic University of Korea; College of Medicine; Seoul Korea
| |
Collapse
|
22
|
Effect of systemic celecoxib on human meningioma after intracranial transplantation into nude mice. Acta Neurochir (Wien) 2013; 155:173-82. [PMID: 23143216 DOI: 10.1007/s00701-012-1534-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2012] [Accepted: 10/16/2012] [Indexed: 10/27/2022]
Abstract
BACKGROUND Meningiomas are mostly benign, but they may have a notorious tendency to recur when total resection is not possible. Systemic chemotherapeutical treatment has been largely disappointing. The treatment of meningiomas with the cyclooxygenase-2 (COX-2) inhibitor celecoxib showed inhibitory-growth effects in vitro and in vivo after subcutaneous transplantation into mouse. So far, celecoxib has never been tested in an orthotopic model of meningioma. In this work, we tested the effects of celecoxib on the growth of human benign meningiomas after transplantation into the prefrontal cortex of nude mice after confirming the inhibitory in vitro effect on these cells. METHODS Primary cell cultures were stereotactically implanted into mice and were treated with 0, 750, or 1,500 ppm celecoxib for 3 months. The mice were then killed and blood was analyzed for celecoxib concentration. The mice brains were histologically processed for measurement of tumor volume, COX-2 expression, proliferation index (PI), intratumoral microvessel density (iMVD), and vascular endothelial growth factor (VEGF) expression. RESULTS Treatment with celecoxib had no effect on tumor volume, despite the fact that we found a dose-dependent inhibitory effect on cell cultures and there was a sufficiently high celecoxib concentration in blood plasma and brain tissue. Additionally, celecoxib had neither an effect on COX-2 and VEGF expression nor on the PI and iMVD. CONCLUSIONS Our findings suggest that celecoxib may not be effective on meningioma growth in clinical settings. In general, these results may indicate that the effect of treatment on brain tumors should not only be tested in a heterotopic environment but also in the orthotopic location of these tumors.
Collapse
|
23
|
Clinicopathological evaluation of cyclooxygenase-2 expression in meningioma: immunohistochemical analysis of 76 cases of low and high-grade meningioma. Brain Tumor Pathol 2012; 31:23-30. [PMID: 23250387 DOI: 10.1007/s10014-012-0127-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 11/27/2012] [Indexed: 12/21/2022]
Abstract
Tumorigenic activity of cyclooxygenase-2 (COX-2), a rate-limiting enzyme in the production of prostaglandins (PGs), has been proved for some types of cancer, including brain tumors. We evaluated expression of COX-2 in meningioma, one of the most common intracranial tumors in adults which accounts for 24-30 % of intracranial tumors. We performed immunostaining for COX-2 in 76 cases of meningioma consisting of 44 cases of low-grade (WHO Grade I) and 32 cases of high-grade (29 cases of Grade II and 3 cases of Grade III) meningioma, and evaluated COX-2 expression levels on the basis of staining intensity and proportion in tumor cells. The expression level of COX-2 in meningioma cells was significantly correlated with WHO grade (P = 0.0153). In addition, COX-2 expression was significantly correlated with MIB-1 labeling index for all 76 cases of meningioma (P = 0.0075), suggesting tumor promotion by COX-2 in meningioma progression. Our results may indicate the therapeutic value of non-steroidal anti-inflammatory drugs against meningioma, especially for patients with elevated proliferation, to regulate the tumorigenic activity of COX-2 in meningioma cells.
Collapse
|
24
|
Li W, Xu XL, Zhang J, Cai JH, Tang YX. Effects of cyclooxygenase inhibitors on survival time in ovarian cancer xenograft-bearing mice. Oncol Lett 2012. [PMID: 23205124 DOI: 10.3892/ol.2012.929] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The present study was designed to investigate whether cyclooxygenase (COX) inhibitors (coxibs) could prolong survival time by attenuating the tumor growth of ovarian cancer xenograft-bearing mice. Tumor growth and survival time were observed and compared in mice which were treated with a COX-1 inhibitor (SC-560) and a COX-2 inhibitor (celecoxib) every other day for a 21 day period from the day of tumor formation. The trial lasted a total of 121 days. The combination therapy resulted in statistically significant inhibition of tumor size compared with the control group (P<0.05). Additionally, single treatment of SC-560 or celecoxib significantly prolonged the mean survival time of mice compared with the control group (P<0.05). We suggest that COX-1 and COX-2 inhibitors may improve survival and inhibit tumor growth, and that the tumor growth inhibition by coxibs may be the contributing factor for the prolonged survival time in mouse xenograft models.
Collapse
Affiliation(s)
- Wei Li
- Department of Gynecology, Nanjing Medical University of Hangzhou Hospital, Hangzhou, Zhejiang 310006, P.R. China
| | | | | | | | | |
Collapse
|
25
|
Lao HC, Akunda JK, Chun KS, Flake GP, Yuspa SH, Langenbach R. Genetic ablation of cyclooxygenase-2 in keratinocytes produces a cell-autonomous defect in tumor formation. Carcinogenesis 2012; 33:2293-300. [PMID: 22902545 DOI: 10.1093/carcin/bgs267] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Using a mouse skin tumor model, we reported previously that cyclooxygenase-2 (COX-2) deficiency reduced papilloma formation. However, this model did not differentiate between the effects of systemic COX-2-deficiency and keratinocyte-specific COX-2 deficiency on tumor formation. To determine whether keratinocyte-specific COX-2 deficiency reduced papilloma formation, v-H-ras-transformed COX-2+/+ and COX-2-/- keratinocytes were grafted onto nude mice and tumor development was compared. Transformed COX-2+/+ and COX-2-/- keratinocytes expressed similar levels of H-ras, epidermal growth factor receptor and phospho-extracellular signal-regulated kinase 1/2 in vitro; and COX-2-deficiency did not reduce uninfected or v-H-ras infected keratinocyte replication. In contrast, tumors arising from grafted transformed COX-2+/+ and COX-2-/- keratinocytes expressed similar levels of H-ras, but COX-2 deficiency reduced phospho-extracellular signal-regulated kinase 1/2 and epidermal growth factor receptor levels 50-60% and tumor volume by 80% at 3 weeks. Two factors appeared to account for the reduced papilloma size. First, papillomas derived from COX-2-/- keratinocytes showed about 70% decreased proliferation, as measured by bromodeoxyuridine incorporation, compared with papillomas derived from COX-2+/+ keratinocytes. Second, keratin 1 immunostaining of papillomas indicated that COX-2-/- keratinocytes prematurely initiated terminal differentiation. Differences in the levels of apoptosis and vascularization did not appear to be contributing factors as their levels were similar in tumors derived from COX-2-/- and COX-2+/+ keratinocytes. Overall, the data are in agreement with our previous observations that decreased papilloma number and size on COX-2-/- mice resulted from reduced keratinocyte proliferation and accelerated keratinocyte differentiation. Furthermore, the data indicate that deficiency/inhibition of COX-2 in the initiated keratinocyte is an important determinant of papilloma forming ability.
Collapse
Affiliation(s)
- Huei-Chen Lao
- Laboratory of Toxicology and Pharmacology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | | | | | | | | | | |
Collapse
|
26
|
Ginat DT, Mangla R, Yeaney G, Schaefer PW, Wang H. Correlation between dynamic contrast-enhanced perfusion MRI relative cerebral blood volume and vascular endothelial growth factor expression in meningiomas. Acad Radiol 2012; 19:986-90. [PMID: 22591719 DOI: 10.1016/j.acra.2012.04.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 04/03/2012] [Accepted: 04/04/2012] [Indexed: 12/23/2022]
Abstract
PURPOSE To determine whether there is a correlation between vascular endothelial growth factor (VEGF) expression and cerebral blood flow (CBV) measurements in dynamic contrast-enhanced susceptibility perfusion magnetic resonance imaging (MRI) and to correlate the perfusion characteristics in high- versus low-grade meningiomas. METHODS AND MATERIALS A total of 48 (24 high-grade and 24 low-grade) meningiomas with available dynamic susceptibility-weighted MRI were retrospectively reviewed for maximum CBV and semiquantitative VEGF immunoreactivity. Correlation between normalized CBV and VEGF was made using the Spearman rank test and comparison between CBV in high- versus low-grade meningiomas was made using the Wilcoxon test. RESULTS There was a significant (P = .01) correlation between normalized maximum CBV and VEGF scores with a Spearman correlation coefficient of 0.37. In addition, there was a significant (P < .01) difference in normalized maximum CBV ratios between high-grade meningiomas (mean 12.6; standard deviation 5.2) and low-grade meningiomas (mean 8.2; standard deviation 5.2). CONCLUSION The data suggest that CBV accurately reflects VEGF expression and tumor grade in meningiomas. Perfusion-weighted MRI can potentially serve as a useful biomarker for meningiomas, pending prospective studies.
Collapse
|
27
|
Alternative splicing of CHEK2 and codeletion with NF2 promote chromosomal instability in meningioma. Neoplasia 2012; 14:20-8. [PMID: 22355270 DOI: 10.1593/neo.111574] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Revised: 12/23/2011] [Accepted: 01/03/2012] [Indexed: 12/13/2022] Open
Abstract
Mutations of the NF2 gene on chromosome 22q are thought to initiate tumorigenesis in nearly 50% of meningiomas, and 22q deletion is the earliest and most frequent large-scale chromosomal abnormality observed in these tumors. In aggressive meningiomas, 22q deletions are generally accompanied by the presence of large-scale segmental abnormalities involving other chromosomes, but the reasons for this association are unknown. We find that large-scale chromosomal alterations accumulate during meningioma progression primarily in tumors harboring 22q deletions, suggesting 22q-associated chromosomal instability. Here we show frequent codeletion of the DNA repair and tumor suppressor gene, CHEK2, in combination with NF2 on chromosome 22q in a majority of aggressive meningiomas. In addition, tumor-specific splicing of CHEK2 in meningioma leads to decreased functional Chk2 protein expression. We show that enforced Chk2 knockdown in meningioma cells decreases DNA repair. Furthermore, Chk2 depletion increases centrosome amplification, thereby promoting chromosomal instability. Taken together, these data indicate that alternative splicing and frequent codeletion of CHEK2 and NF2 contribute to the genomic instability and associated development of aggressive biologic behavior in meningiomas.
Collapse
|
28
|
Hussein BH, Azab HA, El-Azab MF, El-Falouji AI. A novel anti-tumor agent, Ln(III) 2-thioacetate benzothiazole induces anti-angiogenic effect and cell death in cancer cell lines. Eur J Med Chem 2012; 51:99-109. [PMID: 22424613 DOI: 10.1016/j.ejmech.2012.02.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2011] [Revised: 02/11/2012] [Accepted: 02/13/2012] [Indexed: 10/28/2022]
|
29
|
Comparative morphological and immunohistochemical study of human meningioma after intracranial transplantation into nude mice. J Neurosci Methods 2011; 205:1-9. [PMID: 22209769 DOI: 10.1016/j.jneumeth.2011.12.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Revised: 12/14/2011] [Accepted: 12/15/2011] [Indexed: 01/25/2023]
Abstract
Although surgical resection of benign human meningiomas is the primary goal, in case of relapse or when they are not fully resectable, other strategies including chemotherapeutical treatment would be appropriate. The initial evaluation of chemotherapeutical agents requires an appropriate tumor model, where the natural characteristics of the original benign tumor is reflected. We here tested, whether primary cell cultures of benign human meningiomas would reliably grow after intracranial transplantation into mice, and whether they would show histomorphological and immunohistochemical characteristics of the original human tumor. Cells of 11 benign human meningiomas were transplanted into the prefrontal cortex of nude mice. After 3 months, the mice were sacrificed and their brains were histologically processed for morphological characterization and measurement of tumor volume. Additionally, the proliferation index (PI), the microvessel density, and epithelial membrane antigen (EMA) were compared between human meningiomas and tumors grown in mice by using immunohistochemical methods. Further, cyclooxygenase-2 (COX-2) expression, a possible target for pharmacological manipulation, was examined. The results showed in almost all mice (93%) a tumor formation with meningothelial histomorphology comparable to the original human tumors. The PI, vascular density and COX-2 expression were similar between human and mice meningiomas, but EMA expression was reduced in mice (P<0.01). In conclusion an implantation of benign human meningioma primary cell cultures in mice reliably results in tumor formation with morphological and immunohistological features comparable to the original human tumor. This model may therefore be suitable to test novel therapeutic agents.
Collapse
|
30
|
R-flurbiprofen, a novel nonsteroidal anti-inflammatory drug, decreases cell proliferation and induces apoptosis in pituitary adenoma cells in vitro. J Neurooncol 2011; 106:561-9. [DOI: 10.1007/s11060-011-0712-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Accepted: 09/12/2011] [Indexed: 12/21/2022]
|
31
|
Choy W, Kim W, Nagasawa D, Stramotas S, Yew A, Gopen Q, Parsa AT, Yang I. The molecular genetics and tumor pathogenesis of meningiomas and the future directions of meningioma treatments. Neurosurg Focus 2011; 30:E6. [DOI: 10.3171/2011.2.focus1116] [Citation(s) in RCA: 124] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Meningiomas are mostly benign, slow-growing tumors of the CNS that originate from arachnoidal cap cells. While monosomy 22 is the most frequent genetic abnormality found in meningiomas, a multitude of other aberrant chromosomal alterations, signaling pathways, and growth factors have been implicated in its pathogenesis. Losses on 22q12.2, a region encoding the tumor suppressor gene merlin, represent the most common genetic alterations in early meningioma formation. Malignant meningioma progression, however, is associated with more complex karyotypes and greater genetic instability. Cytogenetic studies of atypical and anaplastic meningiomas revealed gains and losses on chromosomes 9, 10, 14, and 18, with amplifications on chromosome 17. However, the specific gene targets in a majority of these chromosomal abnormalities remain elusive.
Studies have also implicated a myriad of aberrant signaling pathways involved with meningioma tumorigenesis, including those involved with proliferation, angiogenesis, and autocrine loops. Understanding these disrupted pathways will aid in deciphering the relationship between various genetic changes and their downstream effects on meningioma pathogenesis.
Despite advancements in our understanding of meningioma pathogenesis, the conventional treatments, including surgery, radiotherapy, and stereotactic radiosurgery, have remained largely stagnant. Surgery and radiation therapy are curative in the majority of lesions, yet treatment remains challenging for meningiomas that are recurrent, aggressive, or refractory to conventional treatments. Future therapies will include combinations of targeted molecular agents as a result of continued progress in the understanding of genetic and biological changes associated with meningiomas.
Collapse
Affiliation(s)
| | - Won Kim
- 1Department of Neurological Surgery, and
| | | | | | - Andrew Yew
- 1Department of Neurological Surgery, and
| | - Quinton Gopen
- 2Division of Otolaryngology, University of California Los Angeles; and
| | - Andrew T. Parsa
- 3Department of Neurological Surgery, University of California, San Francisco, California
| | - Isaac Yang
- 1Department of Neurological Surgery, and
| |
Collapse
|
32
|
Genetic and molecular alterations in meningiomas. Clin Neurol Neurosurg 2011; 113:261-7. [DOI: 10.1016/j.clineuro.2010.12.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2010] [Revised: 12/05/2010] [Accepted: 12/09/2010] [Indexed: 11/22/2022]
|
33
|
Cyclin D1 expression and the inhibitory effect of celecoxib on ovarian tumor growth in vivo. Int J Mol Sci 2010; 11:3999-4013. [PMID: 21152316 PMCID: PMC2996781 DOI: 10.3390/ijms11103999] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Revised: 09/28/2010] [Accepted: 10/08/2010] [Indexed: 11/17/2022] Open
Abstract
The report aims to investigate the relationship between the expression of cyclin D1 and Cyclooxgenase-2 (COX-2), thus to explore the molecular mechanisms of the antitumor efficacy of Celecoxib, a COX-2 inhibitor. Human ovarian SKOV-3 carcinoma cell xenograft-bearing mice were treated with Celecoxib by infusing gaster (i.g.) twice/day for 21 days. The mRNA levels of COX-2 and cyclin D1 were determined by RT-PCR. The expression of cyclin D1 at the protein level was detected by immunohistochemistry, while COX-2 protein expression was determined by Western blot. A high-dose of Celecoxib (100 mg/kg) significantly inhibited tumor growth (P < 0.05), and the expression of cyclin D1 was reduced by 61%. Celecoxib decreased the proliferation cell index by 40% (P < 0.001) and increased apoptotic index by 52% (P < 0.05) in high-dose Celecoxib treated group. Our results suggest that the antitumor efficacy of Celecoxib against ovarian cancer in mice may in part be mediated through suppression of cyclin D1, which may contribute to its ability to suppress proliferation.
Collapse
|
34
|
Aberrant signaling pathways in meningiomas. J Neurooncol 2010; 99:315-24. [DOI: 10.1007/s11060-010-0381-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Accepted: 08/20/2010] [Indexed: 12/24/2022]
|
35
|
|
36
|
Tang JY, Aszterbaum M, Athar M, Barsanti F, Cappola C, Estevez N, Hebert J, Hwang J, Khaimskiy Y, Kim A, Lu Y, So PL, Tang X, Kohn MA, McCulloch CE, Kopelovich L, Bickers DR, Epstein EH. Basal cell carcinoma chemoprevention with nonsteroidal anti-inflammatory drugs in genetically predisposed PTCH1+/- humans and mice. Cancer Prev Res (Phila) 2010; 3:25-34. [PMID: 20051370 DOI: 10.1158/1940-6207.capr-09-0200] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In vitro and epidemiologic studies favor the efficacy of nonsteroidal anti-inflammatory drugs (NSAID) in preventing skin squamous photocarcinogenesis, but there has been relatively little study of their efficacy in preventing the more common skin basal cell carcinoma (BCC) carcinogenesis. We first compared the relative anti-BCC effects of genetic deletion and NSAID pharmacologic inhibition of cyclooxygenase (COX) enzymes in the skin of Ptch1(+/-) mice. We then assessed the effects of celecoxib on the development of BCCs in a 3-year, double-blinded, randomized clinical trial in 60 (PTCH1(+/-)) patients with the basal cell nevus syndrome. In Ptch1(+/-) mice, genetic deletion of COX1 or COX2 robustly decreased (75%; P < 0.05) microscopic BCC tumor burden, but pharmacologic inhibition with celecoxib reduced microscopic BCCs less efficaciously (35%; P < 0.05). In the human trial, we detected a trend for oral celecoxib reducing BCC burden in all subjects (P = 0.069). Considering only the 60% of patients with less severe disease (<15 BCCs at study entry), celecoxib significantly reduced BCC number and burden: subjects receiving placebo had a 50% increase in BCC burden per year, whereas subjects in the celecoxib group had a 20% increase (P(difference) = 0.024). Oral celecoxib treatment inhibited BCC carcinogenesis in PTCH1(+/-) mice and had a significant anti-BCC effect in humans with less severe disease.
Collapse
Affiliation(s)
- Jean Y Tang
- The Children's Hospital of Oakland Research Institute, CA 94609, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Kim CK, Joe YA, Lee SK, Kim EK, O E, Kim HK, Oh BJ, Hong SH, Hong YK. Enhancement of anti-tumor activity by low-dose combination of the recombinant urokinase kringle domain and celecoxib in a glioma model. Cancer Lett 2010; 288:251-60. [DOI: 10.1016/j.canlet.2009.07.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Revised: 07/09/2009] [Accepted: 07/10/2009] [Indexed: 11/24/2022]
|
38
|
Wang D, Stockard CR, Harkins L, Lott P, Salih C, Yuan K, Buchsbaum D, Hashim A, Zayzafoon M, Hardy RW, Hameed O, Grizzle W, Siegal GP. Immunohistochemistry in the evaluation of neovascularization in tumor xenografts. Biotech Histochem 2009; 83:179-89. [PMID: 18846440 DOI: 10.1080/10520290802451085] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Angiogenesis, or neovascularization, is known to play an important role in the neoplastic progression leading to metastasis. CD31 or Factor VIII-related antigen (F VIII RAg) immunohistochemistry is widely used in experimental studies for quantifying tumor neovascularization in immunocompromised animal models implanted with transformed human cell lines. Quantification, however, can be affected by variations in the methodology used to measure vascularization including antibody selection, antigen retrieval (AR) pretreatment, and evaluation techniques. To examine this further, we investigated the microvessel density (MVD) and the intensity of microvascular staining among five different human tumor xenografts and a mouse syngeneic tumor using anti-CD31 and F VIII RAg immunohistochemical staining. Different AR methods also were evaluated. Maximal retrieval of CD31 was achieved using 0.5 M Tris (pH 10) buffer, while maximum retrieval of F VIII RAg was achieved using 0.05% pepsin treatment of tissue sections. For each optimized retrieval condition, anti-CD31 highlighted small vessels better than F VIII RAg. Furthermore, the MVD of CD31 was significantly greater than that of F VIII RAg decorated vessels (p<0.001). The choice of antibody and AR method has a significant affect on immunohistochemical findings when studying angiogenesis. One also must use caution when comparing studies in the literature that use different techniques and reagents.
Collapse
Affiliation(s)
- D Wang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Walsh MT, Couldwell WT. Management options for cavernous sinus meningiomas. J Neurooncol 2009; 92:307-16. [DOI: 10.1007/s11060-009-9824-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2008] [Accepted: 02/23/2009] [Indexed: 02/04/2023]
|
40
|
Maussang D, Langemeijer E, Fitzsimons CP, Stigter-van Walsum M, Dijkman R, Borg MK, Slinger E, Schreiber A, Michel D, Tensen CP, van Dongen GAMS, Leurs R, Smit MJ. The human cytomegalovirus-encoded chemokine receptor US28 promotes angiogenesis and tumor formation via cyclooxygenase-2. Cancer Res 2009; 69:2861-9. [PMID: 19318580 DOI: 10.1158/0008-5472.can-08-2487] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The human cytomegalovirus (HCMV), potentially associated with the development of malignancies, encodes the constitutively active chemokine receptor US28. Previously, we have shown that US28 expression induces an oncogenic phenotype both in vitro and in vivo. Microarray analysis revealed differential expression of genes involved in oncogenic signaling in US28-expressing NIH-3T3 cells. In particular, the expression of cyclooxygenase-2 (COX-2), a key mediator of inflammatory diseases and major determinant in several forms of cancer, was highly up-regulated. US28 induced increases in COX-2 expression via activation of nuclear factor-kappaB, driving the production of vascular endothelial growth factor. Also, in HCMV-infected cells, US28 contributed to the viral induction of COX-2. Finally, the involvement of COX-2 in US28-mediated tumor formation was evaluated using the COX-2 selective inhibitor Celecoxib. Targeting COX-2 in vivo with Celecoxib led to a marked delay in the onset of tumor formation in nude mice injected with US28-transfected NIH-3T3 cells and a reduction of subsequent growth by repressing the US28-induced angiogenic activity. Hence, the development of HCMV-related proliferative diseases may partially be ascribed to the ability of US28 to activate COX-2.
Collapse
Affiliation(s)
- David Maussang
- Leiden/Amsterdam Center for Drug Research, Division of Medicinal Chemistry, Faculty of Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Chemotherapy, hormonal therapy, and immunotherapy for recurrent meningiomas. J Neurooncol 2008; 92:1-6. [DOI: 10.1007/s11060-008-9734-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2008] [Accepted: 11/03/2008] [Indexed: 11/26/2022]
|
42
|
Expression of cell adhesion proteins and proteins related to angiogenesis and fatty acid metabolism in benign, atypical, and anaplastic meningiomas. J Neurooncol 2008; 89:73-87. [PMID: 18418552 DOI: 10.1007/s11060-008-9588-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2007] [Accepted: 03/28/2008] [Indexed: 02/05/2023]
Abstract
Most meningiomas are benign tumours of arachnoidal origin, although a small number have high proliferative rates and invasive properties which complicate complete surgical resection and are associated with increased recurrence rates. Few prognostic indicators exist for meningiomas and further research is necessary to identify factors that influence tumour invasion, oedema and recurrence. Paraffin sections from 25 intracranial meningiomas were analysed for expression of the proteins vascular endothelial growth factor (VEGF), VEGF receptors Flt1 and Flk1, E-cadherin, metalloproteinases 2 and 9 (MMP2, MMP9), CD44, receptor for hyaluronic acid-mediated motility (RHAMM), hyaluronic acid (HA), CD45, cyclooxygenase 2 (COX2), brain fatty acid binding protein (BFABP), Ki67, and proliferating cell nuclear antigen (PCNA). Correlations among protein expression were found for several markers of proliferation (Ki67, PCNA, MI) and microvessel density (MVD). COX2 expression increased with increasing with tumour grade and correlated with Ki67, PCNA, MI, MVD, and BFABP. BFABP expression also correlated with Ki67 and PCNA expression. Relationships were also identified among angiogenic factors (VEGF, Flt1, Flk1) and proliferation markers. Oedema was found to correlate with MMP9 expression and MMP9 also correlated with proliferation markers. No correlations were found for MMP2, E-cadherin, or CD44 in meningiomas. In conclusion Ki67, PCNA, MI, MVD, BFABP, and COX2 were significantly correlated with meningioma tumour grade and with each other. These findings, by correlating both intracellular fatty acid transport and eicosanoid metabolism with tumour proliferation, as determined by Ki67 labelling and mitotic index, suggest fatty acids are involved in the progression of meningiomas.
Collapse
|
43
|
Ragel BT, Elam IL, Gillespie DL, Flynn JR, Kelly DA, Mabey D, Feng H, Couldwell WT, Jensen RL. A novel model of intracranial meningioma in mice using luciferase-expressing meningioma cells. Laboratory investigation. J Neurosurg 2008; 108:304-10. [PMID: 18240927 DOI: 10.3171/jns/2008/108/2/0304] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Meningioma research has been hindered by the inability to sequentially measure intracranial tumor growth in a cost-effective, efficient manner. Recently, the luciferase gene has been transfected into cancer lines to obtain cells that express the luciferase enzyme, which oxidizes luciferin in a reaction that releases photon energy that can be measured noninvasively by bioluminescence imaging (BLI) systems. The authors describe a mouse model of intracranial meningioma that uses this novel BLI system. METHODS The immortal meningioma cell lines CH-157-MN and IOMM-Lee were transfected with luciferase and neomycin phosphotransferase (LucNeo) and selected with G418. These cells were stereotactically implanted at skull base and cerebral convexity locations in nude mice. Animals were imaged for bioluminescence biweekly, and 5 mice underwent magnetic resonance (MR) imaging. Tumors were harvested for immunohistochemical and ultrastructural analysis. RESULTS The CH-157-MN-LucNeo and IOMM-Lee-LucNeo cell lines were successfully implanted intracranially in mice. The tumor induction rate for CH-157-MN-LucNeo skull base tumors was 90% (36 of 40 procedures). Statistical analysis of CH-157-MN-LucNeo skull base tumor volume measured on MR imaging correlated with the results of BLI showed an R value of 0.900. The tumors exhibited characteristics of aggressive meningiomas by insinuating along arachnoid planes and invading brain. CONCLUSIONS Noninvasive BLI was successfully used to image intracranial meningiomas in mice. The tumors grew in a fashion similar to that of aggressive meningiomas in humans, and exhibited the microscopic, immunohistochemical, and ultrastructural features characteristic of meningiomas. This animal model overcomes the main obstacle in studying intracranial meningiomas by enabling sequential noninvasive tumor measurement in a cost-effective manner.
Collapse
Affiliation(s)
- Brian T Ragel
- Department of Neurosurgery, University of Utah, Salt Lake City 84132, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Baia GS, Dinca EB, Ozawa T, Kimura ET, McDermott MW, James CD, VandenBerg SR, Lal A. An orthotopic skull base model of malignant meningioma. Brain Pathol 2007; 18:172-9. [PMID: 18093250 DOI: 10.1111/j.1750-3639.2007.00109.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Meningioma tumor growth involves the subarachnoid space that contains the cerebrospinal fluid. Modeling tumor growth in this microenvironment has been associated with widespread leptomeningeal dissemination, which is uncharacteristic of human meningiomas. Consequently, survival times and tumor properties are varied, limiting their utility in testing experimental therapies. We report the development and characterization of a reproducible orthotopic skull-base meningioma model in athymic mice using the IOMM-Lee cell line. Localized tumor growth was obtained by using optimal cell densities and matrigel as the implantation medium. Survival times were within a narrow range of 17-21 days. The xenografts grew locally compressing surrounding brain tissue. These tumors had histopathologic characteristics of anaplastic meningiomas including high cellularity, nuclear pleomorphism, cellular pattern loss, necrosis and conspicuous mitosis. Similar to human meningiomas, considerable invasion of the dura and skull and some invasion of adjacent brain along perivascular tracts were observed. The pattern of hypoxia was also similar to human malignant meningiomas. We use bioluminescent imaging to non-invasively monitor the growth of the xenografts and determine the survival benefit from temozolomide treatment. Thus, we describe a malignant meningioma model system that will be useful for investigating the biology of meningiomas and for preclinical assessment of therapeutic agents.
Collapse
Affiliation(s)
- Gilson S Baia
- Brain Tumor Research Center, Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Ragel BT, Jensen RL, Couldwell WT. Inflammatory response and meningioma tumorigenesis and the effect of cyclooxygenase-2 inhibitors. Neurosurg Focus 2007; 23:E7. [PMID: 17961044 DOI: 10.3171/foc-07/10/e7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
In this article the authors discuss the rationale and research supporting the hypothesis that meningioma tumorigenesis may, in part, be driven by overexpression of cyclooxygenase-2 (Cox-2) and that treatment with celecoxib, a selective Cox-2 inhibitor, may hold therapeutic promise. Because therapies for recurrent or aggressive meningiomas (atypical or malignant subtypes) such as chemotherapy and radiotherapy generally offer little therapeutic benefit, interest in targeting Cox-2 has grown. This rate-limiting enzyme of prostaglandin synthesis can be inhibited with nonsteroidal antiinflammatory drugs (NSAIDs) such as ibuprofen and celecoxib. Treatment with NSAIDs has been shown to curb the tumorigenic properties of prostaglandins in several cancer models via both Cox-2-dependent and -independent mechanisms. In addition, celecoxib is well tolerated in humans, making its use as a chronic therapy for meningiomas attractive.
Collapse
Affiliation(s)
- Brian T Ragel
- Department of Neurosurgery, University of Utah, Salt Lake City, Utah 94132, USA
| | | | | |
Collapse
|
46
|
Pfister C, Ritz R, Pfrommer H, Bornemann A, Tatagiba MS, Roser F. Are there attacking points in the eicosanoid cascade for chemotherapeutic options in benign meningiomas? Neurosurg Focus 2007; 23:E8. [DOI: 10.3171/foc-07/10/e8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Object
The current treatment for recurrent or malignant meningiomas with adjuvant therapies has not been satisfactory, and there is an intense interest in evaluating new molecular markers to act as therapeutic targets. Enzymes of the arachidonic acid (AA) cascade such as cyclooxygenase (COX)–2 or 5-lipoxygenase (5-LO) are upregulated in a number of epithelial tumors, but to date there are hardly any data about the expression of these markers in meningiomas. To find possible targets for chemotherapeutic intervention, the authors evaluated the expression of AA derivatives at different molecular levels in meningiomas.
Methods
One hundred and twenty-four meningioma surgical specimens and normal human cortical tissue samples were immunohistochemically and cytochemically stained for COX-2, COX-1, 5-LO, and prostaglandin E receptor 4 (PTGER4). In addition, Western blot and polymerase chain reaction (PCR) analyses were performed to detect the presence of eicosanoids in vivo and in vitro.
Results
Sixty (63%) of 95 benign meningiomas, 21 (88%) of 24 atypical meningiomas, all five malignant meningiomas, and all normal human cortex samples displayed high COX-2 immunoreactivity. All cultured specimens and IOMM-Lee cells stained positive for COX-2, COX-1, 5-LO, and PTGER4. The PCR analysis demonstrated no changes in eicosanoid expression among meningiomas of different World Health Organization grades and in normal human cortical and dura mater tissue.
Conclusions
Eicosanoid derivatives COX-1, COX-2, 5-LO, and PTGER4 enzymes show a high universal expression in meningiomas but are not upregulated in normal human cortex and dura tissue. This finding of the ubiquitous presence of these enzymes in meningiomas offers an excellent baseline for testing upcoming chemotherapeutic treatments.
Collapse
Affiliation(s)
| | | | | | - Antje Bornemann
- 2the Institute of Brain Research, University of Tübingen, Germany
| | | | | |
Collapse
|