1
|
Ruiz-Garcia H, Ramirez-Loera C, Malouff TD, Seneviratne DS, Palmer JD, Trifiletti DM. Novel Strategies for Nanoparticle-Based Radiosensitization in Glioblastoma. Int J Mol Sci 2021; 22:9673. [PMID: 34575840 PMCID: PMC8465220 DOI: 10.3390/ijms22189673] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 01/09/2023] Open
Abstract
Radiotherapy (RT) is one of the cornerstones in the current treatment paradigm for glioblastoma (GBM). However, little has changed in the management of GBM since the establishment of the current protocol in 2005, and the prognosis remains grim. Radioresistance is one of the hallmarks for treatment failure, and different therapeutic strategies are aimed at overcoming it. Among these strategies, nanomedicine has advantages over conventional tumor therapeutics, including improvements in drug delivery and enhanced antitumor properties. Radiosensitizing strategies using nanoparticles (NP) are actively under study and hold promise to improve the treatment response. We aim to describe the basis of nanomedicine for GBM treatment, current evidence in radiosensitization efforts using nanoparticles, and novel strategies, such as preoperative radiation, that could be synergized with nanoradiosensitizers.
Collapse
Affiliation(s)
- Henry Ruiz-Garcia
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL 32224, USA; (H.R.-G.); (T.D.M.); (D.S.S.)
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL 32224, USA;
| | | | - Timothy D. Malouff
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL 32224, USA; (H.R.-G.); (T.D.M.); (D.S.S.)
| | - Danushka S. Seneviratne
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL 32224, USA; (H.R.-G.); (T.D.M.); (D.S.S.)
| | - Joshua D. Palmer
- Department of Radiation Oncology, Ohio State University, Columbus, OH 43210, USA;
| | - Daniel M. Trifiletti
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL 32224, USA; (H.R.-G.); (T.D.M.); (D.S.S.)
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL 32224, USA;
| |
Collapse
|
2
|
Spiess BD. Oxygen therapeutic agents to target hypoxia in cancer treatment. Curr Opin Pharmacol 2020; 53:146-151. [PMID: 33086188 DOI: 10.1016/j.coph.2020.09.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/15/2020] [Accepted: 09/15/2020] [Indexed: 12/20/2022]
Abstract
Solid tumors have abnormal microcirculation that limits oxygen delivery and leads to a hypoxic tumor microenvironment. Tumor hypoxia stabilizes the transcription factor HIF-1α that can trigger immunosuppression through A2A adenosine receptors which prevents immune attack on tumors. In addition, success of chemotherapy and radiation therapy appears to be dependent on oxygen levels. Two main pharmaceutical classes of agents (hemoglobin based and perfluorocarbon man-made carbon oils) have been tested in tumor models as enhanced oxygen therapeutics. This article will review how these agents function as well as examine work to date with both drug classes.
Collapse
Affiliation(s)
- Bruce D Spiess
- Department of Anesthesiology, College of Medicine, University of Florida, Gainesville, FL, United States.
| |
Collapse
|
3
|
Yu W, Su X, Zhang D, Qiao F, Wang H, Jiang J, Xu H. Dual-Tracer Assessment of Dynamic Changes in Reoxygenation and Proliferation Decrease During Fractionated Radiotherapy in Murine Tumors. Front Oncol 2020; 10:1046. [PMID: 32766135 PMCID: PMC7379890 DOI: 10.3389/fonc.2020.01046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 05/27/2020] [Indexed: 11/15/2022] Open
Abstract
Objective: The present work aimed to assess reoxygenation and tumor inhibition during fractionated radiotherapy (FRT) in murine tumors using 18F-fluoromisonidazole (18F-FMISO) and 18F-fluorothymidine (18F-FLT) based micro positron emission tomography/computed tomography (PET/CT). Materials and Methods: A nude mouse xenograft model was established with the head and neck squamous carcinoma cell (FaDu), followed by administration of FRT. Imaging was carried out with both 18F-FMISO and 18F-FLT PET/CT, prior to FRT (Pre-FRT, 0 Gy), during FRT (Inter-FRT, 21 Gy), and after FRT (Post-FRT, 40 Gy). The maximum standardized uptake (SUVmax) and tumor-to-normal muscle ratio (TNR) were determined in regions of interest (ROIs) in 18F-FMISO and 18F-FLT PET/CT images. Then, hypoxic (HV) and proliferative tumor (PTV) volumes obtained by PET/CT were analyzed. Immunohistochemistry was performed to analyze the changes of hypoxia-inducible factor- (HIF)-1α, carbonic anhydrase 9 (CAIX), Ki67 and proliferating cell nuclear antigen (PCNA). Associations of the levels of these biomarkers with PET/CT parameters were analyzed. Results:18F-FMISO PET/CT demonstrated markedly elevated reduction rates of SUVmax (30.3 vs. 14.5%, p = 0.012), TNR (27.9 vs. 18.3%, p = 0.032) and HV (85.0 vs. 71.4%, p = 0.047) from Pre-FRT to Inter-FRT compared with values from Inter-FRT to Post-FRT. Meanwhile, PTV reduction rate in 18F-FLT PET/CT from Pre-FRT to Inter-FRT was significantly decreased compared with that from Inter-FRT to Post-FRT (21.2 vs. 82.7%, p = 0.012). Tumor HIF-1α, CAIX, Ki67, and PCNA amounts were continuously down-regulated during radiotherapy. TNR (FMISO) showed significant correlations with HIF-1α (r = 0.692, p = 0.015) and CAIX (r = 0.801, p = 0.006) amounts in xenografts, while associations of SUVmax (FMISO) with hypoxia markers were weak (r = 0.418, p = 0.041 and r = 0.389, p = 0.037, respectively). SUVmax (FLT) was significantly correlated with Ki67 (r = 0.792, p = 0.003) and PCNA (r = 0.837, p = 0.004). Conclusions: Tumor reoxygenation occurs early during radiotherapy, while inhibition of cell proliferation by tumoricidal effects mainly takes place gradually with the course of radiotherapy. 18F-FMISO and 18F-FLT PET/CT are sensitive and non-invasive tools for the monitoring of tumor reoxygenation and proliferation during radiotherapy.
Collapse
Affiliation(s)
- Wenjing Yu
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaoyu Su
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dan Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Feng Qiao
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hui Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jinhui Jiang
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Huiqin Xu
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
4
|
Li R, Zhang J, Guo J, Xu Y, Duan K, Zheng J, Wan H, Yuan Z, Chen H. Application of Nitroimidazole-Carbobane-Modified Phenylalanine Derivatives as Dual-Target Boron Carriers in Boron Neutron Capture Therapy. Mol Pharm 2019; 17:202-211. [PMID: 31763850 DOI: 10.1021/acs.molpharmaceut.9b00898] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Boron neutron capture therapy (BNCT) has received extensive attention as noninvasive cell-level oncotherapy for treating solid cancer tumors. However, boron-containing drugs such as l-boronophenylalanine (BPA) and sodium borocaptate have low boron content and/or poor tumor-targeting ability, limiting their application. In this study, we designed and synthesized a series of nontoxic, dual-target boron carriers (B139, B142, and B151) with the ability to accumulate specifically in tumor cells. We found that the B139 uptake into hypoxic tumor regions was high, with a 70-fold boron content compared to BPA. In addition, in vivo observation showed that B139 can be trapped in tumor cells for a prolonged period and maintains an effective therapeutic concentration, with a peak boron concentration of 50.7 μg/g and a high tumor: blood boron ratio of >3, achieving ideal BNCT conditions. Cytotoxicity evaluation in mice further proved that B139 is safe and reliable. Therefore, B139 has great potential for BNCT application as a dual-target, safe, and efficient boron carrier.
Collapse
Affiliation(s)
- Ruixi Li
- Department of Biomedical Engineering, School of Engineering, State Key Laboratory of Natural Medicines , China Pharmaceutical University , 24 Tongjia Lane , Gulou District, Nanjing 210009 , China
| | - Juanjuan Zhang
- Department of Biomedical Engineering, School of Engineering, State Key Laboratory of Natural Medicines , China Pharmaceutical University , 24 Tongjia Lane , Gulou District, Nanjing 210009 , China
| | - Jingxuan Guo
- Department of Biomedical Engineering, School of Engineering, State Key Laboratory of Natural Medicines , China Pharmaceutical University , 24 Tongjia Lane , Gulou District, Nanjing 210009 , China
| | - Yue Xu
- Department of Biomedical Engineering, School of Engineering, State Key Laboratory of Natural Medicines , China Pharmaceutical University , 24 Tongjia Lane , Gulou District, Nanjing 210009 , China
| | - Kunyuan Duan
- Department of Pharmacy , Qujing Medical College , Qujing 655000 , China
| | - Jinrong Zheng
- Department of Biomedical Engineering, School of Engineering, State Key Laboratory of Natural Medicines , China Pharmaceutical University , 24 Tongjia Lane , Gulou District, Nanjing 210009 , China
| | - Hao Wan
- Department of Biomedical Engineering, School of Engineering, State Key Laboratory of Natural Medicines , China Pharmaceutical University , 24 Tongjia Lane , Gulou District, Nanjing 210009 , China
| | - Zhenwei Yuan
- Department of Biomedical Engineering, School of Engineering, State Key Laboratory of Natural Medicines , China Pharmaceutical University , 24 Tongjia Lane , Gulou District, Nanjing 210009 , China
| | - Haiyan Chen
- Department of Biomedical Engineering, School of Engineering, State Key Laboratory of Natural Medicines , China Pharmaceutical University , 24 Tongjia Lane , Gulou District, Nanjing 210009 , China
| |
Collapse
|
5
|
Yu W, Qiao F, Su X, Zhang D, Wang H, Jiang J, Xu H. 18F-HX4/18F-FMISO-based micro PET for imaging of tumor hypoxia and radiotherapy-associated changes in mice. Biomed Pharmacother 2019; 119:109454. [DOI: 10.1016/j.biopha.2019.109454] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/03/2019] [Accepted: 09/09/2019] [Indexed: 10/26/2022] Open
|
6
|
Luderer MJ, Muz B, Alhallak K, Sun J, Wasden K, Guenthner N, de la Puente P, Federico C, Azab AK. Thermal Sensitive Liposomes Improve Delivery of Boronated Agents for Boron Neutron Capture Therapy. Pharm Res 2019; 36:144. [PMID: 31392417 DOI: 10.1007/s11095-019-2670-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 07/09/2019] [Indexed: 11/26/2022]
Abstract
PURPOSE Boron neutron capture therapy (BNCT) has the potential to become a viable cancer treatment modality, but its clinical translation requires sufficient tumor boron delivery while minimizing nonspecific accumulation. METHODS Thermal sensitive liposomes (TSLs) were designed to have a stable drug payload at physiological temperatures but engineered to have high permeability under mild hyperthermia. RESULTS We found that TSLs improved the tumor-specific delivery of boronophenylalanine (BPA) and boronated 2-nitroimidazole derivative B-381 in D54 glioma cells. Uniquely, the 2-nitroimidazole moiety extended the tumor retention of boron content compared to BPA. CONCLUSION This is the first study to show the delivery of boronated compounds using TSLs for BNCT, and these results will provide the basis of future clinical trials using TSLs for BNCT.
Collapse
Affiliation(s)
- Micah John Luderer
- Department of Radiation Oncology, Cancer Biology Division, Washington University School of Medicine, 4511 Forest Park Ave., Room 3103, St. Louis, MO, 63108, USA
| | - Barbara Muz
- Department of Radiation Oncology, Cancer Biology Division, Washington University School of Medicine, 4511 Forest Park Ave., Room 3103, St. Louis, MO, 63108, USA
| | - Kinan Alhallak
- Department of Radiation Oncology, Cancer Biology Division, Washington University School of Medicine, 4511 Forest Park Ave., Room 3103, St. Louis, MO, 63108, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| | - Jennifer Sun
- Department of Radiation Oncology, Cancer Biology Division, Washington University School of Medicine, 4511 Forest Park Ave., Room 3103, St. Louis, MO, 63108, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| | - Katherine Wasden
- Department of Radiation Oncology, Cancer Biology Division, Washington University School of Medicine, 4511 Forest Park Ave., Room 3103, St. Louis, MO, 63108, USA
| | - Nicole Guenthner
- Department of Radiation Oncology, Cancer Biology Division, Washington University School of Medicine, 4511 Forest Park Ave., Room 3103, St. Louis, MO, 63108, USA
| | - Pilar de la Puente
- Department of Radiation Oncology, Cancer Biology Division, Washington University School of Medicine, 4511 Forest Park Ave., Room 3103, St. Louis, MO, 63108, USA
| | - Cinzia Federico
- Department of Radiation Oncology, Cancer Biology Division, Washington University School of Medicine, 4511 Forest Park Ave., Room 3103, St. Louis, MO, 63108, USA
| | - Abdel Kareem Azab
- Department of Radiation Oncology, Cancer Biology Division, Washington University School of Medicine, 4511 Forest Park Ave., Room 3103, St. Louis, MO, 63108, USA.
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA.
| |
Collapse
|
7
|
Hua L, Wang Z, Zhao L, Mao H, Wang G, Zhang K, Liu X, Wu D, Zheng Y, Lu J, Yu R, Liu H. Hypoxia-responsive lipid-poly-(hypoxic radiosensitized polyprodrug) nanoparticles for glioma chemo- and radiotherapy. Theranostics 2018; 8:5088-5105. [PMID: 30429888 PMCID: PMC6217062 DOI: 10.7150/thno.26225] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 09/19/2018] [Indexed: 02/06/2023] Open
Abstract
Treatment of malignant glioma is a challenge facing cancer therapy. In addition to surgery, and chemotherapy, radiotherapy (RT) is one of the most effective modalities of glioma treatment. However, there are two crucial challenges for RT facing malignant glioma therapy: first, gliomas are known to be resistant to radiation due to their intratumoral hypoxia; second, radiosensitizers may exhibit a lack of target specificity, which may cause a lower concentration of radiosensitizers in tumors and toxic side effects in normal tissues. Thus, novel angiopep-2-lipid-poly-(metronidazoles)n (ALP-(MIs)n) hypoxic radiosensitizer-polyprodrug nanoparticles (NPs) were designed to enhance the radiosensitizing effect on gliomas. Methods: In this study, different degrees and biodegradabilites of hypoxic radiosensitizer MIs-based polyprodrug (P-(MIs)n) were synthesized as a hydrophobic core. P-(MIs)n were mixed with DSPE-PEG2000, angiopep-2-DSPE-PEG2000 and lecithin to self-assemble ALP-(MIs)n through a single-step nanoprecipitation method. The ALP-(MIs)n encapsulate doxorubicin (DOX) (ALP-(MIs)n/DOX) and provoke the release of DOX under hypoxic conditions for glioma chemo- and radiotherapy. In vivo glioma targeting was tested in an orthotopic glioma using live animal fluorescence/bioluminescence imaging. The effect on sensitization to RT of ALP-(MIs)n and the combination of chemotherapy and RT of ALP-(MIs)n/DOX for glioma treatment were also investigated both in vitro and in vivo. Results: ALP-(MIs)n/DOX effectively accumulated in gliomas and could reach the hypoxic glioma site after systemic in vivo administration. These ALP-(MIs)n showed a significant radiosensitizing effect on gliomas and realized combination chemotherapy and RT for glioma treatment both in vitro and in vivo. Conclusions: In summary, we constructed a lipid-poly-(hypoxic radiosensitized polyprodrug) nanoparticles for enhancing the RT sensitivity of gliomas and achieving the combination of radiation and chemotherapy for gliomas.
Collapse
|
8
|
Yasui H, Kawai T, Matsumoto S, Saito K, Devasahayam N, Mitchell JB, Camphausen K, Inanami O, Krishna MC. Quantitative imaging of pO 2 in orthotopic murine gliomas: hypoxia correlates with resistance to radiation. Free Radic Res 2018; 51:861-871. [PMID: 29076398 DOI: 10.1080/10715762.2017.1388506] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Hypoxia is considered one of the microenvironmental factors associated with the malignant nature of glioblastoma. Thus, evaluating intratumoural distribution of hypoxia would be useful for therapeutic planning as well as assessment of its effectiveness during the therapy. Electron paramagnetic resonance imaging (EPRI) is an imaging technique which can generate quantitative maps of oxygen in vivo using the exogenous paramagnetic compound, triarylmethyl and monitoring its line broadening caused by oxygen. In this study, the feasibility of EPRI for assessment of oxygen distribution in the glioblastoma using orthotopic U87 and U251 xenograft model is examined. Heterogeneous distribution of pO2 between 0 and 50 mmHg was observed throughout the tumours except for the normal brain tissue. U251 glioblastoma was more likely to exhibit hypoxia than U87 for comparable tumour size (median pO2; 29.7 and 18.2 mmHg, p = .028, in U87 and U251, respectively). The area with pO2 under 10 mmHg on the EPR oximetry (HF10) showed a good correlation with pimonidazole staining among tumours with evaluated size. In subcutaneous xenograft model, irradiation was relatively less effective for U251 compared with U87. In conclusion, EPRI is a feasible method to evaluate oxygen distribution in the brain tumour.
Collapse
Affiliation(s)
- Hironobu Yasui
- a Central Institute of Isotope Science, Hokkaido University , Sapporo , Japan
| | - Tatsuya Kawai
- b Radiation Oncology Branch , Center for Cancer Research, National Cancer Institute, National Health Institutes , Bethesda , MD , USA
| | - Shingo Matsumoto
- c Division of Bioengineering and Bioinformatics , Graduate School of Information Science and Technology, Hokkaido University , Sapporo , Japan
| | - Keita Saito
- d Radiation Biology Branch , Center for Cancer Research, National Cancer Institute, National Health Institutes , Bethesda , MD , USA
| | - Nallathamby Devasahayam
- d Radiation Biology Branch , Center for Cancer Research, National Cancer Institute, National Health Institutes , Bethesda , MD , USA
| | - James B Mitchell
- d Radiation Biology Branch , Center for Cancer Research, National Cancer Institute, National Health Institutes , Bethesda , MD , USA
| | - Kevin Camphausen
- b Radiation Oncology Branch , Center for Cancer Research, National Cancer Institute, National Health Institutes , Bethesda , MD , USA
| | - Osamu Inanami
- e Laboratory of Radiation Biology, Department of Environmental Veterinary Sciences, Graduate School of Veterinary Medicine , Hokkaido University , Sapporo , Japan
| | - Murali C Krishna
- d Radiation Biology Branch , Center for Cancer Research, National Cancer Institute, National Health Institutes , Bethesda , MD , USA
| |
Collapse
|
9
|
Feldman LA, Fabre MS, Grasso C, Reid D, Broaddus WC, Lanza GM, Spiess BD, Garbow JR, McConnell MJ, Herst PM. Perfluorocarbon emulsions radiosensitise brain tumors in carbogen breathing mice with orthotopic GL261 gliomas. PLoS One 2017; 12:e0184250. [PMID: 28873460 PMCID: PMC5584944 DOI: 10.1371/journal.pone.0184250] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 08/21/2017] [Indexed: 01/02/2023] Open
Abstract
Background Tumour hypoxia limits the effectiveness of radiation therapy. Delivering normobaric or hyperbaric oxygen therapy elevates pO2 in both tumour and normal brain tissue. However, pO2 levels return to baseline within 15 minutes of stopping therapy. Aim To investigate the effect of perfluorocarbon (PFC) emulsions on hypoxia in subcutaneous and intracranial mouse gliomas and their radiosensitising effect in orthotopic gliomas in mice breathing carbogen (95%O2 and 5%CO2). Results PFC emulsions completely abrogated hypoxia in both subcutaneous and intracranial GL261 models and conferred a significant survival advantage orthotopically (Mantel Cox: p = 0.048) in carbogen breathing mice injected intravenously (IV) with PFC emulsions before radiation versus mice receiving radiation alone. Carbogen alone decreased hypoxia levels substantially and conferred a smaller but not statistically significant survival advantage over and above radiation alone. Conclusion IV injections of PFC emulsions followed by 1h carbogen breathing, radiosensitises GL261 intracranial tumors.
Collapse
Affiliation(s)
- Lisa A Feldman
- Department of Neurosurgery, Virginia Commonwealth University, Richmond, VA United States of America.,Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Marie-Sophie Fabre
- School of Biological Sciences, Victoria University, Wellington, New Zealand
| | - Carole Grasso
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Dana Reid
- School of Biological Sciences, Victoria University, Wellington, New Zealand
| | - William C Broaddus
- Department of Neurosurgery, Virginia Commonwealth University, Richmond, VA United States of America
| | - Gregory M Lanza
- Division of Cardiovascular Diseases, Washington University School of Medicine, St. Louis, MO United States of America
| | - Bruce D Spiess
- Department of Anesthesiology, College of Medicine, University of Florida, Gainesville, FL United States of America
| | - Joel R Garbow
- Mallinckrodt Institute, Washington University School of Medicine, St. Louis, MO United States of America
| | - Melanie J McConnell
- Malaghan Institute of Medical Research, Wellington, New Zealand.,School of Biological Sciences, Victoria University, Wellington, New Zealand
| | - Patries M Herst
- Malaghan Institute of Medical Research, Wellington, New Zealand.,Department of Radiation Therapy, University of Otago, Wellington, New Zealand
| |
Collapse
|
10
|
Bohn KA, Adkins CE, Nounou MI, Lockman PR. Inhibition of VEGF and Angiopoietin-2 to Reduce Brain Metastases of Breast Cancer Burden. Front Pharmacol 2017; 8:193. [PMID: 28443023 PMCID: PMC5387068 DOI: 10.3389/fphar.2017.00193] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 03/24/2017] [Indexed: 11/13/2022] Open
Abstract
For metastases in the central nervous system, angiogenesis enhances metastatic potential and promotes progression. Primary factors which drive vessel growth are vascular endothelial growth factor (VEGF) and angiopoietin-2. Preclinical models show inhibition of either factor reduces metastases spread and inhibits growth. This work sets out to answer two questions in a preclinical mouse model. First, whether the combined inhibition of VEGF and angiopoietin-2, reduces passive permeability and limits drug uptake into brain metastases; and second, whether this inhibition reduces metastases burden in brain. We observed combinatorial inhibition of VEGF and angiopoietin-2, decreased (p < 0.05) angiogenesis and vascular branching in an aortic ring assay and decreased (p < 0.05) endothelial wound closure times. Using a brain metastases of breast cancer model (induced by intracardiac injections of brain seeking MDA-MB-231Br cells or 4T1Br cells), we observed, similar to VEGF, angiopoetin-2 expression correlates to increased angiogenesis (p < 0.05) and increased lesion permeability. To determine efficacy, animals were administered bevacizumab plus L1-10 (angiopoietin inhibitor) twice per week until neurological symptoms developed. Lesion permeability significantly decreased by ∼50% (p < 0.05) compared to untreated lesions, but remained ∼25% greater (p < 0.0%) than brain. In subsequent experiments, animals were administered similar regimens but sacrificed on day 32. The number of metastatic lesions developed was significantly (p < 0.001) reduced in the bevacizumab group (56%) and combination group (86%). Lesions’ size was reduced in bevacizumab treated lesions (∼67%) and bevacizumab and L1-10 treated lesions (∼78%) developing area < 0.5 mm2. In summary, combinatorial inhibition of VEGF and angiopoietin reduces lesion permeability and brain metastatic burden.
Collapse
Affiliation(s)
- Kaci A Bohn
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, AmarilloTX, USA.,Department of Pharmaceutical Sciences, College of Pharmacy, Harding University, SearcyAR, USA
| | - Chris E Adkins
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, AmarilloTX, USA.,Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University Health Sciences Center, MorgantownWV, USA.,Department of Pharmaceutical Sciences, South University, SavannahGA, USA
| | - Mohamed I Nounou
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, AmarilloTX, USA.,Department of Pharmaceutical Sciences, Appalachian College of Pharmacy, OakwoodVA, USA.,Department of Pharmaceutics, Faculty of Pharmacy, Alexandria UniversityAlexandria, Egypt.,Department of Pharmaceutical Sciences, School of Pharmacy, University of Saint Joseph, HartfordCT, USA
| | - Paul R Lockman
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, AmarilloTX, USA
| |
Collapse
|
11
|
Bekaert L, Valable S, Lechapt-Zalcman E, Ponte K, Collet S, Constans JM, Levallet G, Bordji K, Petit E, Branger P, Emery E, Manrique A, Barré L, Bernaudin M, Guillamo JS. [18F]-FMISO PET study of hypoxia in gliomas before surgery: correlation with molecular markers of hypoxia and angiogenesis. Eur J Nucl Med Mol Imaging 2017; 44:1383-1392. [PMID: 28315948 DOI: 10.1007/s00259-017-3677-5] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 03/09/2017] [Indexed: 12/12/2022]
Abstract
PURPOSE Hypoxia in gliomas is associated with tumor resistance to radio- and chemotherapy. However, positron emission tomography (PET) imaging of hypoxia remains challenging, and the validation of biological markers is, therefore, of great importance. We investigated the relationship between uptake of the PET hypoxia tracer [18F]-FMISO and other markers of hypoxia and angiogenesis and with patient survival. PATIENTS AND METHODS In this prospective single center clinical study, 33 glioma patients (grade IV: n = 24, III: n = 3, and II: n = 6) underwent [18F]-FMISO PET and MRI including relative cerebral blood volume (rCBV) maps before surgery. Maximum standardized uptake values (SUVmax) and hypoxic volume were calculated, defining two groups of patients based on the presence or absence of [18F]-FMISO uptake. After surgery, molecular quantification of CAIX, VEGF, Ang2 (rt-qPCR), and HIF-1α (immunohistochemistry) were performed on tumor specimens. RESULTS [18F]-FMISO PET uptake was closely linked to tumor grade, with high uptake in glioblastomas (GB, grade IV). Expression of biomarkers of hypoxia (CAIX, HIF-1α), and angiogenesis markers (VEGF, Ang2, rCBV) were significantly higher in the [18F]-FMISO uptake group. We found correlations between the degree of hypoxia (hypoxic volume and SUVmax) and expression of HIF-1α, CAIX, VEGF, Ang2, and rCBV (p < 0.01). Patients without [18F]-FMISO uptake had a longer survival time than uptake positive patients (log-rank, p < 0.005). CONCLUSIONS Tumor hypoxia as evaluated by [18F]-FMISO PET is associated with the expression of hypoxia markers on a molecular level and is related to angiogenesis. [18F]-FMISO uptake is a mark of an aggressive tumor, almost always a glioblastoma. Our results underline that [18F]-FMISO PET could be useful to guide glioma treatment, and in particular radiotherapy, since hypoxia is a well-known factor of resistance.
Collapse
Affiliation(s)
- Lien Bekaert
- Department of Neurology, CHU de Caen, Caen, France. .,Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, 14000, Caen, France. .,Department of Neurosurgery, CHU de Caen, Caen, France. .,Service de Neurochirurgie, CHU de Caen, Avenue de la Côte de Nacre, 14000, Caen, France.
| | - Samuel Valable
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, 14000, Caen, France
| | - Emmanuèle Lechapt-Zalcman
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, 14000, Caen, France.,Department of Pathology, CHU de Caen, Caen, France
| | - Keven Ponte
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, 14000, Caen, France.,Department of Neurosurgery, CHU de Caen, Caen, France
| | - Solène Collet
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, 14000, Caen, France
| | - Jean-Marc Constans
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, 14000, Caen, France.,Department of Neuroradiology, CHU de Caen, Caen, France
| | | | - Karim Bordji
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, 14000, Caen, France
| | - Edwige Petit
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, 14000, Caen, France
| | | | - Evelyne Emery
- Department of Neurosurgery, CHU de Caen, Caen, France
| | - Alain Manrique
- Department of Nuclear Medicine, CHU de Caen, Caen, France
| | - Louisa Barré
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/LDM-TEP group, 14000, Caen, France
| | - Myriam Bernaudin
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, 14000, Caen, France
| | - Jean-Sébastien Guillamo
- Department of Neurology, CHU de Caen, Caen, France. .,Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, 14000, Caen, France. .,Department of Neurology, CHU de Nimes, Place du Professeur Robert Debre, 30029, Nimes cedex 9, France.
| |
Collapse
|
12
|
McConnell ED, Wei HS, Reitz KM, Kang H, Takano T, Vates GE, Nedergaard M. Cerebral microcirculatory failure after subarachnoid hemorrhage is reversed by hyaluronidase. J Cereb Blood Flow Metab 2016; 36:1537-52. [PMID: 26661183 PMCID: PMC5012515 DOI: 10.1177/0271678x15608389] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 06/17/2015] [Indexed: 11/15/2022]
Abstract
Aneurysmal subarachnoid hemorrhage remains one of the more devastating forms of stroke due in large part to delayed cerebral ischemia that appears days to weeks following the initial hemorrhage. Therapies exclusively targeting large caliber arterial vasospasm have fallen short, and thus we asked whether capillary dysfunction contributes to delayed cerebral ischemia after subarachnoid hemorrhage. Using a mouse model of subarachnoid hemorrhage and two-photon microscopy we showed capillary dysfunction unrelated to upstream arterial constriction. Subarachnoid hemorrhage decreased RBC velocity by 30%, decreased capillary pulsatility by 50%, and increased length of non-perfusing capillaries by 15%. This was accompanied by severe brain hypoxia and neuronal loss. Hyaluronidase, an enzyme that alters capillary blood flow by removing the luminal glycocalyx, returned RBC velocity and pulsatility to normal. Hyaluronidase also reversed brain hypoxia and prevented neuron loss typically seen after subarachnoid hemorrhage. Thus, subarachnoid hemorrhage causes specific changes in capillary RBC flow independent of arterial spasm, and hyaluronidase treatment that normalizes capillary blood flow can prevent brain hypoxia and injury after subarachnoid hemorrhage. Prevention or treatment of capillary dysfunction after subarachnoid hemorrhage may reduce the incidence or severity of subarachnoid hemorrhage-induced delayed cerebral ischemia.
Collapse
Affiliation(s)
- Evan D McConnell
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, University of Rochester Medical Center, Rochester, NY, USA Department of Neurological Surgery, University of Rochester Medical Center, Rochester, NY, USA
| | - Helen S Wei
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, University of Rochester Medical Center, Rochester, NY, USA Department of Neurological Surgery, University of Rochester Medical Center, Rochester, NY, USA
| | - Katherine M Reitz
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, University of Rochester Medical Center, Rochester, NY, USA Department of Neurological Surgery, University of Rochester Medical Center, Rochester, NY, USA
| | - Hongyi Kang
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, University of Rochester Medical Center, Rochester, NY, USA Department of Neurological Surgery, University of Rochester Medical Center, Rochester, NY, USA
| | - Takahiro Takano
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, University of Rochester Medical Center, Rochester, NY, USA Department of Neurological Surgery, University of Rochester Medical Center, Rochester, NY, USA
| | - G Edward Vates
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, University of Rochester Medical Center, Rochester, NY, USA Department of Neurological Surgery, University of Rochester Medical Center, Rochester, NY, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, University of Rochester Medical Center, Rochester, NY, USA Department of Neurological Surgery, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
13
|
Parikh U, Williams M, Jacobs A, Pineda JA, Brody DL, Friess SH. Delayed Hypoxemia Following Traumatic Brain Injury Exacerbates White Matter Injury. J Neuropathol Exp Neurol 2016; 75:731-747. [PMID: 27288907 PMCID: PMC7299434 DOI: 10.1093/jnen/nlw045] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Indexed: 12/04/2022] Open
Abstract
Hypoxemia immediately following traumatic brain injury (TBI) has been observed to exacerbate injury. However, it remains unclear whether delayed hypoxemia beyond the immediate postinjury period influences white matter injury. In a retrospective clinical cohort of children aged 4-16 years admitted with severe TBI, 28/74 (35%) patients were found to experience delayed normocarbic hypoxemia within 7 days of admission. Based on these clinical findings, we developed a clinically relevant mouse model of TBI with delayed hypoxemia by exposing 5-week old (adolescent) mice to hypoxic conditions for 30 minutes starting 24 hours after moderate controlled cortical impact (CCI). Injured mice with hypoxemia had increased axonal injury using both β-amyloid precursor protein and NF200 immunostaining in peri-contusional white matter compared with CCI alone. Furthermore, we detected increased peri-contusional white matter tissue hypoxia with pimonidazole and augmented astrogliosis with anti-glial fibrillary acidic protein staining in CCI + delayed hypoxemia compared with CCI alone or sham surgery + delayed hypoxemia. Microglial activation as evidenced by Iba1 staining was not significantly altered by delayed hypoxemia. These clinical and experimental data indicate the prevention or amelioration of delayed hypoxemia effects following TBI may provide a unique opportunity for the development of therapeutic interventions to reduce axonal injury and improve clinical outcomes.
Collapse
Affiliation(s)
- Umang Parikh
- From the Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri (UP, MW, AJ, JAP, SHF)Department of Neurology, Washington University School of Medicine, St. Louis, Missouri (DLB)
| | - Melissa Williams
- From the Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri (UP, MW, AJ, JAP, SHF)Department of Neurology, Washington University School of Medicine, St. Louis, Missouri (DLB)
| | - Addison Jacobs
- From the Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri (UP, MW, AJ, JAP, SHF)Department of Neurology, Washington University School of Medicine, St. Louis, Missouri (DLB)
| | - Jose A Pineda
- From the Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri (UP, MW, AJ, JAP, SHF)Department of Neurology, Washington University School of Medicine, St. Louis, Missouri (DLB)
| | - David L Brody
- From the Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri (UP, MW, AJ, JAP, SHF)Department of Neurology, Washington University School of Medicine, St. Louis, Missouri (DLB)
| | - Stuart H Friess
- From the Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri (UP, MW, AJ, JAP, SHF)Department of Neurology, Washington University School of Medicine, St. Louis, Missouri (DLB).
| |
Collapse
|
14
|
Luderer MJ, Muz B, de la Puente P, Chavalmane S, Kapoor V, Marcelo R, Biswas P, Thotala D, Rogers B, Azab AK. A Hypoxia-Targeted Boron Neutron Capture Therapy Agent for the Treatment of Glioma. Pharm Res 2016; 33:2530-9. [PMID: 27401411 DOI: 10.1007/s11095-016-1977-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 06/20/2016] [Indexed: 10/21/2022]
Abstract
PURPOSE Boron neutron capture therapy (BNCT) has the potential to become a viable cancer treatment modality, but its clinical translation has been limited by the poor tumor selectivity of agents. To address this unmet need, a boronated 2-nitroimidazole derivative (B-381) was synthesized and evaluated for its capability of targeting hypoxic glioma cells. METHODS B-381 has been synthesized from a 1-step reaction. Using D54 and U87 glioma cell lines, the in vitro cytotoxicity and cellular accumulation of B-381 has been evaluated under normoxic and hypoxic conditions compared to L-boronophenylalanine (BPA). Furthermore, tumor retention of B-381 was evaluated in vivo. RESULTS B-381 had low cytotoxicity in normal and cancer cells. Unlike BPA, B-381 illustrated preferential retention in hypoxic glioma cells compared to normoxic glioma cells and normal tissues in vitro. In vivo, B-381 illustrated significantly higher long-term tumor retention compared to BPA, with 9.5-fold and 6.5-fold higher boron levels at 24 and 48 h, respectively. CONCLUSIONS B-381 represents a new class of BNCT agents in which their selectivity to tumors is based on hypoxic tumor metabolism. Further studies are warranted to evaluate B-381 and similar compounds as preclinical candidates for future BNCT clinical trials for the treatment of glioma.
Collapse
Affiliation(s)
- Micah John Luderer
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, 4511 Forest Park Ave., Room 3103, St. Louis, Missouri, 63108, USA
| | - Barbara Muz
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, 4511 Forest Park Ave., Room 3103, St. Louis, Missouri, 63108, USA
| | - Pilar de la Puente
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, 4511 Forest Park Ave., Room 3103, St. Louis, Missouri, 63108, USA
| | - Sanmathi Chavalmane
- Department of Energy, Environmental and Chemical Engineering, Washington University in St. Louis, St. Louis, Missouri, 63130, USA
| | - Vaishali Kapoor
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, 4511 Forest Park Ave., Room 3103, St. Louis, Missouri, 63108, USA
| | - Raymundo Marcelo
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, 4511 Forest Park Ave., Room 3103, St. Louis, Missouri, 63108, USA
| | - Pratim Biswas
- Department of Energy, Environmental and Chemical Engineering, Washington University in St. Louis, St. Louis, Missouri, 63130, USA
| | - Dinesh Thotala
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, 4511 Forest Park Ave., Room 3103, St. Louis, Missouri, 63108, USA
| | - Buck Rogers
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, 4511 Forest Park Ave., Room 3103, St. Louis, Missouri, 63108, USA
| | - Abdel Kareem Azab
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, 4511 Forest Park Ave., Room 3103, St. Louis, Missouri, 63108, USA.
| |
Collapse
|
15
|
Bürgi S, Seuwen A, Keist R, Vom Berg J, Grandjean J, Rudin M. In vivo imaging of hypoxia-inducible factor regulation in a subcutaneous and orthotopic GL261 glioma tumor model using a reporter gene assay. Mol Imaging 2015; 13. [PMID: 25248521 DOI: 10.2310/7290.2014.00029] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Intratumoral hypoxia changes the metabolism of gliomas, leading to a more aggressive phenotype with increased resistance to radio- and chemotherapy. Hypoxia triggers a signaling cascade with hypoxia-inducible factor (HIF) as a key regulator. We monitored activation of the HIF pathway longitudinally in murine glioma tumors. GL261 cells, stably transfected with a luciferase reporter driven under the control of a promoter comprising the HIF target gene motive hypoxia response element, were implanted either subcutaneously or orthotopically. In vivo experiments were carried out using bioluminescence imaging. Tumors were subsequently analyzed using immunofluorescence staining for hypoxia, endothelial cells, tumor perfusion, and glucose transporter expression. Transient upregulation of the HIF signaling was observed in both subcutaneous and orthotopic gliomas. Immunofluorescence staining confirmed hypoxic regions in subcutaneous and, to a lesser extent, intracranial tumors. Subcutaneous tumors showed substantial necrosis, which might contribute to the decreased bioluminescence output observed toward the end of the experiment. Orthotopic tumors were less hypoxic than subcutaneous ones and did not develop extensive necrotic areas. Although this may be the result of the overall smaller size of orthotopic tumors, it might also reflect differences in the local environment, such as the better intrinsic vascularization of brain tissue compared to the subcutaneous tissue compartment.
Collapse
|
16
|
Jensen RL, Mumert ML, Gillespie DL, Kinney AY, Schabel MC, Salzman KL. Preoperative dynamic contrast-enhanced MRI correlates with molecular markers of hypoxia and vascularity in specific areas of intratumoral microenvironment and is predictive of patient outcome. Neuro Oncol 2013; 16:280-91. [PMID: 24305704 DOI: 10.1093/neuonc/not148] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Measures of tumor vascularity and hypoxia have been correlated with glioma grade and outcome. Dynamic contrast-enhanced (DCE) MRI can noninvasively map tumor blood flow, vascularity, and permeability. In this prospective observational cohort pilot study, preoperative imaging was correlated with molecular markers of hypoxia, vascularity, proliferation, and progression-free and overall patient survival. METHODS Pharmacokinetic modeling methods were used to generate maps of tumor blood flow, extraction fraction, permeability-surface area product, transfer constant, washout rate, interstitial volume, blood volume, capillary transit time, and capillary heterogeneity from preoperative DCE-MRI data in human glioma patients. Tissue was obtained from areas of peritumoral edema, active tumor, hypoxic penumbra, and necrotic core and evaluated for vascularity, proliferation, and expression of hypoxia-regulated molecules. DCE-MRI parameter values were correlated with hypoxia-regulated protein expression at tissue sample sites. RESULTS Patient survival correlated with DCE parameters in 2 cases: capillary heterogeneity in active tumor and interstitial volume in areas of peritumoral edema. Statistically significant correlations were observed between several DCE parameters and tissue markers. In addition, MIB-1 index was predictive of overall survival (P = .044) and correlated with vascular endothelial growth factor expression in hypoxic penumbra (r = 0.7933, P = .0071) and peritumoral edema (r = 0.4546). Increased microvessel density correlated with worse patient outcome (P = .026). CONCLUSIONS Our findings suggest that DCE-MRI may facilitate noninvasive preoperative predictions of areas of tumor with increased hypoxia and proliferation. Both imaging and hypoxia biomarkers are predictive of patient outcome. This has the potential to allow unprecedented prognostic decisions and to guide therapies to specific tumor areas.
Collapse
Affiliation(s)
- Randy L Jensen
- Corresponding author: Randy L. Jensen, MD, PhD, Huntsman Cancer Institute and Departments of Neurosurgery, Radiation Oncology, Oncological Sciences, Clinical Neuroscience Center, University of Utah, 175 North Medical Drive, Salt Lake City, Utah 84132.
| | | | | | | | | | | |
Collapse
|
17
|
Pedron S, Harley BAC. Impact of the biophysical features of a 3D gelatin microenvironment on glioblastoma malignancy. J Biomed Mater Res A 2013; 101:3404-15. [DOI: 10.1002/jbm.a.34637] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 01/31/2013] [Accepted: 02/04/2013] [Indexed: 11/06/2022]
Affiliation(s)
- S. Pedron
- Institute for Genomic Biology; University of Illinois at Urbana-Champaign; Urbana IL 61801
| | - B. A. C. Harley
- Institute for Genomic Biology; University of Illinois at Urbana-Champaign; Urbana IL 61801
- Department of Chemical and Biomolecular Engineering; University of Illinois at Urbana-Champaign; Urbana IL 61801
| |
Collapse
|
18
|
Verreault M, Strutt D, Masin D, Anantha M, Yung A, Kozlowski P, Waterhouse D, Bally MB, Yapp DT. Vascular normalization in orthotopic glioblastoma following intravenous treatment with lipid-based nanoparticulate formulations of irinotecan (Irinophore C™), doxorubicin (Caelyx®) or vincristine. BMC Cancer 2011; 11:124. [PMID: 21477311 PMCID: PMC3080346 DOI: 10.1186/1471-2407-11-124] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Accepted: 04/08/2011] [Indexed: 02/21/2023] Open
Abstract
Background Chemotherapy for glioblastoma (GBM) patients is compromised in part by poor perfusion in the tumor. The present study evaluates how treatment with liposomal formulation of irinotecan (Irinophore C™), and other liposomal anticancer drugs, influence the tumor vasculature of GBM models grown either orthotopically or subcutaneously. Methods Liposomal vincristine (2 mg/kg), doxorubicin (Caelyx®; 15 mg/kg) and irinotecan (Irinophore C™; 25 mg/kg) were injected intravenously (i.v.; once weekly for 3 weeks) in Rag2M mice bearing U251MG tumors. Tumor blood vessel function was assessed using the marker Hoechst 33342 and by magnetic resonance imaging-measured changes in vascular permeability/flow (Ktrans). Changes in CD31 staining density, basement membrane integrity, pericyte coverage, blood vessel diameter were also assessed. Results The three liposomal drugs inhibited tumor growth significantly compared to untreated control (p < 0.05-0.001). The effects on the tumor vasculature were determined 7 days following the last drug dose. There was a 2-3 fold increase in the delivery of Hoechst 33342 observed in subcutaneous tumors (p < 0.001). In contrast there was a 5-10 fold lower level of Hoechst 33342 delivery in the orthotopic model (p < 0.01), with the greatest effect observed following treatment with Irinophore C. Following treatment with Irinophore C, there was a significant reduction in Ktrans in the orthotopic tumors (p < 0.05). Conclusion The results are consistent with a partial restoration of the blood-brain barrier following treatment. Further, treatment with the selected liposomal drugs gave rise to blood vessels that were morphologically more mature and a vascular network that was more evenly distributed. Taken together the results suggest that treatment can lead to normalization of GBM blood vessel the structure and function. An in vitro assay designed to assess the effects of extended drug exposure on endothelial cells showed that selective cytotoxic activity against proliferating endothelial cells could explain the effects of liposomal formulations on the angiogenic tumor vasculature.
Collapse
Affiliation(s)
- Maite Verreault
- Experimental Therapeutics, British Columbia Cancer Agency, 675 West 10thAvenue, Vancouver, BC V5Z 1L3, Canada.
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Evans CE, Mattock K, Humphries J, Saha P, Ahmad A, Waltham M, Patel A, Modarai B, Porter L, Premaratne S, Smith A. Techniques of assessing hypoxia at the bench and bedside. Angiogenesis 2011; 14:119-24. [PMID: 21327472 DOI: 10.1007/s10456-011-9205-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Accepted: 02/03/2011] [Indexed: 10/18/2022]
Abstract
Tissues require an adequate supply of oxygen in order to maintain normal cell function. Low oxygen tension (hypoxia) is characteristic of a number of conditions, including cancer, atherosclerosis, rheumatoid arthritis, critical limb ischaemia, peripheral vascular disease, and ischaemic heart disease. Tissue hypoxia is found in tumours, atherosclerotic plaque, and ischaemic myocardium. There is a growing interest in methods to detect and assess hypoxia, given that hypoxia is important in the progression of these diseases. Hypoxia can be assessed at the level of the whole organ, tissue, or cell, using both invasive and non-invasive methods, and by a range of immunohistochemical, biochemical, or imaging techniques. This review describes and critiques current methods of assessing hypoxia that are used at the bench and in clinical practice.
Collapse
Affiliation(s)
- C E Evans
- Academic Department of Surgery, St Thomas' Hospital, King's College London, BHF Centre of Research Excellence and NIHR Biomedical Research Centre at King's Health Partners, London, UK.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Allard E, Huynh NT, Vessières A, Pigeon P, Jaouen G, Benoit JP, Passirani C. Dose effect activity of ferrocifen-loaded lipid nanocapsules on a 9L-glioma model. Int J Pharm 2009; 379:317-23. [PMID: 19467309 DOI: 10.1016/j.ijpharm.2009.05.031] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2009] [Revised: 04/27/2009] [Accepted: 05/18/2009] [Indexed: 11/16/2022]
Abstract
Ferrociphenol (Fc-diOH) is a new molecule belonging to the fast-growing family of organometallic anti-cancer drugs. In a previous study, we showed promising in vivo results obtained after the intratumoural subcutaneous administration of the new drug-carrier system Fc-diOH-LNCs on a 9L-glioma model. To further increase the dose of this lipophilic entity, we have created a series of prodrugs of Fc-diOH. The phenol groups were protected by either an acetyl (Fc-diAc) or by the long fatty-acid chain of a palmitate (Fc-diPal). LNCs loaded with Fc-diOH prodrugs have to be activated in situ by enzymatic hydrolysis. We show here that the protection of diphenol groups with palmitoyl results in the loss of Fc-diOH in vitro activity, probably due to a lack of in situ hydrolysis. On the contrary, protection with an acetate group does not affect the strong, in vitro, antiproliferative effect of ferrocifen-loaded-LNCs neither the reduction of tumour volume observed on an ectopic model, confirming that acetate is easily cleaved by cell hydrolases. Moreover, the cytostatic activity of Fc-diOH-LNCs is confirmed on an orthotopic glioma model since the difference in survival time between the infusion of 0.36 mg/rat Fc-diOH-LNCs and blank LNCs is statistically significant. By using LNCs or Labrafac to carry the drug, a dose-effect ranging from 0.005 to 2.5mg of Fc-diOH per animal can be evidenced.
Collapse
Affiliation(s)
- E Allard
- INSERM, U646, Angers, F-49100 France
| | | | | | | | | | | | | |
Collapse
|
21
|
Homing properties of adipose-derived stem cells to intracerebral glioma and the effects of adenovirus infection. Cancer Lett 2008; 274:78-87. [PMID: 18842332 DOI: 10.1016/j.canlet.2008.08.035] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2008] [Revised: 08/28/2008] [Accepted: 08/29/2008] [Indexed: 01/14/2023]
Abstract
The inevitable clinical recurrence of high grade gliomas after standard treatment is due to the highly diffuse infiltrating parts of these tumors, which remain after surgery and respond poorly to radiation and chemotherapy. It has been proposed to employ the homing capacity of neural stem cells (NSCs) to different types of intracerebral pathology for selective targeting of glioma cells, and delivery of transgenic expressed therapeutics. This approach has been successful in a number of preclinical experimental studies, however, a major drawback for clinical translation has been the limitation of harvesting and ex vivo expansion of NSCs in patients. Here we demonstrate that adipose derived stem cells (ASCs), which are easily harvested in relatively large quantities in humans, display the same tropism for gliomas as NSCs in vitro and in vivo. Both ipsilateral as well as contralateral injection of these cells in brains of glioma-bearing mice, led to extensive homing to the tumor by the ASCs. The potential of loading these cellular vehicles with transgenes was assessed using adenoviral vectors. ASCs could be infected with adenoviral vectors, albeit at very high MOI. Insertion of the arg-gly-asp (RGD) motif into the adenovirus fiber knob, thereby redirecting primary attachment of the virus to integrins, resulted in a striking 7000-fold increase in infection efficiency. However, in vivo migration of adenovirus-infected ASCs was not observed, most likely due to an inflammatory response to these cells which was not observed with control non-infected ASCs. These results indicate that ASCs are an interesting candidate for further development for cell-based therapy of gliomas, however adenoviruses are not appropriate vectors for delivery of transgenes in this context.
Collapse
|
22
|
Thorsen F, Jirak D, Wang J, Sykova E, Bjerkvig R, Enger PØ, van der Kogel A, Hajek M. Two distinct tumor phenotypes isolated from glioblastomas show different MRS characteristics. NMR IN BIOMEDICINE 2008; 21:830-838. [PMID: 18613001 DOI: 10.1002/nbm.1263] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
We have developed a human brain tumor model in immunodeficient rats that gradually changes its phenotype by serial passages in vivo, from a highly infiltrative, non-angiogenic one with numerous stem cell markers [low-generation (LG) tumor] to a more typical glioblastoma one with extensive angiogenesis and necrosis [high-generation (HG) tumor]. In this study we determined the metabolic properties of these two phenotypes, using (1)H MRS. The LG tumors showed an intact blood-brain barrier and normal vascular morphology, as shown by MRI and Hoechst staining. In contrast, the HG tumors exhibited vascular leakage and necrosis. The animals with HG tumor had raised concentrations of choline and myo-inositol, and decreased concentrations of glutamate and N-acetylaspartate. In the LG tumor group, similar changes in metabolic concentrations were detected, although the alterations were more pronounced. The LG tumors also had higher concentrations of choline, taurine, and lactate. Subdividing the LG and HG tumors into large and small tumors revealed a significant increase in choline and decrease in glutamate as the LG tumors increased in size. Our results show that metabolic profiles produced by (1)H MRS can be used to distinguish between two distinct glioblastoma phenotypes. More pronounced anaerobic metabolism was present in the LG stem-cell-like tumors, suggesting a more malignant phenotype.
Collapse
Affiliation(s)
- Frits Thorsen
- Department of Biomedicine, University of Bergen, 5009 Bergen, Norway
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Efficacy of suicide gene therapy in hypoxic rat 9L glioma cells. J Neurooncol 2008; 90:19-24. [PMID: 18594766 DOI: 10.1007/s11060-008-9635-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2007] [Accepted: 06/06/2008] [Indexed: 10/21/2022]
Abstract
Viral vector mediated suicide gene therapy (SGT) involving thymidine kinase (TK) or cytosine deaminase (CD) have considerable promise in the treatment of malignant brain tumors. An unresolved issue is to what extent tumor hypoxia influences the outcome of SGT since brain tumors characterized by regions of hypoxia have potentially reduced cellular metabolism and SGT's cytotoxicity is manifest through cellular metabolism. We studied in vitro and in vivo, the effect of hypoxia on the cytotoxicity of SGT in rat 9L glioma cells. Neither acute nor chronic hypoxia affected the cell killing of SGT by TK or CD. In vivo confirmation that SGT efficacy was not adversely affected by tumor hypoxia using the hypoxic cell marker pimonidazole was shown by the absence of a change in tumor hypoxia by SGT. These studies support the use of SGT utilizing either TK or CD gene strategies even when tumors are characterized by a hypoxic microenvironment.
Collapse
|
24
|
Claes A, Wesseling P, Jeuken J, Maass C, Heerschap A, Leenders WPJ. Antiangiogenic compounds interfere with chemotherapy of brain tumors due to vessel normalization. Mol Cancer Ther 2008; 7:71-8. [PMID: 18187807 DOI: 10.1158/1535-7163.mct-07-0552] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Glioblastomas are highly aggressive primary brain tumors. Curative treatment by surgery and radiotherapy is generally impossible due to the presence of diffusely infiltrating tumor cells. Furthermore, the blood-brain barrier (BBB) in infiltrative tumor areas is largely intact, and this hampers chemotherapy as well. The occurrence of angiogenesis in these tumors makes these tumors attractive candidates for antiangiogenic therapies. Because antiangiogenic compounds have been shown to synergize with chemotherapeutic compounds in other tumor types, based on vessel normalization, there is a tendency toward such combination therapies for primary brain tumors also. However, vessel normalization in brain may result in restoration of the BBB with consequences for the efficacy of chemotherapeutic agents. In this study, we investigated this hypothesis. BALB/c nude mice with intracerebral xenografts of the human glioblastoma lines E98 or U87 were subjected to therapy with different dosages of vandetanib (an angiogenesis inhibitor), temozolomide (a DNA alkylating agent), or a combination (n>8 in each group). Vandetanib selectively inhibited angiogenic growth aspects of glioma and restored the BBB. It did not notably affect diffuse infiltrative growth and survival. Furthermore, vandetanib antagonized the effects of temozolomide presumably by restoration of the BBB and obstruction of chemodistribution to tumor cells. The tumor microenvironment is an extremely important determinant for the response to antiangiogenic therapy. Particularly in brain, antiangiogenic compounds may have adverse effects when combined with chemotherapy. Thus, use of such compounds in neuro-oncology should be reconsidered.
Collapse
Affiliation(s)
- An Claes
- Department of Pathology, Radboud University Nijmegen Medical Centre, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | | | | | | | | | | |
Collapse
|
25
|
Hoogsteen IJ, Marres HAM, van der Kogel AJ, Kaanders JHAM. The hypoxic tumour microenvironment, patient selection and hypoxia-modifying treatments. Clin Oncol (R Coll Radiol) 2007; 19:385-96. [PMID: 17433637 DOI: 10.1016/j.clon.2007.03.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2007] [Accepted: 03/02/2007] [Indexed: 01/18/2023]
Abstract
Tumour hypoxia has been found to be a characteristic feature in many solid tumours. It has been shown to decrease the therapeutic efficacy of radiation treatment, surgery and some forms of chemotherapy. Successful approaches have been developed to counteract this resistance mechanism, although usually at the cost of increased short- and long-term side-effects. New methods for qualitative and quantitative assessment of tumour oxygenation have made it possible to establish the prognostic significance of tumour hypoxia. The ability to determine the degree and extent of hypoxia in solid tumours is not only important prognostically, but also in the selection of patients for hypoxia-modifying treatments. To provide the best attainable quality of life for individual patients it is of increasing importance that tools be developed that allow a better selection of patients for these intensified treatment strategies. Several genes and proteins involved in the response to hypoxia have been identified as potential candidates for future use in predictive assays. Although some markers and combinations have shown potential benefit and are associated with treatment outcome, their clinical usefulness needs to be validated in prospective trials. A review of published studies was carried out, focusing on the assessment of tumour hypoxia, patient selection and the possibilities to overcome hypoxia during treatment.
Collapse
Affiliation(s)
- I J Hoogsteen
- Department of Radiation Oncology, Radboud University, Nijmegen Medical Centre, Nijmegen, The Netherlands.
| | | | | | | |
Collapse
|
26
|
Parmar K, Mauch P, Vergilio JA, Sackstein R, Down JD. Distribution of hematopoietic stem cells in the bone marrow according to regional hypoxia. Proc Natl Acad Sci U S A 2007; 104:5431-6. [PMID: 17374716 PMCID: PMC1838452 DOI: 10.1073/pnas.0701152104] [Citation(s) in RCA: 631] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The interaction of stem cells with their bone marrow microenvironment is a critical process in maintaining normal hematopoiesis. We applied an approach to resolve the spatial organization that underlies these interactions by evaluating the distribution of hematopoietic cell subsets along an in vivo Hoechst 33342 (Ho) dye perfusion gradient. Cells isolated from different bone marrow regions according to Ho fluorescence intensity contained the highest concentration of hematopoietic stem cell (HSC) activity in the lowest end of the Ho gradient (i.e., in the regions reflecting diminished perfusion). Consistent with the ability of Ho perfusion to simulate the level of oxygenation, bone marrow fractions separately enriched for HSCs were found to be the most positive for the binding of the hypoxic marker pimonidazole. Moreover, the in vivo administration of the hypoxic cytotoxic agent tirapazamine exhibited selective toxicity to the primitive stem cell subset. These data collectively indicate that HSCs and the supporting cells of the stem cell niche are predominantly located at the lowest end of an oxygen gradient in the bone marrow with the implication that regionally defined hypoxia plays a fundamental role in regulating stem cell function.
Collapse
Affiliation(s)
- Kalindi Parmar
- *Department of Radiation Oncology, Dana–Farber Cancer Institute
- To whom correspondence may be addressed at:
Department of Radiation Oncology, Jimmy Fund Building, Room 518B, Dana–Farber Cancer Institute, 44 Binney Street, Boston, MA 02115. E-mail:
| | - Peter Mauch
- *Department of Radiation Oncology, Dana–Farber Cancer Institute
- Departments of Radiation Oncology and
- To whom correspondence may be addressed at:
Department of Radiation Oncology, ASB1-L2, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115. E-mail:
| | | | - Robert Sackstein
- Departments of Dermatology and Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115; and
| | | |
Collapse
|
27
|
Jensen RL, Ragel BT, Whang K, Gillespie D. Inhibition of hypoxia inducible factor-1α (HIF-1α) decreases vascular endothelial growth factor (VEGF) secretion and tumor growth in malignant gliomas. J Neurooncol 2006; 78:233-47. [PMID: 16612574 DOI: 10.1007/s11060-005-9103-z] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2005] [Accepted: 12/07/2005] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Hypoxia inducible factor-1alpha (HIF-1alpha) regulates vascular endothelial growth factor (VEGF), the presumed principal mediator of angiogenesis in malignant gliomas, under normal physiologic conditions. We examined the effect of HIF-1alpha on VEGF secretion, tumor growth, and angiogenesis in malignant gliomas. METHODS We examined 175 human gliomas for expression of HIF-1alpha and its downstream-regulated proteins. HIF-1alpha expression and VEGF secretion in glioma cell lines under normoxia and hypoxia were examined using ELISA and Western blot. Malignant glioma cell lines were transfected with dominant-negative HIF-1alpha (DN-HIF-1alpha) expression vector or siRNA constructs against the HIF-1alpha gene. Growth studies were conducted on cells with the highest VEGF/HIF-1alpha inhibition isolated from stable transfected cell lines. MIB-1-labeling index and microvascular density (MVD) measurements were performed on the in vivo tumors. RESULTS HIF-1 expression correlates with malignant glioma phenotype and was not confined to perinecrotic, pseudopalisading cells. VEGF and HIF-1 expression was high in glioma cell lines even under normoxia, and increased after exposure to hypoxia or growth factor stimulation. Cells transfected with DN-HIF-1alpha or HIF-1alpha siRNA demonstrated decreased HIF-1alpha and VEGF secretion. In vivo but not in vitro growth decreased in response to VEGF and HIF-1 inhibition. HIF-1 siRNA studies showed decreased VEGF secretion and in vitro and in vivo growth of glioma cell lines. MVD was unchanged but MIB-1 proliferation index decreased for both types of HIF-1 inhibition. CONCLUSIONS VEGF and HIF-1alpha are elevated in malignant gliomas. HIF-1alpha inhibition results in VEGF secretion inhibition. HIF-1alpha expression affects glioma tumor growth, suggesting clinical applications for malignant glioma treatment.
Collapse
Affiliation(s)
- Randy L Jensen
- Department of Neurosurgery, University of Utah, Salt Lake City, Utah 84132-2303, USA.
| | | | | | | |
Collapse
|
28
|
Gijtenbeek JMM, Wesseling P, Maass C, Burgers L, van der Laak JAWM. Three-dimensional reconstruction of tumor microvasculature: simultaneous visualization of multiple components in paraffin-embedded tissue. Angiogenesis 2005; 8:297-305. [PMID: 16328157 DOI: 10.1007/s10456-005-9019-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2005] [Revised: 09/06/2005] [Accepted: 09/21/2005] [Indexed: 10/25/2022]
Abstract
Three-dimensional (3D) visualization of microscopic structures may provide useful information about the exact 3D configuration, and offers a useful tool to examine the spatial relationship between different components in tissues. A promising field for 3D investigation is the microvascular architecture in normal and pathological tissue, especially because pathological angiogenesis plays a key role in tumor growth and metastasis formation. This paper describes an improved method for 3D reconstruction of microvessels and other microscopic structures in transmitted light microscopy. Serial tissue sections were stained for the endothelial marker CD34 to highlight microvessels and corresponding images were selected and aligned. Alignment of stored images was further improved by automated non-rigid image registration, and automated segmentation of microvessels was performed. Using this technique, 3D reconstructions were produced of the vasculature of the normal brain. Also, to illustrate the complexity of tumor vasculature, 3D reconstructions of two brain tumors were performed: a hemangioblastoma and a glioblastoma multiforme. The possibility of multiple component visualization was shown in a 3D reconstruction of endothelium and pericytes of normal cerebellar cortex and a hemangioblastoma using alternate staining for CD34 and alpha-smooth muscle actin in serial sections, and of a GBM using immunohistochemical double staining. In conclusion, the described 3D reconstruction procedure provides a promising tool for simultaneous visualization of microscopic structures.
Collapse
Affiliation(s)
- J M M Gijtenbeek
- Department of Neurology, Radboud University Nijmegen Medical Centre, The Netherlands.
| | | | | | | | | |
Collapse
|
29
|
van Wijngaarden J, de Rooij K, van Beek E, Bernsen H, Que I, van Hinsbergh VWM, Löwik C. Identification of differentially expressed genes in a renal cell carcinoma tumor model after endostatin-treatment. J Transl Med 2004; 84:1472-83. [PMID: 15273700 DOI: 10.1038/labinvest.3700157] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Endostatin is a cleavage product of collagen XVIII that has shown to inhibit tumor-angiogenesis in experimental tumor models. At present, the exact molecular mechanism of action of endostatin is not completely elucidated. In this study, we wanted to identify specific target genes of endostatin. For this purpose, the human renal cell carcinoma RC-9 was subcutaneously implanted in nude mice and treated with endostatin. Tumor growth was inhibited by endostatin after 4 days of treatment. Using immunohistochemistry and the hypoxia marker pimonidazole, we demonstrate disintegration of blood vessels and hypoxia and anoxia as a result of the treatment. Hereafter, we applied the polymerase chain reaction (PCR)-based subtractive suppression hybridization (SSH) method, together with the mirror orientation selection (MOS) technique to identify specifically induced and suppressed genes after endostatin-treatment. We found eight genes to be specifically induced and 11 to be suppressed by the endostatin-treatment. Among other genes, core binding factor a-1/osteoblast-specific factor-2 (cbfa1/osf2) was found to be specifically suppressed by endostatin. Unexpectedly, cbfa1/osf2 was found to be specifically expressed in granulocytes in the tumor, not only in the experimental RC-9 tumor model, but in sections of human breast cancer as well. Since an effect of antiangiogenic therapy on granulocytes has been reported before, this might lead to new insights in the role of granulocytes in antiangiogenic therapy in general. In conclusion, the SSH-PCR implemented with the MOS-technique is a powerful tool to identify differentially expressed genes. Using these techniques, we have identified several target genes of endostatin, of which cbfa1/osf2 was found to be specifically expressed in granulocytes in the tumor.
Collapse
MESH Headings
- Angiogenesis Inhibitors/therapeutic use
- Animals
- Antineoplastic Agents/therapeutic use
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma, Renal Cell/drug therapy
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/metabolism
- Cell Line, Tumor
- Core Binding Factor Alpha 1 Subunit
- Core Binding Factors
- Endostatins/therapeutic use
- Gene Expression Regulation, Neoplastic/drug effects
- Granulocytes/drug effects
- Granulocytes/metabolism
- Humans
- Kidney Neoplasms/drug therapy
- Kidney Neoplasms/genetics
- Kidney Neoplasms/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Mice, Nude
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Neoplasm Transplantation
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/prevention & control
- Transcription Factors/genetics
- Transcription Factors/metabolism
Collapse
Affiliation(s)
- Jens van Wijngaarden
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
30
|
Kaanders JHAM, Bussink J, van der Kogel AJ. Clinical studies of hypoxia modification in radiotherapy. Semin Radiat Oncol 2004; 14:233-40. [PMID: 15254866 DOI: 10.1016/j.semradonc.2004.04.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Hypoxic modification has been the subject of investigations in clinical radiation oncology since the early 60s. To date, this has not yet resulted in a treatment that has been widely accepted. Logistics and technical difficulties limit the routine use of hyperbaric oxygen in radiotherapy. The nitroimidazoles have not gained general acceptance, initially because of their toxicity and later because of doubts about the effectiveness of the newer generation of less toxic drugs. Nevertheless, there is good evidence from these studies that improving clinical outcome by hypoxic modulation is an achievable goal. Newer approaches including combinations of radiotherapy with tirapazamine, erythropoietin, and carbogen and nicotinamide (ARCON) are currently in phase III trial. For these new strategies to be successful, it is important that the proper patient categories are selected. Various methods to assess tumor oxygenation are now becoming available in the clinic. These potential predictive assays must be incorporated and validated in current and future large-scale clinical trials. Modifiers that target other aspects of tumor biology may also have indirect effects on tumor oxygenation. These aspects require further study in preclinical and early clinical settings.
Collapse
|
31
|
van Bree C, Franken NAP, Rodermond HM, Stalpers LJA, Haveman J. Repair of Potentially Lethal Damage does not Depend on Functional TP53 in Human Glioblastoma Cells. Radiat Res 2004; 161:511-6. [PMID: 15161373 DOI: 10.1667/3154] [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: 11/03/2022]
Abstract
The functionality of G(1)-phase arrest was investigated in relation to repair of potentially lethal damage (PLD) in human glioblastoma Gli-06 cells. Confluent cultures were irradiated and plated for clonogenic survival either immediately or 24 h after gamma irradiation. Bivariate flow cytometry was performed to assess the distribution over the cell cycle. Levels of TP53 and CDKN1A protein were assessed with Western blotting and levels of CDKN1A mRNA with RT-PCR. Confluence significantly reduced the number of proliferating cells. Marked PLD repair was found in the absence of an intact G(1) arrest. No accumulation of TP53 was observed, and the protein was smaller than the wild-type TP53 of RKO cells. No increased expression of CDKN1A at the mRNA or protein levels was found in Gli-06 cells. The TP53 of Gli-06 cells was unable to transactivate the CDKN1A gene. From this study, it is evident that PLD repair may be present without a functional TP53 or G(1) arrest.
Collapse
Affiliation(s)
- C van Bree
- Academic Medical Center, University of Amsterdam, Department of Radiotherapy, 1100 DE Amsterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
32
|
Abstract
Two mechanisms of radiotherapy resistance which are of major importance in various tumour types are tumour-cell repopulation and hypoxia. ARCON (accelerated radiotherapy with carbogen and nicotinamide) is a new therapeutic strategy that combines radiation treatment modifications, with the aim of counteracting these resistance mechanisms. To limit clonogenic repopulation during therapy, the overall duration of the radiotherapy is reduced, generally by delivering several fractions per day. This accelerated radiotherapy is combined with inhalation of hyperoxic gas to decrease diffusion-limited hypoxia, and nicotinamide, a vasoactive agent, to decrease perfusion-limited hypoxia. Preclinical studies have been done to test the enhancing effects of these three components of ARCON, individually and in combination, in several experimentally induced tumours and normal tissues. In a mouse mammary carcinoma, the tumour-control rate obtained with ARCON was the same as that with conventional treatment, but with a radiation dose almost 50% lower. Phase 1 and 2 clinical trials have shown the feasibility and tolerability of ARCON, and have produced promising results in terms of tumour control. In particular in cancers of the head and neck and bladder, the local tumour-control rates are higher than in other studies, and phase 3 trials for these tumour types are underway. In conjunction with these trials, hypoxia markers detectable by immunohistochemistry are being tested for their potential use in predictive assays to select patients for ARCON and other hypoxia-modifying therapies.
Collapse
Affiliation(s)
- Johannes H A M Kaanders
- Department of Radiation Ocology of the University Medical Centre Nijmegen, Nijmegen, Netherlands.
| | | | | |
Collapse
|
33
|
Abstract
Assessment of the oxygenation status of brain tumors has been studied increasingly with imaging techniques in light of recent advances in oncology. Tumor oxygen tension is a critical factor influencing the effectiveness of radiation and chemotherapy and malignant progression. Hypoxic tumors are resistant to treatment, and prognostic value of tumor oxygen status is shown in head and neck tumors. Strategies increasing the tumor oxygenation are being investigated to overcome the compromising [figure: see text] effect of hypoxia on tumor treatment. Administration of nicotinamide and inhalation of various high oxygen concentrations have been implemented. Existing methods for assessment of tissue oxygen level are either invasive or insufficient. Accurate and noninvasive means to measure tumor oxygenation are needed for treatment planning, identification of patients who might benefit from oxygenation strategies, and assessing the efficacy of interventions aimed to increase the radiosensitivity of tumors. Of the various imaging techniques used to assess tissue oxygenation, MR spectroscopy and MR imaging are widely available, noninvasive, and clinically applicable techniques. Tumor hypoxia is related closely to insufficient blood flow through chaotic and partially nonfunctional tumor vasculature and the distance between the capillaries and the tumor cells. Information on characteristics of tumor vasculature such as blood volume, perfusion, and increased capillary permeability can be provided with MR imaging. MR imaging techniques can provide a measure of capillary permeability based on contrast enhancement and relative cerebral blood volume estimates using dynamic susceptibility MR imaging. Blood oxygen level dependent contrast MR imaging using gradient echo sequence is intrinsically sensitive to changes in blood oxygen level. Animal models using blood oxygen level-dependent contrast imaging reveal the different responses of normal and tumor vasculature under hyperoxia. Normobaric hyperoxia is used in MR studies as a method to produce MR contrast in tissues. Increased T2* signal intensity of brain tissue has been observed using blood oxygen level-dependent contrast MR imaging. Dynamic blood oxygen level-dependent contrast MR imaging during hyperoxia is suggested to image tumor oxygenation. Quantification of cerebral oxygen saturation using blood oxygen level-dependent MR imaging also has been reported. Quantification of cerebral blood oxygen saturation using MR imaging has promising clinical applications; however, technical difficulties have to be resolved. Blood oxygen level dependent MR imaging is an emerging technique to evaluate the cerebral blood oxygen saturation, and it has the potential and versatility to assess oxygenation status of brain tumors. Upon improvement and validation of current MR techniques, better diagnostic, prognostic, and treatment monitoring capabilities can be provided for patients with brain tumors.
Collapse
Affiliation(s)
- F Zerrin Yetkin
- Division of Neuroradiology, Department of Radiology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390-8896, USA.
| | | |
Collapse
|
34
|
Lapidot T, Petit I. Current understanding of stem cell mobilization: the roles of chemokines, proteolytic enzymes, adhesion molecules, cytokines, and stromal cells. Exp Hematol 2002; 30:973-81. [PMID: 12225788 DOI: 10.1016/s0301-472x(02)00883-4] [Citation(s) in RCA: 568] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Mobilization of hematopoietic stem and progenitor cells from the bone marrow into the circulation by repetitive, daily stimulations with G-CSF alone, or in combination with cyclophosphamide, is increasingly used clinically; however, the mechanism is not fully understood. Moreover, following mobilization stem cells also home back to the bone marrow, suggesting that stem cell release/mobilization and homing are sequential events with physiological roles. Previously, a role for cytokines such as G-CSF and SCF, and adhesion molecules such as VLA-4 and P/E selectins, was determined for stem cell mobilization. Recent results using experimental animal models and samples from clinical mobilization protocols demonstrate major involvement of chemokines such as stromal derived factor-1 (SDF-1) and IL-8, as well as proteolytic enzymes such as elastase, cathepsin G, and various MMPs in the mobilization process. These results will be reviewed together with the central roles of SDF-1 and CXCR4 interactions in G-CSF or G-CSF in combination with cyclophosphamide-induced mobilization. Furthermore, the central role of this chemokine in stem cell homing to the bone marrow as well as retention of undifferentiated cells within this tissue will also be discussed.
Collapse
Affiliation(s)
- Tsvee Lapidot
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel.
| | | |
Collapse
|
35
|
Oldham M, Sapareto SA, Li XA, Allen J, Sutlief S, Wong OC, Wong JW. Practical aspects of in situ 16O (gamma,n) 15O activation using a conventional medical accelerator for the purpose of perfusion imaging. Med Phys 2001; 28:1669-78. [PMID: 11548936 DOI: 10.1118/1.1386777] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
We report investigations into the feasibility of generating radioactive oxygen (15O, a positron emitter, with half-life 2.05 min) using a tuned Elekta SL25 accelerator, for the end purpose of imaging tumor perfusion. 15O is produced by the "gamma, neutron," (gamma,n) reaction between high-energy photons and normal oxygen (16O) in the body. As most in vivo 16O is bound in water molecules the 15O radio-marker is produced in proportion to water content in tissue. Imaging the washout of the 15O distribution using sensitive positron-emission-tomography (PET) technology can yield spatial information about blood perfusion in the tissue. The aim of this article was to determine the amount of 15O activity that could be produced by the tuned medical accelerator. A further aim was to model the activation process using Monte Carlo and to investigate ways to optimize the amount of 15O that could be generated. Increased activation was achieved by (i) tuning the beam to give higher-energy electrons incident on the target of the accelerator, (ii) increasing dose rate by removing the conventional filtration in the beam and reducing the source to object distance, and (iii) reducing low-energy photons by means of a carbon block absorber. The activity per-unit-dose produced by the tuned beam was measured by irradiating spheres of water to known doses and placing the spheres in a calibrated coincidence-counting apparatus. Peak energy of the tuned bremsstrahlung beam was estimated at 29 MeV, and generated activity up to 0.24/microCi/cc/3Gy in water. The measured amount of 15O agreed to within 10% of the prediction from the Monte-Carlo-computed spectrum, indicating reasonable ability to model the activation process. The optimal thickness of the carbon absorber was found to be about 25 cm. The insertion of a carbon absorber improved spectral quality for activation purposes but at the cost of reduced dose rate. In conclusion, the viability of generating 15O with an Elekta SL25 has been demonstrated. In conjunction with recent advances in high-sensitivity portable PET imaging devices, real potential exists for imaging in situ activated 15O washout as a surrogate measurement of macroscopic tumor perfusion.
Collapse
Affiliation(s)
- M Oldham
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, Michigan 48073, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Rijken PF, Bernsen HJ, Peters JP, Hodgkiss RJ, Raleigh JA, van der Kogel AJ. Spatial relationship between hypoxia and the (perfused) vascular network in a human glioma xenograft: a quantitative multi-parameter analysis. Int J Radiat Oncol Biol Phys 2000; 48:571-82. [PMID: 10974478 DOI: 10.1016/s0360-3016(00)00686-6] [Citation(s) in RCA: 98] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
PURPOSE To quantitatively study the spatial distribution of tumor hypoxia in relation to the perfused vasculature. METHODS AND MATERIALS Using a human glioma xenograft model, nude mice were administered two different hypoxia markers (NITP or pimonidazole) and the perfusion marker Hoechst 33342. Frozen tumor sections were sequentially scanned for perfusion, hypoxia, and vasculature, respectively, to quantitate perfusion, vasculature, and hypoxia parameters in the same section. RESULTS All tumors showed incomplete perfusion. Both NITP and pimonidazole stained the same hypoxic tumor areas. No statistically significant differences between the two markers were observed. The density of the perfused vessels was inversely related to the hypoxic fraction. At critical distances from perfused vessels, hypoxia occurred. These data suggest that predominantly diffusion-limited hypoxia was detected, based on the spatial distribution of nearby vessels. Also, the proportion of hypoxia distributed over arbitrary zones of 50 microm around perfused vessels was calculated. The largest proportion of hypoxia was found at distances beyond 100 microm from perfused vessels. CONCLUSION With the multiple staining and functional microscopic imaging technique described here, the spatial relationship between perfused vessels and hypoxia was quantified in whole tumor cross-sections. The usefulness of this histologically-based method to quantitate morphological and physiological aspects of the tumor microenvironment was evaluated.
Collapse
Affiliation(s)
- P F Rijken
- Department of Radiotherapy, University of Nijmegen, Nijmegen, The Netherlands.
| | | | | | | | | | | |
Collapse
|