1
|
The Extension of the LeiCNS-PK3.0 Model in Combination with the "Handshake" Approach to Understand Brain Tumor Pathophysiology. Pharm Res 2022; 39:1343-1361. [PMID: 35258766 PMCID: PMC9246813 DOI: 10.1007/s11095-021-03154-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 12/10/2021] [Indexed: 12/22/2022]
Abstract
Micrometastatic brain tumor cells, which cause recurrence of malignant brain tumors, are often protected by the intact blood–brain barrier (BBB). Therefore, it is essential to deliver effective drugs across not only the disrupted blood-tumor barrier (BTB) but also the intact BBB to effectively treat malignant brain tumors. Our aim is to predict pharmacokinetic (PK) profiles in brain tumor regions with the disrupted BTB and the intact BBB to support the successful drug development for malignant brain tumors. LeiCNS-PK3.0, a comprehensive central nervous system (CNS) physiologically based pharmacokinetic (PBPK) model, was extended to incorporate brain tumor compartments. Most pathophysiological parameters of brain tumors were obtained from literature and two missing parameters of the BTB, paracellular pore size and expression level of active transporters, were estimated by fitting existing data, like a “handshake”. Simultaneous predictions were made for PK profiles in extracellular fluids (ECF) of brain tumors and normal-appearing brain and validated on existing data for six small molecule anticancer drugs. The LeiCNS-tumor model predicted ECF PK profiles in brain tumor as well as normal-appearing brain in rat brain tumor models and high-grade glioma patients within twofold error for most data points, in combination with estimated paracellular pore size of the BTB and active efflux clearance at the BTB. Our model demonstrated a potential to predict PK profiles of small molecule drugs in brain tumors, for which quantitative information on pathophysiological alterations is available, and contribute to the efficient and successful drug development for malignant brain tumors.
Collapse
|
2
|
Wu Z, Nakamura M, Krauss JK, Schwabe K, John N. Intracranial rat glioma model for tumor resection and local treatment. J Neurosci Methods 2018; 299:1-7. [PMID: 29425709 DOI: 10.1016/j.jneumeth.2018.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 01/25/2018] [Accepted: 02/02/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND Although tumor resection is among the most important prognostic factors, high grade gliomas regrow in most cases. Also, resection of glial tumors in eloquent brain regions is not or only partially possible. Despite these severe restraints, however, only a few in-vivo models have been established to investigate tumor recurrence and local treatment. Here we characterize the intracranial BT4Ca rat glioma as a model for these aspects. NEW METHOD BT4Ca cells were stereotaxically implanted into the frontal cortex of BDIX rats. Rats were than allocated to (1) a control group, which received no further treatment; (2) a catheter group, where a catheter was implanted for repeated microinjection of vehicle every 3rd day as catheter-control; (3) a resection group, where the tumor was microsurgically removed eight days after cell injection. Postoperatively, survival time, weight and general health condition were scored and the tumor size was histologically assessed. RESULTS Injection of BT4Ca cells induced fast-growing tumors with a mean survival time of 16 days in the control and catheter groups. Resection significantly prolonged survival time whereby the tumor regrew in all rats. Tumor size was similar between all groups. COMPARISON WITH EXISTING METHOD(S) We here present a robust and reliable intracranial rat glioma model, which is suitable to simulate tumor recurrence after surgical resection and local treatment. Importantly, this model does not require advanced imaging or elaborate surgical techniques. CONCLUSIONS The intracranial BT4Ca glioma model appears to be a feasible tool to investigate tumor recurrence after resection and to test local treatment.
Collapse
Affiliation(s)
- Zhiqun Wu
- Department of Neurosurgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Makoto Nakamura
- Department of Neurosurgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Joachim K Krauss
- Department of Neurosurgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Kerstin Schwabe
- Department of Neurosurgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Nadine John
- Department of Neurosurgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| |
Collapse
|
3
|
Wang X, Zhan RY, Wang YY, Yan XI, Cao D, Li Y, Wang YQ, Luo F. Endostatin improves cancer-associated systemic syndrome in a lung cancer model. Oncol Lett 2015; 9:2023-2030. [PMID: 26137006 DOI: 10.3892/ol.2015.3049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 02/18/2015] [Indexed: 02/05/2023] Open
Abstract
Cancer-associated systemic syndrome (CASS) is characterized by a constellation of symptoms, including progressive weight loss, anemia, endocrine disorders, gastrointestinal dysfunction, muscle and adipose atrophy, hepatic peliosis and kidney failure. The present study assesses the effects of endostatin on CASS and any possible underlying mechanism in tumor-bearing mice. The results showed that the inoculation of Lewis lung carcinoma cells into mice led to CASS that was characterized by a notable decrease in body weight, severe anemia phenotype, disordered biochemistry, hepatosplenomegaly, and a marked increase in serum vascular endothelial growth factor (VEGF), tumor necrosis factor α and interleukin-6 (IL-6). The continuous injection of 10 mg/kg/day endostatin suppressed tumor growth and alleviated CASS in the tumor-bearing mice, as shown by weight gain, improvement in biochemistry and anemia, and the preservation of organ function. The effects of endostatin on CASS in the tumor-bearing mice were accompanied by the downregulation of serum VEGF and IL-6. Collectively, these findings indicate that endostatin improves CASS in tumor-bearing mice by decreasing the serum levels of VEGF and IL-6.
Collapse
Affiliation(s)
- Xia Wang
- Department of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Rui-Yu Zhan
- Department of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yu-Yi Wang
- Department of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - X I Yan
- Department of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Dan Cao
- Department of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yan Li
- Department of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yi-Qin Wang
- Department of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Feng Luo
- Department of Medical Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
4
|
Wang K, Wang K, Shen B, Huang T, Sun X, Li W, Jin G, Li L, Bu L, Li R, Wang D, Chen X. MR reporter gene imaging of endostatin expression and therapy. Mol Imaging Biol 2009; 12:520-9. [PMID: 19957205 DOI: 10.1007/s11307-009-0286-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Revised: 06/03/2009] [Accepted: 06/11/2009] [Indexed: 10/20/2022]
Abstract
PURPOSE The aim of this study is to monitor endostatin gene expression and therapy using transferrin receptor (TfR) as reporter gene and transferrin conjugate of ultrasmall supramagnetic iron oxide nanoparticle (Tf-USPIO) as magnetic resonance (MR) reporter probe. PROCEDURE A retroviral plasmid (pLP-LNCX) encoding mouse endostatin and TfR was constructed, and packaged with a titer of 4 × 10(7)colony-forming units per millimeter. MDA-MB-231 breast tumors were established in BALB/c mice by subcutaneous injection of 2 × 10(6) MDA-MB-231 cells. Mice were intratumorally injected with recombinant retrovirus and imaged with MR using Tf-USPIO. Western blot, Prussian blue, and immunohistochemical staining were performed to validate the magnetic resonance imaging results. The antitumor effect of retro-endostatin (ES)-TfR was also evaluated by intratumoral injection of the viral vector. RESULTS The expression of both endostatin and TfR genes in MDA-MB-231 cells after retroviral transfection was confirmed by Western blot and flow cytometry. Tf-USPIO conjugate binds specifically to cells stably transfected with retro-ES-TfR. After intravenous injection of the Tf-USPIO conjugate, there was a more pronounced decrease in T2 relaxation time in tumors treated with retro-ES-TfR than in tumors treated with empty retrovirus retro-LNCX. The expression of ES gene significantly delayed the growth of MDA-MB-231 tumor and reduction of microvessel density and VEGF level as compared to those without viral transfection or transfected with empty retro-LNCX vector. CONCLUSIONS Endostatin therapeutic gene expression was visualized successfully using TfR reporter gene and Tf-USPIO MR reporter probe, which indicates that MR reporter gene imaging may be valuable in gene therapy to evaluate therapeutic gene expression and treatment efficacy.
Collapse
Affiliation(s)
- Kai Wang
- Department of Medical Imaging and Nuclear Medicine, 4th Affiliated Hospital, Harbin Medical University, Harbin, 150001, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Barth RF, Kaur B. Rat brain tumor models in experimental neuro-oncology: the C6, 9L, T9, RG2, F98, BT4C, RT-2 and CNS-1 gliomas. J Neurooncol 2009; 94:299-312. [PMID: 19381449 PMCID: PMC2730996 DOI: 10.1007/s11060-009-9875-7] [Citation(s) in RCA: 309] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2009] [Accepted: 03/16/2009] [Indexed: 02/08/2023]
Abstract
In this review we will describe eight commonly used rat brain tumor models and their application for the development of novel therapeutic and diagnostic modalities. The C6, 9L and T9 gliomas were induced by repeated injections of methylnitrosourea (MNU) to adult rats. The C6 glioma has been used extensively for a variety of studies, but since it arose in an outbred Wistar rat, it is not syngeneic to any inbred strain, and its potential to evoke an alloimmune response is a serious limitation. The 9L gliosarcoma has been used widely and has provided important information relating to brain tumor biology and therapy. The T9 glioma, although not generally recognized, was and probably still is the same as the 9L. Both of these tumors arose in Fischer rats and can be immunogenic in syngeneic hosts, a fact that must be taken into consideration when used in therapy studies, especially if survival is the endpoint. The RG2 and F98 gliomas were both chemically induced by administering ethylnitrosourea (ENU) to pregnant rats, the progeny of which developed brain tumors that subsequently were propagated in vitro and cloned. They are either weakly or non-immunogenic and have an invasive pattern of growth and uniform lethality, which make them particularly attractive models to test new therapeutic modalities. The CNS-1 glioma was induced by administering MNU to a Lewis rat. It has an infiltrative pattern of growth and is weakly immunogenic, which should make it useful in experimental neuro-oncology. Finally, the BT4C glioma was induced by administering ENU to a BD IX rat, following which brain cells were propagated in vitro until a tumorigenic clone was isolated. This tumor has been used for a variety of studies to evaluate new therapeutic modalities. The Avian Sarcoma Virus (ASV) induced tumors, and a continuous cell line derived from one of them designated RT-2, have been useful for studies in which de novo tumor induction is an important requirement. These tumors also are immunogenic and this limits their usefulness for therapy studies. It is essential to recognize the limitations of each of the models that have been described, and depending upon the nature of the study to be conducted, it is important that the appropriate model be selected.
Collapse
Affiliation(s)
- R. F. Barth
- Department of Pathology, The Ohio State University, 165 Hamilton Hall, 1645 Neil Avenue, Columbus, OH 43210, USA, e-mail:
| | - B. Kaur
- Department of Neurological Surgery, Dardinger Laboratory for Neuro-Oncology and Neurosciences, The Ohio State University, 385-D OSUCCC, 410 West 12th Avenue, OSUCCC, Columbus, OH 43210, USA, e-mail:
| |
Collapse
|
6
|
Szentirmai O, Baker CH, Bullain SS, Lin N, Takahashi M, Folkman J, Mulligan RC, Carter BS. Successful inhibition of intracranial human glioblastoma multiforme xenograft growth via systemic adenoviral delivery of soluble endostatin and soluble vascular endothelial growth factor receptor-2: laboratory investigation. J Neurosurg 2008; 108:979-88. [PMID: 18447716 PMCID: PMC4459889 DOI: 10.3171/jns/2008/108/5/0979] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
OBJECT Glioblastoma multiforme (GBM) is characterized by neovascularization, raising the question of whether angiogenic blockade may be a useful therapeutic strategy for this disease. It has been suggested, however, that, to be useful, angiogenic blockade must be persistent and at levels sufficient to overcome proangiogenic signals from tumor cells. In this report, the authors tested the hypothesis that sustained high concentrations of 2 different antiangiogenic proteins, delivered using a systemic gene therapy strategy, could inhibit the growth of established intracranial U87 human GBM xenografts in nude mice. METHODS Mice harboring established U87 intracranial tumors received intravenous injections of adenoviral vectors encoding either the extracellular domain of vascular endothelial growth factor receptor-2-Fc fusion protein (Ad-VEGFR2-Fc) alone, soluble endostatin (Ad-ES) alone, a combination of Ad-VEGFR2-Fc and Ad-ES, or immunoglobulin 1-Fc (Ad-Fc) as a control. RESULTS Three weeks after treatment, magnetic resonance imaging-based determination of tumor volume showed that treatment with Ad-VEGFR2-Fc, Ad-ES, or Ad-VEGFR2-Fc in combination with Ad-ES, produced 69, 59, and 74% growth inhibition, respectively. Bioluminescent monitoring of tumor growth revealed growth inhibition in the same treatment groups to be 62, 74, and 72%, respectively. Staining with proliferating cell nuclear antigen and with terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling showed reduced tumor cell proliferation and increased apoptosis in all antiangiogenic treatment groups. CONCLUSIONS These results suggest that systemic delivery and sustained production of endostatin and soluble VEGFR2 can slow intracranial glial tumor growth by both reducing cell proliferation and increasing tumor apoptosis. This work adds further support to the concept of using antiangiogenesis therapy for intracranial GBM.
Collapse
Affiliation(s)
- Oszkar Szentirmai
- Department of Genetics, Harvard Institutes of Medicine and Harvard Medical School, and Department of Pediatrics, Children's Hospital, Boston, Massachusetts
- Neurosurgical Service, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Cheryl H. Baker
- Department of Genetics, Harvard Institutes of Medicine and Harvard Medical School, and Department of Pediatrics, Children's Hospital, Boston, Massachusetts
| | - Szofia S. Bullain
- Department of Genetics, Harvard Institutes of Medicine and Harvard Medical School, and Department of Pediatrics, Children's Hospital, Boston, Massachusetts
- Neurosurgical Service, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ning Lin
- Neurosurgical Service, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Masaya Takahashi
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Institutes of Medicine, Boston, Massachusetts
| | - Judah Folkman
- Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Richard C. Mulligan
- Department of Genetics, Harvard Institutes of Medicine and Harvard Medical School, and Department of Pediatrics, Children's Hospital, Boston, Massachusetts
| | - Bob S. Carter
- Neurosurgical Service, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
7
|
Xu YF, Zhu LP, Hu B, Rong ZG, Zhu HW, Xu JM, Wu ZW, Wang JJ, Xu GX. A quantitative method for measuring the antitumor potency of recombinant human endostatin in vivo. Eur J Pharmacol 2007; 564:1-6. [PMID: 17346697 DOI: 10.1016/j.ejphar.2007.01.086] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2006] [Revised: 12/11/2006] [Accepted: 01/30/2007] [Indexed: 11/24/2022]
Abstract
Recombinant human endostatin (rhEndostatin) has been shown to inhibit tumor growth, but the variable antitumor activity of different rhEndostatin preparations has necessitated the development of an accurate, reproducible in vivo bioassay for evaluating the rhEndostatin activity. To assess the in vivo antitumor efficacy of rhEndostatin, H22 tumor-bearing mice received three doses of rhEndostatin and the potency of rhEndostatin preparations in inhibiting tumor growth was determined by ED(50)-potency assay and validated by dose-response parallel-line assay. There was a consistent and highly reproducible linear regression relationship between rhEndostatin dosage and tumor growth inhibition rate. The ED(50) values were determined from dose-response regression lines for seven rhEndostatin preparations with high reproducibility. On the basis of the current study, the potency of rhEndostatin preparations was assigned a value of 6.09 x 10(5) U/ampoule and a 95% confidence limit of 5.96 x 10(5)-6.22 x 10(5). We consider that this procedure can be served as a potential candidate pharmacopoeial method for potency measurement of different rhEndostatin preparations.
Collapse
Affiliation(s)
- Ye-Fen Xu
- Department of Biological Science and Technology and State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Mailbox 426, Nanjing University, 22 Hankou Road, Nanjing 210093, China
| | | | | | | | | | | | | | | | | |
Collapse
|