1
|
La Torre D, Della Torre A, Lo Turco E, Longo P, Pugliese D, Lacroce P, Raudino G, Romano A, Lavano A, Tomasello F. Primary Intracranial Gliosarcoma: Is It Really a Variant of Glioblastoma? An Update of the Clinical, Radiological, and Biomolecular Characteristics. J Clin Med 2023; 13:83. [PMID: 38202090 PMCID: PMC10779593 DOI: 10.3390/jcm13010083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/14/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Gliosarcomas (GS) are sporadic malignant tumors classified as a Glioblastoma (GBM) variant with IDH-wild type phenotype. It appears as a well-circumscribed lesion with a biphasic, glial, and metaplastic mesenchymal component. The current knowledge about GS comes from the limited literature. Furthermore, recent studies describe peculiar characteristics of GS, such as hypothesizing that it could be a clinical-pathological entity different from GBM. Here, we review radiological, biomolecular, and clinical data to describe the peculiar characteristics of PGS, treatment options, and outcomes in light of the most recent literature. A comprehensive literature review of PubMed and Web of Science databases was conducted for articles written in English focused on gliosarcoma until 2023. We include relevant data from a few case series and only a single meta-analysis. Recent evidence describes peculiar characteristics of PGS, suggesting that it might be a specific clinical-pathological entity different from GBM. This review facilitates our understanding of this rare malignant brain tumor. However, in the future we recommend multi-center studies and large-scale metanalyses to clarify the biomolecular pathways of PGS to develop new specific therapeutic protocols, different from conventional GBM therapy in light of the new therapeutic opportunities.
Collapse
Affiliation(s)
- Domenico La Torre
- Department of Medical and Surgery Sciences, School of Medicine, AOU “Renato Dulbecco”, University of Catanzaro, 88100 Catanzaro, Italy; (A.D.T.); (P.L.); (P.L.); (A.L.)
| | - Attilio Della Torre
- Department of Medical and Surgery Sciences, School of Medicine, AOU “Renato Dulbecco”, University of Catanzaro, 88100 Catanzaro, Italy; (A.D.T.); (P.L.); (P.L.); (A.L.)
| | - Erica Lo Turco
- Department of Medical and Surgery Sciences, School of Medicine, AOU “Renato Dulbecco”, University of Catanzaro, 88100 Catanzaro, Italy; (A.D.T.); (P.L.); (P.L.); (A.L.)
| | - Prospero Longo
- Department of Medical and Surgery Sciences, School of Medicine, AOU “Renato Dulbecco”, University of Catanzaro, 88100 Catanzaro, Italy; (A.D.T.); (P.L.); (P.L.); (A.L.)
| | - Dorotea Pugliese
- Humanitas, Istituto Clinico Catanese, 95045 Catania, Italy; (D.P.); (G.R.); (A.R.); (F.T.)
| | - Paola Lacroce
- Department of Medical and Surgery Sciences, School of Medicine, AOU “Renato Dulbecco”, University of Catanzaro, 88100 Catanzaro, Italy; (A.D.T.); (P.L.); (P.L.); (A.L.)
| | - Giuseppe Raudino
- Humanitas, Istituto Clinico Catanese, 95045 Catania, Italy; (D.P.); (G.R.); (A.R.); (F.T.)
| | - Alberto Romano
- Humanitas, Istituto Clinico Catanese, 95045 Catania, Italy; (D.P.); (G.R.); (A.R.); (F.T.)
| | - Angelo Lavano
- Department of Medical and Surgery Sciences, School of Medicine, AOU “Renato Dulbecco”, University of Catanzaro, 88100 Catanzaro, Italy; (A.D.T.); (P.L.); (P.L.); (A.L.)
| | - Francesco Tomasello
- Humanitas, Istituto Clinico Catanese, 95045 Catania, Italy; (D.P.); (G.R.); (A.R.); (F.T.)
| |
Collapse
|
2
|
Suda T, Yokoo T, Kanefuji T, Kamimura K, Zhang G, Liu D. Hydrodynamic Delivery: Characteristics, Applications, and Technological Advances. Pharmaceutics 2023; 15:pharmaceutics15041111. [PMID: 37111597 PMCID: PMC10141091 DOI: 10.3390/pharmaceutics15041111] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/20/2023] [Accepted: 03/23/2023] [Indexed: 04/03/2023] Open
Abstract
The principle of hydrodynamic delivery was initially used to develop a method for the delivery of plasmids into mouse hepatocytes through tail vein injection and has been expanded for use in the delivery of various biologically active materials to cells in various organs in a variety of animal species through systemic or local injection, resulting in significant advances in new applications and technological development. The development of regional hydrodynamic delivery directly supports successful gene delivery in large animals, including humans. This review summarizes the fundamentals of hydrodynamic delivery and the progress that has been made in its application. Recent progress in this field offers tantalizing prospects for the development of a new generation of technologies for broader application of hydrodynamic delivery.
Collapse
|
3
|
Li Y, Zhang Z, Huang J, Xing H, Wang L, Huang L, Sui X, Luo Y, Shen L, Wang Y, Yang J. A fast-acting brain-targeted nano-delivery system with ultra-simple structure for brain emergency poisoning rescue. NANOSCALE 2023; 15:4852-4862. [PMID: 36790243 DOI: 10.1039/d2nr05093e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Treatment for acute brain conditions remains a major challenge owing to the unavailability of antidotes, especially for organophosphorus compounds, exposure to which leads to rapid death. Despite recent advances in brain-targeted nano delivery systems (BTNDS), the traditional ones which have been developed will likely not lead to the quick release of an antidote, which is essential to counteract fast neurotoxic effects. Herein, we present a BTNDS using thermosensitive liposomes, without the need for functionalization, to obtain a platform for brain-targeted delivery, which has a simple structure and thus can be easily synthesized and scaled-up. The brain-targeting effect of BTNDS was amplified by phospholipase A2 (PLA2), an inflammatory biomarker. The combination of PLA2 and BTNDS significantly improved brain targeting, leading to an excellent emergency rescue effect - 83- and 4.8-fold better cerebral AChE reactivation response and survival time, respectively. These findings provide a promising strategy to generate a facile, druggable, and effective BTNDS.
Collapse
Affiliation(s)
- Yao Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Institutes of Pharmacology and Toxicology, Beijing, 100850, China.
| | - Zinan Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Institutes of Pharmacology and Toxicology, Beijing, 100850, China.
| | - Jingyi Huang
- State Key Laboratory of Toxicology and Medical Countermeasures, Institutes of Pharmacology and Toxicology, Beijing, 100850, China.
| | - Huanchun Xing
- State Key Laboratory of Toxicology and Medical Countermeasures, Institutes of Pharmacology and Toxicology, Beijing, 100850, China.
| | - Lin Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Institutes of Pharmacology and Toxicology, Beijing, 100850, China.
| | - Lijuan Huang
- State Key Laboratory of Toxicology and Medical Countermeasures, Institutes of Pharmacology and Toxicology, Beijing, 100850, China.
| | - Xin Sui
- State Key Laboratory of Toxicology and Medical Countermeasures, Institutes of Pharmacology and Toxicology, Beijing, 100850, China.
| | - Yuan Luo
- State Key Laboratory of Toxicology and Medical Countermeasures, Institutes of Pharmacology and Toxicology, Beijing, 100850, China.
| | - Liao Shen
- State Key Laboratory of Toxicology and Medical Countermeasures, Institutes of Pharmacology and Toxicology, Beijing, 100850, China.
| | - Yongan Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Institutes of Pharmacology and Toxicology, Beijing, 100850, China.
| | - Jun Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Institutes of Pharmacology and Toxicology, Beijing, 100850, China.
| |
Collapse
|
4
|
Pinkiewicz M, Pinkiewicz M, Walecki J, Zawadzki M. A systematic review on intra-arterial cerebral infusions of chemotherapeutics in the treatment of glioblastoma multiforme: The state-of-the-art. Front Oncol 2022; 12:950167. [PMID: 36212394 PMCID: PMC9539841 DOI: 10.3389/fonc.2022.950167] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 09/02/2022] [Indexed: 12/24/2022] Open
Abstract
Objective To provide a comprehensive review of intra-arterial cerebral infusions of chemotherapeutics in glioblastoma multiforme treatment and discuss potential research aims. We describe technical aspects of the intra-arterial delivery, methods of blood-brain barrier disruption, the role of intraoperative imaging and clinical trials involving intra-arterial cerebral infusions of chemotherapeutics in the treatment of glioblastoma multiforme. Method 159 articles in English were reviewed and used as the foundation for this paper. The Medline/Pubmed, Cochrane databases, Google Scholar, Scielo and PEDro databases have been used to select the most relevant and influential papers on the intra-arterial cerebral infusions of chemotherapeutics in the treatment of glioblastoma multiforme. Additionally, we have included some relevant clinical trials involving intra-arterial delivery of chemotherapeutics to other than GBM brain tumours. Conclusion Considering that conventional treatments for glioblastoma multiforme fall short of providing a significant therapeutic benefit, with a majority of patients relapsing, the neuro-oncological community has considered intra-arterial administration of chemotherapeutics as an alternative to oral or intravenous administration. Numerous studies have proven the safety of IA delivery of chemotherapy and its ability to ensure higher drug concentrations in targeted areas, simultaneously limiting systemic toxicity. Nonetheless, the scarcity of phase III trials prevents any declaration of a therapeutic benefit. Given that the likelihood of a single therapeutic agent which will be effective for the treatment of glioblastoma multiforme is extremely low, it is paramount to establish an adequate multimodal therapy which will have a synergistic effect on the diverse pathogenesis of GBM. Precise quantitative and spatial monitoring is necessary to guarantee the accurate delivery of the therapeutic to the tumour. New and comprehensive pharmacokinetic models, a more elaborate understanding of glioblastoma biology and effective methods of diminishing treatment-related neurotoxicity are paramount for intra-arterial cerebral infusion of chemotherapeutics to become a mainstay treatment for glioblastoma multiforme. Additional use of other imaging methods like MRI guidance during the procedure could have an edge over X-ray alone and aid in selecting proper arteries as well as infusion parameters of chemotherapeutics making the procedure safer and more effective.
Collapse
Affiliation(s)
- Mateusz Pinkiewicz
- Department of Diagnostic Imaging, Mazowiecki Regional Hospital in Siedlce, Siedlce, Poland
| | - Milosz Pinkiewicz
- English Division, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Jerzy Walecki
- Division of Interventional Neuroradiology of the Central Clinical Hospital of the Ministry of Interior and Administration, Department of Radiology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Michał Zawadzki
- Division of Interventional Neuroradiology of the Central Clinical Hospital of the Ministry of Interior and Administration, Department of Radiology, Centre of Postgraduate Medical Education, Warsaw, Poland
- *Correspondence: Michał Zawadzki,
| |
Collapse
|
5
|
Dorrell MI, Kast-Woelbern HR, Botts RT, Bravo SA, Tremblay JR, Giles S, Wada JF, Alexander M, Garcia E, Villegas G, Booth CB, Purington KJ, Everett HM, Siles EN, Wheelock M, Silva JA, Fortin BM, Lowey CA, Hale AL, Kurz TL, Rusing JC, Goral DM, Thompson P, Johnson AM, Elson DJ, Tadros R, Gillette CE, Coopwood C, Rausch AL, Snowbarger JM. A novel method of screening combinations of angiostatics identifies bevacizumab and temsirolimus as synergistic inhibitors of glioma-induced angiogenesis. PLoS One 2021; 16:e0252233. [PMID: 34077449 PMCID: PMC8172048 DOI: 10.1371/journal.pone.0252233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/11/2021] [Indexed: 12/15/2022] Open
Abstract
Tumor angiogenesis is critical for the growth and progression of cancer. As such, angiostasis is a treatment modality for cancer with potential utility for multiple types of cancer and fewer side effects. However, clinical success of angiostatic monotherapies has been moderate, at best, causing angiostatic treatments to lose their early luster. Previous studies demonstrated compensatory mechanisms that drive tumor vascularization despite the use of angiostatic monotherapies, as well as the potential for combination angiostatic therapies to overcome these compensatory mechanisms. We screened clinically approved angiostatics to identify specific combinations that confer potent inhibition of tumor-induced angiogenesis. We used a novel modification of the ex ovo chick chorioallantoic membrane (CAM) model that combined confocal and automated analyses to quantify tumor angiogenesis induced by glioblastoma tumor onplants. This model is advantageous due to its low cost and moderate throughput capabilities, while maintaining complex in vivo cellular interactions that are difficult to replicate in vitro. After screening multiple combinations, we determined that glioblastoma-induced angiogenesis was significantly reduced using a combination of bevacizumab (Avastin®) and temsirolimus (Torisel®) at doses below those where neither monotherapy demonstrated activity. These preliminary results were verified extensively, with this combination therapy effective even at concentrations further reduced 10-fold with a CI value of 2.42E-5, demonstrating high levels of synergy. Thus, combining bevacizumab and temsirolimus has great potential to increase the efficacy of angiostatic therapy and lower required dosing for improved clinical success and reduced side effects in glioblastoma patients.
Collapse
Affiliation(s)
- Michael I. Dorrell
- Department of Biology, Point Loma Nazarene University, San Diego, CA, United States of America
- * E-mail:
| | - Heidi R. Kast-Woelbern
- Department of Biology, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Ryan T. Botts
- Department of Mathematical, Information, and Computer Sciences, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Stephen A. Bravo
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Jacob R. Tremblay
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Sarah Giles
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Jessica F. Wada
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - MaryAnn Alexander
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Eric Garcia
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
- Department of Biology, Dr. Heidi R. Kast-Woelbern’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Gabriel Villegas
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Caylor B. Booth
- Department of Mathematical, Information, and Computer Sciences, Dr. Ryan Bott’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Kaitlyn J. Purington
- Department of Mathematical, Information, and Computer Sciences, Dr. Ryan Bott’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Haylie M. Everett
- Department of Mathematical, Information, and Computer Sciences, Dr. Ryan Bott’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Erik N. Siles
- Department of Mathematical, Information, and Computer Sciences, Dr. Ryan Bott’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Michael Wheelock
- Department of Mathematical, Information, and Computer Sciences, Dr. Ryan Bott’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Jordan A. Silva
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
- Department of Biology, Dr. Heidi R. Kast-Woelbern’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Bridget M. Fortin
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
- Department of Biology, Dr. Heidi R. Kast-Woelbern’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Connor A. Lowey
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
- Department of Biology, Dr. Heidi R. Kast-Woelbern’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Allison L. Hale
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
- Department of Biology, Dr. Heidi R. Kast-Woelbern’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Troy L. Kurz
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Jack C. Rusing
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Dawn M. Goral
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Paul Thompson
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Alec M. Johnson
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Daniel J. Elson
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Roujih Tadros
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
- Department of Biology, Dr. Heidi R. Kast-Woelbern’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Charisa E. Gillette
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
- Department of Biology, Dr. Heidi R. Kast-Woelbern’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Carley Coopwood
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
- Department of Biology, Dr. Heidi R. Kast-Woelbern’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Amy L. Rausch
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
- Department of Biology, Dr. Heidi R. Kast-Woelbern’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| | - Jeffrey M. Snowbarger
- Department of Biology, Dr. Michael Dorrell’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
- Department of Biology, Dr. Heidi R. Kast-Woelbern’s Lab, Point Loma Nazarene University, San Diego, CA, United States of America
| |
Collapse
|
6
|
Huang R, Boltze J, Li S. Strategies for Improved Intra-arterial Treatments Targeting Brain Tumors: a Systematic Review. Front Oncol 2020; 10:1443. [PMID: 32983974 PMCID: PMC7479245 DOI: 10.3389/fonc.2020.01443] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 07/08/2020] [Indexed: 12/16/2022] Open
Abstract
Conventional treatments for brain tumors relying on surgery, radiation, and systemic chemotherapy are often associated with high recurrence and poor prognosis. In recent decades, intra-arterial administration of anti-cancer drugs has been considered a suitable alternative drug delivery route to intravenous and oral administration. Intra-arterial administration is believed to offer increasing drug responses by primary and metastatic brain tumors, and to be associated with better median overall survival. By directly injecting therapeutic agents into carotid or vertebral artery, intra-arterial administration rapidly increases intra-tumoral drug concentration but lowers systemic exposure. However, unexpected vascular or neural toxicity has questioned the therapeutic safety of intra-arterial drug administration and limits its widespread clinical application. Therefore, improving targeting and accuracy of intra-arterial administration has become a major research focus. This systematic review categorizes strategies for optimizing intra-arterial administration into five categories: (1) transient blood-brain barrier (BBB)/blood-tumor barrier (BTB) disruption, (2) regional cerebral hypoperfusion for peritumoral hemodynamic changes, (3) superselective endovascular intervention, (4) high-resolution imaging techniques, and (5) others such as cell and gene therapy. We summarize and discuss both preclinical and clinical research, focusing on advantages and disadvantages of different treatment strategies for a variety of cerebral tumor types.
Collapse
Affiliation(s)
- Rui Huang
- Department of Neurology, Dalian Municipal Central Hospital Affiliated With Dalian Medical University, Dalian, China
| | - Johannes Boltze
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Shen Li
- Department of Neurology, Dalian Municipal Central Hospital Affiliated With Dalian Medical University, Dalian, China
| |
Collapse
|
7
|
Singh SP, Ravoori MK, Dixon KA, Han L, Gupta S, Uthamanthil R, Wright KC, Kundra V. Angiotensin II increases gene expression after selective intra-arterial adenovirus delivery in a rabbit model assessed using in vivo SSTR2-based reporter imaging. EJNMMI Res 2016; 6:25. [PMID: 26983635 PMCID: PMC4794473 DOI: 10.1186/s13550-016-0183-x] [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: 10/30/2015] [Accepted: 03/08/2016] [Indexed: 12/02/2022] Open
Abstract
Background Gene therapy has been hampered by low expression upon in vivo delivery. Using a somatostatin receptor type 2 (SSTR2)-based reporter, we assessed whether angiotensin II (AII) can improve gene expression by adenovirus upon intra-arterial (IA) delivery in a large animal model. Methods A SSTR2-based reporter that can be imaged by a clinically approved radiopharmaceutical was used to assess gene expression. Eight rabbits bearing VX2 tumors in each thigh were randomly injected IA with adenovirus containing a human SSTR2 (Ad-CMV-HA-SSTR2) gene chimera ± AII or control adenovirus containing green fluorescent protein (Ad-CMV-GFP). Three days later, 111In-octreotide was given IV after computed tomography (CT) imaging using a clinical CT scanner and intravenous contrast. Tumor uptake of 111In-octreotide was evaluated the next day using a clinical gamma camera. Gene expression was normalized to tumor weight and morphology from CT to obtain in vivo biodistribution. Results SSTR2-based expression was readily visualized. VX2 tumors infected with Ad-CMV-HA-SSTR2 upon intra-arterial delivery with AII had greater in vivo biodistribution, thus greater gene expression, than those without AII (p < 0.01, n = 6). VX2 tumors infected with Ad-CMV-HA-SSTR2 upon IA delivery had greater biodistribution, thus greater gene expression, than those with the negative control Ad-CMV-GFP (p < 0.02). Similarly, VX2 tumors infected with Ad-CMV-HA-SSTR2 upon IA delivery with AII had greater biodistribution, thus greater gene expression, than those with the negative control Ad-CMV-GFP (p < 0.01). Conclusions Angiotensin II improves in vivo gene expression by adenovirus upon intra-arterial delivery and thus may improve gene therapy efficacy. In vivo SSTR2-based reporter imaging can be used to compare methodologies for improving gene expression.
Collapse
Affiliation(s)
- Sheela P Singh
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Murali K Ravoori
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Katherine A Dixon
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Lin Han
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Sanjay Gupta
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Rajesh Uthamanthil
- Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Kenneth C Wright
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Vikas Kundra
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA. .,Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA. .,UT MD Anderson Cancer Center, 1400 Pressler St., Unit 1473, Houston, TX, 77030, USA.
| |
Collapse
|
8
|
Ananta JS, Paulmurugan R, Massoud TF. Temozolomide-loaded PLGA nanoparticles to treat glioblastoma cells: a biophysical and cell culture evaluation. Neurol Res 2016; 38:51-9. [DOI: 10.1080/01616412.2015.1133025] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
9
|
Poluzzi C, Iozzo RV, Schaefer L. Endostatin and endorepellin: A common route of action for similar angiostatic cancer avengers. Adv Drug Deliv Rev 2016; 97:156-73. [PMID: 26518982 DOI: 10.1016/j.addr.2015.10.012] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 10/15/2015] [Accepted: 10/16/2015] [Indexed: 12/17/2022]
Abstract
Traditional cancer therapy typically targets the tumor proper. However, newly-formed vasculature exerts a major role in cancer development and progression. Autophagy, as a biological mechanism for clearing damaged proteins and oxidative stress products released in the tumor milieu, could help in tumor resolution by rescuing cells undergoing modifications or inducing autophagic-cell death of tumor blood vessels. Cleaved fragments of extracellular matrix proteoglycans are emerging as key players in the modulation of angiogenesis and endothelial cell autophagy. An essential characteristic of cancer progression is the remodeling of the basement membrane and the release of processed forms of its constituents. Endostatin, generated from collagen XVIII, and endorepellin, the C-terminal segment of the large proteoglycan perlecan, possess a dual activity as modifiers of both angiogenesis and endothelial cell autophagy. Manipulation of these endogenously-processed forms, located in the basement membrane within tumors, could represent new therapeutic approaches for cancer eradication.
Collapse
Affiliation(s)
- Chiara Poluzzi
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Renato V Iozzo
- Department of Pathology, Anatomy and Cell Biology, and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Liliana Schaefer
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany.
| |
Collapse
|
10
|
Abstract
Hydrodynamic delivery (HD) is a broadly used procedure for DNA and RNA delivery in rodents, serving as a powerful tool for gene/protein drug discovery, gene function analysis, target validation, and identification of elements in regulating gene expression in vivo. HD involves a pressurized injection of a large volume of solution into a vasculature. New procedures are being developed to satisfy the need for a safe and efficient gene delivery in clinic. Here, we summarize the fundamentals of HD, its applications, and future perspectives for clinical use.
Collapse
Affiliation(s)
- Takeshi Suda
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Dexi Liu
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, School of Pharmacy, Athens, GA, USA
| |
Collapse
|
11
|
Abstract
The key impediment to the successful application of gene therapy in clinics is not the paucity of therapeutic genes. It is rather the lack of nontoxic and efficient strategies to transfer therapeutic genes into target cells. Over the past few decades, considerable progress has been made in gene transfer technologies, and thus far, three different delivery systems have been developed with merits and demerits characterizing each system. Viral and chemical methods of gene transfer utilize specialized carrier to overcome membrane barrier and facilitate gene transfer into cells. Physical methods, on the other hand, utilize various forms of mechanical forces to enforce gene entry into cells. Starting in 1980s, physical methods have been introduced as alternatives to viral and chemical methods to overcome various extra- and intracellular barriers that limit the amount of DNA reaching the intended cells. Accumulating evidence suggests that it is quite feasible to directly translocate genes into cytoplasm or even nuclei of target cells by means of mechanical force, bypassing endocytosis, a common pathway for viral and nonviral vectors. Indeed, several methods have been developed, and the majority of them share the same underlying mechanism of gene transfer, i.e., physically created transient pores in cell membrane through which genes get into cells. Here, we provide an overview of the current status and future research directions in the field of physical methods of gene transfer.
Collapse
|
12
|
Gatson NN, Chiocca EA, Kaur B. Anti-angiogenic gene therapy in the treatment of malignant gliomas. Neurosci Lett 2012; 527:62-70. [PMID: 22906922 DOI: 10.1016/j.neulet.2012.08.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Accepted: 08/03/2012] [Indexed: 11/15/2022]
Abstract
More than four decades ago, Dr. Judah Folkman hypothesized that angiogenesis was a critical process in tumor growth. Since that time, there have been significant advances in understanding tumor biology and groundbreaking research in cancer therapy that have validated his hypothesis. However, in spite of extensive research, glioblastoma multiforme (GBM), the most common and malignant primary brain tumor, has gained little in the way of improved median survival. There have been several angiogenesis targets that have resulted in drugs that are in clinical trials or FDA approved for clinical use in several cancers. GBM is a highly angiogenic tumor and several drugs are showing promise in clinical trials with one (bevacizumab), clinically approved for use. We will review several possible angiogenic targets in GBM as well as the vector methodologies used for delivery. In addition, GBMs present several therapeutic challenges related to structure, tumor immune microenvironment and resistance to angiogenesis. To overcome these challenges will require novel approaches to improve therapeutic gene expression and vector biodistribution in the glioma.
Collapse
Affiliation(s)
- NaTosha N Gatson
- Dardinger Center for Neuro-oncology and Neurosciences, N-1017 Doan Hall, 410 W. 10th Avenue, James Cancer Hospital/Solove Research Institute and The Ohio State University Wexner Medical Center, Columbus, OH 43210-1240, USA
| | | | | |
Collapse
|
13
|
EDL-291, a novel isoquinoline, presents antiglioblastoma effects in vitro and in vivo. Anticancer Drugs 2012; 23:494-504. [DOI: 10.1097/cad.0b013e328351ee4f] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
14
|
Adhim Z, Lin X, Huang W, Morishita N, Nakamura T, Yasui H, Otsuki N, Shigemura K, Fujisawa M, Nibu K, Shirakawa T. E10A, an adenovirus-carrying endostatin gene, dramatically increased the tumor drug concentration of metronomic chemotherapy with low-dose cisplatin in a xenograft mouse model for head and neck squamous-cell carcinoma. Cancer Gene Ther 2012; 19:144-52. [PMID: 22116375 DOI: 10.1038/cgt.2011.79] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Revised: 09/29/2011] [Accepted: 10/23/2011] [Indexed: 12/24/2022]
Abstract
Most cancer chemotherapeutic agents are administered at the maximum-tolerated dose (MTD) in short cycles with treatment breaks. However, MTD-based chemotherapies are often associated with significant toxicity and treatment breaks allow the opportunity for tumor regrowth and acquisition of chemoresistance. To minimize these drawbacks, a metronomic strategy, in which chemotherapeutics are administered at doses significantly below the MTD without treatment breaks, has been suggested by many investigators. The antitumor effect of metronomic chemotherapy may be partially due to inhibition of tumor angiogenesis, and it could be enhanced by a combination therapy, including antiangiogenic agents. In this study, we evaluated the synergistic effect of E10A, an adenovirus carrying the endostatin gene, the most potent inhibitors of tumor angiogenesis, in combination with weekly low-dose cisplatin in a xenograft mouse model for head and neck squamous-cell carcinoma. The E10A induced mRNA and protein expressions of endostatin in H891 cells in vitro. E10A significantly enhanced the in vivo tumor growth inhibitory effect of cisplatin. Immunohistochemical analysis with a TUNEL (terminal deoxynucleotidyl transferase-mediated nick-end labeling) assay and anti-CD31 antibodies revealed that the combination of E10A and cisplatin induced high levels of cell apoptosis and inhibited tumor angiogenesis. Importantly, E10A increased the platinum concentrations in tumors to fivefold higher than that induced by cisplatin alone.
Collapse
Affiliation(s)
- Z Adhim
- Division of Otolaryngology-Head and Neck Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Effect of photodynamic therapy and endostatin on human glioma xenografts in nude mice. Photodiagnosis Photodyn Ther 2011; 8:314-20. [DOI: 10.1016/j.pdpdt.2011.04.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Revised: 04/13/2011] [Accepted: 04/19/2011] [Indexed: 11/17/2022]
|
16
|
van Putten EH, Dirven CM, van den Bent MJ, Lamfers ML. Sitimagene ceradenovec: a gene-based drug for the treatment of operable high-grade glioma. Future Oncol 2011; 6:1691-710. [PMID: 21142657 DOI: 10.2217/fon.10.134] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The field of gene therapy for malignant glioma has made important advances since the first gene transfer studies were performed 20 years ago. Multiple Phase I/II trials and two Phase III trials have been performed and have demonstrated the feasibility and safety of intratumoral vector delivery in the brain. Sitimagene ceradenovec is an adenoviral vector encoding the herpes simplex thymidine kinase gene, developed by Ark Therapeutics Group plc (UK and Finland) for the treatment of patients with operable high-grade glioma. In preclinical and Phase I/II clinical studies, sitimagene ceradenovec exhibited a significant increase in survival. Although the preliminary results of a Phase III clinical study demonstrated a significant positive effect of sitimagene ceradenovec treatment on time to reintervention or death when compared with standard care treatment (hazard ratio: 1.43; 95% CI: 1.06-1.93; p < 0.05), the European Committee for Medicinal Products for Human Use did not consider the data to provide sufficient evidence of clinical benefit. Further clinical evaluation, powered to demonstrate a benefit on a robust end point, is required. This article focuses on sitimagene ceradenovec and provides an overview of the developments in the field of gene therapy for malignant glioma.
Collapse
Affiliation(s)
- Erik Hp van Putten
- Department of Neurosurgery, Erasmus University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands.
| | | | | | | |
Collapse
|
17
|
Hydrodynamic gene delivery and its applications in pharmaceutical research. Pharm Res 2010; 28:694-701. [PMID: 21191634 DOI: 10.1007/s11095-010-0338-9] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Accepted: 11/24/2010] [Indexed: 02/01/2023]
Abstract
Hydrodynamic delivery has emerged as the simplest and most effective method for intracellular delivery of membrane-impermeable substances in rodents. The system employs a physical force generated by a rapid injection of large volume of solution into a blood vessel to enhance the permeability of endothelium and the plasma membrane of the parenchyma cells to allow delivery of substance into cells. The procedure was initially established for gene delivery in mice, and its applications have been extended to the delivery of proteins, oligo nucleotides, genomic DNA and RNA sequences, and small molecules. The focus of this review is on applications of hydrodynamic delivery in pharmaceutical research. Examples are provided to highlight the use of hydrodynamic delivery for study of transcriptional regulation of CYP enzymes, for establishment of animal model for viral infections, and for gene drug discovery and gene function analysis.
Collapse
|
18
|
Tang MM, Zhu QE, Fan WZ, Zhang SL, Li DZ, Liu LZ, Chen M, Zhang M, Zhou J, Wei CJ. Intra-arterial targeted islet-specific expression of Sirt1 protects β cells from streptozotocin-induced apoptosis in mice. Mol Ther 2010; 19:60-6. [PMID: 20842108 DOI: 10.1038/mt.2010.187] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Gene therapy provides a promising approach to curing diabetes. However, an effective route for islet-specific targeting has yet to be established. Toward this end, the pancreatic blood circulation system in Balb/c mice was determined by the injection of rhodamine-containing beads. The efficiency of islet targeting was then measured by the injection of adenoviral vectors carrying a green fluorescence gene via the celiac trunk (C.T.). The results showed that >95% of islets and about 60% of β cells within the pancreatic body and tail could be labeled 3 days after surgery. α-Cell labeling was not as efficient, whereas labeling of nonendocrine tissues was barely detectable. For proof of principle, adenoviral vectors carrying a Sirtuin transgene were injected similarly to test the islet protection effect in the streptozotocin (STZ)-induced type 1 diabetic model. The results demonstrated that overexpression of Sirtuin in STZ-treated mice reduced the level of β-cell death and extent of glucose intolerance. This study reports on efficient islet-specific targeting by using adenoviral injection. This procedure could be invaluable to the treatment of diabetes and the study of islet biology.
Collapse
Affiliation(s)
- Mi-mi Tang
- Multidisciplinary Research Center, Shantou University, Shantou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
Improving the transfection efficiencies of nonviral gene delivery requires properly engineered nanoscaled delivery carriers that can overcome the multiple barriers associated with the delivery of oligonucleotides from the site of administration to the nucleus or cytoplasm of the target cell. This article reviews the current advantages and limitation of polyplex nonviral delivery systems, including the apparent barriers that limit gene expression efficiency compared to physical methods such as hydrodynamic dosing and electroporation. An emphasis is placed on engineered nanoscaled polyplexes (NSPs) of modular design that both self-assemble and systematically disassemble at the desired stage of delivery. It is suggested that NSPs of increasingly sophisticated designs are necessary to improve the efficiency of the rate limiting steps in gene delivery.
Collapse
Affiliation(s)
- Christian A Fernandez
- Division of Pharmaceutics, College of Pharmacy, University of Iowa, Iowa City, Iowa 52242, USA
| | | |
Collapse
|
20
|
Thaker NG, Pollack IF. Molecularly targeted therapies for malignant glioma: rationale for combinatorial strategies. Expert Rev Neurother 2009; 9:1815-36. [PMID: 19951140 PMCID: PMC2819818 DOI: 10.1586/ern.09.116] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Median survival of patients with malignant glioma (MG) from time of diagnosis is approximately 1 year, despite surgery, irradiation and conventional chemotherapy. Improving patient outcome relies on our ability to develop more effective therapies that are directed against the unique molecular aberrations within a patient's tumor. Such molecularly targeted therapies may provide novel treatments that are more effective than conventional chemotherapeutics. Recently developed therapeutic strategies have focused on targeting several core glioma signaling pathways, including pathways mediated by growth-factors, PI3K/Akt/PTEN/mTOR, Ras/Raf/MEK/MAPK and other vital pathways. However, given the molecular diversity, heterogeneity and diverging and converging signaling pathways associated with MG, it is unlikely that any single agent will have efficacy in more than a subset of tumors. Overcoming these therapeutic barriers will require multiple agents that can simultaneously inhibit these processes, providing a rationale for combination therapies. This review summarizes the currently implemented single-agent and combination molecularly targeted therapies for MG.
Collapse
Affiliation(s)
- Nikhil G Thaker
- Doris Duke Clinical Research Fellow, Departments of Neurosurgery, Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15260 and 6 Oakwood Place, Voorhees, NJ 08043, USA Tel.: +1 856 392 4727 Fax: +1 412 692 5921
| | - Ian F Pollack
- Department of Neurosurgery, Children’s Hospital of Pittsburgh, University of Pittsburgh Brain Tumor Center, University of Pittsburgh School of Medicine, Biomedical Science Tower 3, 3501 Fifth Avenue, University of Pittsburgh, Pittsburgh, PA 15213, USA Tel.: +1 412 692 5881 Fax: +1 412 692 5921
| |
Collapse
|
21
|
Tate MC, Aghi MK. Biology of angiogenesis and invasion in glioma. Neurotherapeutics 2009; 6:447-57. [PMID: 19560735 PMCID: PMC5084181 DOI: 10.1016/j.nurt.2009.04.001] [Citation(s) in RCA: 160] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2009] [Revised: 03/30/2009] [Accepted: 04/09/2009] [Indexed: 01/12/2023] Open
Abstract
Treatment of adult brain tumors, in particular glioblastoma, remains a significant clinical challenge, despite modest advances in surgical technique, radiation, and chemotherapeutics. The formation of abnormal, dysfunctional tumor vasculature and glioma cell invasion along white matter tracts are believed to be major components of the inability to treat these tumors effectively. Recent insight into the fundamental processes governing glioma angiogenesis and invasion provide a renewed hope for development of novel strategies aimed at reducing the morbidity of this uniformly fatal disease. In this review, we discuss background biology of the blood brain barrier and its pertinence to blood vessel formation and tumor invasion. We will then focus our attention on the biology of glioma angiogenesis and invasion, and the key mediators of these processes. Last, we will briefly discuss recent and ongoing clinical trials targeting mediators of angiogenesis or invasion in glioma patients. The findings provide a renewed hope for those endeavoring to improve treatment of patients with glioma by providing a novel set of rational targets for translational drug discovery.
Collapse
Affiliation(s)
- Matthew C. Tate
- grid.266102.10000000122976811Department of Neurological Surgery, University of California, 505 Parnassus Avenue, Room M779, 94143-0112 San Francisco, CA
| | - Manish K. Aghi
- grid.266102.10000000122976811Department of Neurological Surgery, University of California, 505 Parnassus Avenue, Room M779, 94143-0112 San Francisco, CA
| |
Collapse
|
22
|
Bai RZ, Wu Y, Liu Q, Xie K, Wei YQ, Wang YS, Liu K, Luo Y, Su JM, Hu B, Liu JY, Li Q, Niu T, Zhao ZW, Yang L. Suppression of lung cancer in murine model: treated by combination of recombinant human endostsatin adenovirus with low-dose cisplatin. J Exp Clin Cancer Res 2009; 28:31. [PMID: 19265510 PMCID: PMC2657125 DOI: 10.1186/1756-9966-28-31] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2008] [Accepted: 03/05/2009] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The sustained growth of tumors necessitates neovascularization. As one of the potent endogenous vascular inhibitors, endostatin has been widely used in antiangiogenesis therapy for tumor. Cisplatin is normally administered in chemotherapy for lung cancer but accompanied with serious side effects. In the current study, we investigated a novel chemo-antiangiogenesis therapeutic strategy to both improve toxic effects on lung cancer cells and reduce damages to normal cells in the anti-tumor therapy. METHODS In vitro, we transduced LLC cells with Ad-hEndo and collected supernatants. Western blotting analysis of the supernatants revealed expression of endostatin. In vivo, to fully investigate the suppression effect on murine lung cancer of the combination therapy, we injected recombinant human endostatin adenovirus intratumorally plus a low dose of cisplatin intraperitoneally routinely. The tumor volume and survival time were observed. Angiogenesis was apparently inhibited within the tumor tissues and on the alginate beads. Assessment of apoptotic cells by the TUNEL assay was conducted in the tumor tissues. RESULTS The combination treatment significantly suppressed the tumor growth and prolonged survival time of the murine LLC tumor model. This anti-tumor activity was associated with decreased microvessel density and increased apoptotic index of tumor cells. CONCLUSION According to the results in this study, recombinant human endostatin adenovirus in combination with a low dose of cisplatin demonstrated apparent synergistic anti-tumor activity without marked toxicity. Thus, these observations may provide a rational alternative for lung cancer treatment.
Collapse
MESH Headings
- Adenoviridae/genetics
- Animals
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Apoptosis/genetics
- Carcinoma, Lewis Lung/blood supply
- Carcinoma, Lewis Lung/drug therapy
- Carcinoma, Lewis Lung/genetics
- Carcinoma, Lewis Lung/therapy
- Cell Line, Tumor
- Cisplatin/pharmacology
- Combined Modality Therapy
- Endostatins/biosynthesis
- Endostatins/genetics
- Female
- Genetic Therapy/methods
- Humans
- Lung Neoplasms/blood supply
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/therapy
- Mice
- Mice, Inbred C57BL
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/therapy
- Random Allocation
- Recombinant Proteins/biosynthesis
- Recombinant Proteins/genetics
- Transduction, Genetic
Collapse
Affiliation(s)
- Rui Z Bai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Keyuan Fourth Road, Chengdu, Sichuan, PR China
| | - Yang Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Keyuan Fourth Road, Chengdu, Sichuan, PR China
| | - Quan Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Keyuan Fourth Road, Chengdu, Sichuan, PR China
| | - Ke Xie
- Department of Oncology, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, Chengdu, 610072, PR China
| | - Yu Q Wei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Keyuan Fourth Road, Chengdu, Sichuan, PR China
| | - Yong S Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Keyuan Fourth Road, Chengdu, Sichuan, PR China
| | - Kang Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Keyuan Fourth Road, Chengdu, Sichuan, PR China
| | - Yan Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Keyuan Fourth Road, Chengdu, Sichuan, PR China
| | - Jing M Su
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Keyuan Fourth Road, Chengdu, Sichuan, PR China
| | - Bing Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Keyuan Fourth Road, Chengdu, Sichuan, PR China
| | - Ji Y Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Keyuan Fourth Road, Chengdu, Sichuan, PR China
| | - Qiu Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Keyuan Fourth Road, Chengdu, Sichuan, PR China
| | - Ting Niu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Keyuan Fourth Road, Chengdu, Sichuan, PR China
| | - Zhi W Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Keyuan Fourth Road, Chengdu, Sichuan, PR China
| | - Li Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Keyuan Fourth Road, Chengdu, Sichuan, PR China
| |
Collapse
|
23
|
Abstract
Antiangiogenesis approaches have the potential to be particularly effective in the treatment of glioblastoma tumours. These tumours exhibit extremely high levels of neovascularisation, which may contribute to their extremely aggressive behaviour, not only by providing oxygenation and nutrition, but also by establishing a leaky vasculature that lacks a blood-brain barrier. This leaky vasculature enables migration of tumour cells, as well as the build up of fluid, which exacerbates tissue damage due to increased intracranial pressure. Here, we discuss the considerable progress that has been made in the identification of the pro- and antiangiogenic factors produced by glioblastoma tumours and the effects of these molecules in animal models of the disease. The safety and efficacy of some of these approaches have now been demonstrated in clinical trials. However, the ability of tumours to overcome these therapies and to re-establish angiogenesis requires further clinical research regarding potential multimodality therapies, as well as basic research into the regulation of angiogenesis by as yet unidentified factors. Optimisation of noninvasive procedures for monitoring of angiogenesis would greatly facilitate such research.
Collapse
|
24
|
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 DOI: 10.3171/jns/2008/108/5/0979] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [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, MA, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
Efficient and safe methods for delivering genetic materials into cells must be developed before the clinical potential of gene therapy can be fully realized. Recently, hydrodynamic gene delivery using a rapid injection of a relatively large volume of DNA solution has opened up a new avenue for gene therapy studies in vivo. This method is superior to the existing delivery systems because of its simplicity, efficiency, and versatility. Wide success in applying hydrodynamic principles to delivery of DNA, RNA, proteins, and synthetic compounds, into the cells in various tissues of small animals, has inspired the recent attempts at establishing a hydrodynamic procedure for clinical use. In this review, we provide an overview of the theory and practice of hydrodynamic gene delivery so as to aid researchers for the use of this method in their pre-clinical and translational gene therapy studies.
Collapse
Affiliation(s)
- Takeshi Suda
- 1Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania 15261, USA
| | | |
Collapse
|
26
|
Honma K, Miyata T, Ochiya T. Type I collagen gene suppresses tumor growth and invasion of malignant human glioma cells. Cancer Cell Int 2007; 7:12. [PMID: 17578585 PMCID: PMC1925056 DOI: 10.1186/1475-2867-7-12] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2007] [Accepted: 06/20/2007] [Indexed: 11/10/2022] Open
Abstract
Background Invasion is a hallmark of a malignant tumor, such as a glioma, and the progression is followed by the interaction of tumor cells with an extracellular matrix (ECM). This study examined the role of type I collagen in the invasion of the malignant human glioma cell line T98G by the introduction of the human collagen type I α1 (HCOL1A1) gene. Results The cells overexpressing HCOL1A1 were in a cluster, whereas the control cells were scattered. Overexpression of HCOL1A1 significantly suppressed the motility and invasion of the tumor cells. The glioma cell growth was markedly inhibited in vitro and in vivo by the overexpression of HCOL1A1; in particular, tumorigenicity completely regressed in nude mice. Furthermore, the HCOL1A1 gene induced apoptosis in glioma cells. Conclusion These results indicate that HCOL1A1 have a suppressive biological function in glioma progression and that the introduction of HCOL1A1 provides the basis of a novel therapeutic approach for the treatment of malignant human glioma.
Collapse
Affiliation(s)
- Kimi Honma
- Section for Studies on Metastasis, National Cancer Center Research Institute, Tokyo, Japan
- Koken Bioscience Institute, Koken Co., Ltd., Tokyo, Japan
| | - Teruo Miyata
- Koken Bioscience Institute, Koken Co., Ltd., Tokyo, Japan
| | - Takahiro Ochiya
- Section for Studies on Metastasis, National Cancer Center Research Institute, Tokyo, Japan
| |
Collapse
|
27
|
Dorrell MI, Aguilar E, Scheppke L, Barnett FH, Friedlander M. Combination angiostatic therapy completely inhibits ocular and tumor angiogenesis. Proc Natl Acad Sci U S A 2007; 104:967-72. [PMID: 17210921 PMCID: PMC1764763 DOI: 10.1073/pnas.0607542104] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2006] [Indexed: 01/06/2023] Open
Abstract
Angiostatic therapies designed to inhibit neovascularization associated with multiple pathological conditions have only been partially successful; complete inhibition has not been achieved. We demonstrate synergistic effects of combining angiostatic molecules that target distinct aspects of the angiogenic process, resulting in the complete inhibition of neovascular growth associated with development, ischemic retinopathy, and tumor growth, with little or no effect on normal, mature tissue vasculature. Tumor vascular obliteration using combination angiostatic therapy was associated with reduced tumor mass and increased survival in a rat 9L gliosarcoma model, whereas individual monotherapies were ineffective. Significant compensatory up-regulation of several proangiogenic factors was observed after treatment with a single angiostatic agent. In contrast, treatment with combination angiostatic therapy significantly reduced compensatory up-regulation. Therapies that combine angiostatic molecules targeting multiple, distinct aspects of the angiogenic process may represent a previously uncharacterized paradigm for the treatment of many devastating diseases with associated pathological neovascularization.
Collapse
Affiliation(s)
- Michael I. Dorrell
- Department of Cell Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037
| | - Edith Aguilar
- Department of Cell Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037
| | - Lea Scheppke
- Department of Cell Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037
| | - Faith H. Barnett
- Department of Cell Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037
| | - Martin Friedlander
- Department of Cell Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037
| |
Collapse
|
28
|
Abstract
Over the last few years, hydrodynamic tail vein delivery has established itself as a simple, yet very effective method for gene transfer into small rodents. Hydrodynamic delivery of plasmid DNA expression vectors or small interfering RNA allows for a broad range of in vivo experiments, including the testing of regulatory elements, antibody generation, evaluation of gene therapy approaches, basic biology and disease model creation (non-heritable transgenics). The recent development of the hydrodynamic limb vein procedure provides a safe nucleic acid delivery technique with equally high efficiency in small and large research animals and, importantly, the prospects for clinical translation.
Collapse
|
29
|
Reardon DA, Wen PY. Therapeutic advances in the treatment of glioblastoma: rationale and potential role of targeted agents. Oncologist 2006; 11:152-64. [PMID: 16476836 DOI: 10.1634/theoncologist.11-2-152] [Citation(s) in RCA: 154] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Despite advances in standard therapy, including surgical resection followed by radiation and chemotherapy, the prognosis for patients with glioblastoma multiforme (GBM) remains poor. Unfortunately, most patients die within 2 years of diagnosis of their disease. Molecular abnormalities vary among individual patients and also within each tumor. Indeed, one of the distinguishing features of GBM is its marked genetic heterogeneity. Nonetheless, recent developments in the field of tumor biology have elucidated signaling pathways and genes involved in the development of GBM, and several novel agents that target these signaling pathways are being developed. As new details on the genetic characteristics of this disease become available, innovative treatment regimens, including a variety of traditional treatment modalities such as surgery, radiation, and cytotoxic chemotherapy, will be combined with newer targeted therapies. This review introduces these new targeted therapies in the context of current treatment options for patients with GBM. It is hoped that this combined approach will overcome the current limitations in the treatment of patients with GBM and result in a better prognosis for these patients.
Collapse
Affiliation(s)
- David A Reardon
- Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina 27710, USA.
| | | |
Collapse
|
30
|
Cabrera G, Porvasnik SL, DiCorleto PE, Siemionow M, Goldman CK. Intra-arterial adenoviral mediated tumor transfection in a novel model of cancer gene therapy. Mol Cancer 2006; 5:32. [PMID: 16899125 PMCID: PMC1560393 DOI: 10.1186/1476-4598-5-32] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2006] [Accepted: 08/09/2006] [Indexed: 11/26/2022] Open
Abstract
Background The aim of the present study was to develop and characterize a novel in vivo cancer gene therapy model in which intra-arterial adenoviral gene delivery can be characterized. In this model, the rat cremaster muscle serves as the site for tumor growth and provides convenient and isolated access to the tumor parenchyma with discrete control of arterial and venous access for delivery of agents. Results Utilizing adenovirus encoding the green fluorescent protein we demonstrated broad tumor transfection. We also observed a dose dependant increment in luciferase activity at the tumor site using an adenovirus encoding the luciferase reporter gene. Finally, we tested the intra-arterial adenovirus dwelling time required to achieve optimal tumor transfection and observed a minimum time of 30 minutes. Conclusion We conclude that adenovirus mediated tumor transfection grown in the cremaster muscle of athymic nude rats via an intra-arterial route could be achieved. This model allows definition of the variables that affect intra-arterial tumor transfection. This particular study suggests that allowing a defined intra-tumor dwelling time by controlling the blood flow of the affected organ during vector infusion can optimize intra-arterial adenoviral delivery.
Collapse
Affiliation(s)
- Gustavo Cabrera
- Gene Therapy Laboratory, National Cancer Institute, Mexico City, Mexico
| | - Stacy L Porvasnik
- Powel Gene Therapy Center, The University of Florida, Gainesville, USA
| | - Paul E DiCorleto
- Department of Cell Biology, Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, USA
| | - Maria Siemionow
- Department of Plastic and Reconstructive Surgery, The Cleveland Clinic Foundation, Cleveland, USA
| | - Corey K Goldman
- Department of Vascular Medicine, Ochsner Clinic Foundation, New Orleans, USA
| |
Collapse
|
31
|
Abstract
Gene therapy is a potentially useful approach in the treatment of human brain tumors, which are notoriously refractory to conventional approaches. Most human clinical trials to date have been unsuccessful in terms of improving patient outcome. Recent improvements in viral vectors, the development of stem cell technology, and increased understanding of the mechanism of action of therapeutic transgenes provide hope that the next generation of gene therapeutics may show increased efficacy in treatment of this devastating disease.
Collapse
Affiliation(s)
- S E Lawler
- Department of Neurological Surgery, The Dardinger Family Laboratory for Neuro-oncology and Neurosciences, The Ohio State University Medical Center, Columbus, 43210, USA
| | | | | |
Collapse
|
32
|
Tsukada S, Parsons CJ, Rippe RA. Mechanisms of liver fibrosis. Clin Chim Acta 2005; 364:33-60. [PMID: 16139830 DOI: 10.1016/j.cca.2005.06.014] [Citation(s) in RCA: 275] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2005] [Revised: 06/07/2005] [Accepted: 06/08/2005] [Indexed: 01/18/2023]
Abstract
Liver fibrosis represents a significant health problem worldwide of which no acceptable therapy exists. The most characteristic feature of liver fibrosis is excess deposition of type I collagen. A great deal of research has been performed to understand the molecular mechanisms responsible for the development of liver fibrosis. The activated hepatic stellate cell (HSC) is the primary cell type responsible for the excess production of collagen. Following a fibrogenic stimulus, HSCs change from a quiescent to an activated, collagen-producing cell. Numerous changes in gene expression are associated with HSC activation including the induction of several intracellular signaling cascades, which help maintain the activated phenotype and control the fibrogenic and proliferative state of the cell. Detailed analyses in understanding the molecular basis of collagen gene regulation have revealed a complex process offering the opportunity for multiple potential therapeutic strategies. However, further research is still needed to gain a better understanding of HSC activation and how this cell maintains its fibrogenic nature. In this review we describe many of the molecular events that occur following HSC activation and collagen gene regulation that contribute to the fibrogenic nature of these cells and provide a review of therapeutic strategies to treat this disease.
Collapse
Affiliation(s)
- Shigeki Tsukada
- Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina at Chapel Hill, NC 27599-7032, USA
| | | | | |
Collapse
|