1
|
Mali A, Bhanwase A. In vivo pharmacokinetic study of carmustine in rats after giving single-dose of carmustine API solution, flexible liposomes, in situ nasal gel, optimized flexible liposomes embedded in situ nasal gel, and marketed formulation. ANNALES PHARMACEUTIQUES FRANÇAISES 2024:S0003-4509(24)00135-4. [PMID: 39270836 DOI: 10.1016/j.pharma.2024.09.002] [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: 09/24/2023] [Revised: 05/06/2024] [Accepted: 09/09/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND Carmustine is used in the treatment of glioblastoma (GBM). GBM is a well-known life-threatening type of cancerous tumor. GBM covers 60.00% among all primary brain tumors, with an occurrence of 74,000 cases across the globe. Management for GBM is still very difficult because most of the medicines are unable to cross the blood-brain barrier (BBB). The present work observed that flexible liposomes embedded in situ nasal gel of carmustine is the best brain-targeted medicine delivery system for the management of GBM through the nasal route. AIM To evaluate in vivo pharmacokinetic parameters of carmustine formulations administered through nasal routes in Wistar rats. METHODS In this work, different pharmacokinetic parameters were determined for carmustine formulations viz. carmustine API (Active Pharmaceutical Ingredient) solution, flexible liposomes, in situ thermoreversible intranasal gel, optimized flexible liposomes embedded in situ thermoreversible intranasal gel via intranasal administration in rats, and compared with marketed intravenous injection of carmustine administered through intravenous route. Carmustine was estimated with the help of a validated high-performance liquid chromatography (HPLC) approach. Three to four-months-old normal Wistar rats of either sex, having a weight of 200-250 grams were used in this study. RESULTS Intranasal administration of optimized flexible liposomes embedded in situ nasal gel showed greater Cmax (∼two-fold), AUC0→t (∼three-fold), AUC0→∞ (∼six-fold), and decreased Tmax (1h) data in the brain, than commercial intravenous injection of carmustine. The plasma concentration of carmustine administered through nasal route was found to be comparatively lower than intravenous administration, indicating lower systemic exposure to carmustine via the nasal route. CONCLUSION In vivo pharmacokinetics results revealed that the optimized flexible liposomes embedded in situ nasal gel of carmustine can effectively deliver carmustine to brain by nasal drug delivery system in Wistar rats.
Collapse
Affiliation(s)
- Audumbar Mali
- School of Life Sciences, Punyashlok Ahilyadevi Holkar Solapur University, 413255 Solapur, Maharashtra, India.
| | - Anil Bhanwase
- Department of Pharmaceutical Chemistry, SPM's College of Pharmacy, 413101 Akluj, Maharashtra, India
| |
Collapse
|
2
|
Hoelm M, Porwański S, Jóźwiak P, Krześlak A. Combined Theoretical and Experimental Investigations: Design, Synthesis, Characterization, and In Vitro Cytotoxic Activity Assessment of a Complex of a Novel Ureacellobiose Drug Carrier with the Anticancer Drug Carmustine. Molecules 2024; 29:3359. [PMID: 39064937 PMCID: PMC11280079 DOI: 10.3390/molecules29143359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/15/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Drug delivery systems (DDSs) are used to transport drugs which are characterized by some pharmaceutical problems to the specific target site, enhancing therapeutic efficacy and reducing off-target accumulation in the body. In this work, one of the recently synthesized molecules, 1,10-N,N'-bis-(β-ᴅ-ureidocellobiosyl)-4,7,13,16-tetraoxa-1,10-diazacyclooctadecane (TN), was tested as a potential drug carrier towards the anticancer drug carmustine. For this purpose, different techniques were used, from synthesis and calculations to cytotoxicity assessment. Our results showed that TN is characterized by a very compact geometry, which significantly impacts its complexation properties. Although it forms a very stable complex with carmustine, it adopts a non-inclusion geometry, as verified by both experimental and theoretical NMR analyses. The cytotoxicity study performed for all analyzed molecules (TN; carmustine; TN:carmustine complex) towards normal and cancer (breast and colon) cells revealed that TN is not toxic and that the formation of complexes with carmustine reduces the toxicity of carmustine to normal cells.
Collapse
Affiliation(s)
- Marta Hoelm
- Theoretical and Structural Group, Department of Physical Chemistry, Faculty of Chemistry, University of Lodz, Pomorska 163/165, 90-236 Lodz, Poland
| | - Stanisław Porwański
- Department of Organic and Applied Chemistry, Faculty of Chemistry, University of Lodz, Tamka 12, 91-403 Lodz, Poland;
| | - Paweł Jóźwiak
- Department of Cytobiochemistry, Faculty of Biology, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (P.J.); (A.K.)
| | - Anna Krześlak
- Department of Cytobiochemistry, Faculty of Biology, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (P.J.); (A.K.)
| |
Collapse
|
3
|
Ballestín A, Armocida D, Ribecco V, Seano G. Peritumoral brain zone in glioblastoma: biological, clinical and mechanical features. Front Immunol 2024; 15:1347877. [PMID: 38487525 PMCID: PMC10937439 DOI: 10.3389/fimmu.2024.1347877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 02/14/2024] [Indexed: 03/17/2024] Open
Abstract
Glioblastoma is a highly aggressive and invasive tumor that affects the central nervous system (CNS). With a five-year survival rate of only 6.9% and a median survival time of eight months, it has the lowest survival rate among CNS tumors. Its treatment consists of surgical resection, subsequent fractionated radiotherapy and concomitant and adjuvant chemotherapy with temozolomide. Despite the implementation of clinical interventions, recurrence is a common occurrence, with over 80% of cases arising at the edge of the resection cavity a few months after treatment. The high recurrence rate and location of glioblastoma indicate the need for a better understanding of the peritumor brain zone (PBZ). In this review, we first describe the main radiological, cellular, molecular and biomechanical tissue features of PBZ; and subsequently, we discuss its current clinical management, potential local therapeutic approaches and future prospects.
Collapse
Affiliation(s)
- Alberto Ballestín
- Tumor Microenvironment Laboratory, UMR3347 CNRS/U1021 INSERM, Institut Curie, Orsay, France
| | - Daniele Armocida
- Human Neurosciences Department, Neurosurgery Division, Sapienza University, Rome, Italy
| | - Valentino Ribecco
- Tumor Microenvironment Laboratory, UMR3347 CNRS/U1021 INSERM, Institut Curie, Orsay, France
| | - Giorgio Seano
- Tumor Microenvironment Laboratory, UMR3347 CNRS/U1021 INSERM, Institut Curie, Orsay, France
| |
Collapse
|
4
|
Parikh SD, Wang W, Nelson MT, Sulentic CEW, Mukhopadhyay SM. Bioinspired Hierarchical Carbon Structures as Potential Scaffolds for Wound Healing and Tissue Regeneration Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:nano13111791. [PMID: 37299693 DOI: 10.3390/nano13111791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/22/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023]
Abstract
Engineered bio-scaffolds for wound healing provide an attractive treatment option for tissue engineering and traumatic skin injuries since they can reduce dependence on donors and promote faster repair through strategic surface engineering. Current scaffolds present limitations in handling, preparation, shelf life, and sterilization options. In this study, bio-inspired hierarchical all-carbon structures comprising carbon nanotube (CNT) carpets covalently bonded to flexible carbon fabric have been investigated as a platform for cell growth and future tissue regeneration applications. CNTs are known to provide guidance for cell growth, but loose CNTs are susceptible to intracellular uptake and are suspected to cause in vitro and in vivo cytotoxicity. This risk is suppressed in these materials due to the covalent attachment of CNTs on a larger fabric, and the synergistic benefits of nanoscale and micro-macro scale architectures, as seen in natural biological materials, can be obtained. The structural durability, biocompatibility, tunable surface architecture, and ultra-high specific surface area of these materials make them attractive candidates for wound healing. In this study, investigations of cytotoxicity, skin cell proliferation, and cell migration were performed, and results indicate promise in both biocompatibility and directed cell growth. Moreover, these scaffolds provided cytoprotection against environmental stressors such as Ultraviolet B (UVB) rays. It was seen that cell growth could also be tailored through the control of CNT carpet height and surface wettability. These results support future promise in the design of hierarchical carbon scaffolds for strategic wound healing and tissue regeneration applications.
Collapse
Affiliation(s)
- Soham D Parikh
- Department of Mechanical & Materials Engineering, Wright State University, 3640 Col. Glen Hwy, Dayton, OH 45435, USA
| | - Wenhu Wang
- Frontier Institute for Research in Sensor Technologies (FIRST), University of Maine, United States Air Force Research Laboratory, Orono, ME 04469, USA
| | - M Tyler Nelson
- 711th Human Performance Wing, Airman Systems Directorate, Bioengineering Division, Wright-Patterson Air Force Base, Dayton, OH 45433, USA
| | - Courtney E W Sulentic
- Department of Pharmacology and Toxicology, Wright State University, Boonshoft School of Medicine, 3640 Col. Glen Hwy, Dayton, OH 45435, USA
| | - Sharmila M Mukhopadhyay
- Frontier Institute for Research in Sensor Technologies (FIRST), University of Maine, United States Air Force Research Laboratory, Orono, ME 04469, USA
| |
Collapse
|
5
|
Yang X, Zheng L, Gao L, Zhang L, He J, Wei Y, You J, Li H, Yi C, Luo H. A Local Strategy Toward Concurrent Chemoradiotherapy Based on Fibrin Gel for Postsurgical Cancer Treatment. Int J Radiat Oncol Biol Phys 2023:S0360-3016(23)00150-5. [PMID: 36796499 DOI: 10.1016/j.ijrobp.2023.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/18/2023] [Accepted: 02/03/2023] [Indexed: 02/16/2023]
Abstract
PURPOSE Postoperative cancer recurrence and metastasis have always been huge challenges in cancer therapy. The concurrent cisplatin (CDDP)-based chemoradiotherapy regimen is a standard therapeutic strategy in some cancer treatments after surgical resection. However, severe side effects and unsatisfactory local tumor concentrations of CDDP have hampered the application of this concurrent chemoradiotherapy. Therefore, a superior option that can enhance CDDP-based chemoradiotherapy efficacy with milder concurrent therapy-related toxicity is highly desirable. METHODS AND MATERIALS We developed a platform based on fibrin gel (Fgel) loaded with CDDP to be implanted into the tumor bed after surgery combined with concurrent radiation therapy for the prevention of postoperative local cancer recurrence and distant metastasis. The postoperative subcutaneous tumor mouse models established by incomplete resection of primary tumors were used to evaluate the therapeutic advantages of this chemoradiotherapy regimen for postsurgical treatment. RESULTS The local and sustained release of CDDP from Fgel could enhance the antitumor efficacy of radiation therapy in the residual tumor with lower systemic toxicity. The therapeutic benefits of this approach are demonstrated in breast cancer, anaplastic thyroid carcinoma, and osteosarcoma mouse models. CONCLUSIONS Our work offers a general platform for concurrent chemoradiotherapy to prevent postoperative cancer recurrence and metastasis.
Collapse
Affiliation(s)
- Xi Yang
- Division of Radiotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Lingnan Zheng
- Division of Radiotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ling Gao
- Department of Health Ward, Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lingyun Zhang
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong SAR, China; Department of Thyroid and Parathyroid Surgery, Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jinlan He
- Division of Radiotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - YuanFeng Wei
- Division of Radiotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jia You
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huawei Li
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Yi
- Division of Radiotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Han Luo
- Department of Thyroid and Parathyroid Surgery, Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
6
|
Fabrication and Modelling of a Reservoir-Based Drug Delivery System for Customizable Release. Pharmaceutics 2022; 14:pharmaceutics14040777. [PMID: 35456611 PMCID: PMC9025308 DOI: 10.3390/pharmaceutics14040777] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/29/2022] [Accepted: 03/29/2022] [Indexed: 01/27/2023] Open
Abstract
Localized therapy approaches have emerged as an alternative drug administration route to overcome the limitations of systemic therapies, such as the crossing of the blood–brain barrier in the case of brain tumor treatment. For this, implantable drug delivery systems (DDS) have been developed and extensively researched. However, to achieve an effective localized treatment, the release kinetics of DDS needs to be controlled in a defined manner, so that the concentration at the tumor site is within the therapeutic window. Thus, a DDS, with patient-specific release kinetics, is crucial for the improvement of therapy. Here, we present a computationally supported reservoir-based DDS (rDDS) development towards patient-specific release kinetics. The rDDS consists of a reservoir surrounded by a polydimethylsiloxane (PDMS) microchannel membrane. By tailoring the rDDS, in terms of membrane porosity, geometry, and drug concentration, the release profiles can be precisely adapted, with respect to the maximum concentration, release rate, and release time. The release is investigated using a model dye for varying parameters, leading to different distinct release profiles, with a maximum release of up to 60 days. Finally, a computational simulation, considering exemplary in vivo conditions (e.g., exchange of cerebrospinal fluid), is used to study the resulting drug release profiles, demonstrating the customizability of the system. The establishment of a computationally supported workflow, for development towards a patient-specific rDDS, in combination with the transfer to suitable drugs, could significantly improve the efficacy of localized therapy approaches.
Collapse
|
7
|
Iuchi T, Inoue A, Hirose Y, Morioka M, Horiguchi K, Natsume A, Arakawa Y, Iwasaki K, Fujiki M, Kumabe T, Sakata Y. Long-term effectiveness of Gliadel implant for malignant glioma and prognostic factors for survival: 3-year results of a postmarketing surveillance in Japan. Neurooncol Adv 2022; 4:vdab189. [PMID: 35118382 PMCID: PMC8807118 DOI: 10.1093/noajnl/vdab189] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Adjuvant treatment with Gliadel wafers may prolong overall survival (OS) for malignant glioma patients without increasing toxicity. In Japan, the long-term OS of these patients treated with Gliadel 7.7 mg implants has not been studied. We evaluated OS and prognostic factors that might affect OS in Japanese patients with malignant glioma who received the Gliadel 7.7 mg implant. METHODS This observational, long-term, postmarketing surveillance was an extension of a previous surveillance. Data were collected through case report forms at 2 and 3 years after Gliadel implant. Up to 8 Gliadel wafers (61.6 mg of carmustine) were placed over the tumor resection site. Primary endpoints were OS and prognostic factors that may influence OS. RESULTS Among the 506 patients analyzed, 62.6% had newly diagnosed disease, and 37.4% had recurrent disease; 79.1% had glioblastoma histological type and 79.6% had World Health Organization Grade IV disease. Patients received a median of 8 wafers. The median OS was 18.0 months; OS rates were 39.8% and 31.5% at 2 and 3 years, respectively. Age ≥65 years (hazard ratio [HR]: 1.456; P = .002), lower resection rate (HR: 1.206; P < .001), recurrence (HR: 2.418; P < .001), and concomitant radiotherapy (HR: 0.588; P < .001) were identified as significant prognostic factors. CONCLUSIONS This study confirmed the 2- and 3-year OS of Japanese malignant glioma patients with varied backgrounds after Gliadel implant. With a careful interpretation of indirect comparisons with previously reported data, the results suggest that prognosis could be improved with Gliadel implants. CLINICAL TRIAL REGISTRATION NCT02300506.
Collapse
Affiliation(s)
- Toshihiko Iuchi
- Division of Neurological Surgery, Chiba Cancer Center, Chiba, Japan
| | - Akihiro Inoue
- Department of Neurosurgery, Ehime University School of Medicine, Ehime, Japan
| | - Yuichi Hirose
- Department of Neurosurgery, Fujita Health University, Aichi, Japan
| | - Motohiro Morioka
- Department of Neurosurgery, Kurume University School of Medicine, Fukuoka, Japan
| | - Keishi Horiguchi
- Department of Neurosurgery, Gunma University Hospital, Gunma, Japan
| | - Atsushi Natsume
- Department of Neurosurgery, Nagoya University Hospital, Aichi, Japan
| | - Yoshiki Arakawa
- Department of Neurosurgery, Kyoto University Hospital, Kyoto, Japan
| | - Koichi Iwasaki
- Department of Neurosurgery, Tazuke Kofukai Medical Research Institute, Kitano Hospital, Osaka, Japan
| | - Minoru Fujiki
- Department of Neurosurgery, Oita University Hospital, Oita, Japan
| | - Toshihiro Kumabe
- Department of Neurosurgery, Kitasato University Hospital, Kanagawa, Japan
| | - Yukinori Sakata
- Clinical Planning Department, Medical Headquarters, Eisai Co., Ltd, Tokyo, Japan
| |
Collapse
|
8
|
NISHIKAWA R, IWATA H, SAKATA Y, MURAMOTO K, MATSUOKA T. Safety of Gliadel Implant for Malignant Glioma: Report of Postmarketing Surveillance in Japan. Neurol Med Chir (Tokyo) 2021; 61:536-548. [PMID: 34092748 PMCID: PMC8443968 DOI: 10.2176/nmc.oa.2021-0024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/26/2021] [Indexed: 11/20/2022] Open
Abstract
Clinical trial data of Carmustine implant (Gliadel Wafer) in Japanese patients with malignant glioma are limited; thus, we conducted a postmarketing surveillance study to evaluate the safety of Gliadel in real-world clinical practice in Japan. In this postmarketing surveillance study, all patients who received Gliadel placement for malignant glioma surgeries from its market launch (January 9, 2013) to July 10, 2013 were enrolled from 229 institutions using a central registration system. Up to eight wafers of Gliadel (containing 61.6 mg of carmustine) were used to cover the site of brain tumor resection intraoperatively according to the size and shape of the tumor resection cavity. The observation period lasted 3 months after Gliadel placement. Patients were followed up for 1 year postoperatively. Safety was assessed by the incidence of adverse events (AEs) and adverse drug reactions (ADRs). In total, 558 patients were included. Most patients (66.7%) received eight Gliadel wafers. The percentage of patients with ADRs was 35.7% (365 ADR episodes in 199 patients). Of the AEs of special interest, the most common were cerebral edema (22.2%, 124/558 patients), convulsion (9.9%, 55/558 patients), impaired healing (4.8%, 27/558 patients), and infection (3.4%, 19/558 patients). This first all-case postmarketing surveillance report of the safety of Gliadel in real-world clinical practice in Japan suggests that the risk of toxicity with Gliadel placement is relatively tolerable. The survival benefits of Gliadel placement should be evaluated and considered carefully by the clinician taking into account possible toxicities.
Collapse
Affiliation(s)
- Ryo NISHIKAWA
- Department of Neuro-Oncology and Neurosurgery, Saitama Medical University
International Medical Center, Hidaka, Saitama, Japan
| | | | | | | | | |
Collapse
|
9
|
Alghamdi M, Gumbleton M, Newland B. Local delivery to malignant brain tumors: potential biomaterial-based therapeutic/adjuvant strategies. Biomater Sci 2021; 9:6037-6051. [PMID: 34357362 DOI: 10.1039/d1bm00896j] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Glioblastoma (GBM) is the most aggressive malignant brain tumor and is associated with a very poor prognosis. The standard treatment for newly diagnosed patients involves total tumor surgical resection (if possible), plus irradiation and adjuvant chemotherapy. Despite treatment, the prognosis is still poor, and the tumor often recurs within two centimeters of the original tumor. A promising approach to improving the efficacy of GBM therapeutics is to utilize biomaterials to deliver them locally at the tumor site. Local delivery to GBM offers several advantages over systemic administration, such as bypassing the blood-brain barrier and increasing the bioavailability of the therapeutic at the tumor site without causing systemic toxicity. Local delivery may also combat tumor recurrence by maintaining sufficient drug concentrations at and surrounding the original tumor area. Herein, we critically appraised the literature on local delivery systems based within the following categories: polymer-based implantable devices, polymeric injectable systems, and hydrogel drug delivery systems. We also discussed the negative effect of hypoxia on treatment strategies and how one might utilize local implantation of oxygen-generating biomaterials as an adjuvant to enhance current therapeutic strategies.
Collapse
Affiliation(s)
- Majed Alghamdi
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK. and Faculty of Pharmacy, King Abdulaziz University, Jeddah, 22522, Kingdom of Saudi Arabia
| | - Mark Gumbleton
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK.
| | - Ben Newland
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK. and Leibniz-Institut für Polymerforschung Dresden, Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, D-01069 Dresden, Germany
| |
Collapse
|
10
|
Ak G, Ünal A, Karakayalı T, Özel B, Selvi Günel N, Hamarat Şanlıer Ş. Brain-targeted, drug-loaded solid lipid nanoparticles against glioblastoma cells in culture. Colloids Surf B Biointerfaces 2021; 206:111946. [PMID: 34216850 DOI: 10.1016/j.colsurfb.2021.111946] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/23/2021] [Accepted: 06/24/2021] [Indexed: 12/11/2022]
Abstract
The aim of this study was the preparation of solid lipid nanoparticles (SLN) formed from cetyl palmitate with having targeting molecules for monocarboxylate transporter-1 (MCT-1): β-hydroxybutyric acid and anticancer agents: carmustine (BCNU) and temozolomide (TMZ) for enhanced anti-proliferation against glioblastoma multiforme (GBM). Properties including size, morphology, chemical structure, zeta potential, drug encapsulation efficacy, drug release, biocompatibility, stability were determined, and in vitro studies were done. BCNU and TMZ loaded SLNs had a hydrodynamic size of 227 nm ± 46 a zeta potential of -25 mV ± 4 with biocompatible features. The data showed rapid drug release at first and then continuous release. Nanoparticles could be stored for nine months. BCNU and TMZ loaded SLNs exhibited a remarkable increment in the antitumor activity compared to the free-drugs and induced apoptosis on U87MG cells. In addition, targeted nanoparticles were more uptaken by MCT-1 expressing brain cells. This study indicated that BCNU and TMZ loaded SLNs could act as a useful anticancer system for targeted GBM therapy.
Collapse
Affiliation(s)
- Güliz Ak
- Biochemistry Department, Faculty of Science, Ege University, 35040, Izmir, Turkey; Center for Drug Research, Development and Pharmacokinetic Applications (ARGEFAR), Ege University, 35100, Izmir, Turkey.
| | - Ayşe Ünal
- Biochemistry Department, Faculty of Science, Ege University, 35040, Izmir, Turkey
| | - Tuğba Karakayalı
- Biochemistry Department, Faculty of Science, Ege University, 35040, Izmir, Turkey
| | - Buket Özel
- Center for Drug Research, Development and Pharmacokinetic Applications (ARGEFAR), Ege University, 35100, Izmir, Turkey; Department of Medical Biology, Faculty of Medicine, Ege University, 35100, Izmir, Turkey
| | - Nur Selvi Günel
- Department of Medical Biology, Faculty of Medicine, Ege University, 35100, Izmir, Turkey
| | - Şenay Hamarat Şanlıer
- Biochemistry Department, Faculty of Science, Ege University, 35040, Izmir, Turkey; Center for Drug Research, Development and Pharmacokinetic Applications (ARGEFAR), Ege University, 35100, Izmir, Turkey
| |
Collapse
|
11
|
Erthal LCS, Gobbo OL, Ruiz-Hernandez E. Biocompatible copolymer formulations to treat glioblastoma multiforme. Acta Biomater 2021; 121:89-102. [PMID: 33227487 DOI: 10.1016/j.actbio.2020.11.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/12/2020] [Accepted: 11/17/2020] [Indexed: 12/20/2022]
Abstract
The treatment for glioblastoma multiforme (GBM) has not changed for more than 20 years while the prognosis for the patients is still poor and most of them survive less than 1 year after diagnosis. The standard of care for GBM is comprised of surgical resection followed by radiotherapy and oral chemotherapy with temozolomide. The placement of carmustine wafers in the brain after tumour removal is added in cases of recurrent glioma. Significant research is underway to improve the GBM therapy outcome and patient quality of life. Biomaterials are in the front line of the research focus for new treatment options. Specially, biocompatible polymers have been proposed in hydrogel-based formulations aiming at injectable and localized therapies. These formulations can comprise many different pharmacological agents such as chemotherapeutic drugs, nanoparticles, cells, nucleic acids, and diagnostic agents. In this manuscript, we review the most recent formulations developed and tested both in vitro and in vivo using different types of hydrogels. Firstly, we describe three common types of thermo-responsive polymers addressing the advantages and drawbacks of their formulations. Then, we focus on formulations specifically developed for GBM treatment.
Collapse
Affiliation(s)
- Luiza C S Erthal
- School of Pharmacy and Pharmaceutical Sciences & Trinity St. James's Cancer Institute, Trinity College Dublin, College Green, Dublin 2, Ireland
| | - Oliviero L Gobbo
- School of Pharmacy and Pharmaceutical Sciences & Trinity St. James's Cancer Institute, Trinity College Dublin, College Green, Dublin 2, Ireland
| | - Eduardo Ruiz-Hernandez
- School of Pharmacy and Pharmaceutical Sciences & Trinity St. James's Cancer Institute, Trinity College Dublin, College Green, Dublin 2, Ireland.
| |
Collapse
|
12
|
Li R, Ng TS, Garlin MA, Weissleder R, Miller MA. Understanding the in vivo Fate of Advanced Materials by Imaging. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1910369. [PMID: 38545084 PMCID: PMC10972611 DOI: 10.1002/adfm.201910369] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 02/09/2020] [Indexed: 11/13/2024]
Abstract
Engineered materials are ubiquitous in biomedical applications ranging from systemic drug delivery systems to orthopedic implants, and their actions unfold across multiple time- and length-scales. The efficacy and safety of biologics, nanomaterials, and macroscopic implants are all dictated by the same general principles of pharmacology as apply to small molecule drugs, comprising how the body affects materials (pharmacokinetics, PK) and conversely how materials affect the body (pharmacodynamics, PD). Imaging technologies play an increasingly insightful role in monitoring both of these processes, often simultaneously: translational macroscopic imaging modalities such as MRI and PET/CT offer whole-body quantitation of biodistribution and structural or molecular response, while ex vivo approaches and optical imaging via in vivo (intravital) microscopy reveal behaviors at subcellular resolution. In this review, the authors survey developments in imaging the in situ behavior of systemically and locally administered materials, with a particular focus on using microscopy to understand transport, target engagement, and downstream host responses at a single-cell level. The themes of microenvironmental influence, controlled drug release, on-target molecular action, and immune response, especially as mediated by macrophages and other myeloid cells are examined. Finally, the future directions of how new imaging technologies may propel efficient clinical translation of next-generation therapeutics and medical devices are proposed.
Collapse
Affiliation(s)
- Ran Li
- Center for Systems Biology, Massachusetts General Hospital Research Institute
| | - Thomas S.C. Ng
- Center for Systems Biology, Massachusetts General Hospital Research Institute
| | - Michelle A. Garlin
- Center for Systems Biology, Massachusetts General Hospital Research Institute
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School
- Department of Systems Biology, Harvard Medical School
| | - Miles A. Miller
- Center for Systems Biology, Massachusetts General Hospital Research Institute
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School
| |
Collapse
|
13
|
Parikh SD, Dave S, Huang L, Wang W, Mukhopadhyay SM, Mayes DA. Multi-walled carbon nanotube carpets as scaffolds for U87MG glioblastoma multiforme cell growth. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 108:110345. [PMID: 31924041 DOI: 10.1016/j.msec.2019.110345] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 10/02/2019] [Accepted: 10/19/2019] [Indexed: 10/25/2022]
Abstract
Carbon Nanotubes (CNTs) are known for effective adhesion, growth, and differentiation of bone, muscle, and cardiac cells. CNTs can provide excellent mechanical and electrical properties for cell scaffolding; however, loose CNTs can cause in-vivo toxicity. To suppress this risk, our team has developed biomimetic scaffolds with multiscale hierarchy where carpet-like CNT arrays are covalently bonded to larger biocompatible substrates. In this study, we investigated the interaction between glioblastoma multiforme (GBM) cells (U87MG) and our unique hierarchical CNT-coated scaffolds upon brain tumor cell proliferation. U87MG cells grown on un-modified carbon scaffolds grew in a bi-phasic fashion. Initially, the scaffolds prevented GBM cell growth; however, prolonged growth on such scaffolds significantly increased GBM cell proliferation. We further defined the importance of the hydrophobicity/hydrophilicity of the CNT-coated scaffolds in this cellular response by utilizing sodium-hypochlorite based bleach treatment prior to cellular exposure. This surface modification increased the hydrophilicity of the CNT-coated scaffolds and ameliorated the biphasic response of U87MG cells allowing for a normal growth curve. Findings highlight the importance of surface modification and wettability of the CNT-coated scaffolds for cell growth applications. The focus for this study was to determine whether scaffold surface features could modulate tumor-scaffold interactions, and thus to improve our understanding of and optimize successful development of future scaffold-based chemotherapy applications. Overall, it appears that the wettability of carbon scaffolds coated with CNTs is an important regulator of U87MG cellular growth. These findings will be important to consider when developing a potential chemotherapy-attached implant to be used post-surgical resection for GBM patient treatment.
Collapse
Affiliation(s)
- Soham D Parikh
- Center for Nanoscale Multifunctional Materials, Department of Mechanical & Materials Engineering, Wright State University; 3640 Col. Glen Hwy, Dayton, OH, 45435, USA; Department of Neuroscience, Cell Biology and Physiology, Translational Neuroscience Institute, Wright State University, Boonshoft School of Medicine, College of Science and Math, 3640 Col. Glen Hwy, Dayton, OH, 45435, USA
| | - Soham Dave
- Department of Neuroscience, Cell Biology and Physiology, Translational Neuroscience Institute, Wright State University, Boonshoft School of Medicine, College of Science and Math, 3640 Col. Glen Hwy, Dayton, OH, 45435, USA
| | - Luping Huang
- Department of Neuroscience, Cell Biology and Physiology, Translational Neuroscience Institute, Wright State University, Boonshoft School of Medicine, College of Science and Math, 3640 Col. Glen Hwy, Dayton, OH, 45435, USA
| | - Wenhu Wang
- Center for Nanoscale Multifunctional Materials, Department of Mechanical & Materials Engineering, Wright State University; 3640 Col. Glen Hwy, Dayton, OH, 45435, USA
| | - Sharmila M Mukhopadhyay
- Center for Nanoscale Multifunctional Materials, Department of Mechanical & Materials Engineering, Wright State University; 3640 Col. Glen Hwy, Dayton, OH, 45435, USA.
| | - Debra A Mayes
- Department of Neuroscience, Cell Biology and Physiology, Translational Neuroscience Institute, Wright State University, Boonshoft School of Medicine, College of Science and Math, 3640 Col. Glen Hwy, Dayton, OH, 45435, USA.
| |
Collapse
|
14
|
Yi S, Yang F, Jie C, Zhang G. A novel strategy to the formulation of carmustine and bioactive nanoparticles co-loaded PLGA biocomposite spheres for targeting drug delivery to glioma treatment and nursing care. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:3438-3447. [PMID: 31411066 DOI: 10.1080/21691401.2019.1652628] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Shufeng Yi
- Department of Neurosurgery, Jinan People's Hospital, Jinan, P. R. China
| | - Fan Yang
- EICU, Jinan People's Hospital, Jinan, P. R. China
| | - Cunle Jie
- Dialysis Room, Jinan People's Hospital, Jinan, Shandong Province, P. R. China
| | - Guiqin Zhang
- Department of Science and Education, Jinan People's Hospital, Jinan, P. R. China
| |
Collapse
|
15
|
Zhao M, Bozzato E, Joudiou N, Ghiassinejad S, Danhier F, Gallez B, Préat V. Codelivery of paclitaxel and temozolomide through a photopolymerizable hydrogel prevents glioblastoma recurrence after surgical resection. J Control Release 2019; 309:72-81. [PMID: 31306678 DOI: 10.1016/j.jconrel.2019.07.015] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 07/02/2019] [Accepted: 07/11/2019] [Indexed: 01/03/2023]
Abstract
A photopolymerizable hydrogel-based local drug delivery system was developed for the postsurgical treatment of glioblastoma (GBM). We aimed for a local drug combination therapy with paclitaxel (PTX) and temozolomide (TMZ) within a hydrogel to synergistically inhibit tumor growth. The in vitro cytotoxicity of TMZ was assessed in U87MG cells. We demonstrated the synergistic effect of PTX and TMZ on U87MG cells by clonogenic assay. Treatment with TMZ did not induce O6-methylguanine-DNA methyltransferase related drug resistance in tumor-bearing mice. PTX had sustained release for at least 1 month in vivo in healthy mice brains. The drug combination was tolerable and suppressed tumor growth more efficiently than the single drugs in the U87MG orthotopic tumor model. The PTX and TMZ codelivery hydrogel showed superior antitumor effects and can be considered a promising approach for the postsurgical treatment of GBM.
Collapse
Affiliation(s)
- Mengnan Zhao
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier, 73, B1.73.12, 1200 Brussels, Belgium
| | - Elia Bozzato
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier, 73, B1.73.12, 1200 Brussels, Belgium
| | - Nicolas Joudiou
- Université catholique de Louvain, Louvain Drug Research Institute, Nuclear and Electron Spin Technologies Platform (NEST), Avenue Mounier, 73, B1.73.08, 1200 Brussels, Belgium
| | - Sina Ghiassinejad
- Université catholique de Louvain, Institute of Condensed Matter and Nanoscience (IMCN), Bio and Soft Matter, Croix du Sud 1, B-1348 Louvain-la-Neuve, Belgium
| | - Fabienne Danhier
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier, 73, B1.73.12, 1200 Brussels, Belgium
| | - Bernard Gallez
- Université catholique de Louvain, Louvain Drug Research Institute, Biomedical Magnetic Resonance Research Group, Avenue Mounier, 73, B1.73.08, 1200 Brussels, Belgium
| | - Véronique Préat
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier, 73, B1.73.12, 1200 Brussels, Belgium.
| |
Collapse
|
16
|
Jalota A, Kumar M, Das BC, Yadav AK, Chosdol K, Sinha S. A drug combination targeting hypoxia induced chemoresistance and stemness in glioma cells. Oncotarget 2018; 9:18351-18366. [PMID: 29719610 PMCID: PMC5915077 DOI: 10.18632/oncotarget.24839] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 02/21/2018] [Indexed: 12/18/2022] Open
Abstract
Hypoxia is a characteristic of solid tumors especially Glioblastoma and is critical to chemoresistance. Cancer stem cells present in hypoxic niches are known to be a major cause of the progression, metastasis and relapse. We tried to identify synergistic combinations of drugs effective in both hypoxia and normoxia in tumor cells as well as in cancer stem cells. Since COX-2 is over-expressed in subset of glioblastoma and is also induced in hypoxia, we studied combinations of a prototype Cyclooxygenase (COX-2) inhibitor, NS-398 with various drugs (BCNU, Temozolomide, 2-Deoxy-D-glucose and Cisplatin) for their ability to abrogate chemoresistance under both severe hypoxia (0.2% O2) and normoxia (20% O2) in glioma cells. The only effective combination was of NS-398 and BCNU which showed a synergistic effect in both hypoxia and normoxia. This synergism was evident at sub-lethal doses for either of the single agent. The effectiveness of the combination resulted from increased pro- apoptotic and decreased anti-apoptotic molecules and increased caspase activity. PGE2 levels, a manifestation of COX-2 activity were increased during hypoxia, but were reduced by the combination during both hypoxia and normoxia. The combination reduced the levels of epithelial-mesenchymal transition (EMT) markers. It also resulted in a greater reduction of cell migration. While single drugs could reduce the number of gliomaspheres, the combination successfully abrogated their formation. The combination also resulted in a greater reduction of the cancer stem cell marker CD133. This combination could be a prototype of possible therapy in a tumor with a high degree of hypoxia like glioma.
Collapse
Affiliation(s)
- Akansha Jalota
- National Brain Research Centre, Manesar, Gurgaon-122051, India.,Department of Biochemistry, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Mukesh Kumar
- National Brain Research Centre, Manesar, Gurgaon-122051, India
| | - Bhudev C Das
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi-110007, India.,Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida-201313, India
| | - Ajay K Yadav
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi-110007, India
| | - Kunzang Chosdol
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Subrata Sinha
- National Brain Research Centre, Manesar, Gurgaon-122051, India.,Department of Biochemistry, All India Institute of Medical Sciences, New Delhi-110029, India
| |
Collapse
|
17
|
Miranda A, Blanco-Prieto M, Sousa J, Pais A, Vitorino C. Breaching barriers in glioblastoma. Part I: Molecular pathways and novel treatment approaches. Int J Pharm 2017; 531:372-388. [PMID: 28755993 DOI: 10.1016/j.ijpharm.2017.07.056] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 07/18/2017] [Accepted: 07/19/2017] [Indexed: 12/12/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common primary brain tumour, and the most aggressive in nature. The prognosis for patients with GBM remains poor, with a median survival time of only 1-2 years. The treatment failure relies on the development of resistance by tumour cells and the difficulty of ensuring that drugs effectively cross the dual blood brain barrier/blood brain tumour barrier. The advanced molecular and genetic knowledge has allowed to identify the mechanisms responsible for temozolomide resistance, which represents the standard of care in GBM, along with surgical resection and radiotherapy. Such resistance has motivated the researchers to investigate new avenues for GBM treatment intended to improve patient survival. In this review, we provide an overview of major obstacles to effective treatment of GBM, encompassing biological barriers, cancer stem cells, DNA repair mechanisms, deregulated signalling pathways and autophagy. New insights and potential therapy approaches for GBM are also discussed, emphasizing localized chemotherapy delivered directly to the brain, immunotherapy, gene therapy and nanoparticle-mediated brain drug delivery.
Collapse
Affiliation(s)
- Ana Miranda
- Faculty of Pharmacy, University of Coimbra, Portugal; Pharmacometrics Group of the Centre for Neurosciences and Cell Biology (CNC), University of Coimbra, Portugal
| | - María Blanco-Prieto
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, Spain
| | - João Sousa
- Faculty of Pharmacy, University of Coimbra, Portugal; Pharmacometrics Group of the Centre for Neurosciences and Cell Biology (CNC), University of Coimbra, Portugal
| | - Alberto Pais
- Coimbra Chemistry Center, Department of Chemistry, University of Coimbra, Portugal
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, Portugal; Pharmacometrics Group of the Centre for Neurosciences and Cell Biology (CNC), University of Coimbra, Portugal.
| |
Collapse
|
18
|
Chakroun RW, Zhang P, Lin R, Schiapparelli P, Quinones-Hinojosa A, Cui H. Nanotherapeutic systems for local treatment of brain tumors. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2017; 10. [PMID: 28544801 DOI: 10.1002/wnan.1479] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 04/14/2017] [Accepted: 04/18/2017] [Indexed: 12/31/2022]
Abstract
Malignant brain tumor, including the most common type glioblastoma, are histologically heterogeneous and invasive tumors known as the most devastating neoplasms with high morbidity and mortality. Despite multimodal treatment including surgery, radiotherapy, chemotherapy, and immunotherapy, the disease inevitably recurs and is fatal. This lack of curative options has motivated researchers to explore new treatment strategies and to develop new drug delivery systems (DDSs); however, the unique anatomical, physiological, and pathological features of brain tumors greatly limit the effectiveness of conventional chemotherapy. In this context, we review the recent progress in the development of nanoparticle-based DDSs aiming to address the key challenges in transporting sufficient amount of therapeutic agents into the brain tumor areas while minimizing the potential side effects. We first provide an overview of the standard treatments currently used in the clinic for the management of brain cancers, discussing the effectiveness and limitations of each therapy. We then provide an in-depth review of nanotherapeutic systems that are intended to bypass the blood-brain barrier, overcome multidrug resistance, infiltrate larger tumorous tissue areas, and/or release therapeutic agents in a controlled manner. WIREs Nanomed Nanobiotechnol 2018, 10:e1479. doi: 10.1002/wnan.1479 This article is categorized under: Implantable Materials and Surgical Technologies > Nanomaterials and Implants Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Rami Walid Chakroun
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Pengcheng Zhang
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Ran Lin
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | | | | | - Honggang Cui
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
19
|
Patterns of Recurrence After Resection of Malignant Gliomas With BCNU Wafer Implants: Retrospective Review in a Single Institution. World Neurosurg 2016; 90:340-347. [DOI: 10.1016/j.wneu.2016.02.102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 02/24/2016] [Accepted: 02/25/2016] [Indexed: 11/22/2022]
|
20
|
The ability of hyaluronan fragments to reverse the resistance of C6 rat glioma cell line to temozolomide and carmustine. Contemp Oncol (Pozn) 2014; 18:323-8. [PMID: 25477754 PMCID: PMC4248052 DOI: 10.5114/wo.2014.43493] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 12/11/2013] [Accepted: 04/11/2014] [Indexed: 11/18/2022] Open
Abstract
Aim of the study Hyaluronan (HA) is an extracellular matrix (ECM) polymer that may contribute to the emergence of anti-cancer drug resistance. Attempts to reverse drug resistance using small hyaluronan oligomers (oHA) are being made. The initial reports suggest that the oHA fraction may effectively reverse anti-cancer drug resistance in glioma models. However, the reversal effects of oHA of defined molecular length on glioma cells have not been investigated yet. In this study, we examined HA fragments containing 2 disaccharide units (oHA-2), 5 disaccharide units (oHA-5), and 68 kDa hyaluronan polymer (HA-68k) as agents possibly reversing the resistance of a C6 rat glioma cell line to temozolomide (TMZ) and carmustine (BCNU). Material and methods A 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) viability assay was used to assess the cytotoxicity of TMZ and BCNU in the presence or absence of the hyaluronan fragments. By comparing viability of the cells, the reversal effects of HA fragments on TMZ and BCNU resistance in C6 glioma cells were assessed. Results We found statistically significant decreases in the viability of cells in the presence of TMZ+oHA-5 as compared to TMZ alone (51.2 ±4.5 vs. 74.2 ±5.8, p = 0.0031), BCNU+o-HA5 as compared to BCNU alone (49.3 ±4.4 vs. 65.6 ±5.7, p = 0.0119), and BCNU+HA-68k as compared to BCNU alone (55.2 ±2.3 vs. 65.6 ±5.7, p = 0.0496). Conclusions Conclusions: Hyaluronan oligomers of 5 disaccharide units (oHA-5) significantly reversed the resistance of C6 cells to TMZ and BCNU. The results are only preliminary and a more thorough follow-up investigation is required to assess their actual role.
Collapse
|
21
|
Low-dose fractionated radiotherapy and concomitant chemotherapy for recurrent or progressive glioblastoma: final report of a pilot study. Strahlenther Onkol 2014; 190:370-6. [PMID: 24429479 DOI: 10.1007/s00066-013-0506-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 11/08/2013] [Indexed: 12/22/2022]
Abstract
BACKGROUND Evaluated in this study were the feasibility and the efficacy of concurrent low dose fractionated radiotherapy (LD-FRT) and chemotherapy as palliative treatment for recurrent/progressive glioblastoma multiforme (GBM). PATIENTS AND METHODS Eligible patients had recurrent or progressive GBM, Karnofsky performance status ≥ 70, prior surgery, and standard radiochemotherapy treatment. Recurrence/progression disease during temozolomide (TMZ) received cisplatin (CDDP; 30 mg/m(2) on days 1, 8, 15), fotemustine (FTM; 40 mg/m(2) on days 2, 9, 16), and concurrent LD-FRT (0.3 Gy twice daily); recurrence/progression after 4 months from the end of adjuvant TMZ were treated by TMZ (150/200 mg/m(2) on days 1-5) concomitant with LD-FRT (0.4 Gy twice daily). Primary endpoints were safety and toxicity. RESULTS A total of 32 patients were enrolled. Hematologic toxicity G1-2 was observed in 18.7 % of patients and G3-4 in 9.4 %. One patient (3.1 %) had complete response, 3 (9.4 %) had partial response, 8 (25 %) had stable disease for at least 8 weeks, while 20 patients (62.5 %) experienced progressive disease. The clinical benefit was 37.5 %. Median progression-free survival (PFS) and overall survival (OS) were 5 and 8 months, respectively. Survival rate at 12 months was of 27.8 %. CONCLUSION LD-FRT and chemotherapy for recurrent/progressive GBM have a good toxicity profile and clinical outcomes, even though further investigation of this novel palliative treatment approach is warranted.
Collapse
|
22
|
Stepensky D. Pharmacokinetic and Pharmacodynamic Aspects of Focal and Targeted Delivery of Drugs. ACTA ACUST UNITED AC 2013. [DOI: 10.1007/978-1-4614-9434-8_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2023]
|
23
|
Xu GF, Xie WF. Effect of ERBB2 expression on invasiveness of glioma TJ905 cells. ASIAN PAC J TROP MED 2013; 6:964-7. [DOI: 10.1016/s1995-7645(13)60172-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 10/15/2013] [Accepted: 11/15/2013] [Indexed: 11/16/2022] Open
|
24
|
Abstract
Patients with glioblastoma typically present when tumors are at an advanced stage. Surgical resection, radiotherapy and adjuvant chemotherapy are currently the standard of care for glioblastoma. However, due to the infiltrative and dispersive nature of the tumor, recurrence rate remains high and typically results in very poor prognosis. Efforts to treat the primary tumor are, therefore, palliative rather than curative. From a practical perspective, controlling growth and dispersal of the recurrence may have a greater impact on disease-free survival. In order for cells to disperse, they must first detach from the mass. Preventing detachment may keep tumors that recur more localized and perhaps more amenable to therapy. Here we introduce a new perspective in which a quantifiable mechanical property, namely tissue surface tension, can provide novel information on tumor behavior. The overall theme of the discussion will attempt to integrate how adhesion molecules can alter a tumor's mechanical properties and how, in turn, these properties can be modified to prevent tumor cell detachment and dispersal.
Collapse
Affiliation(s)
- Ramsey A Foty
- Department of Surgery, University of Medicine & Dentistry, New Jersey Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA.
| |
Collapse
|
25
|
Jilani K, Lang F. Carmustine-induced phosphatidylserine translocation in the erythrocyte membrane. Toxins (Basel) 2013; 5:703-16. [PMID: 23604064 PMCID: PMC3705288 DOI: 10.3390/toxins5040703] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 04/07/2013] [Accepted: 04/10/2013] [Indexed: 02/07/2023] Open
Abstract
The nitrosourea alkylating agent, carmustine, is used as chemotherapeutic drug in several malignancies. The substance triggers tumor cell apoptosis. Side effects of carmustine include myelotoxicity with anemia. At least in theory, anemia could partly be due to stimulation of eryptosis, the suicidal death of erythrocytes, characterized by cell shrinkage and breakdown of phosphatidylserine asymmetry of the cell membrane with phosphatidylserine exposure at the erythrocyte surface. Stimulators of eryptosis include increase of cytosolic Ca²⁺ activity ([Ca²⁺]i). The present study tested whether carmustine triggers eryptosis. To this end [Ca²⁺]i was estimated from Fluo3 fluorescence, cell volume from forward scatter, phosphatidylserine exposure from annexin V binding, and hemolysis from hemoglobin release. As a result a 48 h exposure to carmustine (≥25 µM) significantly increased [Ca²⁺]i, decreased forward scatter and increased annexin V binding. The effect on annexin V binding was significantly blunted in the absence of extracellular Ca²⁺. In conclusion, carmustine stimulates eryptosis at least partially by increasing cytosolic Ca²⁺ activity.
Collapse
Affiliation(s)
- Kashif Jilani
- Department of Physiology, University of Tuebingen, Gmelinstr. 5, Tuebingen D-72076, Germany.
| | | |
Collapse
|
26
|
Kleinberg L. Polifeprosan 20, 3.85% carmustine slow-release wafer in malignant glioma: evidence for role in era of standard adjuvant temozolomide. CORE EVIDENCE 2012; 7:115-30. [PMID: 23118709 PMCID: PMC3484478 DOI: 10.2147/ce.s23244] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Indexed: 11/23/2022]
Abstract
The Polifeprosan 20 with carmustine (BCNU, bis-chloroethylnitrosourea, Gliadel®) polymer implant wafer is a biodegradable compound containing 3.85% carmustine which slowly degrades to release carmustine and protects it from exposure to water with resultant hydrolysis until the time of release. The carmustine implant wafer was demonstrated to improve survival in blinded placebo-controlled trials in selected patients with newly diagnosed or recurrent malignant glioma, with little increased risk of adverse events. Based on these trials and other supporting data, US and European regulatory authorities granted approval for its use in recurrent and newly diagnosed malignant glioma, and it remains the only approved local treatment. The preclinical and clinical data suggest that it is optimally utilized primarily in the proportion of patients who may have total or near total removal of gross tumor. The aim of this work was to review the evidence for the use of carmustine implants in the management of malignant astrocytoma (World Health Organization grades III and IV), including newly diagnosed and recurrent disease, especially in the setting of a standard of care that has changed since the randomized trials were completed. Therapy has evolved such that patients now generally receive temozolomide chemotherapy during and after radiotherapy treatment. For patients undergoing repeat resection for malignant glioma, a randomized, blinded, placebo-controlled trial demonstrated a median survival for 110 patients who received carmustine polymers of 31 weeks compared with 23 weeks for 122 patients who only received placebo polymers. The benefit achieved statistical significance only on analysis adjusting for prognostic factors rather than for the randomized groups as a whole (hazard ratio = 0.67, P = 0.006). A blinded, placebo-controlled trial has also been performed for carmustine implant placement in newly diagnosed patients prior to standard radiotherapy. Median survival was improved from 11.6 to 13.9 months (P = 0.03), with a 29% reduction in the risk of death. When patients with glioblastoma multiforme alone were analyzed, the median survival improved from 11.4 to 13.5 months, but this improvement was not statistically significant. When a Cox’s proportional hazard model was utilized to account for other potential prognostic factors, there was a significant 31% reduction in the risk of death (P = 0.04) in this subgroup. Data from other small reports support these results and confirm that the incidence of adverse events does not appear to be increased meaningfully. Given the poor prognosis without possibility of cure, these benefits from a treatment with a favorable safety profile were considered meaningful. There is randomized evidence to support the use of carmustine wafers placed during resection of recurrent disease. Therefore, although there is limited specific evidence, this treatment is likely to be efficacious in an environment when nearly all patients receive temozolomide as part of initial management. Given that half of the patients in the randomized trial assessing the value of carmustine implants in recurrent disease had received prior chemotherapy, it is likely that this remains a valuable treatment at the time of repeat resection, even after temozolomide. There are data from multiple reports to support safety. Although there is randomized evidence to support the use of this therapy in newly diagnosed patients who will receive radiotherapy alone, it is now standard to administer both adjuvant temozolomide and radiotherapy. There are survival outcome reports for small cohorts of patients receiving temozolomide with radiotherapy, but this information is not sufficient to support firm recommendations. Based on the rationale and evidence of safety, this approach appears to be a reasonable option as more information is acquired. Available data support the safety of using carmustine wafers in this circumstance, although special attention to surgical guidelines for implanting the wafers is warranted.
Collapse
Affiliation(s)
- Lawrence Kleinberg
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Oncology Center Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
27
|
Rice KP, Klinkerch EJ, Gerber SA, Schleicher TR, Kraus TJ, Buros CM. Thioredoxin reductase is inhibited by the carbamoylating activity of the anticancer sulfonylhydrazine drug laromustine. Mol Cell Biochem 2012; 370:199-207. [PMID: 22864532 DOI: 10.1007/s11010-012-1411-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 07/25/2012] [Indexed: 12/24/2022]
Abstract
The thioredoxin system facilitates proliferative processes in cells and is upregulated in many cancers. The activities of both thioredoxin (Trx) and its reductase (TrxR) are mediated by oxidation/reduction reactions among cysteine residues. A common target in preclinical anticancer research, TrxR is reported here to be significantly inhibited by the anticancer agent laromustine. This agent, which has been in clinical trials for acute myelogenous leukemia and glioblastoma multiforme, is understood to be cytotoxic principally via interstrand DNA crosslinking that originates from a 2-chloroethylating species generated upon activation in situ. The spontaneous decomposition of laromustine also yields methyl isocyanate, which readily carbamoylates thiols and primary amines. Purified rat liver TrxR was inhibited by laromustine with a clinically relevant IC(50) value of 4.65 μM. A derivative of laromustine that lacks carbamoylating activity did not appreciably inhibit TrxR while another derivative, lacking only the 2-chloroethylating activity, retained its inhibitory potency. Furthermore, in assays measuring TrxR activity in murine cell lysates, a similar pattern of inhibition among these compounds was observed. These data contrast with previous studies demonstrating that glutathione reductase, another enzyme that relies on cysteine-mediated redox chemistry, was not inhibited by methylcarbamoylating agents when measured in cell lysates. Mass spectrometry of laromustine-treated enzyme revealed significant carbamoylation of TrxR, albeit not on known catalytically active residues. However, there was no evidence of 2-chloroethylation anywhere on the protein. The inhibition of TrxR is likely to contribute to the cytotoxic, anticancer mechanism of action for laromustine.
Collapse
Affiliation(s)
- Kevin P Rice
- Department of Chemistry, Colby College, Waterville, ME 04901, USA.
| | | | | | | | | | | |
Collapse
|
28
|
MRI-monitored long-term therapeutic hydrogel system for brain tumors without surgical resection. Biomaterials 2012; 33:4836-42. [DOI: 10.1016/j.biomaterials.2012.03.048] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 03/10/2012] [Indexed: 11/19/2022]
|
29
|
Gulati K, Aw MS, Losic D. Nanoengineered drug-releasing Ti wires as an alternative for local delivery of chemotherapeutics in the brain. Int J Nanomedicine 2012; 7:2069-76. [PMID: 22619543 PMCID: PMC3356184 DOI: 10.2147/ijn.s29917] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
The blood–brain barrier (BBB) blocks the passage of active molecules from the blood which makes drug delivery to the brain a challenging problem. Oral drug delivery using chemically modified drugs to enhance their transport properties or remove the blocking of drug transport across the BBB is explored as a common approach to address these problems, but with limited success. Local delivery of drugs directly to the brain interstitium using implants such as polymeric wafers, gels, and catheters has been recognized as a promising alternative particularly for the treatment of brain cancer (glioma) and neurodegenerative disorders. The aim of this study was to introduce a new solution by engineering a drug-releasing implant for local drug delivery in the brain, based on titanium (Ti) wires with titania nanotube (TNT) arrays on their surfaces. Drug loading and drug release characteristics of this system were explored using two drugs commonly used in oral brain therapy: dopamine (DOPA), a neurotransmitter agent; and doxorubicin (DOXO), an anticancer drug. Results showed that TNT/Ti wires could provide a considerable amount of drugs (>170 μg to 1000 μg) with desirable release kinetics and controllable release time (1 to several weeks) and proved their feasibility for use as drug-releasing implants for local drug delivery in the brain.
Collapse
Affiliation(s)
- Karan Gulati
- Ian Wark Research Institute, The University of South Australia, Adelaide, SA, Australia
| | | | | |
Collapse
|
30
|
Kamaly N, Xiao Z, Valencia PM, Radovic-Moreno AF, Farokhzad OC. Targeted polymeric therapeutic nanoparticles: design, development and clinical translation. Chem Soc Rev 2012; 41:2971-3010. [PMID: 22388185 PMCID: PMC3684255 DOI: 10.1039/c2cs15344k] [Citation(s) in RCA: 1146] [Impact Index Per Article: 88.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Polymeric materials have been used in a range of pharmaceutical and biotechnology products for more than 40 years. These materials have evolved from their earlier use as biodegradable products such as resorbable sutures, orthopaedic implants, macroscale and microscale drug delivery systems such as microparticles and wafers used as controlled drug release depots, to multifunctional nanoparticles (NPs) capable of targeting, and controlled release of therapeutic and diagnostic agents. These newer generations of targeted and controlled release polymeric NPs are now engineered to navigate the complex in vivo environment, and incorporate functionalities for achieving target specificity, control of drug concentration and exposure kinetics at the tissue, cell, and subcellular levels. Indeed this optimization of drug pharmacology as aided by careful design of multifunctional NPs can lead to improved drug safety and efficacy, and may be complimentary to drug enhancements that are traditionally achieved by medicinal chemistry. In this regard, polymeric NPs have the potential to result in a highly differentiated new class of therapeutics, distinct from the original active drugs used in their composition, and distinct from first generation NPs that largely facilitated drug formulation. A greater flexibility in the design of drug molecules themselves may also be facilitated following their incorporation into NPs, as drug properties (solubility, metabolism, plasma binding, biodistribution, target tissue accumulation) will no longer be constrained to the same extent by drug chemical composition, but also become in-part the function of the physicochemical properties of the NP. The combination of optimally designed drugs with optimally engineered polymeric NPs opens up the possibility of improved clinical outcomes that may not be achievable with the administration of drugs in their conventional form. In this critical review, we aim to provide insights into the design and development of targeted polymeric NPs and to highlight the challenges associated with the engineering of this novel class of therapeutics, including considerations of NP design optimization, development and biophysicochemical properties. Additionally, we highlight some recent examples from the literature, which demonstrate current trends and novel concepts in both the design and utility of targeted polymeric NPs (444 references).
Collapse
Affiliation(s)
- Nazila Kamaly
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Zeyu Xiao
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Pedro M. Valencia
- The David H. Koch Institute for Integrative Cancer Research and Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Aleksandar F. Radovic-Moreno
- The David H. Koch Institute for Integrative Cancer Research and Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Omid C. Farokhzad
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
31
|
Ulmer S, Spalek K, Nabavi A, Schultka S, Mehdorn HM, Kesari S, Dörner L. Temporal changes in magnetic resonance imaging characteristics of Gliadel wafers and of the adjacent brain parenchyma. Neuro Oncol 2012; 14:482-90. [PMID: 22319220 DOI: 10.1093/neuonc/nos003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Carmustine is used in the treatment of glioblastomas as locally applied chemotherapy in the form of biodegradable wafers, which are lined on the walls of the resection cavity at the end of the resection, to increase local concentrations and decrease systemic toxicity. A total of 44 patients with glioblastoma with gross macroscopic tumor removal were included. MRIs were performed at various times postoperatively (within 24 hours, 1 week, 1 month, 2 months, 3 months, 6 months, 9 months, and 1 year). MR protocols included a T2-, diffusion-weighted, and T1-weighted sequences with and without intravenous administration of gadolinium. On T1, the wafers change from their initial hypointense to an isointense appearance after a period during which they appear to be hypointense, with a hyperintense rim most prominent less than 1 month postoperatively. On T2 they change from a hypointense to an isointense appearance. Restricted diffusivity reshaping the silhouette of the wafer's surface at the rim of the resection cavity can be found as early as day 1 postoperatively; however, 1 month after implantation, they all show areas of restricted diffusion, which may remain up to 1 year. Contrast enhancement at the rim of the resection cavity can already be found at day 1 postoperatively, with a peak shortly after 1 month after surgery. These changes can easily be mistaken for an abscess and hamper the early differentiation between residual tumor tissue and normal postoperative changes. However, early changes in either appearance do not predict overall survival or the progression free interval.
Collapse
Affiliation(s)
- Stephan Ulmer
- Diagnostic and Interventional Neuroradiology, University Hospital of Basel, Basel, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
32
|
Prognostic factors and survival in a prospective cohort of patients with high-grade glioma treated with carmustine wafers or temozolomide on an intention-to-treat basis. Acta Neurochir (Wien) 2012; 154:211-22; discussion 222. [PMID: 22002506 DOI: 10.1007/s00701-011-1199-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 09/29/2011] [Indexed: 10/17/2022]
Abstract
BACKGROUND Patients with high-grade glioma can be treated with carmustine wafers or following the Stupp protocol. As far as we are aware, no scientific evidence has been published comparing the two treatments. The primary objective of this study was to analyse the survival of groups of patients with each of these treatment modalities. The secondary objective was to assess the influence of the usual prognostic factors on the patients in our hospital. METHODS A prospective cohort of 110 patients with single, supratentorial high-grade glioma treated by craniotomy and tumour resection was retrospectively studied. Half of the patients had carmustine wafers placed during this operation while the others (55) did not, the latter group receiving first-line systemic chemotherapy on an intention-to-treat basis. FINDINGS Patients treated with carmustine wafers had a median survival of 13.414 months compared with 11.047 in the group without implants (p = 0.856). For the overall cohort of patients, the following factors were found to influence survival: age (p < 0.0001), postoperative KPS score (p = 0.001), histological grade (p = 0.004), RPA class (p = 0.001), extent of resection (p = 0.002) and salvage surgery (p = 0.028). CONCLUSIONS In this prospective cohort of patients, analysed on the basis of intention-to-treat at the time of the first surgery, no statistically significant differences in survival were found between the two treatment modalities (carmustine wafers vs. first-line systemic chemotherapy). On the other hand, age, preoperative KPS, histological grade, and RPA class were confirmed to be prognostic factors in this cohort. Finally, the extent of resection was also found to influence survival.
Collapse
|
33
|
Persano L, Rampazzo E, Della Puppa A, Pistollato F, Basso G. The three-layer concentric model of glioblastoma: cancer stem cells, microenvironmental regulation, and therapeutic implications. ScientificWorldJournal 2011; 11:1829-41. [PMID: 22125441 PMCID: PMC3217608 DOI: 10.1100/2011/736480] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Accepted: 09/29/2011] [Indexed: 12/15/2022] Open
Abstract
Tumors arising in the central nervous system are thought to
originate from a sub-population of cells named cancer stem cells
(CSCs) or tumor initiating cells (TICs) that possess an immature
phenotype, combined with self-renewal and chemotherapy resistance
capacity. Moreover, in the last years, these cells have been
identified in particular brain tumor niches fundamental for
supporting their characteristics. In this paper, we report studies
from many authors demonstrating that hypoxia or the so called
“hypoxic niche” plays a crucial role in controlling CSC molecular
and phenotypic profile. We recently investigated the relationship
existing between Glioblastoma (GBM) stem cells and their niche,
defining the theory of three-concentric layers model for GBM mass.
According to this model, GBM stem cells reside preferentially
within the hypoxic core of the tumour mass, while more
differentiated cells are mainly localized along the peripheral and
vascularized part of the tumour. This GBM model provides
explanation of the effects mediated by the tumour microenvironment
on the phenotypic and molecular regulation of GBM stem cells,
describing their spatial distribution in the tumor bulk. Moreover,
we discuss the possible clinical implications of the creation of
this model for future GBM patient management and novel therapeutic
strategies development.
Collapse
Affiliation(s)
- Luca Persano
- Oncohematology Laboratory, Department of Paediatrics, University of Padova, Via Giustiniani 3, Padova 35128, Italy.
| | | | | | | | | |
Collapse
|
34
|
|
35
|
Niyazi M, Siefert A, Schwarz SB, Ganswindt U, Kreth FW, Tonn JC, Belka C. Therapeutic options for recurrent malignant glioma. Radiother Oncol 2010; 98:1-14. [PMID: 21159396 DOI: 10.1016/j.radonc.2010.11.006] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 10/08/2010] [Accepted: 11/07/2010] [Indexed: 01/17/2023]
Abstract
BACKGROUND AND PURPOSE Despite the given advances in neuro-oncology most patients with high grade malignant glioma ultimately fail locally or locoregionally. In parallel with improvements of initial treatment options, several salvage strategies have been elucidated and already entered clinical practice. Aim of this article is to review the current status of salvage strategies in recurrent high grade glioma. MATERIAL AND METHODS Using the following MESH headings and combinations of these terms the pubmed database was searched: "Glioma", "Recurrence", "Neoplasm Recurrence, Local", "Radiosurgery", "Brachytherapy", "Neurosurgical Procedures" and "Drug Therapy". For citation crosscheck the ISI web of science database was used employing the same search terms. In parallel, the abstracts of ASCO 2008-2009 were analyzed accordingly. RESULTS Currently the following options for salvage entered clinical practice: re-resection, re-irradiation (stereotactic radiosurgery, (hypo-)fractionated (stereotactic) radiotherapy, interstitial brachytherapy) or single/poly-chemotherapy schedules including new dose-intensified or alternative treatment protocols employing targeted drugs. Re-operation is associated with high morbidity and mortality, however, is an option in a highly selected patient cohort. Since toxicity has been overestimated, re-irradiation is an increasingly used option with precise fractionated radiotherapy being the most optimal technique. On average, time to secondary progression is in the range of several months. Conventional chemotherapy regimens also improve time to secondary progression; however the efficacy is only modest and treatment-related toxicities like myelo-suppression occur very frequently. Molecular targeted agents/kinases are undergoing clinical testing; however no final recommendations can be made. CONCLUSIONS Currently, several re-treatment options with only modest efficacy exist. The relative value of each approach compared to other options is unknown as well as it remains open which sequence of modalities should be chosen.
Collapse
Affiliation(s)
- Maximilian Niyazi
- Department of Radiation Oncology, Ludwig-Maximilians-University Munich, München, Germany
| | | | | | | | | | | | | |
Collapse
|
36
|
Chen PY, Liu HL, Hua MY, Yang HW, Huang CY, Chu PC, Lyu LA, Tseng IC, Feng LY, Tsai HC, Chen SM, Lu YJ, Wang JJ, Yen TC, Ma YH, Wu T, Chen JP, Chuang JI, Shin JW, Hsueh C, Wei KC. Novel magnetic/ultrasound focusing system enhances nanoparticle drug delivery for glioma treatment. Neuro Oncol 2010; 12:1050-60. [PMID: 20663792 DOI: 10.1093/neuonc/noq054] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Malignant glioma is a common and severe primary brain tumor with a high recurrence rate and an extremely high mortality rate within 2 years of diagnosis, even when surgical, radiological, and chemotherapeutic interventions are applied. Intravenously administered drugs have limited use because of their adverse systemic effects and poor blood-brain barrier penetration. Here, we combine 2 methods to increase drug delivery to brain tumors. Focused ultrasound transiently permeabilizes the blood-brain barrier, increasing passive diffusion. Subsequent application of an external magnetic field then actively enhances localization of a chemotherapeutic agent immobilized on a novel magnetic nanoparticle. Combining these techniques significantly improved the delivery of 1,3-bis(2-chloroethyl)-1-nitrosourea to rodent gliomas. Furthermore, the physicochemical properties of the nanoparticles allowed their delivery to be monitored by magnetic resonance imaging (MRI). The resulting suppression of tumor progression without damaging the normal regions of the brain was verified by MRI and histological examination. This noninvasive, reversible technique promises to provide a more effective and tolerable means of tumor treatment, with lower therapeutic doses and concurrent clinical monitoring.
Collapse
Affiliation(s)
- Pin-Yuan Chen
- Department of Neurosurgery, Chang-Gung University and Memorial Hospital, Taoyuan 333
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Baltes S, Freund I, Lewis AL, Nolte I, Brinker T. Doxorubicin and irinotecan drug-eluting beads for treatment of glioma: a pilot study in a rat model. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2010; 21:1393-402. [PMID: 20162337 DOI: 10.1007/s10856-009-3803-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2009] [Accepted: 06/10/2009] [Indexed: 05/05/2023]
Abstract
Despite some progress in therapy, the prognosis of patients with malignant gliomas remains poor. Local delivery of cytostatics to the tumour has been proven to be an efficacious therapeutic approach but which nevertheless needs further improvements. Drug Eluting Beads (DEB), have been developed as drug delivery embolisation systems for use in trans-arterial chemoembolisation. We tested in a rat model of malignant glioma, whether DEB, loaded with doxorubicin or irinotecan, may be used for local treatment of brain tumours. Unloaded and drug loaded DEB were implanted into the brains of healthy and tumour bearing BD IX rats followed by histological investigations and survival assessment. Intracerebral implantation of unloaded DEB caused no significant local tissue damage, whilst both doxorubicin and irinotecan DEB improved survival time significantly. However, a significant local toxicity was found after the implantation of doxorubicin DEB but not with irinotecan DEB. We concluded that irinotecan appears to be superior in terms of the risk-benefit ratio and that DEB may be used for local treatment of brain tumours.
Collapse
Affiliation(s)
- Steffen Baltes
- International Neuroscience Institute GmbH, Hannover, Germany
| | | | | | | | | |
Collapse
|
38
|
Morales-Ramírez P, Vallarino-Kelly T, Cruz-Vallejo VL. Effect of O6-chloroethylguanine DNA lesions on the kinetics and mechanism of micronucleus induction in vivo. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2010; 51:236-242. [PMID: 19844954 DOI: 10.1002/em.20538] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
The aim of this work was to determine the kinetics of micronucleus production because of an increase in O(6)-chloroethyl guanine (O6-ChlEt-G) DNA lesions in murine bone marrow cells in vivo. We increased the frequency of O6-ChlEt-G lesions by pretreatment with an inhibitor of O(6)-methylguanine-DNA methyltransferase (MGMT), O(6)-benzylguanine (O6BG), and subsequent treatment with bis-chloroethylnitrosourea (BCNU). The kinetics of micronucleated-polychromatic erythrocyte (MN-PCE) induction was established by scoring the frequency of MN-PCEs per 2000 PCEs in peripheral blood at 8-hr intervals from immediately prior to treatment to 72-hr post-treatment. We examined groups of five mice treated with (i) dimethylsulfoxide (DMSO), (ii) O6BG in DMSO, (iii) BCNU, or (iv) O6BG in DMSO plus BCNU. The data indicate that O6BG pretreatment causes: (i) ían increase in MN-PCEs induced by BCNU, (ii) a delay in the time of maximal MN-PCE induction produced by the different BCNU doses, and (iii) an increase in cytotoxicity. These data confirm that O6-ChlEt-G is a lesion involved in DNA break induction and in the subsequent production of micronuclei, and also that these lesions seem to be stoichiometrically reduced by MGMT. These data also show that induction of MN-PCEs by BCNU is delayed by pretreatment with O6BG for more than 6 hr, perhaps due to the time required for repair of crosslinks derived from O6-ChlEt-G and/or for DNA duplication, which is required for adduct transformation into crosslinks.
Collapse
Affiliation(s)
- P Morales-Ramírez
- Instituto Nacional de Investigaciones Nucleares, AP 18-1027 México, DF, México. pedro.morales@ inin.gob.mx
| | | | | |
Collapse
|
39
|
Balossier A, Dörner L, Emery E, Heese O, Mehdorn HM, Menei P, Singh J. Incorporating BCNU Wafers into Malignant Glioma Treatment. Clin Drug Investig 2010; 30:195-204. [DOI: 10.2165/11532900-000000000-00000] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
40
|
Sarin H, Kanevsky AS, Wu H, Brimacombe KR, Fung SH, Sousa AA, Auh S, Wilson CM, Sharma K, Aronova MA, Leapman RD, Griffiths GL, Hall MD. Effective transvascular delivery of nanoparticles across the blood-brain tumor barrier into malignant glioma cells. J Transl Med 2008; 6:80. [PMID: 19094226 PMCID: PMC2639552 DOI: 10.1186/1479-5876-6-80] [Citation(s) in RCA: 182] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2008] [Accepted: 12/18/2008] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Effective transvascular delivery of nanoparticle-based chemotherapeutics across the blood-brain tumor barrier of malignant gliomas remains a challenge. This is due to our limited understanding of nanoparticle properties in relation to the physiologic size of pores within the blood-brain tumor barrier. Polyamidoamine dendrimers are particularly small multigenerational nanoparticles with uniform sizes within each generation. Dendrimer sizes increase by only 1 to 2 nm with each successive generation. Using functionalized polyamidoamine dendrimer generations 1 through 8, we investigated how nanoparticle size influences particle accumulation within malignant glioma cells. METHODS Magnetic resonance and fluorescence imaging probes were conjugated to the dendrimer terminal amines. Functionalized dendrimers were administered intravenously to rodents with orthotopically grown malignant gliomas. Transvascular transport and accumulation of the nanoparticles in brain tumor tissue was measured in vivo with dynamic contrast-enhanced magnetic resonance imaging. Localization of the nanoparticles within glioma cells was confirmed ex vivo with fluorescence imaging. RESULTS We found that the intravenously administered functionalized dendrimers less than approximately 11.7 to 11.9 nm in diameter were able to traverse pores of the blood-brain tumor barrier of RG-2 malignant gliomas, while larger ones could not. Of the permeable functionalized dendrimer generations, those that possessed long blood half-lives could accumulate within glioma cells. CONCLUSION The therapeutically relevant upper limit of blood-brain tumor barrier pore size is approximately 11.7 to 11.9 nm. Therefore, effective transvascular drug delivery into malignant glioma cells can be accomplished by using nanoparticles that are smaller than 11.7 to 11.9 nm in diameter and possess long blood half-lives.
Collapse
Affiliation(s)
- Hemant Sarin
- National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, USA
- Diagnostic Radiology Department, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Ariel S Kanevsky
- Diagnostic Radiology Department, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Haitao Wu
- Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Kyle R Brimacombe
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Steve H Fung
- Neuroradiology Department, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Alioscka A Sousa
- National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Sungyoung Auh
- Biostatistics, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Colin M Wilson
- Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Kamal Sharma
- Metabolism Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
- Division of Biologic Drug Products, Office of Oncology Products, Center for Drug Evaluation and Research, U.S. Food & Drug Administration, Silver Spring, Maryland 20993, USA
| | - Maria A Aronova
- National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Richard D Leapman
- National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Gary L Griffiths
- Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Matthew D Hall
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
41
|
Parvez T. Present trend in the primary treatment of aggressive malignant glioma: glioblastoma multiforme. Technol Cancer Res Treat 2008; 7:241-8. [PMID: 18473496 DOI: 10.1177/153303460800700310] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The standard treatment for glioblastoma multiforme is surgery, radiation, and chemotherapy. Yet this aggressive therapy has only a modest effect on survival with most patients surviving less than 1 year after diagnosis. This poor prognosis has lead scientists to seek alternative molecular approaches for the treatment of glioblastoma multiforme. Among these, gene therapy, vaccine therapy, and immunotherapy are all approaches that are currently being investigated. While these molecular approaches may not herald an immediate change in the prognosis of these aggressive tumors, combining them with existing approaches may bring some progress in the standard of care. This paper reviews current treatments and several newer therapies in preclinical and early clinical studies.
Collapse
Affiliation(s)
- Tariq Parvez
- Medical Director II Oncology, PRA International, Charlottesville, VA 22911, USA.
| |
Collapse
|