1
|
Doron O, Wong T, Ablyazova F, Singha S, Cavallaro J, Ben-Shalom N, D'Amico RS, Harshan M, McKeown A, Zlochower A, Langer DJ, Boockvar JA. Results from a first-in-human phase I safety trial to evaluate the use of a vascularized pericranial/temporoparietal fascial flap to line the resection cavity following resection of newly diagnosed glioblastoma. J Neurooncol 2024; 168:225-235. [PMID: 38664311 PMCID: PMC11147875 DOI: 10.1007/s11060-024-04647-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 03/13/2024] [Indexed: 05/18/2024]
Abstract
PURPOSE The efficacy of systemic therapies for glioblastoma (GBM) remains limited due to the constraints of systemic toxicity and blood-brain barrier (BBB) permeability. Temporoparietal fascial flaps (TPFFs) and vascularized peri cranial flaps (PCF) are not restricted by the blood-brain barrier (BBB), as they derive their vascular supply from branches of the external carotid artery. Transposition of a vascularized TPFF or PCF along a GBM resection cavity may bring autologous tissue not restricted by the BBB in close vicinity to the tumor bed microenvironment, permit ingrowth of vascular channels fed by the external circulation, and offer a mechanism of bypassing the BBB. In addition, circulating immune cells in the vascularized flap may have better access to tumor-associated antigens (TAA) within the tumor microenvironment. We conducted a first-in-human Phase I trial assessing the safety of lining the resection cavity with autologous TPFF/PCF of newly diagnosed patients with GBM. METHODS 12 patients underwent safe, maximal surgical resection of newly diagnosed GBMs, followed by lining of the resection cavity with a pedicled, autologous TPFF or PCF. Safety was assessed by monitoring adverse events. Secondary analysis of efficacy was examined as the proportion of patients experiencing progression-free disease (PFS) as indicated by response assessment in neuro-oncology (RANO) criteria and overall survival (OS). The study was powered to determine whether a Phase II study was warranted based on these early results. For this analysis, subjects who were alive and had not progressed as of the date of the last follow-up were considered censored and all living patients who were alive as of the date of last follow-up were considered censored for overall survival. For simplicity, we assumed that a 70% PFS rate at 6 months would be considered an encouraging response and would make an argument for further investigation of the procedure. RESULTS Median age of included patients was 57 years (range 46-69 years). All patients were Isocitrate dehydrogenase (IDH) wildtype. Average tumor volume was 56.6 cm3 (range 14-145 cm3). Resection was qualified as gross total resection (GTR) of all of the enhancing diseases in all patients. Grade III or above adverse events were encountered in 3 patients. No Grade IV or V serious adverse events occurred in the immediate post-operative period including seizure, infection, stroke, or tumor growing along the flap. Disease progression at the site of the original tumor was identified in only 4 (33%) patients (median 23 months, range 8-25 months), 3 of whom underwent re-operation. Histopathological analyses of those implanted flaps and tumor bed biopsy at repeat surgery demonstrated robust immune infiltrates within the transplanted flap. Importantly, no patient demonstrated evidence of tumor infiltration into the implanted flap. At the time of this manuscript preparation, only 4/12 (33%) of patients have died. Based on the statistical considerations above and including all 12 patients 10/12 (83.3%) had 6-month PFS. The median PFS was 9.10 months, and the OS was 17.6 months. 4/12 (33%) of patients have been alive for more than two years and our longest surviving patient currently is alive at 60 months. CONCLUSIONS This pilot study suggests that insertion of pedicled autologous TPFF/PCF along a GBM resection cavity is safe and feasible. Based on the encouraging response rate in 6-month PFS and OS, larger phase II studies are warranted to assess and reproduce safety, feasibility, and efficacy. TRIAL REGISTRATION NUMBER AND DATE OF REGISTRATION FOR PROSPECTIVELY REGISTERED TRIALS: ClinicalTrials.gov ID NCT03630289, dated: 08/02/2018.
Collapse
Affiliation(s)
- Omer Doron
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, 130 East 77Th Street New York,, New York, NY, 10075, USA
- Department of Biomedical Engineering, The Aldar and Iby Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Tamika Wong
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, 130 East 77Th Street New York,, New York, NY, 10075, USA
| | - Faina Ablyazova
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, 130 East 77Th Street New York,, New York, NY, 10075, USA
| | - Souvik Singha
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, 130 East 77Th Street New York,, New York, NY, 10075, USA
| | - Julianna Cavallaro
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, 130 East 77Th Street New York,, New York, NY, 10075, USA
| | - Netanel Ben-Shalom
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, 130 East 77Th Street New York,, New York, NY, 10075, USA
| | - Randy S D'Amico
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, 130 East 77Th Street New York,, New York, NY, 10075, USA
| | - Manju Harshan
- Department of Pathology, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, 130 East 77Th Street New York,, New York, NY, 10075, USA
| | - Amy McKeown
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, 130 East 77Th Street New York,, New York, NY, 10075, USA
| | - Avraham Zlochower
- Department of Radiology, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, 130 East 77Th Street New York,, New York, NY, 10075, USA
| | - David J Langer
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, 130 East 77Th Street New York,, New York, NY, 10075, USA
| | - John A Boockvar
- Department of Neurosurgery, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, 130 East 77Th Street New York,, New York, NY, 10075, USA.
| |
Collapse
|
2
|
Tian M, Ma Z, Yang GZ. Micro/nanosystems for controllable drug delivery to the brain. Innovation (N Y) 2024; 5:100548. [PMID: 38161522 PMCID: PMC10757293 DOI: 10.1016/j.xinn.2023.100548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 11/26/2023] [Indexed: 01/03/2024] Open
Abstract
Drug delivery to the brain is crucial in the treatment for central nervous system disorders. While significant progress has been made in recent years, there are still major challenges in achieving controllable drug delivery to the brain. Unmet clinical needs arise from various factors, including controlled drug transport, handling large drug doses, methods for crossing biological barriers, the use of imaging guidance, and effective models for analyzing drug delivery. Recent advances in micro/nanosystems have shown promise in addressing some of these challenges. These include the utilization of microfluidic platforms to test and validate the drug delivery process in a controlled and biomimetic setting, the development of novel micro/nanocarriers for large drug loads across the blood-brain barrier, and the implementation of micro-intervention systems for delivering drugs through intraparenchymal or peripheral routes. In this article, we present a review of the latest developments in micro/nanosystems for controllable drug delivery to the brain. We also delve into the relevant diseases, biological barriers, and conventional methods. In addition, we discuss future prospects and the development of emerging robotic micro/nanosystems equipped with directed transportation, real-time image guidance, and closed-loop control.
Collapse
Affiliation(s)
- Mingzhen Tian
- Institute of Medical Robotics, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhichao Ma
- Institute of Medical Robotics, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Guang-Zhong Yang
- Institute of Medical Robotics, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
3
|
Mahvi DA, Korunes-Miller J, Bordeianu C, Chu NQ, Geller AD, Sabatelle R, Berry S, Hung YP, Colson YL, Grinstaff MW, Raut CP. High dose, dual-release polymeric films for extended surgical bed paclitaxel delivery. J Control Release 2023; 363:682-691. [PMID: 37776906 PMCID: PMC10990290 DOI: 10.1016/j.jconrel.2023.09.048] [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: 12/02/2022] [Revised: 08/17/2023] [Accepted: 09/27/2023] [Indexed: 10/02/2023]
Abstract
While surgery represents a major therapy for most solid organ cancers, local recurrence is clinically problematic for cancers such as sarcoma for which adjuvant radiotherapy and systemic chemotherapy provide minimal local control or survival benefit and are dose-limited due to off-target side effects. We describe an implantable, biodegradable poly(1,2-glycerol carbonate) and poly(caprolactone) film with entrapped and covalently-bound paclitaxel enabling safe, controlled, and extended local delivery of paclitaxel achieving concentrations 10,000× tissue levels compared to systemic administration. Films containing entrapped and covalently-bound paclitaxel implanted in the tumor bed, immediately after resection of human cell line-derived chondrosarcoma and patient-derived xenograft liposarcoma and leiomyosarcoma in mice, improve median 90- or 200-day recurrence-free and overall survival compared to control mice. Furthermore, mice in the experimental film arm show no film-related morbidity. Continuous, extended, high-dose paclitaxel delivery via this unique polymer platform safely improves outcomes in three different sarcoma models and provides a rationale for future incorporation into human trials.
Collapse
Affiliation(s)
- David A Mahvi
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States of America
| | - Jenny Korunes-Miller
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States of America
| | - Catalina Bordeianu
- Department of Chemistry, Boston University, Boston, MA 02215, United States of America
| | - Ngoc-Quynh Chu
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States of America; Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States of America
| | - Abraham D Geller
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States of America
| | - Robbie Sabatelle
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States of America
| | - Samantha Berry
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States of America
| | - Yin P Hung
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States of America
| | - Yolonda L Colson
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States of America.
| | - Mark W Grinstaff
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States of America; Department of Chemistry, Boston University, Boston, MA 02215, United States of America.
| | - Chandrajit P Raut
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States of America; Center for Sarcoma and Bone Oncology, Dana Farber Cancer Institute, Boston, MA 02115, United States of America.
| |
Collapse
|
4
|
Gazaille C, Bozzato E, Madadian-Bozorg N, Mellinger A, Sicot M, Farooq U, Saulnier P, Eyer J, Préat V, Bertrand N, Bastiat G. Glioblastoma-targeted, local and sustained drug delivery system based on an unconventional lipid nanocapsule hydrogel. BIOMATERIALS ADVANCES 2023; 153:213549. [PMID: 37453243 DOI: 10.1016/j.bioadv.2023.213549] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/21/2023] [Accepted: 07/01/2023] [Indexed: 07/18/2023]
Abstract
The objective of this work was to develop an implantable therapeutic hydrogel that will ensure continuity in treatment between surgery and radiochemotherapy for patients with glioblastoma (GBM). A hydrogel of self-associated gemcitabine-loaded lipid nanocapsules (LNC) has shown therapeutic efficacy in vivo in murine GBM resection models. To improve the targeting of GBM cells, the NFL-TBS.40-63 peptide (NFL), was associated with LNC. The LNC-based hydrogels were formulated with the NFL. The peptide was totally and instantaneously adsorbed at the LNC surface, without modifying the hydrogel mechanical properties, and remained adsorbed to the LNC surface after the hydrogel dissolution. In vitro studies on GBM cell lines showed a faster internalization of the LNC and enhanced cytotoxicity, in the presence of NFL. Finally, in vivo studies in the murine GBM resection model proved that the gemcitabine-loaded LNC with adsorbed NFL could target the non-resected GBM cells and significantly delay or even inhibit the apparition of recurrences.
Collapse
Affiliation(s)
- Claire Gazaille
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | | | | | - Adélie Mellinger
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | - Marion Sicot
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | - Umer Farooq
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | - Patrick Saulnier
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | - Joël Eyer
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | | | - Nicolas Bertrand
- Univ Laval, Faculty of Pharmacy, CHU Quebec Research Center, Québec, QC, Canada
| | | |
Collapse
|
5
|
Krajcer A, Grzywna E, Lewandowska-Łańcucka J. Strategies increasing the effectiveness of temozolomide at various levels of anti-GBL therapy. Biomed Pharmacother 2023; 165:115174. [PMID: 37459661 DOI: 10.1016/j.biopha.2023.115174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 08/17/2023] Open
Abstract
Glioblastoma (GBL) is the most common (60-70% of primary brain tumours) and the most malignant of the glial tumours. Although current therapies remain palliative, they have been proven to prolong overall survival. Within an optimal treatment regimen (incl. surgical resection, radiation therapy, and chemotherapy) temozolomide as the current anti-GBL first-line chemotherapeutic has increased the median overall survival to 14-15 months, and the percentage of patients alive at two years has been reported to rise from 10.4% to 26.5%. Though, the effectiveness of temozolomide chemotherapy is limited by the serious systemic, dose-related side effects. Therefore, the ponderation regarding novel treatment methods along with innovative formulations is crucial to emerging the therapeutic potential of the widely used drug simultaneously reducing the drawbacks of its use. Herein the complex temozolomide application restrictions present at different levels of therapy as well as, the currently proposed strategies aimed at reducing those limitations are demonstrated. Approaches increasing the efficacy of anti-GBL treatment are addressed. Our paper is focused on the most recent developments in the field of nano/biomaterials-based systems for temozolomide delivery and their functionalization towards more effective blood-brain-barrier crossing and/or tumour targeting. Appropriate designing accounting for the physical and chemical features of formulations along with distinct routes of administration is also discussed. In addition, considering the multiple resistance mechanisms, the molecular heterogeneity and the evolution of tumour the purposely selected delivery methods, the combined therapeutic approaches and specifically focused on GBL cells therapies are reviewed.
Collapse
Affiliation(s)
- Aleksandra Krajcer
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Kraków, Poland
| | - Ewelina Grzywna
- Department of Neurosurgery and Neurotraumatology, Jagiellonian University Medical College, Św. Anny 12, 31-008 Kraków, Poland
| | | |
Collapse
|
6
|
Padmakumar S, Amiji MM. Long-Acting Therapeutic Delivery Systems for the Treatment of Gliomas. Adv Drug Deliv Rev 2023; 197:114853. [PMID: 37149040 DOI: 10.1016/j.addr.2023.114853] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 04/13/2023] [Accepted: 04/23/2023] [Indexed: 05/08/2023]
Abstract
Despite the emergence of cutting-edge therapeutic strategies and tremendous progress in research, a complete cure of glioma remains elusive. The heterogenous nature of tumor, immunosuppressive state and presence of blood brain barrier are few of the major obstacles in this regard. Long-acting depot formulations such as injectables and implantables are gaining attention for drug delivery to brain owing to their ease in administration and ability to elute drug locally for extended durations in a controlled manner with minimal toxicity. Hybrid matrices fabricated by incorporating nanoparticulates within such systems help to enhance pharmaceutical advantages. Utilization of long-acting depots as monotherapy or in conjunction with existing strategies rendered significant survival benefits in many preclinical studies and some clinical trials. The discovery of novel targets, immunotherapeutic strategies and alternative drug administration routes are now coupled with several long-acting systems with an ultimate aim to enhance patient survival and prevent glioma recurrences.
Collapse
Affiliation(s)
- Smrithi Padmakumar
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, 02115
| | - Mansoor M Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, 02115; Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, MA, 02115.
| |
Collapse
|
7
|
Haim O, Agur A, Efrat OT, Valdes P, Ram Z, Grossman R. The clinical significance of radiological changes associated with gliadel implantation in patients with recurrent high grade glioma. Sci Rep 2023; 13:11. [PMID: 36593342 PMCID: PMC9807577 DOI: 10.1038/s41598-022-27128-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 12/26/2022] [Indexed: 01/04/2023] Open
Abstract
Gliadel occasionally induces edema following its implantation. We aimed to correlate such post-surgical radiological changes to its efficacy and subsequent survival. Fifty-six patients with recurrent high grade glioma were treated between 2005 and 2016 with Gliadel implantation. Volumetric measurements of MRI features, including FLAIR abnormalities, tumor bulk (volume of gadolinium enhancement on T1) and resection cavity volumes over time were conducted. To assess dynamics over time, linear regression trendlines for each of these were calculated and examined to correlate with survival. Median follow-up after resection was 21.5 months. Median survival post-Gliadel implantation and overall survival since diagnosis were 12 months and 22 months, respectively. A subgroup of patients (n = 6) with a transient increase in FLAIR changes volume over time survived significantly longer post-Gliadel compared to those who did not demonstrate such change (36 vs 12 months, p = .03). Positive trends, representing overall growth in volume over time, of tumor bulk and resection cavity predicted survival in multivariate analyses (hazard ratios 7.9 and 84, p = .003 and .002, respectively). Increase in tumor bulk and resection cavity over time were associated with decreased survival, while transient FLAIR increase was a favorable prognostic factor. This may represent a transient inflammatory process in the tumor, possibly stemming from a presumed immune-mediated anti-tumor response.
Collapse
Affiliation(s)
- Oz Haim
- grid.12136.370000 0004 1937 0546Department of Neurosurgery, Tel-Aviv Medical Center, affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, 6 Weizman Street, 6423906 Tel-Aviv, Israel
| | - Ariel Agur
- grid.12136.370000 0004 1937 0546Department of Neurosurgery, Tel-Aviv Medical Center, affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, 6 Weizman Street, 6423906 Tel-Aviv, Israel
| | - Or-Tal Efrat
- grid.12136.370000 0004 1937 0546Department of Neurosurgery, Tel-Aviv Medical Center, affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, 6 Weizman Street, 6423906 Tel-Aviv, Israel
| | - Pablo Valdes
- grid.12136.370000 0004 1937 0546Department of Neurosurgery, Tel-Aviv Medical Center, affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, 6 Weizman Street, 6423906 Tel-Aviv, Israel
| | - Zvi Ram
- grid.12136.370000 0004 1937 0546Department of Neurosurgery, Tel-Aviv Medical Center, affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, 6 Weizman Street, 6423906 Tel-Aviv, Israel
| | - Rachel Grossman
- grid.12136.370000 0004 1937 0546Department of Neurosurgery, Tel-Aviv Medical Center, affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, 6 Weizman Street, 6423906 Tel-Aviv, Israel
| |
Collapse
|
8
|
Śledzińska P, Bebyn M, Furtak J, Koper A, Koper K. Current and promising treatment strategies in glioma. Rev Neurosci 2022:revneuro-2022-0060. [PMID: 36062548 DOI: 10.1515/revneuro-2022-0060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/30/2022] [Indexed: 12/14/2022]
Abstract
Gliomas are the most common primary central nervous system tumors; despite recent advances in diagnosis and treatment, glioma patients generally have a poor prognosis. Hence there is a clear need for improved therapeutic options. In recent years, significant effort has been made to investigate immunotherapy and precision oncology approaches. The review covers well-established strategies such as surgery, temozolomide, PCV, and mTOR inhibitors. Furthermore, it summarizes promising therapies: tumor treating fields, immune therapies, tyrosine kinases inhibitors, IDH(Isocitrate dehydrogenase)-targeted approaches, and others. While there are many promising treatment strategies, none fundamentally changed the management of glioma patients. However, we are still awaiting the outcome of ongoing trials, which have the potential to revolutionize the treatment of glioma.
Collapse
Affiliation(s)
- Paulina Śledzińska
- Molecular Oncology and Genetics Department, Innovative Medical Forum, The F. Lukaszczyk Oncology Center, 85-796 Bydgoszcz, Poland
| | - Marek Bebyn
- Molecular Oncology and Genetics Department, Innovative Medical Forum, The F. Lukaszczyk Oncology Center, 85-796 Bydgoszcz, Poland
| | - Jacek Furtak
- Department of Neurosurgery, 10th Military Research Hospital and Polyclinic, 85-681 Bydgoszcz, Poland.,Department of Neurooncology and Radiosurgery, The F. Lukaszczyk Oncology Center, 85-796 Bydgoszcz, Poland
| | - Agnieszka Koper
- Department of Oncology, Nicolaus Copernicus University in Torun, Ludwik Rydygier Collegium Medicum, 85-067 Bydgoszcz, Poland.,Department of Oncology, Franciszek Lukaszczyk Oncology Centre, 85-796 Bydgoszcz, Poland
| | - Krzysztof Koper
- Department of Oncology, Franciszek Lukaszczyk Oncology Centre, 85-796 Bydgoszcz, Poland.,Department of Clinical Oncology, and Nursing, Departament of Oncological Surgery, Nicolaus Copernicus University in Torun, Ludwik Rydygier Collegium Medicum, 85-067 Bydgoszcz, Poland
| |
Collapse
|
9
|
Shibahara I, Shibahara Y, Hagiwara H, Watanabe T, Orihashi Y, Handa H, Inukai M, Hide T, Yasui Y, Kumabe T. Ventricular opening and cerebrospinal fluid circulation accelerate the biodegradation process of carmustine wafers suggesting their immunomodulation potential in the human brain. J Neurooncol 2022; 159:425-435. [PMID: 35802230 DOI: 10.1007/s11060-022-04078-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/25/2022] [Indexed: 11/28/2022]
Abstract
PURPOSE Opening the ventricular system during glioblastoma surgery is often necessary, but the consequent effect on the tumor microenvironment of glioblastoma remains unknown. Implantation of carmustine wafer enables direct drug delivery to the tumor site; however, the exact mechanism of the wafer's biodegradation process is unclear, and the available data is limited to in vivo non-human mammalian studies. We hypothesized that the ventricular opening affects the degradation process of the wafer and the glioblastoma tumor microenvironment. METHODS This study included 30 glioblastoma patients. 21 patients underwent carmustine wafer implantation during initial surgery. All patients underwent repeated surgical resection upon recurrence, allowing for pathological comparison of changes associated with wafer implantation. Immunohistochemical analyses were performed using CD68, TMEM119, CD163, IBA1, BIN1, and CD31 antibodies to highlight microglia, macrophages, and tumor vascularity, and the quantitative scoring results were correlated with clinical, molecular, and surgical variables, including the effect of the ventricular opening. RESULTS The carmustine wafer implanted group presented significantly less TMEM119-positive microglia within the tumor (P = 0.0002). Simple and multiple regression analyses revealed that the decrease in TMEM119-positive microglia was correlated with longer intervals between surgeries and opened ventricular systems. No correlation was observed between age, methylated O6-methylguanine DNA methyltransferase promoter expression, and the extent of surgical resection. CONCLUSIONS Our study findings strongly suggest that biomaterials may possess immunomodulation capacity, which is significantly impacted by the ventricular opening procedure. Furthermore, our data highlights the pathophysiological effects of the ventricular opening within the surrounding human brain, especially after the wafer implantation.
Collapse
Affiliation(s)
- Ichiyo Shibahara
- Department of Neurosurgery, Kitasato University School of Medicine, 1-15-1 Kitasato Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan.
| | - Yukiko Shibahara
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Hiroyuki Hagiwara
- Department of Neurosurgery, Yamato Municipal Hospital, Yamato, Kanagawa, Japan
| | - Takashi Watanabe
- Department of General Internal Medicine, JCHO Sendai Hospital, Sendai, Miyagi, Japan
| | - Yasushi Orihashi
- Division of Clinical Research, Kitasato University Hospital, Sagamihara, Kanagawa, Japan
| | - Hajime Handa
- Department of Neurosurgery, Kitasato University School of Medicine, 1-15-1 Kitasato Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Madoka Inukai
- Department of Neurosurgery, Kitasato University School of Medicine, 1-15-1 Kitasato Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan.,Department of Pathology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Takuichiro Hide
- Department of Neurosurgery, Kitasato University School of Medicine, 1-15-1 Kitasato Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Yoshie Yasui
- Department of Neurosurgery, Kitasato University School of Medicine, 1-15-1 Kitasato Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Toshihiro Kumabe
- Department of Neurosurgery, Kitasato University School of Medicine, 1-15-1 Kitasato Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| |
Collapse
|
10
|
Pardridge WM. A Historical Review of Brain Drug Delivery. Pharmaceutics 2022; 14:1283. [PMID: 35745855 PMCID: PMC9229021 DOI: 10.3390/pharmaceutics14061283] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022] Open
Abstract
The history of brain drug delivery is reviewed beginning with the first demonstration, in 1914, that a drug for syphilis, salvarsan, did not enter the brain, due to the presence of a blood-brain barrier (BBB). Owing to restricted transport across the BBB, FDA-approved drugs for the CNS have been generally limited to lipid-soluble small molecules. Drugs that do not cross the BBB can be re-engineered for transport on endogenous BBB carrier-mediated transport and receptor-mediated transport systems, which were identified during the 1970s-1980s. By the 1990s, a multitude of brain drug delivery technologies emerged, including trans-cranial delivery, CSF delivery, BBB disruption, lipid carriers, prodrugs, stem cells, exosomes, nanoparticles, gene therapy, and biologics. The advantages and limitations of each of these brain drug delivery technologies are critically reviewed.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| |
Collapse
|
11
|
El Kheir W, Marcos B, Virgilio N, Paquette B, Faucheux N, Lauzon MA. Drug Delivery Systems in the Development of Novel Strategies for Glioblastoma Treatment. Pharmaceutics 2022; 14:1189. [PMID: 35745762 PMCID: PMC9227363 DOI: 10.3390/pharmaceutics14061189] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 05/25/2022] [Accepted: 05/30/2022] [Indexed: 02/04/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a grade IV glioma considered the most fatal cancer of the central nervous system (CNS), with less than a 5% survival rate after five years. The tumor heterogeneity, the high infiltrative behavior of its cells, and the blood-brain barrier (BBB) that limits the access of therapeutic drugs to the brain are the main reasons hampering the current standard treatment efficiency. Following the tumor resection, the infiltrative remaining GBM cells, which are resistant to chemotherapy and radiotherapy, can further invade the surrounding brain parenchyma. Consequently, the development of new strategies to treat parenchyma-infiltrating GBM cells, such as vaccines, nanotherapies, and tumor cells traps including drug delivery systems, is required. For example, the chemoattractant CXCL12, by binding to its CXCR4 receptor, activates signaling pathways that play a critical role in tumor progression and invasion, making it an interesting therapeutic target to properly control the direction of GBM cell migration for treatment proposes. Moreover, the interstitial fluid flow (IFF) is also implicated in increasing the GBM cell migration through the activation of the CXCL12-CXCR4 signaling pathway. However, due to its complex and variable nature, the influence of the IFF on the efficiency of drug delivery systems is not well understood yet. Therefore, this review discusses novel drug delivery strategies to overcome the GBM treatment limitations, focusing on chemokines such as CXCL12 as an innovative approach to reverse the migration of infiltrated GBM. Furthermore, recent developments regarding in vitro 3D culture systems aiming to mimic the dynamic peritumoral environment for the optimization of new drug delivery technologies are highlighted.
Collapse
Affiliation(s)
- Wiam El Kheir
- Advanced Dynamic Cell Culture Systems Laboratory, Department of Chemical Engineering and Biotechnology Engineering, Faculty of Engineering, Université de Sherbrooke, 2500 Boul. Université, Sherbrooke, QC J1K 2R1, Canada;
- Laboratory of Cell-Biomaterial Biohybrid Systems, Department of Chemical Engineering and Biotechnology Engineering, Faculty of Engineering, Université de Sherbrooke, 2500 Boul. Université, Sherbrooke, QC J1K 2R1, Canada;
| | - Bernard Marcos
- Department of Chemical Engineering and Biotechnology Engineering, Faculty of Engineering, Université de Sherbrooke, 2500 Boul. Université, Sherbrooke, QC J1K 2R1, Canada;
| | - Nick Virgilio
- Department of Chemical Engineering, Polytechnique Montréal, 2500 Chemin de Polytechnique, Montréal, QC H3T 1J4, Canada;
| | - Benoit Paquette
- Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada;
- Clinical Research Center of the Centre Hospitalier Universitaire de l’Université de Sherbrooke, 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Nathalie Faucheux
- Laboratory of Cell-Biomaterial Biohybrid Systems, Department of Chemical Engineering and Biotechnology Engineering, Faculty of Engineering, Université de Sherbrooke, 2500 Boul. Université, Sherbrooke, QC J1K 2R1, Canada;
- Clinical Research Center of the Centre Hospitalier Universitaire de l’Université de Sherbrooke, 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Marc-Antoine Lauzon
- Advanced Dynamic Cell Culture Systems Laboratory, Department of Chemical Engineering and Biotechnology Engineering, Faculty of Engineering, Université de Sherbrooke, 2500 Boul. Université, Sherbrooke, QC J1K 2R1, Canada;
- Research Center on Aging, 1036 Rue Belvédère Sud, Sherbrooke, QC J1H 4C4, Canada
| |
Collapse
|
12
|
Nance E, Pun SH, Saigal R, Sellers DL. Drug delivery to the central nervous system. NATURE REVIEWS. MATERIALS 2022; 7:314-331. [PMID: 38464996 PMCID: PMC10923597 DOI: 10.1038/s41578-021-00394-w] [Citation(s) in RCA: 158] [Impact Index Per Article: 52.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/05/2021] [Indexed: 03/12/2024]
Abstract
Despite the rising global incidence of central nervous system (CNS) disorders, CNS drug development remains challenging, with high costs, long pathways to clinical use and high failure rates. The CNS is highly protected by physiological barriers, in particular, the blood-brain barrier and the blood-cerebrospinal fluid barrier, which limit access of most drugs. Biomaterials can be designed to bypass or traverse these barriers, enabling the controlled delivery of drugs into the CNS. In this Review, we first examine the effects of normal and diseased CNS physiology on drug delivery to the brain and spinal cord. We then discuss CNS drug delivery designs and materials that are administered systemically, directly to the CNS, intranasally or peripherally through intramuscular injections. Finally, we highlight important challenges and opportunities for materials design for drug delivery to the CNS and the anticipated clinical impact of CNS drug delivery.
Collapse
Affiliation(s)
- Elizabeth Nance
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
- These authors contributed equally: Elizabeth Nance, Suzie H. Pun, Rajiv Saigal, Drew L. Sellers
| | - Suzie H. Pun
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- These authors contributed equally: Elizabeth Nance, Suzie H. Pun, Rajiv Saigal, Drew L. Sellers
| | - Rajiv Saigal
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
- These authors contributed equally: Elizabeth Nance, Suzie H. Pun, Rajiv Saigal, Drew L. Sellers
| | - Drew L. Sellers
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- These authors contributed equally: Elizabeth Nance, Suzie H. Pun, Rajiv Saigal, Drew L. Sellers
| |
Collapse
|
13
|
Gazaille C, Sicot M, Saulnier P, Eyer J, Bastiat G. Local Delivery and Glioblastoma: Why Not Combining Sustained Release and Targeting? FRONTIERS IN MEDICAL TECHNOLOGY 2022; 3:791596. [PMID: 35047971 PMCID: PMC8757870 DOI: 10.3389/fmedt.2021.791596] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 10/28/2021] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma is one of the most aggressive brain tumors and is associated with a very low overall median survival despite the current treatment. The standard of care used in clinic is the Stupp's protocol which consists of a maximal resection of the tumor when possible, followed by radio and chemotherapy using temozolomide. However, in most cases, glioblastoma cells infiltrate healthy tissues and lead to fatal recurrences. There are a lot of hurdles to overcome in the development of new therapeutic strategies such as tumor heterogeneity, cell infiltration, alkylating agent resistance, physiological barriers, etc., and few treatments are on the market today. One of them is particularly appealing because it is a local therapy, which does not bring additional invasiveness since tumor resection is included in the gold standard treatment. They are implants: the Gliadel® wafers, which are deposited post-surgery. Nevertheless, in addition to presenting important undesirable effects, it does not bring any major benefit in the therapy despite the strategy being particularly attractive. The purpose of this review is to provide an overview of recent advances in the development of innovative therapeutic strategies for glioblastoma using an implant-type approach. The combination of this local strategy with effective targeting of the tumor microenvironment as a whole, also developed in this review, may be of interest to alleviate some of the obstacles encountered in the treatment of glioblastoma.
Collapse
Affiliation(s)
| | - Marion Sicot
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, Angers, France
| | | | - Joël Eyer
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, Angers, France
| | | |
Collapse
|
14
|
Design of Biopolymer-Based Interstitial Therapies for the Treatment of Glioblastoma. Int J Mol Sci 2021; 22:ijms222313160. [PMID: 34884965 PMCID: PMC8658694 DOI: 10.3390/ijms222313160] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 12/31/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common form of primary brain cancer and has the highest morbidity rate and current treatments result in a bleak 5-year survival rate of 5.6%. Interstitial therapy is one option to increase survival. Drug delivery by interstitial therapy most commonly makes use of a polymer implant encapsulating a drug which releases as the polymer degrades. Interstitial therapy has been extensively studied as a treatment option for GBM as it provides several advantages over systemic administration of chemotherapeutics. Primarily, it can be applied behind the blood–brain barrier, increasing the number of possible chemotherapeutic candidates that can be used and reducing systemic levels of the therapy while concentrating it near the cancer source. With interstitial therapy, multiple drugs can be released locally into the brain at the site of resection as the polymer of the implant degrades, and the release profile of these drugs can be tailored to optimize combination therapy or maintain synergistic ratios. This can bypass the blood–brain barrier, alleviate systemic toxicity, and resolve drug resistance in the tumor. However, tailoring drug release requires appropriate consideration of the complex relationship between the drug, polymer, and formulation method. Drug physicochemical properties can result in intermolecular bonding with the polymeric matrix and affect drug distribution in the implant depending on the formulation method used. This review is focused on current works that have applied interstitial therapy towards GBM, discusses polymer and formulation methods, and provides design considerations for future implantable biodegradable materials.
Collapse
|
15
|
Rationally designed drug delivery systems for the local treatment of resected glioblastoma. Adv Drug Deliv Rev 2021; 177:113951. [PMID: 34461201 DOI: 10.1016/j.addr.2021.113951] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/26/2021] [Accepted: 08/24/2021] [Indexed: 02/08/2023]
Abstract
Glioblastoma (GBM) is a particularly aggressive brain cancer associated with high recurrence and poor prognosis. The standard of care, surgical resection followed by concomitant radio- and chemotherapy, leads to low survival rates. The local delivery of active agents within the tumor resection cavity has emerged as an attractive means to initiate oncological treatment immediately post-surgery. This complementary approach bypasses the blood-brain barrier, increases the local concentration at the tumor site while reducing or avoiding systemic side effects. This review will provide a global overview on the local treatment for GBM with an emphasis on the lessons learned from past clinical trials. The main parameters to be considered to rationally design fit-of-purpose biomaterials and develop drug delivery systems for local administration in the GBM resection cavity to prevent the tumor recurrence will be described. The intracavitary local treatment of GBM should i) use materials that facilitate translation to the clinic; ii) be characterized by easy GMP effective scaling up and easy-handling application by the neurosurgeons; iii) be adaptable to fill the tumor-resected niche, mold to the resection cavity or adhere to the exposed brain parenchyma; iv) be biocompatible and possess mechanical properties compatible with the brain; v) deliver a therapeutic dose of rationally-designed or repurposed drug compound(s) into the GBM infiltrative margin. Proof of concept with high translational potential will be provided. Finally, future perspectives to facilitate the clinical translation of the local perisurgical treatment of GBM will be discussed.
Collapse
|
16
|
Wu W, Klockow JL, Zhang M, Lafortune F, Chang E, Jin L, Wu Y, Daldrup-Link HE. Glioblastoma multiforme (GBM): An overview of current therapies and mechanisms of resistance. Pharmacol Res 2021; 171:105780. [PMID: 34302977 PMCID: PMC8384724 DOI: 10.1016/j.phrs.2021.105780] [Citation(s) in RCA: 361] [Impact Index Per Article: 90.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 07/18/2021] [Accepted: 07/19/2021] [Indexed: 12/21/2022]
Abstract
Glioblastoma multiforme (GBM) is a WHO grade IV glioma and the most common malignant, primary brain tumor with a 5-year survival of 7.2%. Its highly infiltrative nature, genetic heterogeneity, and protection by the blood brain barrier (BBB) have posed great treatment challenges. The standard treatment for GBMs is surgical resection followed by chemoradiotherapy. The robust DNA repair and self-renewing capabilities of glioblastoma cells and glioma initiating cells (GICs), respectively, promote resistance against all current treatment modalities. Thus, durable GBM management will require the invention of innovative treatment strategies. In this review, we will describe biological and molecular targets for GBM therapy, the current status of pharmacologic therapy, prominent mechanisms of resistance, and new treatment approaches. To date, medical imaging is primarily used to determine the location, size and macroscopic morphology of GBM before, during, and after therapy. In the future, molecular and cellular imaging approaches will more dynamically monitor the expression of molecular targets and/or immune responses in the tumor, thereby enabling more immediate adaptation of tumor-tailored, targeted therapies.
Collapse
Affiliation(s)
- Wei Wu
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| | - Jessica L Klockow
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Michael Zhang
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA; Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Famyrah Lafortune
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| | - Edwin Chang
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| | - Linchun Jin
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL 32611, USA
| | - Yang Wu
- Department of Neuropathology, Institute of Pathology, Technical University of Munich, Munich, Bayern 81675, Germany
| | - Heike E Daldrup-Link
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
17
|
Bastiancich C, Bozzato E, Henley I, Newland B. Does local drug delivery still hold therapeutic promise for brain cancer? A systematic review. J Control Release 2021; 337:296-305. [PMID: 34298055 DOI: 10.1016/j.jconrel.2021.07.031] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 07/16/2021] [Accepted: 07/17/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Glioblastoma (GBM) is the most common and aggressive primary brain tumor in adults. Despite the gold standard treatment combining surgical resection, radiation and adjuvant plus concomitant chemotherapy with the alkylating agent temozolomide (TMZ), the prognosis remains poor (5-year survival rate < 10%). Over the last three decades, a vast array of drug delivery systems (DDS) have been developed for the local treatment of GBM, with the majority of the characterization being undertaken in pre-clinical models. We aimed to gain an overview of the potential efficacy of such local delivery systems in comparison to the systemic drug administration. METHODS In this paper, a systematic search of Pubmed, Web of Science, and Scopus was performed using pre-determined search terms. Studies were assessed for eligibility based on specific inclusion and exclusion criteria. A total of fifteen publications were included for analysis of local vs systemic group median survival, tumor volume and adverse events, with five brought forward for a meta-analysis. RESULTS The majority of studies showed local delivery to be more efficacious than systemic administration, regardless of the drug, animal model, type of DDS used, or duration of the study. The meta-analysis also showed that the mean difference between median survival ratios was statistically significantly in favor of local delivery. CONCLUSION Preclinical evidence shows that there is a firm rationale for further developing DDS for local therapeutic delivery to GBM and other brain cancers.
Collapse
Affiliation(s)
- C Bastiancich
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France.
| | - E Bozzato
- Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium
| | - I Henley
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff CF10 3NB, UK
| | - B Newland
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff CF10 3NB, UK.
| |
Collapse
|
18
|
Zero-order drug delivery: State of the art and future prospects. J Control Release 2020; 327:834-856. [PMID: 32931897 DOI: 10.1016/j.jconrel.2020.09.020] [Citation(s) in RCA: 165] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 01/21/2023]
Abstract
Pharmaceutical drugs are an important part of the global healthcare system, with some estimates suggesting over 50% of the world's population takes at least one medication per day. Most drugs are delivered as immediate-release formulations that lead to a rapid increase in systemic drug concentration. Although these formulations have historically played an important role, they can be limited by poor patient compliance, adverse side effects, low bioavailability, or undesirable pharmacokinetics. Drug delivery systems featuring first-order release kinetics have been able to improve pharmacokinetics but are not ideal for drugs with short biological half-lives or small therapeutic windows. Zero-order drug delivery systems have the potential to overcome the issues facing immediate-release and first-order systems by releasing drug at a constant rate, thereby maintaining drug concentrations within the therapeutic window for an extended period of time. This release profile can be used to limit adverse side effects, reduce dosing frequency, and potentially improve patient compliance. This review covers strategies being employed to attain zero-order release or alter traditionally first-order release kinetics to achieve more consistent release before discussing opportunities for improving device performance based on emerging materials and fabrication methods.
Collapse
|
19
|
Wang F, Su H, Lin R, Chakroun RW, Monroe MK, Wang Z, Porter M, Cui H. Supramolecular Tubustecan Hydrogel as Chemotherapeutic Carrier to Improve Tumor Penetration and Local Treatment Efficacy. ACS NANO 2020; 14:10083-10094. [PMID: 32806082 DOI: 10.1021/acsnano.0c03286] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Local chemotherapy is a clinically proven strategy in treating malignant brain tumors. Its benefits, however, are largely limited by the rapid release and clearance of therapeutic agents and the inability to penetrate through tumor tissues. We report here on a supramolecular tubustecan (TT) hydrogel as both a therapeutic and drug carrier that enables long-term, sustained drug release and improved tumor tissue penetration. Covalent linkage of a tissue penetrating cyclic peptide to two camptothecin drug units creates a TT prodrug amphiphile that can associate into tubular supramolecular polymers and subsequently form a well-defined sphere-shaped hydrogel after injection into tumor tissues. The hollow nature of the resultant tubular assemblies allows for encapsulation of doxorubicin or curcumin for combination therapy. Our in vitro and in vivo studies reveal that these dual drug-bearing supramolecular hydrogels enhance tumor retention and penetration, serving as a local therapeutic depot for potent tumor regression, inhibition of tumor metastasis and recurrence, and mitigation of the off-target side effects.
Collapse
Affiliation(s)
- Feihu Wang
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Institute for NanoBiotechnology (INBT), Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Hao Su
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Institute for NanoBiotechnology (INBT), Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Ran Lin
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Institute for NanoBiotechnology (INBT), Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Rami W Chakroun
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Institute for NanoBiotechnology (INBT), Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Maya K Monroe
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Institute for NanoBiotechnology (INBT), Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Zongyuan Wang
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Institute for NanoBiotechnology (INBT), Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Michael Porter
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Institute for NanoBiotechnology (INBT), Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Honggang Cui
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Institute for NanoBiotechnology (INBT), Johns Hopkins University, Baltimore, Maryland 21218, United States
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| |
Collapse
|
20
|
Targeting Glioblastoma: Advances in Drug Delivery and Novel Therapeutic Approaches. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000124] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
21
|
Graham-Gurysh EG, Murthy AB, Moore KM, Hingtgen SD, Bachelder EM, Ainslie KM. Synergistic drug combinations for a precision medicine approach to interstitial glioblastoma therapy. J Control Release 2020; 323:282-292. [PMID: 32335153 DOI: 10.1016/j.jconrel.2020.04.028] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 04/18/2020] [Indexed: 01/12/2023]
Abstract
Glioblastoma (GBM) is a highly aggressive and heterogeneous form of brain cancer. Genotypic and phenotypic heterogeneity drives drug resistance and tumor recurrence. Combination chemotherapy could overcome drug resistance; however, GBM's location behind the blood-brain barrier severely limits chemotherapeutic options. Interstitial therapy, delivery of chemotherapy locally to the tumor site, via a biodegradable polymer implant can overcome the blood-brain barrier and increase the range of drugs available for therapy. Ideal drug candidates for interstitial therapy are those that are potent against GBM and work in combination with both standard-of-care therapy and new precision medicine targets. Herein we evaluated paclitaxel for interstitial therapy, investigating the effect of combination with both temozolomide, a clinical standard-of-care chemotherapy for GBM, and everolimus, a mammalian target of rapamycin (mTOR) inhibitor that modulates aberrant signaling present in >80% of GBM patients. Tested against a panel of GBM cell lines in vitro, paclitaxel was found to be effective at nanomolar concentrations, complement therapy with temozolomide, and synergize strongly with everolimus. The strong synergism seen with paclitaxel and everolimus was then explored in vivo. Paclitaxel and everolimus were separately formulated into fibrous scaffolds composed of acetalated dextran, a biodegradable polymer with tunable degradation rates, for implantation in the brain. Acetalated dextran degradation rates were tailored to attain matching release kinetics (~3% per day) of both paclitaxel and everolimus to maintain a fixed combination ratio of the two drugs. Combination interstitial therapy of both paclitaxel and everolimus significantly reduced GBM growth and improved progression free survival in two clinically relevant orthotopic models of GBM resection and recurrence. This work illustrates the advantages of synchronized interstitial therapy of paclitaxel and everolimus for post-surgical tumor control of GBM.
Collapse
Affiliation(s)
- Elizabeth G Graham-Gurysh
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Ananya B Murthy
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Kathryn M Moore
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA
| | - Shawn D Hingtgen
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Eric M Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Kristy M Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA; Department of Microbiology and Immunology, UNC School of Medicine, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
22
|
El Demerdash N, Kedda J, Ram N, Brem H, Tyler B. Novel therapeutics for brain tumors: current practice and future prospects. Expert Opin Drug Deliv 2020; 17:9-21. [DOI: 10.1080/17425247.2019.1676227] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Nagat El Demerdash
- Department of Neurosurgery, Hunterian Neurosurgical Research Laboratory, Johns Hopkins University, Baltimore, MD, USA
| | - Jayanidhi Kedda
- Department of Neurosurgery, Hunterian Neurosurgical Research Laboratory, Johns Hopkins University, Baltimore, MD, USA
| | - Nivi Ram
- Department of Neurosurgery, Hunterian Neurosurgical Research Laboratory, Johns Hopkins University, Baltimore, MD, USA
| | - Henry Brem
- Department of Neurosurgery, Hunterian Neurosurgical Research Laboratory, Johns Hopkins University, Baltimore, MD, USA
- Departments of Biomedical Engineering, Oncology, and Ophthalmology, Johns Hopkins University, Baltimore, MD, USA
| | - Betty Tyler
- Department of Neurosurgery, Hunterian Neurosurgical Research Laboratory, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
23
|
Belousov A, Titov S, Shved N, Garbuz M, Malykin G, Gulaia V, Kagansky A, Kumeiko V. The Extracellular Matrix and Biocompatible Materials in Glioblastoma Treatment. Front Bioeng Biotechnol 2019; 7:341. [PMID: 31803736 PMCID: PMC6877546 DOI: 10.3389/fbioe.2019.00341] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 10/30/2019] [Indexed: 02/06/2023] Open
Abstract
During cancer genesis, the extracellular matrix (ECM) in the human brain undergoes important transformations, starting to resemble embryonic brain cell milieu with a much denser structure. However, the stiffness of the tumor ECM does not preclude cancer cells from migration. The importance of the ECM role in normal brain tissue as well as in tumor homeostasis has engaged much effort in trials to implement ECM as a target and an instrument in the treatment of brain cancers. This review provides a detailed analysis of both experimental and applied approaches in combined therapy for gliomas in adults. In general, matrix materials for glioma treatment should have properties facilitating the simplest delivery into the body. Hence, to deliver an artificial implant directly into the operation cavity it should be packed into a gel form, while for bloodstream injections matrix needs to be in the form of polymer micelles, nanoparticles, etc. Furthermore, the delivered material should mimic biomechanical properties of the native tissue, support vital functions, and slow down or stop the proliferation of surrounding cells for a prolonged period. The authors propose a two-step approach aimed, on the one hand, at elimination of remaining cancer cells and on the other hand, at restoring normal brain tissue. Thereby, the first bioartificial matrix to be applied should have relatively low elastic modulus should be loaded with anticancer drugs, while the second material with a higher elastic modulus for neurite outgrowth support should contain specific factors stimulating neuroregeneration.
Collapse
Affiliation(s)
- Andrei Belousov
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Sergei Titov
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
- School of Natural Sciences, Far Eastern Federal University, Vladivostok, Russia
| | - Nikita Shved
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch of Russian Academy of Sciences, Vladivostok, Russia
| | - Mikhail Garbuz
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Grigorii Malykin
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch of Russian Academy of Sciences, Vladivostok, Russia
| | - Valeriia Gulaia
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Alexander Kagansky
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Vadim Kumeiko
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
- School of Natural Sciences, Far Eastern Federal University, Vladivostok, Russia
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch of Russian Academy of Sciences, Vladivostok, Russia
| |
Collapse
|
24
|
Electrospray for generation of drug delivery and vaccine particles applied in vitro and in vivo. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 105:110070. [PMID: 31546372 PMCID: PMC10366704 DOI: 10.1016/j.msec.2019.110070] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/17/2019] [Accepted: 08/09/2019] [Indexed: 12/16/2022]
Abstract
Also known as electrospray, electrohydrodynamic atomization has been used extensively in the last 15 years to develop polymer-based particles for drug delivery in cell and animal models. More recently, novel core-shell, multi-axial, and other electrospray particles have been developed from an array of polymers for a variety of biomedical applications. This review focuses on electrospray as a novel method of particle fabrication for drug delivery, specifically highlighting the applications of these particle systems in cell culture and animal models while also discussing polymers used for particle fabrication. Applications of electrospray particles to treat glioma, ovarian cancer, and breast cancer are reviewed. Additionally, delivery of antibiotics, gene therapy, and bacterial cells formulated in electrospray particles is discussed. Finally, vaccines as well as drug eluting particles for differentiation of stem cells and tissue engineering are highlighted. The article concludes with a discussion of where the future of electrospray technology can go to strengthen its foothold in the biomedical field.
Collapse
|
25
|
Maharajan K. Feasibility of local administration of chemotherapeutic drugs as an effective adjuvant therapy in primary, recurrent and metastatic extradural tumours of the spine-review. JOURNAL OF SPINE SURGERY (HONG KONG) 2019; 5:273-284. [PMID: 31380482 PMCID: PMC6626732 DOI: 10.21037/jss.2019.04.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 04/17/2019] [Indexed: 06/10/2023]
Abstract
Present day multimodality treatment with advances in systemic chemotherapy and radiotherapy has increased the survival of patients significantly even in those primary tumours which were once considered to have a poor prognosis. However, local recurrence can severely jeopardise the quality of life and even reduce survival. Hence local recurrence is considered as the worst complication in the management of spinal tumours and the need to achieving adequate local tumour control cannot be overemphasised. Techniques like en bloc resections which significantly reduce the chances of local recurrence are always not possible due to anatomical and technical reasons and sometimes, not feasible in debilitated patients. Local administration of chemotherapeutic drugs has already been recognised as a treatment strategy in the management of bladder and brain tumours. In this literature review, an attempt is made to explore the available evidence in the English literature for local administration of chemotherapeutic drugs in the surgical management of primary, recurrent and metastatic spinal tumours.
Collapse
|
26
|
Mirani B, Pagan E, Shojaei S, Duchscherer J, Toyota BD, Ghavami S, Akbari M. A 3D bioprinted hydrogel mesh loaded with all-trans retinoic acid for treatment of glioblastoma. Eur J Pharmacol 2019; 854:201-212. [PMID: 30974104 DOI: 10.1016/j.ejphar.2019.04.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 04/01/2019] [Accepted: 04/02/2019] [Indexed: 12/27/2022]
Abstract
Treatment of glioblastoma (GBM), as the most lethal type of brain tumor, still remains a major challenge despite the various therapeutic approaches developed over the recent decades. GBM is considered as one of the most therapy-resistant human tumors. Treatment with temozolomide (TMZ) chemotherapy and radiotherapy in GBM patients has led to 30% of two-year survival rate (American Brain Tumor Association), representing a demanding field to develop more effective therapeutic strategies. This study presents a novel method for local delivery of all-trans retinoic acid (ATRA) for targeting GBM cells as a possible adjuvant therapeutic strategy for this disease. We have used 3D bioprinting to fabricate hydrogel meshes laden with ATRA-loaded polymeric particles. The ATRA-loaded meshes have been shown to facilitate a sustained release of ATRA with tunable release rate. Cell viability assay was used to demonstrate the ability of fabricated meshes in reducing cell growth in U-87 MG cell line. We later showed that the developed meshes induced apoptotic cell death in U-87 MG. Furthermore, the use of hydrogel for embedding the ATRA-loaded particles can facilitate the immobilization of the drug next to the tumor site. Our current innovative approach has shown the potential to open up new avenues for treatment of GBM, benefiting patients who suffer from this debilitating disease.
Collapse
Affiliation(s)
- Bahram Mirani
- Laboratory for Innovations in Microengineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada; Centre for Advanced Materials and Related Technologies (CAMTEC), University of Victoria, Victoria, BC, V8P 5C2, Canada; Centre for Biomedical Research (CBR), University of Victoria, Victoria, BC, V8P 5C2, Canada
| | - Erik Pagan
- Laboratory for Innovations in Microengineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Shahla Shojaei
- Laboratory for Innovations in Microengineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada; Centre for Advanced Materials and Related Technologies (CAMTEC), University of Victoria, Victoria, BC, V8P 5C2, Canada; Centre for Biomedical Research (CBR), University of Victoria, Victoria, BC, V8P 5C2, Canada
| | - Jade Duchscherer
- Laboratory for Innovations in Microengineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Brian D Toyota
- Division of Neurosurgery, Faculty of Medicine, Queen's University, Kingston, ON, K7K 1G8, Canada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Sciences, Faculty of Health Science, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, R3E 0J9, Canada; Autophagy Research Center, Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran; Research Institute of Oncology and Hematology, CancerCare Manitoba, University of Manitoba, Winnipeg, MB, Canada
| | - Mohsen Akbari
- Laboratory for Innovations in Microengineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada; Centre for Advanced Materials and Related Technologies (CAMTEC), University of Victoria, Victoria, BC, V8P 5C2, Canada; Centre for Biomedical Research (CBR), University of Victoria, Victoria, BC, V8P 5C2, Canada.
| |
Collapse
|
27
|
Tabet A, Jensen MP, Parkins CC, Patil PG, Watts C, Scherman OA. Designing Next-Generation Local Drug Delivery Vehicles for Glioblastoma Adjuvant Chemotherapy: Lessons from the Clinic. Adv Healthc Mater 2019; 8:e1801391. [PMID: 30632715 DOI: 10.1002/adhm.201801391] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/03/2018] [Indexed: 12/11/2022]
Abstract
To date, the clinical outcomes and survival rates for patients with glioblastoma (GB) remain poor. A promising approach to disease-modification involves local delivery of adjuvant chemotherapy into the resection cavity, thus circumventing the restrictions imposed by the blood-brain barrier. The clinical performance of the only FDA-approved local therapy for GB [carmustine (BCNU)-loaded polyanhydride wafers], however, has been disappointing. There is an unmet medical need in the local treatment of GB for drug delivery vehicles that provide sustained local release of small molecules and combination drugs over several months. Herein, key quantitative lessons from the use of local and systemic adjuvant chemotherapy for GB in the clinic are outlined, and it is discussed how these can inform the development of next-generation therapies. Several recent approaches are highlighted, and it is proposed that long-lasting soft materials can capture the value of stiff BCNU-loaded wafers while addressing a number of unmet medical needs. Finally, it is suggested that improved communication between materials scientists, biomedical scientists, and clinicians may facilitate translation of these materials into the clinic and ultimately lead to improved clinical outcomes.
Collapse
Affiliation(s)
- Anthony Tabet
- Melville Laboratory for Polymer Synthesis; Department of Chemistry; University of Cambridge; Lensfield Road Cambridge CB2 1EW UK
| | - Melanie P. Jensen
- Division of Neurosurgery; Department of Clinical Neurosciences; Addenbrooke's Hospital; University of Cambridge; Hills Road Cambridge CB2 0QQ UK
| | - Christopher C. Parkins
- Melville Laboratory for Polymer Synthesis; Department of Chemistry; University of Cambridge; Lensfield Road Cambridge CB2 1EW UK
| | - Parag G. Patil
- Department of Neurosurgery; University of Michigan Medical School; Ann Arbor MI 48109 USA
| | - Colin Watts
- Division of Neurosurgery; Department of Clinical Neurosciences; Addenbrooke's Hospital; University of Cambridge; Hills Road Cambridge CB2 0QQ UK
- Department of Neurosurgery; Birmingham Brain Cancer Program; Institute of Cancer and Genomic Sciences; University of Birmingham; Birmingham B15 2TT UK
| | - Oren A. Scherman
- Melville Laboratory for Polymer Synthesis; Department of Chemistry; University of Cambridge; Lensfield Road Cambridge CB2 1EW UK
| |
Collapse
|
28
|
Barnabas W. Drug targeting strategies into the brain for treating neurological diseases. J Neurosci Methods 2019; 311:133-146. [DOI: 10.1016/j.jneumeth.2018.10.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 10/08/2018] [Accepted: 10/10/2018] [Indexed: 12/17/2022]
|
29
|
Yu F, Asghar S, Zhang M, Zhang J, Ping Q, Xiao Y. Local strategies and delivery systems for the treatment of malignant gliomas. J Drug Target 2018; 27:367-378. [PMID: 30101621 DOI: 10.1080/1061186x.2018.1509982] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Glioma is one of the most common type of malignant tumours with high morbidity and mortality rates. Due to the particular features of the brain, such as blood-brain barrier or blood-tumour barrier, therapeutic agents are ineffective by systemic administration. The tumour inevitably recurs and devitalises patients. Herein, an overview of the localised gliomas treatment strategies is provided, including direct intratumoural/intracerebral injection, convection-enhanced delivery, and the implant of biodegradable polymer systems. The advantages and disadvantages of each therapy are discussed. Subsequently, we have reviewed the recent developments of therapeutic delivery systems aimed at transporting sufficient amounts of antineoplastic drugs into the brain tumour sites while minimising the potential side effects. To treat gliomas, localised and controlled delivery of drugs at their desired site of action is preferred as it reduces toxicity and increases treatment efficiency. Simultaneously, various drug delivery systems (DDS) have been used to enhance drug delivery to the brain. Use of non-conventional DDS for localised therapy has greatly expanded the spectrum of drugs available for the treatment of malignant tumours. Use smart DDS via localised delivery strategies, in combination with radiotherapy and multiple drug loading would serve as a promising approach to treat gliomas.
Collapse
Affiliation(s)
- Feng Yu
- a Department of Pharmaceutics, State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Sajid Asghar
- b Faculty of Pharmaceutical Sciences , Government College University Faisalabad , Faisalabad , Pakistan
| | - Mei Zhang
- a Department of Pharmaceutics, State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Jingwei Zhang
- a Department of Pharmaceutics, State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Qineng Ping
- a Department of Pharmaceutics, State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Yanyu Xiao
- a Department of Pharmaceutics, State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing , People's Republic of China
| |
Collapse
|
30
|
Renfrow JJ, Strowd RE, Laxton AW, Tatter SB, Geer CP, Lesser GJ. Surgical Considerations in the Optimal Management of Patients with Malignant Brain Tumors. Curr Treat Options Oncol 2018; 18:46. [PMID: 28681208 DOI: 10.1007/s11864-017-0487-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OPINION STATEMENT Advances in technology are revolutionizing medicine and the limits of what we can offer to our patients. In neurosurgery, technology continues to reduce morbidity, increase surgical accuracy, facilitate tissue acquisition, and promote novel techniques for prolonging survival in patients with neuro-oncologic disease. Surgery has been the backbone of glioma diagnosis and treatment by providing adequate, high quality material for precise histologic diagnosis, and genomic characterization in the setting of significant intratumoral heterogeneity, thus allowing personalized treatment selection in the clinic. The ability to obtain and accurately measure the maximal extent of resection in glioma surgery also remains a central role of the neurosurgeon in managing this cancer. To meet these goals, today's operating room has transformed from the traditional operating table and anesthesia machine to include neuronavigation instrumentation, intraoperative computed tomography, and magnetic resonance imaging scanners, advanced surgical microscopes fitted with fluorescent light filters, and electrocorticography machines. While surgeons, oncologists, and radiation oncologists all play unique critical roles in the care of patients with malignant gliomas, familiarity with developing techniques in complimentary subspecialties can enhance coordination of patient care, research productivity, professional interactions, and patient confidence and comfort with the physician team. Herein, we provide a summary of the advances in the field of neurosurgical oncology which allow more precise and optimal surgical resection for patients with malignant gliomas.
Collapse
Affiliation(s)
- Jaclyn J Renfrow
- Department of Neurosurgery, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157-1082, USA.
| | - Roy E Strowd
- Department of Neurology, Wake Forest School of Medicine, Winston-Salem, NC, USA.,Department of Internal Medicine - Section on Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Adrian W Laxton
- Department of Neurosurgery, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157-1082, USA
| | - Stephen B Tatter
- Department of Neurosurgery, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157-1082, USA
| | - Carol P Geer
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Glenn J Lesser
- Department of Internal Medicine - Section on Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
31
|
Liu SJ, Yang TC, Yang ST, Chen YC, Tseng YY. Biodegradable hybrid-structured nanofibrous membrane supported chemoprotective gene therapy enhances chemotherapy tolerance and efficacy in malignant glioma rats. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:515-526. [PMID: 29658349 DOI: 10.1080/21691401.2018.1460374] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Chemotherapy is ineffective for treating malignant glioma (MG) because of the low therapeutic levels of pharmaceuticals in tumour tissues and the well-known tumour resistance. The resistance to alkylators is modulated by the DNA repair protein O6-alkylguanine-DNA alkyltransferase (AGT). O6-benzylguanine (O6-BG) can irreversibly inactivate AGT by competing with O6-methylguanine and has been confirmed to increase the therapeutic activity of alkylators. We developed hybrid-structured poly[(d,l)-lactide-co-glycolide] nanofibrous membranes (HSNMs) that enable the sequential and sustained release of O6-BG and two alkylators (carmustine and temozolomide [TMZ]). HSNMs were surgically instilled into the cerebral cavity of pathogen-free rats and F98 glioma-bearing rats. The release behaviours of loaded drugs were quantified by using high-performance liquid chromatography. The treatment results were compared with the rats treated with intraperitoneal injection of O6-BG combined with surgical implantation of carmustine wafer and oral TMZ. The HSNMs revealed a sequential drug release behaviour with the elution of high drug concentrations of O6-BG in the early phase, followed by high levels of two alkylators. All drug concentrations remained high for over 14 weeks. Tumour growth was slower and the mean survival time was significantly prolonged in the HSNM-treated group. Biodegradable HSNMs can enhance therapeutic efficacy and prevent toxic systemic effects.
Collapse
Affiliation(s)
- Shih-Jung Liu
- a Department of Mechanical Engineering , Chang Gung University , Tao-Yuan , Taiwan, ROC.,b Department of Orthopedic Surgery , Chang Gung Memorial Hospital , Tao-Yuan , Taiwan, ROC
| | - Tao-Chieh Yang
- c Department of Neurosurgery , Asia University Hospital , Taichung , Taiwan, ROC
| | - Shun-Tai Yang
- d Division of Neurosurgery, Department of Surgery , Shuang Ho Hospital, Taipei Medical University , Taipei , Taiwan, ROC.,e Department of Surgery, School of Medicine, College of Medicine , Taipei Medical University , Taipei , Taiwan, ROC
| | - Ying-Chun Chen
- a Department of Mechanical Engineering , Chang Gung University , Tao-Yuan , Taiwan, ROC
| | - Yuan-Yun Tseng
- d Division of Neurosurgery, Department of Surgery , Shuang Ho Hospital, Taipei Medical University , Taipei , Taiwan, ROC.,e Department of Surgery, School of Medicine, College of Medicine , Taipei Medical University , Taipei , Taiwan, ROC
| |
Collapse
|
32
|
Graham-Gurysh E, Moore KM, Satterlee AB, Sheets KT, Lin FC, Bachelder EM, Miller CR, Hingtgen SD, Ainslie KM. Sustained Delivery of Doxorubicin via Acetalated Dextran Scaffold Prevents Glioblastoma Recurrence after Surgical Resection. Mol Pharm 2018; 15:1309-1318. [PMID: 29342360 DOI: 10.1021/acs.molpharmaceut.7b01114] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The primary cause of mortality for glioblastoma (GBM) is local tumor recurrence following standard-of-care therapies, including surgical resection. With most tumors recurring near the site of surgical resection, local delivery of chemotherapy at the time of surgery is a promising strategy. Herein drug-loaded polymer scaffolds with two distinct degradation profiles were fabricated to investigate the effect of local drug delivery rate on GBM recurrence following surgical resection. The novel biopolymer, acetalated dextran (Ace-DEX), was compared with commercially available polyester, poly(l-lactide) (PLA). Steady-state doxorubicin (DXR) release from Ace-DEX scaffolds was found to be faster when compared with scaffolds composed of PLA, in vitro. This increased drug release rate translated to improved therapeutic outcomes in a novel surgical model of orthotopic glioblastoma resection and recurrence. Mice treated with DXR-loaded Ace-DEX scaffolds (Ace-DEX/10DXR) resulted in 57% long-term survival out to study completion at 120 days compared with 20% survival following treatment with DXR-loaded PLA scaffolds (PLA/10DXR). Additionally, all mice treated with PLA/10DXR scaffolds exhibited disease progression by day 38, as defined by a 5-fold growth in tumor bioluminescent signal. In contrast, 57% of mice treated with Ace-DEX/10DXR scaffolds displayed a reduction in tumor burden, with 43% exhibiting complete remission. These results underscore the importance of polymer choice and drug release rate when evaluating local drug delivery strategies to improve prognosis for GBM patients undergoing tumor resection.
Collapse
Affiliation(s)
- Elizabeth Graham-Gurysh
- Division of Pharmacoengineering and Molecular Pharmaceutics , Eshelman School of Pharmacy, University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599 , United States
| | - Kathryn M Moore
- Joint Department of Biomedical Engineering , University of North Carolina at Chapel Hill and North Carolina State University , Raleigh , North Carolina 27695 , United States
| | - Andrew B Satterlee
- Division of Pharmacoengineering and Molecular Pharmaceutics , Eshelman School of Pharmacy, University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599 , United States
| | - Kevin T Sheets
- Division of Pharmacoengineering and Molecular Pharmaceutics , Eshelman School of Pharmacy, University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599 , United States
| | - Feng-Chang Lin
- Department of Biostatistics and North Carolina Translational and Clinical Sciences Institute , University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599 , United States
| | - Eric M Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics , Eshelman School of Pharmacy, University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599 , United States
| | - C Ryan Miller
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, Departments of Neurology and Pharmacology, Lineberger Comprehensive Cancer Center, and Neuroscience Center, School of Medicine , University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599 , United States
| | - Shawn D Hingtgen
- Division of Pharmacoengineering and Molecular Pharmaceutics , Eshelman School of Pharmacy, University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599 , United States
| | - Kristy M Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics , Eshelman School of Pharmacy, University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599 , United States
| |
Collapse
|
33
|
Roux A, Caire F, Guyotat J, Menei P, Metellus P, Pallud J. Carmustine wafer implantation for high-grade gliomas: Evidence-based safety efficacy and practical recommendations from the Neuro-oncology Club of the French Society of Neurosurgery. Neurochirurgie 2017; 63:433-443. [PMID: 29122306 DOI: 10.1016/j.neuchi.2017.07.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 06/21/2017] [Accepted: 07/28/2017] [Indexed: 11/16/2022]
Abstract
There is a growing body of evidence that carmustine wafer implantation during surgery is an effective therapeutic adjunct to the standard combined radio-chemotherapy regimen using temozolomide in newly diagnosed and recurrent high-grade glioma patient management with a statistically significant survival benefit demonstrated across several randomized clinical trials, as well as prospective and retrospective studies (grade A recommendation). Compelling clinical data also support the safety of carmustine wafer implantation (grade A recommendation) in these patients and suggest that observed adverse events can be avoided in experienced neurosurgeon hands. Furthermore, carmustine wafer implantation does not seem to impact negatively on the quality of life and the completion of adjuvant oncological treatments (grade C recommendation). Moreover, emerging findings support the potential of high-grade gliomas molecular status, especially the O(6)-Methylguanine-DNA Methyltransferase promoter methylation status, in predicting the efficacy of such a surgical strategy, especially at recurrence (grade B recommendation). Finally, carmustine wafer implantation appears to be cost-effective in high-grade glioma patients when performed by an experienced team and when total or subtotal resection can be achieved. Altogether, these data underline the current need for a new randomized clinical trial to assess the impact of a maximal safe resection with carmustine wafer implantation followed by the standard combined chemoradiation protocol stratified by molecular status in high-grade glioma patients.
Collapse
Affiliation(s)
- A Roux
- Department of Neurosurgery, Sainte-Anne Hospital, 1, rue Cabanis, 75674 Paris cedex 14, France; Paris Descartes University, Sorbonne Paris Cité, 75006 Paris, France; Inserm, U894, Centre de psychiatrie et neurosciences, 75006 Paris, France
| | - F Caire
- Department of Neurosurgery, CHU de Limoges, Limoges, France
| | - J Guyotat
- Lyon Civil Hospitals, Pierre Wertheimer Neurological and Neurosurgical Hospital, Service of Neurosurgery D, Lyon, France
| | - P Menei
- Department of Neurosurgery, CHU d'Angers, Angers, France; Inserm 1232/CRCINA, France
| | - P Metellus
- Department of Neurosurgery, Clairval Private Hospital, Marseille, France
| | - J Pallud
- Department of Neurosurgery, Sainte-Anne Hospital, 1, rue Cabanis, 75674 Paris cedex 14, France; Paris Descartes University, Sorbonne Paris Cité, 75006 Paris, France; Inserm, U894, Centre de psychiatrie et neurosciences, 75006 Paris, France.
| | | |
Collapse
|
34
|
Wadajkar AS, Dancy JG, Roberts NB, Connolly NP, Strickland DK, Winkles JA, Woodworth GF, Kim AJ. Decreased non-specific adhesivity, receptor targeted (DART) nanoparticles exhibit improved dispersion, cellular uptake, and tumor retention in invasive gliomas. J Control Release 2017; 267:144-153. [PMID: 28887134 DOI: 10.1016/j.jconrel.2017.09.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 08/26/2017] [Accepted: 09/04/2017] [Indexed: 12/15/2022]
Abstract
The most common and deadly form of primary brain cancer, glioblastoma (GBM), is characterized by significant intratumoral heterogeneity, microvascular proliferation, immune system suppression, and brain tissue invasion. Delivering effective and sustained treatments to the invasive GBM cells intermixed with functioning neural elements is a major goal of advanced therapeutic systems for brain cancer. Previously, we investigated the nanoparticle characteristics that enable targeting of invasive GBM cells. This revealed the importance of minimizing non-specific binding within the relatively adhesive, 'sticky' microenvironment of the brain and brain tumors in particular. We refer to such nanoformulations with decreased non-specific adhesivity and receptor targeting as 'DART' therapeutics. In this work, we applied this information toward the design and characterization of biodegradable nanocarriers, and in vivo testing in orthotopic experimental gliomas. We formulated particulate nanocarriers using poly(lactic-co-glycolic acid) (PLGA) and PLGA-polyethylene glycol (PLGA-PEG) polymers to generate sub-100nm nanoparticles with minimal binding to extracellular brain components and strong binding to the Fn14 receptor - an upregulated, conserved component in invasive GBM. Multiple particle tracking in brain tissue slices and in vivo testing in orthotopic murine malignant glioma revealed preserved nanoparticle diffusivity and increased uptake in brain tumor cells. These combined characteristics also resulted in longer retention of the DART nanoparticles within the orthotopic tumors compared to non-targeted versions. Taken together, these results and nanoparticle design considerations offer promising new methods to optimize therapeutic nanocarriers for improving drug delivery and treatment for invasive brain tumors.
Collapse
Affiliation(s)
- Aniket S Wadajkar
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jimena G Dancy
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Nathan B Roberts
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Nina P Connolly
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Dudley K Strickland
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jeffrey A Winkles
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Graeme F Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Anthony J Kim
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
35
|
Yang Y, Du T, Zhang J, Kang T, Luo L, Tao J, Gou Z, Chen S, Du Y, He J, Jiang S, Mao Q, Gou M. A 3D-Engineered Conformal Implant Releases DNA Nanocomplexs for Eradicating the Postsurgery Residual Glioblastoma. ADVANCED SCIENCE 2017; 4:1600491. [PMID: 28852611 PMCID: PMC5566247 DOI: 10.1002/advs.201600491] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 02/01/2017] [Indexed: 02/05/2023]
Abstract
Gene therapy has great promise for glioblastoma treatment; however, it remains a great challenge to efficiently deliver genes to the brain. The incomplete resection of glioblastoma always leads to poor prognosis. Here, a 3D‐engineered conformal implant for eradicating the postsurgery residual glioblastoma is designed. This implant is constructed by 3D‐printing technology to match the tumor cavity and release an oncolytic virus‐inspired DNA nanocomplex to kill glioblastoma cells through apoptosis induction. Meanwhile, a 3D‐engineered subcutaneous glioblastoma xenograft is built to mimic the resection tumor cavity in mice. Insertion of the implant into the glioblastoma resection cavity efficiently delays tumor recurrence and significantly prolongs overall survival. This study provides a proof‐of‐concept of glioblastoma therapy using a conformal implant that releases oncolytic DNA nanocomplexs. This strategy can lead to the development of future precision therapy for eradicating postsurgery residual tumors.
Collapse
Affiliation(s)
- Yuan Yang
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University and Collaborative Innovation Center of Biotherapy; Chengdu P. R. China
- Department of Neurosurgery; West China Hospital; Sichuan University; Chengdu P. R. China
| | - Ting Du
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University and Collaborative Innovation Center of Biotherapy; Chengdu P. R. China
| | - Jiumeng Zhang
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University and Collaborative Innovation Center of Biotherapy; Chengdu P. R. China
| | - Tianyi Kang
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University and Collaborative Innovation Center of Biotherapy; Chengdu P. R. China
| | - Li Luo
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University and Collaborative Innovation Center of Biotherapy; Chengdu P. R. China
| | - Jie Tao
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University and Collaborative Innovation Center of Biotherapy; Chengdu P. R. China
- School of Materials Science and Engineering; Sichuan University; Chengdu Sichuan 610065 P. R. China
| | - Zhiyuan Gou
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University and Collaborative Innovation Center of Biotherapy; Chengdu P. R. China
| | - Shaochen Chen
- Department of Nanoengineering; Institute of Engineering in Medicine; 245B SME Building; MC 0448; University of California; San Diego 9500 Gilman Drive La Jolla CA 92093 USA
| | - Yanan Du
- Department of Biomedical Engineering; Tsinghua University School of Medicine; Beijing P. R. China
| | - Jiankang He
- State key laboratory for manufacturing systems engineering; Xi'an Jiaotong University; Xi'an 710049 China
| | - Shu Jiang
- Department of Neurosurgery; West China Hospital; Sichuan University; Chengdu P. R. China
| | - Qing Mao
- Department of Neurosurgery; West China Hospital; Sichuan University; Chengdu P. R. China
| | - Maling Gou
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University and Collaborative Innovation Center of Biotherapy; Chengdu P. R. China
| |
Collapse
|
36
|
Nanotechnology and nanocarrier-based approaches on treatment of degenerative diseases. INTERNATIONAL NANO LETTERS 2017. [DOI: 10.1007/s40089-017-0208-0] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
37
|
Anticancer drug-loaded hydrogels as drug delivery systems for the local treatment of glioblastoma. J Control Release 2016; 243:29-42. [DOI: 10.1016/j.jconrel.2016.09.034] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 09/15/2016] [Accepted: 09/25/2016] [Indexed: 12/16/2022]
|
38
|
James NS, Joshi P, Ohulchanskyy TY, Chen Y, Tabaczynski W, Durrani F, Shibata M, Pandey RK. Photosensitizer (PS)-cyanine dye (CD) conjugates: Impact of the linkers joining the PS and CD moieties and their orientation in tumor-uptake and photodynamic therapy (PDT). Eur J Med Chem 2016; 122:770-785. [PMID: 27543778 PMCID: PMC5720162 DOI: 10.1016/j.ejmech.2016.06.045] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 06/24/2016] [Accepted: 06/25/2016] [Indexed: 11/22/2022]
Abstract
To investigate the impact of linker(s) joining the photosensitizer HPPH [3-(1'-hexyloxy) ethyl-3-devinylpyropheophorbide-a] and the cyanine dye (CD) in tumor-imaging and photodynamic therapy (dual-function agents), a series of HPPH-CD conjugates were synthesized. The modifications were done in an attempt to minimize Forster Resonance Energy Transfer (FRET) between the two chromophores and maximize singlet oxygen production. Among the conjugates containing variable length of linkers, the HPPH-CD conjugate, in which the photosensitizer (PS) and the CD was joined by four Carbon [(CH2)4] units showed higher tumor uptake, improved tumor contrast and limited skin uptake in mice bearing Colon-26 (BALB/c) or U87 tumors in Nude mice. The bi-functional agents in which the HPPH was linked at the meta-position of phenyl-substituted CD 5, 6 and 7 showed longer tumor response (cure) than the corresponding para-substituted analogs 2, 3, and 4, which suggests that the orientation of the PS and CD moieties within the conjugate also makes a substantial difference in tumor-specificity. Compared to HPPH, the singlet oxygen yields of all the HPPH-CD conjugates were significantly low, and required a higher therapeutic dose to achieve the same in vivo response obtained by HPPH-PDT alone. However, conjugate 6 produced a higher singlet oxygen yield with reduced FRET and exhibited enhanced long-term PDT efficacy in mice bearing Colon-26 (BALB/c) and U87 tumors (nude) than its counterparts, including our lead compound (HPPH-CD), making it the most efficacious of the series. Thus, these conjugates bearing cyanine dye moiety (CD) provide an opportunity of imaging deeply seated tumors for fluorescence-guided surgery with an option of PDT.
Collapse
Affiliation(s)
- Nadine S James
- PDT Center, Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, 14263, USA
| | - Penny Joshi
- PDT Center, Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, 14263, USA
| | - Tymish Y Ohulchanskyy
- College of Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong, 518060, China; Institute for Lasers, Photonics and Biophotonics, University at Buffalo, Buffalo, NY, 14260, USA
| | - Yihui Chen
- PDT Center, Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, 14263, USA
| | - Walter Tabaczynski
- PDT Center, Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, 14263, USA
| | - Farukh Durrani
- PDT Center, Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, 14263, USA
| | - Masayuki Shibata
- Health Informatics, Rutgers School of Health Related Professions, Newark, NJ, 07107, USA
| | - Ravindra K Pandey
- PDT Center, Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, 14263, USA.
| |
Collapse
|
39
|
Tobin MK, Geraghty JR, Engelhard HH, Linninger AA, Mehta AI. Intramedullary spinal cord tumors: a review of current and future treatment strategies. Neurosurg Focus 2016; 39:E14. [PMID: 26235012 DOI: 10.3171/2015.5.focus15158] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Intramedullary spinal cord tumors have low incidence rates but are associated with difficult treatment options. The majority of patients with these tumors can be initially treated with an attempted resection. Unfortunately, those patients who cannot undergo gross-total resection or have subtotal resection are left with few treatment options, such as radiotherapy and chemotherapy. These adjuvant treatments, however, are associated with the potential for significant adverse side effects and still leave patients with a poor prognosis. To successfully manage these patients and improve both their quality of life and prognosis, novel treatment options must be developed to supplement subtotal resection. New research is underway investigating alternative therapeutic approaches for these patients, including directed, localized drug delivery and nanomedicine techniques. These and other future investigations will hopefully lead to promising new therapies for these devastating diseases.
Collapse
|
40
|
Tseng YY, Kau YC, Liu SJ. Advanced interstitial chemotherapy for treating malignant glioma. Expert Opin Drug Deliv 2016; 13:1533-1544. [DOI: 10.1080/17425247.2016.1193153] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Yuan-Yun Tseng
- Department of Neurosurgery, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Chuan Kau
- Department of Anesthesiology, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
| | - Shih-Jung Liu
- Department of Mechanical Engineering, Chang Gung University, Tao-Yuan, Taiwan
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
| |
Collapse
|
41
|
Ene CI, Nerva JD, Morton RP, Barkley AS, Barber JK, Ko AL, Silbergeld DL. Safety and efficacy of carmustine (BCNU) wafers for metastatic brain tumors. Surg Neurol Int 2016; 7:S295-9. [PMID: 27217968 PMCID: PMC4866053 DOI: 10.4103/2152-7806.181987] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 02/22/2016] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Carmustine (BCNU) wafers (Gliadel) prolongs local disease control and progression-free survival (PFS) in patients with malignant gliomas. However, in metastatic brain tumors, there is a paucity of evidence in support of its safety and efficacy. The goal of this study was to assess the safety and efficacy of Gliadel wafers in patients with metastatic brain tumors. METHODS We retrospectively reviewed the University of Washington experience with Gliadel wafers for metastatic brain tumors between 2000 and 2015. RESULTS Gliadel wafers were used in 14 patients with metastatic brain tumors during the period reviewed. There were no postoperative seizures, strokes, or hemorrhages. There was one postoperative wound infection necessitating return to the operating room. The mean time to tumor progression (n = 7) and death (n = 5) after Gliadel wafer implantation was 2.5 and 2.9 years, respectively. Age was the only variable affecting PFS in patients receiving Gliadel wafers. Patients <53 years old (n = 7) had a PFS of 0.52 years, whereas patients >53 years old (n = 7) had a PFS of 4.29 years (P = 0.02). There was no significant difference in PFS in relation to presenting Karnofsky Performance Status (P = 0.26), number of brain metastasis (P = 0.82), tumor volume (P = 0.54), prior surgery (P = 0.57), or prior radiation (P = 0.41). There were no significant differences in the mean survival in relationship to any variable including age. CONCLUSIONS BCNU wafers are a safe and a potentially efficacious adjunct to surgery and radiation for improving local disease control in metastatic brain tumors. Larger studies, however, are needed to examine overall efficacy and tumor specific efficacy.
Collapse
Affiliation(s)
- Chibawanye I Ene
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - John D Nerva
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Ryan P Morton
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Ariana S Barkley
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Jason K Barber
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Andrew L Ko
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Daniel L Silbergeld
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| |
Collapse
|
42
|
Kleinberg L. Polifeprosan 20, 3.85% carmustine slow release wafer in malignant glioma: patient selection and perspectives on a low-burden therapy. Patient Prefer Adherence 2016; 10:2397-2406. [PMID: 27920506 PMCID: PMC5125766 DOI: 10.2147/ppa.s93020] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Polifeprosan 20 with carmustine (GLIADEL®) polymer implant wafer is a biodegradable compound containing 3.85% carmustine (BCNU, bischloroethylnitrosourea) implanted in the brain at the time of planned tumor surgery, which then slowly degrades to release the BCNU chemotherapy directly into the brain thereby bypassing the blood-brain barrier. Carmustine implant wafers were demonstrated to improve survival in randomized placebo-controlled trials in patients undergoing a near total resection of newly diagnosed or recurrent malignant glioma. Based on these trials and other supporting data, carmustine wafer therapy was approved for use for newly diagnosed and recurrent malignant glioma in the United States and the European Union. Adverse events are uncommon, and as this therapy is placed at the time of surgery, it does not add to patient treatment burden. Nevertheless, this therapy appears to be underutilized. This article reviews the evidence for a favorable therapeutic ratio for the patient and the potential barriers. Consideration of these issues is important for optimal use of this therapeutic approach and may be important as this technology and other local therapies are further developed in the future.
Collapse
Affiliation(s)
- Lawrence Kleinberg
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, MD, USA
- Correspondence: Lawrence Kleinberg, Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, 401 North Broadway, Suite 1440, Baltimore, MD 21231, USA, Email
| |
Collapse
|
43
|
Wait SD, Prabhu RS, Burri SH, Atkins TG, Asher AL. Polymeric drug delivery for the treatment of glioblastoma. Neuro Oncol 2015; 17 Suppl 2:ii9-ii23. [PMID: 25746091 DOI: 10.1093/neuonc/nou360] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) remains an almost universally fatal diagnosis. The current therapeutic mainstay consists of maximal safe surgical resection followed by radiation therapy (RT) with concomitant temozolomide (TMZ), followed by monthly TMZ (the "Stupp regimen"). Several chemotherapeutic agents have been shown to have modest efficacy in the treatment of high-grade glioma (HGG), but blood-brain barrier impermeability remains a major delivery obstacle. Polymeric drug-delivery systems, developed to allow controlled local release of biologically active substances for a variety of conditions, can achieve high local concentrations of active agents while limiting systemic toxicities. Polymerically delivered carmustine (BCNU) wafers, placed on the surface of the tumor-resection cavity, can potentially provide immediate chemotherapy to residual tumor cells during the standard delay between surgery and chemoradiotherapy. BCNU wafer implantation as monochemotherapy (with RT) in newly diagnosed HGG has been investigated in 2 phase III studies that reported significant increases in median overall survival. A number of studies have investigated the tumoricidal synergies of combination chemotherapy with BCNU wafers in newly diagnosed or recurrent HGG, and a primary research focus has been the integration of BCNU wafers into multimodality therapy with the standard Stupp regimen. Overall, the results of these studies have been encouraging in terms of safety and efficacy. However, the data must be qualified by the nature of the studies conducted. Currently, there are no phase III studies of BCNU wafers with the standard Stupp regimen. We review the rationale, biochemistry, pharmacokinetics, and research history (including toxicity profile) of this modality.
Collapse
Affiliation(s)
- Scott D Wait
- Carolina Neurosurgery and Spine Associates, Charlotte, North Carolina (S.D.W., A.L.A.); Levine Children's Hospital, Carolinas Medical Center, Charlotte, North Carolina (S.D.W.); Department of Neurosurgery, Levine Cancer Institute, and Neuroscience Institute, Carolinas Medical Center, Charlotte, North Carolina (S.D.W., T.G.A., A.L.A.); Southeast Radiation Oncology, Charlotte, North Carolina (R.S.P., S.H.B.); Department of Radiation Oncology, Levine Cancer Institute, Carolinas Medical Center, Charlotte, North Carolina (R.S.P., S.H.B.)
| | - Roshan S Prabhu
- Carolina Neurosurgery and Spine Associates, Charlotte, North Carolina (S.D.W., A.L.A.); Levine Children's Hospital, Carolinas Medical Center, Charlotte, North Carolina (S.D.W.); Department of Neurosurgery, Levine Cancer Institute, and Neuroscience Institute, Carolinas Medical Center, Charlotte, North Carolina (S.D.W., T.G.A., A.L.A.); Southeast Radiation Oncology, Charlotte, North Carolina (R.S.P., S.H.B.); Department of Radiation Oncology, Levine Cancer Institute, Carolinas Medical Center, Charlotte, North Carolina (R.S.P., S.H.B.)
| | - Stuart H Burri
- Carolina Neurosurgery and Spine Associates, Charlotte, North Carolina (S.D.W., A.L.A.); Levine Children's Hospital, Carolinas Medical Center, Charlotte, North Carolina (S.D.W.); Department of Neurosurgery, Levine Cancer Institute, and Neuroscience Institute, Carolinas Medical Center, Charlotte, North Carolina (S.D.W., T.G.A., A.L.A.); Southeast Radiation Oncology, Charlotte, North Carolina (R.S.P., S.H.B.); Department of Radiation Oncology, Levine Cancer Institute, Carolinas Medical Center, Charlotte, North Carolina (R.S.P., S.H.B.)
| | - Tyler G Atkins
- Carolina Neurosurgery and Spine Associates, Charlotte, North Carolina (S.D.W., A.L.A.); Levine Children's Hospital, Carolinas Medical Center, Charlotte, North Carolina (S.D.W.); Department of Neurosurgery, Levine Cancer Institute, and Neuroscience Institute, Carolinas Medical Center, Charlotte, North Carolina (S.D.W., T.G.A., A.L.A.); Southeast Radiation Oncology, Charlotte, North Carolina (R.S.P., S.H.B.); Department of Radiation Oncology, Levine Cancer Institute, Carolinas Medical Center, Charlotte, North Carolina (R.S.P., S.H.B.)
| | - Anthony L Asher
- Carolina Neurosurgery and Spine Associates, Charlotte, North Carolina (S.D.W., A.L.A.); Levine Children's Hospital, Carolinas Medical Center, Charlotte, North Carolina (S.D.W.); Department of Neurosurgery, Levine Cancer Institute, and Neuroscience Institute, Carolinas Medical Center, Charlotte, North Carolina (S.D.W., T.G.A., A.L.A.); Southeast Radiation Oncology, Charlotte, North Carolina (R.S.P., S.H.B.); Department of Radiation Oncology, Levine Cancer Institute, Carolinas Medical Center, Charlotte, North Carolina (R.S.P., S.H.B.)
| |
Collapse
|
44
|
Promising approaches to circumvent the blood-brain barrier: progress, pitfalls and clinical prospects in brain cancer. Ther Deliv 2015; 6:989-1016. [PMID: 26488496 DOI: 10.4155/tde.15.48] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Brain drug delivery is a major challenge for therapy of central nervous system (CNS) diseases. Biochemical modifications of drugs or drug nanocarriers, methods of local delivery, and blood-brain barrier (BBB) disruption with focused ultrasound and microbubbles are promising approaches which enhance transport or bypass the BBB. These approaches are discussed in the context of brain cancer as an example in CNS drug development. Targeting to receptors enabling transport across the BBB offers noninvasive delivery of small molecule and biological cancer therapeutics. Local delivery methods enable high dose delivery while avoiding systemic exposure. BBB disruption with focused ultrasound and microbubbles offers local and noninvasive treatment. Clinical trials show the prospects of these technologies and point to challenges for the future.
Collapse
|
45
|
Burri SH, Prabhu RS, Sumrall AL, Brick W, Blaker BD, Heideman BE, Boltes P, Kelly R, Symanowski JT, Wiggins WF, Ashby L, Norton HJ, Judy K, Asher AL. BCNU wafer placement with temozolomide (TMZ) in the immediate postoperative period after tumor resection followed by radiation therapy with TMZ in patients with newly diagnosed high grade glioma: final results of a prospective, multi-institutional, phase II trial. J Neurooncol 2015; 123:259-66. [DOI: 10.1007/s11060-015-1793-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 04/25/2015] [Indexed: 12/20/2022]
|
46
|
Warren KE. Novel therapeutic delivery approaches in development for pediatric gliomas. CNS Oncol 2015; 2:427-35. [PMID: 24511389 DOI: 10.2217/cns.13.37] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Pediatric gliomas are a heterogeneous group of diseases, ranging from relatively benign pilocytic astrocytomas with >90% 5-year survival, to glioblastomas and diffuse intrinsic pontine gliomas with <20% 5-year survival. Chemotherapy plays an important role in the management of these tumors, particularly in low-grade gliomas, but many high-grade tumors are resistant to chemotherapy. A major obstacle and contributor to this resistance is the blood–brain barrier, which protects the CNS by limiting entry of potential toxins, including chemotherapeutic agents. Several novel delivery approaches that circumvent the blood–brain barrier have been developed, including some currently in clinical trials. This review describes several of these novel approaches to improve delivery of chemotherapeutic agents to their site of action at the tumor, in attempts to improve their efficacy and the prognosis of children with this disease.
Collapse
|
47
|
Kuramitsu S, Motomura K, Natsume A, Wakabayashi T. Double-edged Sword in the Placement of Carmustine (BCNU) Wafers along the Eloquent Area: A Case Report. NMC Case Rep J 2014; 2:40-45. [PMID: 28663961 PMCID: PMC5364933 DOI: 10.2176/nmccrj.2014-0025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 06/22/2014] [Indexed: 11/28/2022] Open
Abstract
Although direct Gliadel® wafer implantation into the resection cavity has been shown to significantly improve survival in patients with high-grade gliomas, several complications have been associated with the implantations of Gliadel wafers, including brain edema, healing delay, cerebral spinal fluid leak, intracranial infections, and cyst formation. The brain edema that is associated with Gliadel wafer implantation might result in neurological deficits and significant morbidities and mortalities. In particular, it is not clear if they should be placed in the eloquent areas, such as language areas, motor areas, and areas related to cognitive function, even if these areas contain a remnant tumor. Here, we present a case of profound brain edema along the pyramidal tract due to Gliadel wafer implantation, which resulted in severe neurological deficits. This treatment represents a double-edged sword due to the possibility of severe symptomatic brain edema along the eloquent area, even though Gliadel wafers might be effective in controlling local tumor growth. We should keep in mind that Gliadel wafer placement in eloquent areas may result in severe disadvantages to patients and a loss of their quality of life.
Collapse
Affiliation(s)
- Shunichiro Kuramitsu
- Department of Neurosurgery, Nagoya University School of Medicine, Showa-ku, Nagoya, Aichi
| | - Kazuya Motomura
- Department of Neurosurgery, Nagoya University School of Medicine, Showa-ku, Nagoya, Aichi
| | - Atsushi Natsume
- Department of Neurosurgery, Nagoya University School of Medicine, Showa-ku, Nagoya, Aichi
| | - Toshihiko Wakabayashi
- Department of Neurosurgery, Nagoya University School of Medicine, Showa-ku, Nagoya, Aichi
| |
Collapse
|
48
|
Wolbers JG. Novel strategies in glioblastoma surgery aim at safe, supra-maximum resection in conjunction with local therapies. CHINESE JOURNAL OF CANCER 2014; 33:8-15. [PMID: 24384236 PMCID: PMC3905085 DOI: 10.5732/cjc.013.10219] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The biggest challenge in neuro-oncology is the treatment of glioblastoma, which exhibits poor prognosis and is increasing in incidence in an increasing aging population. Diverse treatment strategies aim at maximum cytoreduction and ensuring good quality of life. We discuss multimodal neuronavigation, supra-maximum tumor resection, and the postoperative treatment gap. Multimodal neuronavigation allows the integration of preoperative anatomic and functional data with intraoperative information. This approach includes functional magnetic resonance imaging (MRI) and diffusion tensor imaging in preplanning and ultrasound, computed tomography (CT), MRI and direct (sub)cortical stimulation during surgery. The practice of awake craniotomy decreases postoperative neurologic deficits, and an extensive supra-maximum resection appears to be feasible, even in eloquent areas of the brain. Intraoperative MRI- and fluorescence-guided surgery assist in achieving this goal of supra-maximum resection and have been the subject of an increasing number of reports. Photodynamic therapy and local chemotherapy are properly positioned to bridge the gap between surgery and chemoradiotherapy. The photosensitizer used in fluorescence-guided surgery persists in the remaining peripheral tumor extensions. Additionally, blinded randomized clinical trials showed firm evidence of extra cytoreduction by local chemotherapy in the tumor cavity. The cutting-edge promise is gene therapy although both the delivery and efficacy of the numerous transgenes remain under investigation. Issues such as the choice of (cell) vector, the choice of therapeutic transgene, the optimal route of administration, and biosafety need to be addressed in a systematic way. In this selective review, we present various evidence and promises to improve survival of glioblastoma patients by supra-maximum cytoreduction via local procedures while minimizing the risk of new neurologic deficit.
Collapse
Affiliation(s)
- John G Wolbers
- Department of Neurosurgery, Erasmus University Medical Centre, Rotterdam, The Netherlands.
| |
Collapse
|
49
|
Woodworth GF, Dunn GP, Nance EA, Hanes J, Brem H. Emerging insights into barriers to effective brain tumor therapeutics. Front Oncol 2014; 4:126. [PMID: 25101239 PMCID: PMC4104487 DOI: 10.3389/fonc.2014.00126] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 05/13/2014] [Indexed: 12/27/2022] Open
Abstract
There is great promise that ongoing advances in the delivery of therapeutics to the central nervous system (CNS) combined with rapidly expanding knowledge of brain tumor patho-biology will provide new, more effective therapies. Brain tumors that form from brain cells, as opposed to those that come from other parts of the body, rarely metastasize outside of the CNS. Instead, the tumor cells invade deep into the brain itself, causing disruption in brain circuits, blood vessel and blood flow changes, and tissue swelling. Patients with the most common and deadly form, glioblastoma (GBM) rarely live more than 2 years even with the most aggressive treatments and often with devastating neurological consequences. Current treatments include maximal safe surgical removal or biopsy followed by radiation and chemotherapy to address the residual tumor mass and invading tumor cells. However, delivering effective and sustained treatments to these invading cells without damaging healthy brain tissue is a major challenge and focus of the emerging fields of nanomedicine and viral and cell-based therapies. New treatment strategies, particularly those directed against the invasive component of this devastating CNS disease, are sorely needed. In this review, we (1) discuss the history and evolution of treatments for GBM, (2) define and explore three critical barriers to improving therapeutic delivery to invasive brain tumors, specifically, the neuro-vascular unit as it relates to the blood brain barrier, the extra-cellular space in regard to the brain penetration barrier, and the tumor genetic heterogeneity and instability in association with the treatment efficacy barrier, and (3) identify promising new therapeutic delivery approaches that have the potential to address these barriers and create sustained, meaningful efficacy against GBM.
Collapse
Affiliation(s)
- Graeme F Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine , Baltimore, MD , USA ; Department of Anatomy and Neurobiology, University of Maryland School of Medicine , Baltimore, MD , USA
| | - Gavin P Dunn
- Department of Neurosurgery, Pathology and Immunology, Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine , St. Louis, MO , USA
| | - Elizabeth A Nance
- Center for Nanomedicine, Johns Hopkins University School of Medicine , Baltimore, MD , USA
| | - Justin Hanes
- Center for Nanomedicine, Johns Hopkins University School of Medicine , Baltimore, MD , USA ; Department of Ophthalmology, Johns Hopkins University School of Medicine , Baltimore, MD , USA ; Department of Neurosurgery, Johns Hopkins University School of Medicine , Baltimore, MD , USA
| | - Henry Brem
- Department of Neurosurgery, Johns Hopkins University School of Medicine , Baltimore, MD , USA
| |
Collapse
|
50
|
Saenz del Burgo L, Hernández RM, Orive G, Pedraz JL. Nanotherapeutic approaches for brain cancer management. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2014; 10:905-19. [DOI: 10.1016/j.nano.2013.10.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 09/10/2013] [Accepted: 10/01/2013] [Indexed: 10/26/2022]
|