1
|
Kisby T, Borst GR, Coope DJ, Kostarelos K. Targeting the glioblastoma resection margin with locoregional nanotechnologies. Nat Rev Clin Oncol 2025:10.1038/s41571-025-01020-2. [PMID: 40369318 DOI: 10.1038/s41571-025-01020-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2025] [Indexed: 05/16/2025]
Abstract
Surgical resection is the first stage of treatment for patients diagnosed with resectable glioblastoma and is followed by a combination of adjuvant radiotherapy and systemic single-agent chemotherapy, which is typically commenced 4-6 weeks after surgery. This delay creates an interval during which residual tumour cells residing in the resection margin can undergo uninhibited proliferation and further invasion, even immediately after surgery, thus limiting the effectiveness of adjuvant therapies. Recognition of the postsurgical resection margin and peri-marginal zones as important anatomical clinical targets and the need to rethink current strategies can galvanize opportunities for local, intraoperative approaches, while also generating a new landscape of innovative treatment modalities. In this Perspective, we discuss opportunities and challenges for developing locoregional therapeutic strategies to target the glioblastoma resection margin as well as emerging opportunities offered by nanotechnology in this clinically transformative setting. We also discuss how persistent barriers to clinical translation can be overcome to offer a potential path forward towards broader acceptability of such advanced technologies.
Collapse
Affiliation(s)
- Thomas Kisby
- Centre for Nanotechnology in Medicine, Faculty of Biology & Medicine and Health, University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
| | - Gerben R Borst
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health & Manchester Cancer Research Centre, Manchester Academic Health Science Centre (MAHSC), University of Manchester, Manchester, UK
- Department of Clinical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - David J Coope
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
- Department of Neurosurgery, Manchester Centre for Clinical Neurosciences, Northern Care Alliance NHS Foundation Trust, Salford Royal, Salford, UK
| | - Kostas Kostarelos
- Centre for Nanotechnology in Medicine, Faculty of Biology & Medicine and Health, University of Manchester, Manchester, UK.
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK.
- Nanomedicine Lab, Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Barcelona, Spain.
- Institute of Neuroscience, Universitat Autònoma de Barcelona, Barcelona, Spain.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|
2
|
Zhou S, Zhang M, Wang J, Chen X, Xu Z, Yan Y, Li Y. Nanofibers in Glioma Therapy: Advances, Applications, and Overcoming Challenges. Int J Nanomedicine 2025; 20:4677-4703. [PMID: 40255668 PMCID: PMC12008729 DOI: 10.2147/ijn.s510363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/31/2025] [Indexed: 04/22/2025] Open
Abstract
Despite relentless effort to study glioma treatment, the prognosis for glioma patients remains poor. The main obstacles include the high rate of recurrence and the difficulty of passing the blood-brain barrier (BBB) for therapeutic drugs. Nanomaterials owing to their special physicochemical properties have been used in a wide range of fields thus far. The nanodrug delivery system (NDDS) with the ability of crossing the BBB, targeting glioma site, maintaining drug stability and controlling drug release, has significantly enhanced the anti-tumor therapeutic effect, improving the prognosis of glioma patients. Aligned nanofibers (NFs) are ideal materials to establish in vitro models of glioma microenvironment (GME), enabling the exploration of the mechanism of glioma cell migration and invasion to discover novel therapeutic targets. Moreover, NFs are now widely used in glioma applications such as radiotherapy, phototherapy, thermotherapy and immunotherapy. Despite the absolute dominance of NFs in anti-glioma applications, there are still some problems such as the further optimization of NDDS, and the impact of interactions between nanofibers and the protein corona (PC) on glioma therapy. This paper will shed light on the latest glioma applications of NFs in drug delivery systems and mimicking the tumor microenvironment (TME), and discuss how to further optimize the NDDS and eliminate or utilize the nanomedicine-PC interactions.
Collapse
Affiliation(s)
- Shangjun Zhou
- Department of Pediatric Surgery, Hunan Children’s Hospital, Changsha, Hunan, People’s Republic of China
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Mingcheng Zhang
- Center of Endoscopy, The Second Affiliated Hospital of Shandong First Medical University Tai’an, Shandong, People’s Republic of China
| | - Jiayu Wang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Xi Chen
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Yong Li
- Department of Pediatric Surgery, Hunan Children’s Hospital, Changsha, Hunan, People’s Republic of China
| |
Collapse
|
3
|
Jindal A, Mainuddin, Kumar A, Ratnesh RK, Singh J. Nanotechnology Driven Lipid and Metalloid Based Formulations Targeting Blood-Brain Barrier (3B) for Brain Tumor. Indian J Microbiol 2025; 65:92-119. [PMID: 40371021 PMCID: PMC12069182 DOI: 10.1007/s12088-024-01330-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 06/08/2024] [Indexed: 05/16/2025] Open
Abstract
The evolution of nanotechnology-driven lipid and metalloid-based nanoformulations has garnered significant attention for developing effective drug delivery systems with position/time precision and efficacy. This study focuses on challenges of blood-brain barrier (BBB) and their pivotal role in drug targeting in chronic diseases such as brain tumors (BTs). These formulations encapsulate therapeutic agents within lipidic matrices, enhancing drug solubility, bioavailability, and targeted delivery. The diverse lipid materials used in these nanoformulations highlight their biocompatibility and versatility, covering a wide range of drugs. Emphasis is placed on metal nanoparticles, liposomes, ethosomes, quantum dots, carbon nanotubes, nanorobots, and micelles. The analysis explores their drug loading, stability, release characteristics, and bioavailability modulation. It also delves into the enhanced-permeability and retention (EPR) effect, crucial for passive targeting of tumors. Recent nanocarrier systems enable better penetration of therapeutic compounds through the BBB, addressing treatment failures in invasive BTs.This review highlights the latest nanotechnology developments and potential therapeutic approaches, serving as a valuable resource for researchers, clinicians, and pharmaceutical scientists.
Collapse
Affiliation(s)
- Amulya Jindal
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, Meerut, Uttar Pradesh 250005 India
- SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology (Deemed to Be University), Delhi-NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh 201204 India
| | - Mainuddin
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, Meerut, Uttar Pradesh 250005 India
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Sector 125, Noida, Uttar Pradesh 201301 India
| | - Anoop Kumar
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, Meerut, Uttar Pradesh 250005 India
| | - Ratneshwar Kumar Ratnesh
- Department of Electronics and Communication Engineering, Meerut Institute of Engineering and Technology, Meerut, Uttar Pradesh 250005 India
| | - Jay Singh
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005 India
| |
Collapse
|
4
|
Tripathy NS, Sahoo L, Paikray S, Dilnawaz F. Emerging nanoplatforms towards microenvironment-responsive glioma therapy. Med Oncol 2025; 42:46. [PMID: 39812745 DOI: 10.1007/s12032-024-02596-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/28/2024] [Indexed: 01/16/2025]
Abstract
Gliomas are aggressive intracranial tumors of the central nervous system with a poor prognosis, high risk of recurrence, and low survival rates. Radiation, surgery, and chemotherapy are traditional cancer therapies. It is very challenging to accurately image and differentiate the malignancy grade of gliomas due to their heterogeneous and infiltrating nature and the obstruction of the blood-brain barrier. Imaging plays a crucial role in gliomas which significantly plays an important role in the accuracy of the diagnosis followed by any subsequent surgery or therapy. Other diagnostic methods (such as biopsies or surgery) are often very invasive. Preoperative imaging and intraoperative image-guided surgery perform the most significant safe resection. In recent years, the rapid growth of nanotechnology has opened up new avenues for glioma diagnosis and treatment. For better therapeutic efficacy, developing microenvironment-responsive nanoplatforms, including novel nanotherapeutic platforms of sonodynamic therapy, photodynamic therapy, and photothermal treatments, are employed for improved patient survival and better clinical control outcome. In this review recent advancement of multifunctional nanoplatforms leading toward treatment of glioma is discussed.
Collapse
Affiliation(s)
- Nigam Sekhar Tripathy
- School of Biotechnology, Centurion University of Technology and Management, Jatni, Bhubaneswar, Odisha, 752050, India
| | - Liza Sahoo
- School of Biotechnology, Centurion University of Technology and Management, Jatni, Bhubaneswar, Odisha, 752050, India
| | - Safal Paikray
- School of Biotechnology, Centurion University of Technology and Management, Jatni, Bhubaneswar, Odisha, 752050, India
| | - Fahima Dilnawaz
- School of Biotechnology, Centurion University of Technology and Management, Jatni, Bhubaneswar, Odisha, 752050, India.
| |
Collapse
|
5
|
Matsuda R, Maeoka R, Morimoto T, Nakazawa T, Tokuda N, Kotsugi M, Takeshima Y, Tamura K, Yamada S, Nishimura F, Park YS, Nakagawa I. Effect of Intraoperative Ventricular Opening on Recurrence Patterns Following Bis-Chloroethyl-Nitrosourea Wafer Implantation for Newly Diagnosed Glioblastoma. J Korean Neurosurg Soc 2025; 68:60-66. [PMID: 39081094 PMCID: PMC11725460 DOI: 10.3340/jkns.2024.0030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/15/2024] [Accepted: 05/20/2024] [Indexed: 01/11/2025] Open
Abstract
OBJECTIVE To evaluate the effect of ventricular opening (VO) on recurrence patterns in patients with newly diagnosed glioblastoma (GBM) treated with bis-chloroethyl-nitrosourea (BCNU) wafer implantation. METHODS This single-center retrospective study included 40 patients with newly diagnosed GBM who received BCNU wafer implantation after tumor resection between March 2013 and February 2022. The patients were categorized into two groups based on whether VO occurred during the GBM resection. While 18 patients had VO, 22 did not have VO. In cases with VO, the ventricular wall defect is closed with gelatin or oxidized regenerated cellulose and fibrin glue before BCNU wafer implantation. Recurrence patterns-classified as local, diffuse, distant, or multifocal-and time to recurrence were compared between patients with and without VO. RESULTS The median follow-up period for the entire cohort was 32.2 months (interquartile range, 16.7-38 months). Median survival time was comparable between patients with VO and patients without VO (38 vs. 26 months, p=0.53). Recurrence occurred in 31/40 patients (77.5%) in entire cohort. The incidence of recurrence was comparable between patients with VO and patients without VO (14 [77.8%] vs. 17 [77.3%], p=1.0). No significant differences were seen between the two groups in time to recurrence (p=0.59) or recurrence patterns (p=0.35). CONCLUSION Ventricular opening during surgery with BCNU wafer implantation does not seem to influence the recurrence patterns. Ventricular opening does not induce distant recurrence if appropriate ventricular closure is performed.
Collapse
Affiliation(s)
- Ryosuke Matsuda
- Department of Neurosurgery, Nara Medical University, Kashihara, Japan
| | - Ryosuke Maeoka
- Department of Neurosurgery, Nara Medical University, Kashihara, Japan
| | - Takayuki Morimoto
- Department of Neurosurgery, Nara Medical University, Kashihara, Japan
- Department of Neurosurgery, Nara City Hospital, Nara, Japan
| | - Tsutomu Nakazawa
- Department of Neurosurgery, Nara Medical University, Kashihara, Japan
| | - Noriaki Tokuda
- Department of Neurosurgery, Nara Medical University, Kashihara, Japan
| | - Masashi Kotsugi
- Department of Neurosurgery, Nara Medical University, Kashihara, Japan
| | | | - Kentaro Tamura
- Department of Neurosurgery, Nara Medical University, Kashihara, Japan
| | - Shuichi Yamada
- Department of Neurosurgery, Nara Medical University, Kashihara, Japan
| | | | - Young-Soo Park
- Department of Neurosurgery, Nara Medical University, Kashihara, Japan
| | - Ichiro Nakagawa
- Department of Neurosurgery, Nara Medical University, Kashihara, Japan
| |
Collapse
|
6
|
Serra R, Smith SJ, Rowlinson J, Gorelick N, Moloney C, McCrorie P, Veal GJ, Berry P, Chalmers AJ, Suk I, Shakesheff KM, Alexander C, Grundy RG, Brem H, Tyler BM, Rahman R. Neurosurgical application of olaparib from a thermo-responsive paste potentiates DNA damage to prolong survival in malignant glioma. Br J Cancer 2024; 131:1858-1868. [PMID: 39433869 PMCID: PMC11589713 DOI: 10.1038/s41416-024-02878-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND There is increased pan-cancer specific interest in repurposing the poly adenosine diphosphate-ribose polymerase-1 (PARP-1) inhibitor, olaparib, for newly diagnosed or recurrent isocitrate dehydrogenase wild type glioblastoma. We explore whether intra-cavity delivery of olaparib confers a survival benefit in a pre-clinical high-grade glioma model. METHODS Primary tumor RNA sequencing data was used to determine PARP-1 as a target in the glioblastoma infiltrative margin. We assessed radiosensitization conferred by olaparib alone and concomitant to genotoxic insults in vitro using clonal growth assays, cell cycle analysis and immunocytochemistry, and in vivo upon post-surgical delivery from a temperature-sensitive polymeric paste. RESULTS RNA-sequencing confirmed PARP-1 as a viable therapy target in glioblastoma infiltrative disease. Acute exposure of glioma cells to olaparib impaired proliferation and induced late-stage apoptosis associated with DNA damage in vitro, potentiated by radiation. Using high-grade glioma orthotopic allografts, a long-term overall survival benefit was observed upon interstitial olaparib delivery concomitant with radiotherapy, compared to systemic olaparib and standard glioblastoma treatment. Combined delivery of olaparib with either temozolomide or etoposide increased long-term survival, suggestive of olaparib functioning as DNA damage sensitizer. CONCLUSIONS Collectively, our data support a rationale for localized olaparib delivery concomitant with the current clinical regimen for malignant glioma treatment.
Collapse
Affiliation(s)
- Riccardo Serra
- Department of Neurosurgery, Johns Hopkins University, Baltimore, USA
| | - Stuart J Smith
- Children's Brain Tumour Research Centre, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Jonathan Rowlinson
- Children's Brain Tumour Research Centre, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Noah Gorelick
- Department of Neurosurgery, Johns Hopkins University, Baltimore, USA
| | - Cara Moloney
- Children's Brain Tumour Research Centre, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Phoebe McCrorie
- Children's Brain Tumour Research Centre, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Gareth J Veal
- Newcastle University Centre for Cancer, Newcastle University, Newcastle, UK
| | - Philip Berry
- Newcastle University Centre for Cancer, Newcastle University, Newcastle, UK
| | | | - Ian Suk
- Department of Neurosurgery, Johns Hopkins University, Baltimore, USA
| | | | | | - Richard G Grundy
- Children's Brain Tumour Research Centre, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Henry Brem
- Department of Neurosurgery, Johns Hopkins University, Baltimore, USA
- Departments of Biomedical Engineering, Oncology and Ophthalmology, Johns Hopkins University, Baltimore, USA
| | - Betty M Tyler
- Department of Neurosurgery, Johns Hopkins University, Baltimore, USA.
| | - Ruman Rahman
- Children's Brain Tumour Research Centre, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK.
| |
Collapse
|
7
|
Gkasdaris G, Berthiller J, Guyotat J, Jouanneau E, Gallet C, Meyronet D, Thomas L, Cartalat S, Seyve A, Honnorat J, Ducray F, Picart T. Is Carmustine Wafer Implantation in Progressive High-Grade Gliomas a Relevant Therapeutic Option? Complication Rate, Predictors of Complications and Onco-Functional Outcomes in a Series of 53 Cases. Cancers (Basel) 2024; 16:3465. [PMID: 39456559 PMCID: PMC11506748 DOI: 10.3390/cancers16203465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: The aim was to determine the complication rate and the predictors of complications and survival in high-grade glioma surgically managed at progression with implantation of Carmustine wafers. Methods: A retrospective series of 53 consecutive patients operated on between 2017 and 2022 was built. Results: The median age was 55 ± 10.9 years. The rates of global and infectious complications were 35.8% and 18.9%, respectively. In multivariate analysis, patients with a preoperative neurological deficit were more prone to develop a postoperative complication (HR = 5.35 95% CI 1.49-19.26, p = 0.01). No predictor of infectious complication was identified. In the grade 4 glioma subgroup (n = 44), progression-free and overall survival (calculated starting from the reresection) reached 3.95 months, 95% CI 2.92-5.21 and 11.51 months, 95% CI 9.11-17.18, respectively. Preoperative KPS > 80% (HR = 0.97 95% CI 0.93-0.99, p = 0.04), Gross Total Resection (HR = 0.38 95% CI 0.18-0.80, p = 0.01), and 3-month postoperative KPS > 80% (HR = 0.35 95% CI 0.17-0.72, p = 0.004) were predictors of prolonged overall survival. Conclusions: Surgical resection is a relevant option in high-grade gliomas at progression, especially in patients with a preoperative KPS > 80%, without preoperative neurological deficit, and amenable to complete resection. In patients elected for surgery, Carmustine wafer implantation is associated with a high rate of complications. It is consequently critical to closely monitor the patients for whom this option is chosen.
Collapse
Affiliation(s)
- Grigorios Gkasdaris
- Department of Neurosurgery, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, 69677 Bron, France; (J.G.); (E.J.); (C.G.)
| | - Julien Berthiller
- Department of Research and Clinical Epidemiology—Public Health, Hospices Civils de Lyon, 69677 Bron, France;
| | - Jacques Guyotat
- Department of Neurosurgery, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, 69677 Bron, France; (J.G.); (E.J.); (C.G.)
| | - Emmanuel Jouanneau
- Department of Neurosurgery, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, 69677 Bron, France; (J.G.); (E.J.); (C.G.)
- Faculty of Medicine, University Claude Bernard Lyon I, 69100 Villeurbanne, France; (D.M.); (A.S.); (J.H.); (F.D.)
- Cancer Initiation and Tumoral Cell Identity Department, Cancer Research Centre of Lyon (CRCL) INSERM 1052, CNRS 5286, 69008 Lyon, France
| | - Clémentine Gallet
- Department of Neurosurgery, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, 69677 Bron, France; (J.G.); (E.J.); (C.G.)
| | - David Meyronet
- Faculty of Medicine, University Claude Bernard Lyon I, 69100 Villeurbanne, France; (D.M.); (A.S.); (J.H.); (F.D.)
- Cancer Initiation and Tumoral Cell Identity Department, Cancer Research Centre of Lyon (CRCL) INSERM 1052, CNRS 5286, 69008 Lyon, France
- Department of Neuropathology, Groupement Hospitalier Est, Hospices Civils de Lyon, 69677 Bron, France
| | - Laure Thomas
- Department of Neuro-Oncology, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, 69677 Bron, France; (L.T.); (S.C.)
| | - Stéphanie Cartalat
- Department of Neuro-Oncology, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, 69677 Bron, France; (L.T.); (S.C.)
| | - Antoine Seyve
- Faculty of Medicine, University Claude Bernard Lyon I, 69100 Villeurbanne, France; (D.M.); (A.S.); (J.H.); (F.D.)
- Department of Neuro-Oncology, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, 69677 Bron, France; (L.T.); (S.C.)
| | - Jérôme Honnorat
- Faculty of Medicine, University Claude Bernard Lyon I, 69100 Villeurbanne, France; (D.M.); (A.S.); (J.H.); (F.D.)
- Department of Neuro-Oncology, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, 69677 Bron, France; (L.T.); (S.C.)
- MELIS Institute—Team Synaptopathies and Autoantibodies, INSERM U1314, UMR CNRS 5284, 69677 Bron, France
| | - François Ducray
- Faculty of Medicine, University Claude Bernard Lyon I, 69100 Villeurbanne, France; (D.M.); (A.S.); (J.H.); (F.D.)
- Cancer Initiation and Tumoral Cell Identity Department, Cancer Research Centre of Lyon (CRCL) INSERM 1052, CNRS 5286, 69008 Lyon, France
- Department of Neuro-Oncology, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, 69677 Bron, France; (L.T.); (S.C.)
| | - Thiebaud Picart
- Department of Neurosurgery, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, 69677 Bron, France; (J.G.); (E.J.); (C.G.)
- Faculty of Medicine, University Claude Bernard Lyon I, 69100 Villeurbanne, France; (D.M.); (A.S.); (J.H.); (F.D.)
- Cancer Initiation and Tumoral Cell Identity Department, Cancer Research Centre of Lyon (CRCL) INSERM 1052, CNRS 5286, 69008 Lyon, France
| |
Collapse
|
8
|
Valerius AR, Webb LM, Thomsen A, Lehrer EJ, Breen WG, Campian JL, Riviere-Cazaux C, Burns TC, Sener U. Review of Novel Surgical, Radiation, and Systemic Therapies and Clinical Trials in Glioblastoma. Int J Mol Sci 2024; 25:10570. [PMID: 39408897 PMCID: PMC11477105 DOI: 10.3390/ijms251910570] [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: 08/23/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Glioblastoma (GBM) is the most common malignant primary brain tumor in adults. Despite an established standard of care including surgical resection, radiation therapy, and chemotherapy, GBM unfortunately is associated with a dismal prognosis. Therefore, researchers are extensively evaluating avenues to expand GBM therapy and improve outcomes in patients with GBM. In this review, we provide a broad overview of novel GBM therapies that have recently completed or are actively undergoing study in clinical trials. These therapies expand across medical, surgical, and radiation clinical trials. We additionally review methods for improving clinical trial design in GBM.
Collapse
Affiliation(s)
| | - Lauren M. Webb
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA (U.S.)
| | - Anna Thomsen
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA (U.S.)
| | - Eric J. Lehrer
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - William G. Breen
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Jian L. Campian
- Department of Medical Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Terry C. Burns
- Department of Neurosurgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Ugur Sener
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA (U.S.)
- Department of Medical Oncology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
9
|
McConville C, Lastakchi S, Al Amri A, Ngoga D, Fayeye O, Cruickshank G. Local Delivery of Irinotecan to Recurrent GBM Patients at Reoperation Offers a Safe Route of Administration. Cancers (Basel) 2024; 16:3008. [PMID: 39272866 PMCID: PMC11393903 DOI: 10.3390/cancers16173008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/14/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
Glioblastomas are impossible to completely resect and almost always recur at the borders of the resection margin. There is no established chemotherapy regimen available to patients who recur, while systemic treatment is hampered by the blood-brain barrier. Here, we report on the first evaluation in humans of the intraparenchymal injection of irinotecan into the resection cavity after surgical resection of recurrent glioblastoma patients. The cytotoxicity of irinotecan was compared to SN-38 in primary cells from recurrent glioblastoma patients. Irinotecan was injected at multiple (~30) sites of the resection cavity wall at a depth of 3 to 5 mm. SN-38 was more cytotoxic than irinotecan at concentrations below 1 µM due to enzyme kinetics. The intraparenchymal administration of irinotecan was safe, with good wound healing and an absence of swelling, inflammation, or pseudo-abscess formation. The median survival post irinotecan administration was 32.6 weeks. The median overall survival was 30.5 months, with a two-year survival rate of 56%. This study demonstrates that local delivery of irinotecan into the brain parenchyma offers a safe route of administration over systemic delivery in the treatment of recurrent glioblastoma.
Collapse
Affiliation(s)
- Christopher McConville
- School of Pharmacy, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Sarah Lastakchi
- School of Pharmacy, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Ali Al Amri
- School of Pharmacy, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Desire Ngoga
- Pediatric Neurosurgery, The Bristol Royal Hospital for Children, Bristol BS2 8BJ, UK
| | - Oluwafikayo Fayeye
- Department of Neurosurgery, University Hospitals Birmingham, NHS Foundation Trust, Birmingham B15 2GW, UK
| | - Garth Cruickshank
- Department of Neurosurgery, University Hospitals Birmingham, NHS Foundation Trust, Birmingham B15 2GW, UK
| |
Collapse
|
10
|
Ullah Z, Abbas Y, Gu J, Ko Soe S, Roy S, Peng T, Guo B. Chemodynamic Therapy of Glioblastoma Multiforme and Perspectives. Pharmaceutics 2024; 16:942. [PMID: 39065639 PMCID: PMC11280080 DOI: 10.3390/pharmaceutics16070942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/08/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Glioblastoma multiforme (GBM), a potential public health issue, is a huge challenge for the advanced scientific realm to solve. Chemodynamic therapy (CDT) based on the Fenton reaction emerged as a state-of-the-art therapeutic modality to treat GBM. However, crossing the blood-brain barrier (BBB) to reach the GBM is another endless marathon. In this review, the physiology of the BBB has been elaborated to understand the mechanism of crossing these potential barriers to treat GBM. Moreover, the designing of Fenton-based nanomaterials has been discussed for the production of reactive oxygen species in the tumor area to eradicate the cancer cells. For effective tumor targeting, biological nanomaterials that can cross the BBB via neurovascular transport channels have also been explored. To overcome the neurotoxicity caused by inorganic nanomaterials, the use of smart nanoagents having both enhanced biocompatibility and effective tumor targeting ability to enhance the efficiency of CDT are systematically summarized. Finally, the advancements in intelligent Fenton-based nanosystems for a multimodal therapeutic approach in addition to CDT are demonstrated. Hopefully, this systematic review will provide a better understanding of Fenton-based CDT and insight into GBM treatment.
Collapse
Affiliation(s)
- Zia Ullah
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen 518055, China; (Z.U.); (Y.A.); (S.K.S.); (S.R.)
| | - Yasir Abbas
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen 518055, China; (Z.U.); (Y.A.); (S.K.S.); (S.R.)
| | - Jingsi Gu
- Education Center and Experiments and Innovations, Harbin Institute of Technology, Shenzhen 518055, China;
| | - Sai Ko Soe
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen 518055, China; (Z.U.); (Y.A.); (S.K.S.); (S.R.)
| | - Shubham Roy
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen 518055, China; (Z.U.); (Y.A.); (S.K.S.); (S.R.)
| | - Tingting Peng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 511436, China
| | - Bing Guo
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen 518055, China; (Z.U.); (Y.A.); (S.K.S.); (S.R.)
| |
Collapse
|
11
|
Andrade-Andrade P, Ordóñez-Rubiano EG, Hakim F. Letter: Efficacy and Safety of Carmustine Wafer Implantation After Ventricular Opening in Glioblastomas, Isocitrate Dehydrogenase-Wildtype, in Adults. Neurosurgery 2024; 95:e26-e27. [PMID: 38712932 DOI: 10.1227/neu.0000000000002984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 03/13/2024] [Indexed: 05/08/2024] Open
Affiliation(s)
- Pedro Andrade-Andrade
- Department of Neurosurgery, Fundación Santa Fe de Bogotá, Bogotá , Colombia
- Faculty of Medicine, Universidad de los Andes, Bogotá , Colombia
| | - Edgar G Ordóñez-Rubiano
- Faculty of Medicine, Universidad de los Andes, Bogotá , Colombia
- Department of Neurosurgery, Fundación Universitaria de Ciencias de la Salud (FUCS) - Hospital de San José, Bogotá , Colombia
| | - Fernando Hakim
- Department of Neurosurgery, Fundación Santa Fe de Bogotá, Bogotá , Colombia
| |
Collapse
|
12
|
White J, White MPJ, Wickremesekera A, Peng L, Gray C. The tumour microenvironment, treatment resistance and recurrence in glioblastoma. J Transl Med 2024; 22:540. [PMID: 38844944 PMCID: PMC11155041 DOI: 10.1186/s12967-024-05301-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/13/2024] [Indexed: 06/10/2024] Open
Abstract
The adaptability of glioblastoma (GBM) cells, encouraged by complex interactions with the tumour microenvironment (TME), currently renders GBM an incurable cancer. Despite intensive research, with many clinical trials, GBM patients rely on standard treatments including surgery followed by radiation and chemotherapy, which have been observed to induce a more aggressive phenotype in recurrent tumours. This failure to improve treatments is undoubtedly a result of insufficient models which fail to incorporate components of the human brain TME. Research has increasingly uncovered mechanisms of tumour-TME interactions that correlate to worsened patient prognoses, including tumour-associated astrocyte mitochondrial transfer, neuronal circuit remodelling and immunosuppression. This tumour hijacked TME is highly implicated in driving therapy resistance, with further alterations within the TME and tumour resulting from therapy exposure inducing increased tumour growth and invasion. Recent developments improving organoid models, including aspects of the TME, are paving an exciting future for the research and drug development for GBM, with the hopes of improving patient survival growing closer. This review focuses on GBMs interactions with the TME and their effect on tumour pathology and treatment efficiency, with a look at challenges GBM models face in sufficiently recapitulating this complex and highly adaptive cancer.
Collapse
Affiliation(s)
- Jasmine White
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand
- Centre for Biodiscovery and School of Biological Sciences, Victoria University of Wellington, Kelburn, Wellington, 6021, New Zealand
| | | | - Agadha Wickremesekera
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand
- Department of Neurosurgery, Wellington Regional Hospital, Wellington, New Zealand
| | - Lifeng Peng
- Centre for Biodiscovery and School of Biological Sciences, Victoria University of Wellington, Kelburn, Wellington, 6021, New Zealand.
| | - Clint Gray
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand.
- Centre for Biodiscovery and School of Biological Sciences, Victoria University of Wellington, Kelburn, Wellington, 6021, New Zealand.
| |
Collapse
|
13
|
Kawaguchi Y, Hanakita S, Yoshida S, Ikemoto T, Oya S. Unmanageable Cerebrospinal Fluid Leakage With Eosinophilic Meningitis in a Gliadel Wafer Implant Patient. Cureus 2024; 16:e59718. [PMID: 38841004 PMCID: PMC11151347 DOI: 10.7759/cureus.59718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2024] [Indexed: 06/07/2024] Open
Abstract
Gliadel wafer implants (Eisai Inc., Woodcliff Lake, NJ, USA) have shown their efficacy in prolonging survival in patients with malignant gliomas. The safety of Gliadel wafers has also been reported; however, there is a certain risk of adverse events. We present a rare case of refractory cerebrospinal fluid (CSF) leakage with eosinophilic meningitis in a patient with glioblastoma who underwent tumor resection with Gliadel wafer implants. A 60-year-old man presented with a glioblastoma in the right temporal lobe. The patient underwent tumor resection with Gliadel wafer implants. During the postoperative course, the patient presented with intractable CSF leakage and the development of a pseudomeningocele. A delayed rise in blood and CSF eosinophil count (a few weeks after the primary operation) and positive drug-induced lymphocyte stimulation test (DLST) results against the Gliadel wafer led to the diagnosis of an allergic reaction to these implants. Removal of the Gliadel wafers resolved the eosinophilic reaction; however, the patient subsequently required a shunt procedure for persistent hydrocephalus. This case highlights the importance of investigating rare causes of refractory CSF leakage and hydrocephalus due to allergic reactions to Gliadel wafers. Delayed elevations of eosinophils in blood and CSF tests may lead to a diagnosis of eosinophilic meningitis. DLST against Gliadel wafers is also useful for diagnosis when it is available. To control the hydrocephalus, not only the shunt procedure but also wafer removal must be considered; however, patients with limited life expectancy are generally hesitant to undergo such additional procedures.
Collapse
Affiliation(s)
- Yuki Kawaguchi
- Neurosurgery, Saitama Medical Center, Saitama Medical University, Kawagoe, JPN
| | - Shunya Hanakita
- Neurosurgery, Saitama Medical Center, Saitama Medical University, Kawagoe, JPN
| | - Shinsuke Yoshida
- Neurosurgery, Saitama Medical Center, Saitama Medical University, Kawagoe, JPN
| | - Tomoko Ikemoto
- Neurosurgery, Saitama Medical Center, Saitama Medical University, Kawagoe, JPN
| | - Soichi Oya
- Neurosurgery, Saitama Medical Center, Saitama Medical University, Kawagoe, JPN
| |
Collapse
|
14
|
WADHWA KARAN, CHAUHAN PAYAL, KUMAR SHOBHIT, PAHWA RAKESH, VERMA RAVINDER, GOYAL RAJAT, SINGH GOVIND, SHARMA ARCHANA, RAO NEHA, KAUSHIK DEEPAK. Targeting brain tumors with innovative nanocarriers: bridging the gap through the blood-brain barrier. Oncol Res 2024; 32:877-897. [PMID: 38686045 PMCID: PMC11056000 DOI: 10.32604/or.2024.047278] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/22/2024] [Indexed: 05/02/2024] Open
Abstract
Background Glioblastoma multiforme (GBM) is recognized as the most lethal and most highly invasive tumor. The high likelihood of treatment failure arises from the presence of the blood-brain barrier (BBB) and stem cells around GBM, which avert the entry of chemotherapeutic drugs into the tumor mass. Objective Recently, several researchers have designed novel nanocarrier systems like liposomes, dendrimers, metallic nanoparticles, nanodiamonds, and nanorobot approaches, allowing drugs to infiltrate the BBB more efficiently, opening up innovative avenues to prevail over therapy problems and radiation therapy. Methods Relevant literature for this manuscript has been collected from a comprehensive and systematic search of databases, for example, PubMed, Science Direct, Google Scholar, and others, using specific keyword combinations, including "glioblastoma," "brain tumor," "nanocarriers," and several others. Conclusion This review also provides deep insights into recent advancements in nanocarrier-based formulations and technologies for GBM management. Elucidation of various scientific advances in conjunction with encouraging findings concerning the future perspectives and challenges of nanocarriers for effective brain tumor management has also been discussed.
Collapse
Affiliation(s)
- KARAN WADHWA
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India
| | - PAYAL CHAUHAN
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India
| | - SHOBHIT KUMAR
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology (MIET) NH-58, Delhi-Roorkee Highway, Meerut, 250005, India
| | - RAKESH PAHWA
- Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra, 136119, India
| | - RAVINDER VERMA
- Department of Pharmaceutical Sciences, Chaudhary Bansi Lal University, Bhiwani, 127021, India
| | - RAJAT GOYAL
- MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, 133207, India
| | - GOVIND SINGH
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India
| | - ARCHANA SHARMA
- Delhi Pharmaceutical Sciences and Research University (DIPSAR), Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - NEHA RAO
- Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra, 136119, India
| | - DEEPAK KAUSHIK
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India
| |
Collapse
|
15
|
Singh RR, Mondal I, Janjua T, Popat A, Kulshreshtha R. Engineered smart materials for RNA based molecular therapy to treat Glioblastoma. Bioact Mater 2024; 33:396-423. [PMID: 38059120 PMCID: PMC10696434 DOI: 10.1016/j.bioactmat.2023.11.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 10/19/2023] [Accepted: 11/14/2023] [Indexed: 12/08/2023] Open
Abstract
Glioblastoma (GBM) is an aggressive malignancy of the central nervous system (CNS) that remains incurable despite the multitude of improvements in cancer therapeutics. The conventional chemo and radiotherapy post-surgery have only been able to improve the prognosis slightly; however, the development of resistance and/or tumor recurrence is almost inevitable. There is a pressing need for adjuvant molecular therapies that can successfully and efficiently block tumor progression. During the last few decades, non-coding RNAs (ncRNAs) have emerged as key players in regulating various hallmarks of cancer including that of GBM. The levels of many ncRNAs are dysregulated in cancer, and ectopic modulation of their levels by delivering antagonists or overexpression constructs could serve as an attractive option for cancer therapy. The therapeutic potential of several types of ncRNAs, including miRNAs, lncRNAs, and circRNAs, has been validated in both in vitro and in vivo models of GBM. However, the delivery of these RNA-based therapeutics is highly challenging, especially to the tumors of the brain as the blood-brain barrier (BBB) poses as a major obstacle, among others. Also, since RNA is extremely fragile in nature, careful considerations must be met while designing a delivery agent. In this review we have shed light on how ncRNA therapy can overcome the limitations of its predecessor conventional therapy with an emphasis on smart nanomaterials that can aide in the safe and targeted delivery of nucleic acids to treat GBM. Additionally, critical gaps that currently exist for successful transition from viral to non-viral vector delivery systems have been identified. Finally, we have provided a perspective on the future directions, potential pathways, and target areas for achieving rapid clinical translation of, RNA-based macromolecular therapy to advance the effective treatment of GBM and other related diseases.
Collapse
Affiliation(s)
- Ravi Raj Singh
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, India
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4072, Australia
- University of Queensland –IIT Delhi Academy of Research (UQIDAR)
| | - Indranil Mondal
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, India
| | - Taskeen Janjua
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Amirali Popat
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4072, Australia
- Department of Functional Materials and Catalysis, Faculty of Chemistry, University of Vienna, Währinger Straße 42, 1090 Vienna, Austria
| | - Ritu Kulshreshtha
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, India
| |
Collapse
|
16
|
Rajaratnam V, Islam MM, Kub EF, Rajaratnam S, Kim KB, Rahman MT, Rashid F, Benko AM, Cook JM, Arnold LA, Mirza SP. Development and validation of an LC-MS/MS method for the determination of ARN14988, an acid ceramidase inhibitor, and its application to a pharmacokinetic study in a mouse model. Biomed Chromatogr 2024; 38:e5754. [PMID: 37750452 DOI: 10.1002/bmc.5754] [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: 06/07/2022] [Revised: 08/31/2023] [Accepted: 09/07/2023] [Indexed: 09/27/2023]
Abstract
Despite aggressive treatment approaches, the overall survival of glioblastoma (GBM) patients remained poor with a strong need for more effective chemotherapeutic agents. A previous study has shown that ARN14988 is more cytotoxic to GBM cells compared to US Food and Drug Administration-approved temozolomide. This finding makes ARN14988 a desirable candidate for further pharmacological assessment. Therefore, an efficient analytical method is needed to quantify ARN14988. Herein, we have developed and validated sample preparation and LC-MS/MS triple quadrupole (QQQ) method for quantification of ARN14988 in mouse plasma. In this method, the liquid-liquid extraction of ARN14988 from mouse plasma was performed using 5% ethyl acetate in hexane. The chromatographic separation was achieved using a C18 -column with mobile phases of 10 mm ammonium acetate (pH 5) and 0.1% formic acid in methanol, within a runtime of 10 min. The monitored transitions were m/z 391.20 → m/z 147.00 for ARN14988, and m/z 455.30 → m/z 165.00 for verapamil (internal standard) in positive electrospray ionization. The developed method for ARN14988 showed linearity over the range of 10-5,000 ng/ml (r2 > 0.99). The selectivity, sensitivity, matrix effect, recovery, stability, inter-day and intraday accuracy and precision were determined using four quality control samples. This validated method was successfully applied to the pharmacokinetic study of ARN14988 in mice.
Collapse
Affiliation(s)
- Vilashini Rajaratnam
- Department of Chemistry & Biochemistry, University of Wisconsin-, Milwaukee, WI, USA
| | | | - Ethan F Kub
- Department of Chemistry & Biochemistry, University of Wisconsin-, Milwaukee, WI, USA
| | - Shaarwin Rajaratnam
- Department of Chemistry & Biochemistry, University of Wisconsin-, Milwaukee, WI, USA
| | - Kyu Bum Kim
- Department of Chemistry & Biochemistry, University of Wisconsin-, Milwaukee, WI, USA
| | - Md Toufiqur Rahman
- Department of Chemistry & Biochemistry, University of Wisconsin-, Milwaukee, WI, USA
| | - Farjana Rashid
- Department of Chemistry & Biochemistry, University of Wisconsin-, Milwaukee, WI, USA
| | - Anna M Benko
- Department of Chemistry & Biochemistry, University of Wisconsin-, Milwaukee, WI, USA
| | - James M Cook
- Department of Chemistry & Biochemistry, University of Wisconsin-, Milwaukee, WI, USA
| | - Leggy A Arnold
- Department of Chemistry & Biochemistry, University of Wisconsin-, Milwaukee, WI, USA
| | - Shama P Mirza
- Department of Chemistry & Biochemistry, University of Wisconsin-, Milwaukee, WI, USA
| |
Collapse
|
17
|
Bérard C, Truillet C, Larrat B, Dhermain F, Estève MA, Correard F, Novell A. Anticancer drug delivery by focused ultrasound-mediated blood-brain/tumor barrier disruption for glioma therapy: From benchside to bedside. Pharmacol Ther 2023; 250:108518. [PMID: 37619931 DOI: 10.1016/j.pharmthera.2023.108518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
The therapeutic management of gliomas remains particularly challenging. Brain tumors present multiple obstacles that make therapeutic innovation complex, mainly due to the presence of blood-tumor and blood-brain barriers (BTB and BBB, respectively) which prevent penetration of anticancer agents into the brain parenchyma. Focused ultrasound-mediated BBB disruption (FUS-BBBD) provides a physical method for non-invasive, local, and reversible BBB disruption. The safety of this technique has been demonstrated in small and large animal models. This approach promises to enhance drug delivery into the brain tumor and therefore to improve survival outcomes by repurposing existing drugs. Several clinical trials continue to be initiated in the last decade. In this review, we provide an overview of the rationale behind the use of FUS-BBBD in gliomas and summarize the preclinical studies investigating different approaches (free drugs, drug-loaded microbubbles and drug-loaded nanocarriers) in combination with this technology in in vivo glioma models. Furthermore, we discuss the current state of clinical trials and devices developed and review the challenges to overcome for clinical use of FUS-BBBD in glioma therapy.
Collapse
Affiliation(s)
- Charlotte Bérard
- Aix Marseille Univ, APHM, CNRS, INP, Inst Neurophysiopathol, Hôpital Timone, Service Pharmacie, 13005 Marseille, France.
| | - Charles Truillet
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, 91401 Orsay, France.
| | - Benoit Larrat
- Université Paris-Saclay, CEA, CNRS, NeuroSpin/BAOBAB, Centre d'études de Saclay, 91191 Gif-sur-Yvette, France.
| | - Frédéric Dhermain
- Radiation Oncology Department, Gustave Roussy University Hospital, 94805 Villejuif, France.
| | - Marie-Anne Estève
- Aix Marseille Univ, APHM, CNRS, INP, Inst Neurophysiopathol, Hôpital Timone, Service Pharmacie, 13005 Marseille, France.
| | - Florian Correard
- Aix Marseille Univ, APHM, CNRS, INP, Inst Neurophysiopathol, Hôpital Timone, Service Pharmacie, 13005 Marseille, France.
| | - Anthony Novell
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, 91401 Orsay, France.
| |
Collapse
|
18
|
Zhang Z, Zhu Z, Zhou P, Zou Y, Yang J, Haick H, Wang Y. Soft Bioelectronics for Therapeutics. ACS NANO 2023; 17:17634-17667. [PMID: 37677154 DOI: 10.1021/acsnano.3c02513] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
Soft bioelectronics play an increasingly crucial role in high-precision therapeutics due to their softness, biocompatibility, clinical accuracy, long-term stability, and patient-friendliness. In this review, we provide a comprehensive overview of the latest representative therapeutic applications of advanced soft bioelectronics, ranging from wearable therapeutics for skin wounds, diabetes, ophthalmic diseases, muscle disorders, and other diseases to implantable therapeutics against complex diseases, such as cardiac arrhythmias, cancer, neurological diseases, and others. We also highlight key challenges and opportunities for future clinical translation and commercialization of soft therapeutic bioelectronics toward personalized medicine.
Collapse
Affiliation(s)
- Zongman Zhang
- Department of Chemical Engineering, Guangdong Technion-Israel Institute of Technology, 241 Daxue Road, Shantou, Guangdong 515063, China
- The Wolfson Department of Chemical Engineering, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Zhongtai Zhu
- Department of Chemical Engineering, Guangdong Technion-Israel Institute of Technology, 241 Daxue Road, Shantou, Guangdong 515063, China
| | - Pengcheng Zhou
- Department of Chemical Engineering, Guangdong Technion-Israel Institute of Technology, 241 Daxue Road, Shantou, Guangdong 515063, China
- The Wolfson Department of Chemical Engineering, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Yunfan Zou
- Department of Biotechnology and Food Engineering, Guangdong Technion-Israel Institute of Technology, 241 Daxue Road, Shantou, Guangdong 515063, China
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Jiawei Yang
- Department of Chemical Engineering, Guangdong Technion-Israel Institute of Technology, 241 Daxue Road, Shantou, Guangdong 515063, China
- The Wolfson Department of Chemical Engineering, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Hossam Haick
- The Wolfson Department of Chemical Engineering, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Yan Wang
- Department of Chemical Engineering, Guangdong Technion-Israel Institute of Technology, 241 Daxue Road, Shantou, Guangdong 515063, China
- The Wolfson Department of Chemical Engineering, Technion-Israel Institute of Technology, Haifa 3200003, Israel
- Guangdong Provincial Key Laboratory of Materials and Technologies for Energy Conversion, Guangdong Technion-Israel Institute of Technology, 241 Daxue Road, Shantou, Guangdong 515063, China
| |
Collapse
|
19
|
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
|
20
|
Lenders V, Koutsoumpou X, Phan P, Soenen SJ, Allegaert K, de Vleeschouwer S, Toelen J, Zhao Z, Manshian BB. Modulation of engineered nanomaterial interactions with organ barriers for enhanced drug transport. Chem Soc Rev 2023; 52:4672-4724. [PMID: 37338993 DOI: 10.1039/d1cs00574j] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2023]
Abstract
The biomedical use of nanoparticles (NPs) has been the focus of intense research for over a decade. As most NPs are explored as carriers to alter the biodistribution, pharmacokinetics and bioavailability of associated drugs, the delivery of these NPs to the tissues of interest remains an important topic. To date, the majority of NP delivery studies have used tumor models as their tool of interest, and the limitations concerning tumor targeting of systemically administered NPs have been well studied. In recent years, the focus has also shifted to other organs, each presenting their own unique delivery challenges to overcome. In this review, we discuss the recent advances in leveraging NPs to overcome four major biological barriers including the lung mucus, the gastrointestinal mucus, the placental barrier, and the blood-brain barrier. We define the specific properties of these biological barriers, discuss the challenges related to NP transport across them, and provide an overview of recent advances in the field. We discuss the strengths and shortcomings of different strategies to facilitate NP transport across the barriers and highlight some key findings that can stimulate further advances in this field.
Collapse
Affiliation(s)
- Vincent Lenders
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium.
| | - Xanthippi Koutsoumpou
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium.
| | - Philana Phan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Stefaan J Soenen
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium.
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Karel Allegaert
- Department of Hospital Pharmacy, Erasmus MC University Medical Center, CN Rotterdam, 3015, The Netherlands
- Clinical Pharmacology and Pharmacotherapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, B3000 Leuven, Belgium
- Leuven Child and Youth Institute, KU Leuven, 3000 Leuven, Belgium
- Woman and Child, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Steven de Vleeschouwer
- Department of Neurosurgery, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Experimental Neurosurgery and Neuroanatomy, Department of Neurosciences, KU Leuven, Leuven, Belgium
- Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| | - Jaan Toelen
- Leuven Child and Youth Institute, KU Leuven, 3000 Leuven, Belgium
- Woman and Child, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
- Department of Pediatrics, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Zongmin Zhao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Bella B Manshian
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium.
| |
Collapse
|
21
|
Roux A, Aboubakr O, Elia A, Moiraghi A, Benevello C, Fathallah H, Parraga E, Oppenheim C, Chretien F, Dezamis E, Zanello M, Pallud J. Carmustine wafer implantation for supratentorial glioblastomas, IDH-wildtype in "extreme" neurosurgical conditions. Neurosurg Rev 2023; 46:140. [PMID: 37329341 DOI: 10.1007/s10143-023-02052-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 05/15/2023] [Accepted: 06/10/2023] [Indexed: 06/19/2023]
Abstract
We assessed the feasibility of Carmustine wafer implantation in "extreme" conditions (i.e. patients > 80 years and Karnofsky Performance Status score < 50) and of implantation ≥ 12 Carmustine wafers in adult patients harbouring a newly diagnosed supratentorial glioblastoma, IDH-wildtype. We performed an observational, retrospective single-centre cohort study at a tertiary surgical neuro-oncological centre between January 2006 and December 2021. Four hundred eighty patients who benefited from a surgical resection at first-line treatment were included. We showed that Carmustine wafer implantation in patients > 80 years, in patients with a Karnofsky performance status score < 50, and that implantation ≥ 12 Carmustine wafers (1) did not increase overall postoperative complication rates, (2) did not affect the completion of standard radiochemotherapy protocol, (3) did not worsen the postoperative Karnofsky Performance Status scores, and (4) did not significantly affect the time to oncological treatment. We showed that the implantation of ≥ 12 Carmustine wafers improved progression-free survival (31.0 versus 10.0 months, p = 0.025) and overall survival (39.0 versus 16.5 months, p = 0.041) without increasing postoperative complication rates. Carmustine wafer implantation during the surgical resection of a newly diagnosed supratentorial glioblastoma, IDH-wildtype is safe and efficient in patients > 80 years and in patients with preoperative Karnofsky Performance Status score < 50. The number of Carmustine wafers should be adapted (up to 16 in our experience) to the resection cavity to improve survival without increasing postoperative overall complication rates.
Collapse
Affiliation(s)
- Alexandre Roux
- Service de Neurochirurgie, GHU Paris Psychiatrie Et Neurosciences, Site Sainte Anne, 75014, Paris, France.
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014, Paris, France.
| | - Oumaima Aboubakr
- Service de Neurochirurgie, GHU Paris Psychiatrie Et Neurosciences, Site Sainte Anne, 75014, Paris, France
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014, Paris, France
| | - Angela Elia
- Service de Neurochirurgie, GHU Paris Psychiatrie Et Neurosciences, Site Sainte Anne, 75014, Paris, France
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014, Paris, France
| | - Alessandro Moiraghi
- Service de Neurochirurgie, GHU Paris Psychiatrie Et Neurosciences, Site Sainte Anne, 75014, Paris, France
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014, Paris, France
| | - Chiara Benevello
- Service de Neurochirurgie, GHU Paris Psychiatrie Et Neurosciences, Site Sainte Anne, 75014, Paris, France
| | - Houssem Fathallah
- Service de Neurochirurgie, GHU Paris Psychiatrie Et Neurosciences, Site Sainte Anne, 75014, Paris, France
| | - Eduardo Parraga
- Service de Neurochirurgie, GHU Paris Psychiatrie Et Neurosciences, Site Sainte Anne, 75014, Paris, France
| | - Catherine Oppenheim
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014, Paris, France
- Service de Neuroradiologie, GHU Paris Psychiatrie Et Neurosciences, Site Sainte Anne, 75014, Paris, France
| | - Fabrice Chretien
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014, Paris, France
- Service de Neuropathologie, GHU Paris Psychiatrie Et Neurosciences, Site Sainte Anne, 75014, Paris, France
| | - Edouard Dezamis
- Service de Neurochirurgie, GHU Paris Psychiatrie Et Neurosciences, Site Sainte Anne, 75014, Paris, France
| | - Marc Zanello
- Service de Neurochirurgie, GHU Paris Psychiatrie Et Neurosciences, Site Sainte Anne, 75014, Paris, France
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014, Paris, France
| | - Johan Pallud
- Service de Neurochirurgie, GHU Paris Psychiatrie Et Neurosciences, Site Sainte Anne, 75014, Paris, France
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014, Paris, France
| |
Collapse
|
22
|
Rončević A, Koruga N, Soldo Koruga A, Rončević R, Rotim T, Šimundić T, Kretić D, Perić M, Turk T, Štimac D. Personalized Treatment of Glioblastoma: Current State and Future Perspective. Biomedicines 2023; 11:1579. [PMID: 37371674 DOI: 10.3390/biomedicines11061579] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/24/2023] [Accepted: 05/27/2023] [Indexed: 06/29/2023] Open
Abstract
Glioblastoma (GBM) is the most aggressive glial tumor of the central nervous system. Despite intense scientific efforts, patients diagnosed with GBM and treated with the current standard of care have a median survival of only 15 months. Patients are initially treated by a neurosurgeon with the goal of maximal safe resection of the tumor. Obtaining tissue samples during surgery is indispensable for the diagnosis of GBM. Technological improvements, such as navigation systems and intraoperative monitoring, significantly advanced the possibility of safe gross tumor resection. Usually within six weeks after the surgery, concomitant radiotherapy and chemotherapy with temozolomide are initiated. However, current radiotherapy regimens are based on population-level studies and could also be improved. Implementing artificial intelligence in radiotherapy planning might be used to individualize treatment plans. Furthermore, detailed genetic and molecular markers of the tumor could provide patient-tailored immunochemotherapy. In this article, we review current standard of care and possibilities of personalizing these treatments. Additionally, we discuss novel individualized therapeutic options with encouraging results. Due to inherent heterogeneity of GBM, applying patient-tailored treatment could significantly prolong survival of these patients.
Collapse
Affiliation(s)
- Alen Rončević
- Department of Neurosurgery, University Hospital Center Osijek, 31000 Osijek, Croatia
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Nenad Koruga
- Department of Neurosurgery, University Hospital Center Osijek, 31000 Osijek, Croatia
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Anamarija Soldo Koruga
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Neurology, University Hospital Center Osijek, 31000 Osijek, Croatia
| | - Robert Rončević
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Diagnostic and Interventional Radiology, University Hospital Center Osijek, 31000 Osijek, Croatia
| | - Tatjana Rotim
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Diagnostic and Interventional Radiology, University Hospital Center Osijek, 31000 Osijek, Croatia
| | - Tihana Šimundić
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Nephrology, University Hospital Center Osijek, 31000 Osijek, Croatia
| | - Domagoj Kretić
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Diagnostic and Interventional Radiology, University Hospital Center Osijek, 31000 Osijek, Croatia
| | - Marija Perić
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Cytology, University Hospital Center Osijek, 31000 Osijek, Croatia
| | - Tajana Turk
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Diagnostic and Interventional Radiology, University Hospital Center Osijek, 31000 Osijek, Croatia
| | - Damir Štimac
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Radiology, National Memorial Hospital Vukovar, 32000 Vukovar, Croatia
| |
Collapse
|
23
|
Wang F, Huang Q, Su H, Sun M, Wang Z, Chen Z, Zheng M, Chakroun R, Monroe M, Chen D, Wang Z, Gorelick N, Serra R, Wang H, Guan Y, Suk J, Tyler B, Brem H, Hanes J, Cui H. Self-assembling paclitaxel-mediated stimulation of tumor-associated macrophages for postoperative treatment of glioblastoma. Proc Natl Acad Sci U S A 2023; 120:e2204621120. [PMID: 37098055 PMCID: PMC10161130 DOI: 10.1073/pnas.2204621120] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 03/09/2023] [Indexed: 04/26/2023] Open
Abstract
The unique cancer-associated immunosuppression in brain, combined with a paucity of infiltrating T cells, contributes to the low response rate and poor treatment outcomes of T cell-based immunotherapy for patients diagnosed with glioblastoma multiforme (GBM). Here, we report on a self-assembling paclitaxel (PTX) filament (PF) hydrogel that stimulates macrophage-mediated immune response for local treatment of recurrent glioblastoma. Our results suggest that aqueous PF solutions containing aCD47 can be directly deposited into the tumor resection cavity, enabling seamless hydrogel filling of the cavity and long-term release of both therapeutics. The PTX PFs elicit an immune-stimulating tumor microenvironment (TME) and thus sensitizes tumor to the aCD47-mediated blockade of the antiphagocytic "don't eat me" signal, which subsequently promotes tumor cell phagocytosis by macrophages and also triggers an antitumor T cell response. As adjuvant therapy after surgery, this aCD47/PF supramolecular hydrogel effectively suppresses primary brain tumor recurrence and prolongs overall survivals with minimal off-target side effects.
Collapse
Affiliation(s)
- Feihu Wang
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD21218
- Whiting School of Engineering, Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD21218
- Center for Nanomedicine, Wilmer Eye Institute, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
| | - Qian Huang
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, The Johns Hopkins University, Baltimore, MD21205
| | - Hao Su
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD21218
- Whiting School of Engineering, Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD21218
| | - Mingjiao Sun
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD21218
- Whiting School of Engineering, Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD21218
- Center for Nanomedicine, Wilmer Eye Institute, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
| | - Zeyu Wang
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD21218
| | - Ziqi Chen
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD21218
- Whiting School of Engineering, Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD21218
| | - Mengzhen Zheng
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD21218
- Whiting School of Engineering, Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD21218
| | - Rami W. Chakroun
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD21218
- Whiting School of Engineering, Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD21218
| | - Maya K. Monroe
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD21218
- Whiting School of Engineering, Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD21218
| | - Daiqing Chen
- Center for Nanomedicine, Wilmer Eye Institute, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
| | - Zongyuan Wang
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD21218
- Whiting School of Engineering, Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD21218
| | - Noah Gorelick
- Department of Neurosurgery, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
| | - Riccardo Serra
- Department of Neurosurgery, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
| | - Han Wang
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD21218
- Whiting School of Engineering, Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD21218
| | - Yun Guan
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, The Johns Hopkins University, Baltimore, MD21205
- Department of Neurological Surgery, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
| | - Jung Soo Suk
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD21218
- Center for Nanomedicine, Wilmer Eye Institute, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
- Department of Neurological Surgery, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
| | - Betty Tyler
- Department of Neurosurgery, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
| | - Henry Brem
- Department of Neurosurgery, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
- Department of Ophthalmology, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
- Department of Biomedical Engineering, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
| | - Justin Hanes
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD21218
- Center for Nanomedicine, Wilmer Eye Institute, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
- Department of Ophthalmology, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
- Department of Biomedical Engineering, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
| | - Honggang Cui
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD21218
- Whiting School of Engineering, Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD21218
- Center for Nanomedicine, Wilmer Eye Institute, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, School of Medicine, The Johns Hopkins University, Baltimore, MD21231
- Department of Materials Science and Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD21218
| |
Collapse
|
24
|
Champeaux-Depond C, Jecko V, Weller J, Constantinou P, Tuppin P, Metellus P. Newly Diagnosed High-Grade Glioma Surgery with Carmustine Wafers Implantation. A Long-Term Nationwide Retrospective Study. World Neurosurg 2023; 173:e778-e786. [PMID: 36906091 DOI: 10.1016/j.wneu.2023.03.015] [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: 02/08/2023] [Revised: 03/03/2023] [Accepted: 03/04/2023] [Indexed: 03/11/2023]
Abstract
BACKGROUND Widespread use of carmustine wafers (CWs) to treat high-grade gliomas (HGG) has been limited by uncertainties about their efficacy. We sought to assess the outcome of patients after newly diagnosed HGG surgery with CW implantation and search for associated factors. METHODS We processed the French medico-administrative national database between 2008 and 2019 to retrieve ad hoc cases. Survival methods were implemented. RESULTS In total, 1608 patients who had CW implantation after HGG resection at 42 different institutions between 2008 and 2019 were identified; 36.7% were female and, median age at HGG resection with CW implantation was 61.5 years, interquartile range (IQR) [52.9-69.1]. A total of 1460 patients (90.8%) had died at data collection at a median age at death of 63.5 years, IQR [55.3-71.2]. Median overall survival (OS) was 1.42 years, 95% confidence interval [CI] 1.35-1.49, i.e., 16.8 months. Median age at death was 63.5 years, IQR [55.3-71.2]. OS at 1, 2, and, 5 years was 67.4%, 95% CI 65.1-69.7; 33.1%, 95% CI 30.9-35.5; and 10.7%, 95% CI 9.2-12.4, respectively. In the adjusted regression, sex (hazard ratio [HR] 0.82, 95% CI 0.74-0.92, P < 0.001), age at HGG surgery with CW implantation (HR 1.02, 95% CI 1.02-1.03, P < 0.001), adjuvant radiotherapy (HR 0.78, 95% CI 0.7-0.86, P < 0.001), chemotherapy by temozolomide (HR 0.7, 95% CI 0.63-0.79, P < 0.001), and redo surgery for HGG recurrence (HR 0.81, 95% CI 0.69-0.94, P = 0.005) remained significantly associated with the outcome. CONCLUSIONS OS of patients with newly diagnosed HGG who underwent surgery with CW implantation is better in young patients, those of the female sex, and for those who complete concomitant chemoradiotherapy. Redo surgery for HGG recurrence also was associated with prolonged survival.
Collapse
Affiliation(s)
| | - Vincent Jecko
- Department of Neurosurgery, Pellegrin Hospital, Bordeaux, France
| | | | - Panayotis Constantinou
- Direction de la Stratégie, des Etudes et des Statistiques, Caisse Nationale de L'Assurance Maladie, Paris, France
| | - Philippe Tuppin
- Direction de la Stratégie, des Etudes et des Statistiques, Caisse Nationale de L'Assurance Maladie, Paris, France
| | - Philippe Metellus
- Department of Neurosurgery, Hôpital privé Clairval - Ramsay Santé, Marseille, France; Institut de Neurophysiopathologie - CNRS UMR 7051, Aix-Marseille Université, Marseille, France
| |
Collapse
|
25
|
Champeaux-Depond C, Jecko V, Weller J, Constantinou P, Tuppin P, Metellus P. Recurrent high grade glioma surgery with carmustine wafers implantation: a long-term nationwide retrospective study. J Neurooncol 2023; 162:343-352. [PMID: 36991304 DOI: 10.1007/s11060-023-04295-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/10/2023] [Indexed: 03/31/2023]
Abstract
PURPOSE Widespread use of carmustine wafers (CW) to treat high-grade gliomas (HGG) has been limited by uncertainties about its efficacy. To assess the outcome of patients after recurrent HGG surgery with CW implantation and, search for associated factors. METHODS We processed the French medico-administrative national database between 2008 and 2019 to retrieve ad hoc cases. Survival methods were implemented. RESULTS 559 patients who had CW implantation after recurrent HGG resection at 41 different institutions between 2008 and 2019 were identified. 35.6% were female and, median age at HGG resection with CW implantation was 58.1 years, IQR [50-65.4]. 520 patients (93%) had died at data collection with a median age at death of 59.7 years, IQR [51.6-67.1]. Median overall survival (OS) was 1.1 years, 95%CI[0.97-1.2], id est 13.2 months. Median age at death was 59.7 years, IQR [51.6-67.1]. OS at 1, 2 and 5 years was 52.1%, 95%CI[48.1-56.4], 24.6%, 95%CI[21.3-28.5] & 8%, 95%CI[5.9-10.7] respectively. In the adjusted regression, bevacizumab given before CW implantation, (HR = 1.98, 95%CI[1.49-2.63], p < 0.001), a longer delay between the first and the second HGG surgery (HR = 1, 95%CI[1-1], p < 0.001), RT given before and after CW implantation (HR = 0.59, 95%CI[0.39-0.87], p = 0.009) and TMZ given before and after CW implantation (HR = 0.81, 95%CI[0.66-0.98], p = 0.034) remained significantly associated with a longer survival. CONCLUSION OS of patients with recurrent HGG that underwent surgery with CW implantation is better in case of prolonged delay between the two resections and, for the patients who had RT and TMZ before and after CW implantation.
Collapse
Affiliation(s)
- Charles Champeaux-Depond
- Department of Neurosurgery, Hôpital Privé Clairval - Ramsay Santé, 317 Bd du Redon, 13009, Marseille, France.
| | - Vincent Jecko
- Department of Neurosurgery, Pellegrin Hospital, 33000, Bordeaux, France
| | - Joconde Weller
- Agence Régionale de Santé, 2 Bis, Avenue Georges Brassens, CS 61002 - 97743, Saint Denis Cedex 9, France
| | - Panayotis Constantinou
- Direction de la Stratégie, des Etudes et des Statistiques, Caisse Nationale de L'Assurance Maladie, 26-50, Avenue du Professeur André Lemierre, 75986, Paris Cedex 20, France
| | - Philippe Tuppin
- Direction de la Stratégie, des Etudes et des Statistiques, Caisse Nationale de L'Assurance Maladie, 26-50, Avenue du Professeur André Lemierre, 75986, Paris Cedex 20, France
| | - Philippe Metellus
- Department of Neurosurgery, Hôpital Privé Clairval - Ramsay Santé, 317 Bd du Redon, 13009, Marseille, France
- Institut de Neurophysiopathologie-CNRS UMR 7051, Aix-Marseille Université, Marseille, France
| |
Collapse
|
26
|
Cruz N, Herculano-Carvalho M, Roque D, Faria CC, Cascão R, Ferreira HA, Reis CP, Matela N. Highlighted Advances in Therapies for Difficult-To-Treat Brain Tumours Such as Glioblastoma. Pharmaceutics 2023; 15:pharmaceutics15030928. [PMID: 36986790 PMCID: PMC10054750 DOI: 10.3390/pharmaceutics15030928] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/25/2023] [Accepted: 03/10/2023] [Indexed: 03/15/2023] Open
Abstract
Glioblastoma multiforme (GBM) remains a challenging disease, as it is the most common and deadly brain tumour in adults and has no curative solution and an overall short survival time. This incurability and short survival time means that, despite its rarity (average incidence of 3.2 per 100,000 persons), there has been an increased effort to try to treat this disease. Standard of care in newly diagnosed glioblastoma is maximal tumour resection followed by initial concomitant radiotherapy and temozolomide (TMZ) and then further chemotherapy with TMZ. Imaging techniques are key not only to diagnose the extent of the affected tissue but also for surgery planning and even for intraoperative use. Eligible patients may combine TMZ with tumour treating fields (TTF) therapy, which delivers low-intensity and intermediate-frequency electric fields to arrest tumour growth. Nonetheless, the blood–brain barrier (BBB) and systemic side effects are obstacles to successful chemotherapy in GBM; thus, more targeted, custom therapies such as immunotherapy and nanotechnological drug delivery systems have been undergoing research with varying degrees of success. This review proposes an overview of the pathophysiology, possible treatments, and the most (not all) representative examples of the latest advancements.
Collapse
Affiliation(s)
- Nuno Cruz
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
- iMED.ULisboa, Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Manuel Herculano-Carvalho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Department of Neurosurgery, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte (CHULN), 1649-028 Lisboa, Portugal
| | - Diogo Roque
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Department of Neurosurgery, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte (CHULN), 1649-028 Lisboa, Portugal
| | - Cláudia C. Faria
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Department of Neurosurgery, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte (CHULN), 1649-028 Lisboa, Portugal
| | - Rita Cascão
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Hugo Alexandre Ferreira
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Catarina Pinto Reis
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
- iMED.ULisboa, Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Correspondence: (C.P.R.); (N.M.); Tel.: +351-217-946-400 (ext. 14244) (C.P.R.); Fax: +351-217-946-470 (C.P.R.)
| | - Nuno Matela
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
- Correspondence: (C.P.R.); (N.M.); Tel.: +351-217-946-400 (ext. 14244) (C.P.R.); Fax: +351-217-946-470 (C.P.R.)
| |
Collapse
|
27
|
Das D, Narayanan D, Ramachandran R, Gowd GS, Manohar M, Arumugam T, Panikar D, Nair SV, Koyakutty M. Intracranial nanomedicine-gel with deep brain-penetration for glioblastoma therapy. J Control Release 2023; 355:474-488. [PMID: 36739909 DOI: 10.1016/j.jconrel.2023.01.085] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/15/2022] [Accepted: 01/31/2023] [Indexed: 02/07/2023]
Abstract
Glioblastoma Multiforme (GBM) is one of the challenging tumors to treat as it recurs, almost 100%, even after surgery, radiation, and chemotherapy. In many cases, recurrence happens within 2-3cm depth of the resected tumor margin, indicating the inefficacy of current anti-glioma drugs to penetrate deep into the brain tissue. Here, we report an injectable nanoparticle-gel system, capable of providing deep brain penetration of drug up to 4 cm, releasing in a sustained manner up to >15 days. The system consists of ∼222 nm sized PLGA nanoparticles (NP-222) loaded with an anti-glioma drug, Carmustine (BCNU), and coated with a thick layer of polyethylene glycol (PEG). Upon release of the drug from PLGA core, it will interact with the outer PEG-layer leading to the formation of PEG-BCNU nanocomplexes of size ∼33 nm (BCNU-NC-33), which could penetrate >4 cm deep into the brain tissue compared to the free drug (< 5 mm). In vitro drug release showed sustained release of drug for 15 days by BCNU-NP gel, and enhanced cytotoxicity by BCNU-NC-33 drug-nanocomplexes in glioma cell lines. Ex vivo goat-brain phantom studies showed drug diffusion up to 4 cm in tissue and in vivo brain-diffusion studies showed almost complete coverage within the rat brain (∼1.2 cm), with ∼55% drug retained in the tissue by day-15, compared to only ∼5% for free BCNU. Rat orthotopic glioma studies showed excellent anti-tumor efficacy by BCNU-NP gel compared to free drug, indicating the potential of the gel-system for anti-glioma therapy. In effect, we demonstrate a unique method of sustained release of drug in the brain using larger PLGA nanoparticles acting as a reservoir while deep-penetration of the released drug was achieved by in situ formation of drug-nanocomplexes of size <50 nm which is less than the native pore size of brain tissue (> 100 nm). This method will have a major impact on a challenging field of brain drug delivery.
Collapse
Affiliation(s)
- Devika Das
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - Dhanya Narayanan
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - Ranjith Ramachandran
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - Genekehal Siddaramana Gowd
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - Maneesh Manohar
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - Thennavan Arumugam
- Central Lab Animal Facility, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - Dilip Panikar
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - Shantikumar V Nair
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
| | - Manzoor Koyakutty
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India.
| |
Collapse
|
28
|
Matsuda R, Maeoka R, Tokuda N, Nakazawa T, Morimoto T, Kotsugi M, Takeshima Y, Tamura K, Yamada S, Nishimura F, Nakagawa I, Park YS, Nakase H. Intraoperative Ventricular Opening has No Effect on Complication Development Following BCNU Wafer Implantation for Malignant Glioma. World Neurosurg 2023; 171:e707-e713. [PMID: 36574919 DOI: 10.1016/j.wneu.2022.12.090] [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: 10/21/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022]
Abstract
OBJECTIVE To evaluate the safety profile of bis-chloroethyl-nitrosourea (BCNU) wafer implantation after malignant glioma resection with or without ventricular opening (VO). METHODS This single-center retrospective study included 66 consecutive patients with BCNU wafer implantation after malignant glioma resection between March 2013 and August 2021. The patients were categorized into 2 groups based on whether VO occurred during the malignant glioma resection. Fifty-eight patients had glioblastoma, and 8 had anaplastic astrocytoma or oligodendroglioma. Forty-eight patients underwent an initial treatment, and 18 underwent recurrent surgeries. Infection, hydrocephalus, subcutaneous fluid collection, chronic subdural hematoma, early seizure after surgery within 1 month, symptomatic edema surrounding the resected cavity, cyst formation, and postoperative hemorrhage were defined as adverse events (AEs). RESULTS Thirty-three patients underwent resection with VO, and 33 without. The median survival time was 28 months in the initial treatment group and 11.5 months in the recurrent treatment group. The with and without VO groups had similar median survival times. Postoperative AEs occurred in 7/33 patients (21.2%) with VO and 10/33 (30.3%) without VO, with no difference between them (P = 0.574). CONCLUSIONS This study showed that VO during surgery with BCNU wafer implantation might not influence the occurrence of postoperative AEs. If VO happens, BCNU wafer implantation can be performed safely with accurate closing of the ventricle.
Collapse
Affiliation(s)
- Ryosuke Matsuda
- Department of Neurosurgery, Nara Medical University, Kashihara, Nara, Japan.
| | - Ryosuke Maeoka
- Department of Neurosurgery, Nara Medical University, Kashihara, Nara, Japan
| | - Noriaki Tokuda
- Department of Neurosurgery, Nara Medical University, Kashihara, Nara, Japan
| | - Tsutomu Nakazawa
- Department of Neurosurgery, Nara Medical University, Kashihara, Nara, Japan
| | - Takayuki Morimoto
- Department of Neurosurgery, Nara Medical University, Kashihara, Nara, Japan; Department of Neurosurgery, Okanami General Hospital, Iga, Mie, Japan
| | - Masashi Kotsugi
- Department of Neurosurgery, Nara Medical University, Kashihara, Nara, Japan
| | - Yasuhiro Takeshima
- Department of Neurosurgery, Nara Medical University, Kashihara, Nara, Japan
| | - Kentaro Tamura
- Department of Neurosurgery, Nara Medical University, Kashihara, Nara, Japan
| | - Shuichi Yamada
- Department of Neurosurgery, Nara Medical University, Kashihara, Nara, Japan
| | - Fumihiko Nishimura
- Department of Neurosurgery, Nara Medical University, Kashihara, Nara, Japan
| | - Ichiro Nakagawa
- Department of Neurosurgery, Nara Medical University, Kashihara, Nara, Japan
| | - Young-Soo Park
- Department of Neurosurgery, Nara Medical University, Kashihara, Nara, Japan
| | - Hiroyuki Nakase
- Department of Neurosurgery, Nara Medical University, Kashihara, Nara, Japan
| |
Collapse
|
29
|
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
|
30
|
Haddad AF, Aghi MK, Butowski N. Novel intraoperative strategies for enhancing tumor control: Future directions. Neuro Oncol 2022; 24:S25-S32. [PMID: 36322096 PMCID: PMC9629473 DOI: 10.1093/neuonc/noac090] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2023] Open
Abstract
Maximal safe surgical resection plays a key role in the care of patients with gliomas. A range of technologies have been developed to aid surgeons in distinguishing tumor from normal tissue, with the goal of increasing tumor resection and limiting postoperative neurological deficits. Technologies that are currently being investigated to aid in improving tumor control include intraoperative imaging modalities, fluorescent tumor makers, intraoperative cell and molecular profiling of tumors, improved microscopic imaging, intraoperative mapping, augmented and virtual reality, intraoperative drug and radiation delivery, and ablative technologies. In this review, we summarize the aforementioned advancements in neurosurgical oncology and implications for improving patient outcomes.
Collapse
Affiliation(s)
- Alexander F Haddad
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Manish K Aghi
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Nicholas Butowski
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
31
|
Ruiz-Molina D, Mao X, Alfonso-Triguero P, Lorenzo J, Bruna J, Yuste VJ, Candiota AP, Novio F. Advances in Preclinical/Clinical Glioblastoma Treatment: Can Nanoparticles Be of Help? Cancers (Basel) 2022; 14:4960. [PMID: 36230883 PMCID: PMC9563739 DOI: 10.3390/cancers14194960] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 11/24/2022] Open
Abstract
Glioblastoma multiforme (GB) is the most aggressive and frequent primary malignant tumor in the central nervous system (CNS), with unsatisfactory and challenging treatment nowadays. Current standard of care includes surgical resection followed by chemotherapy and radiotherapy. However, these treatments do not much improve the overall survival of GB patients, which is still below two years (the 5-year survival rate is below 7%). Despite various approaches having been followed to increase the release of anticancer drugs into the brain, few of them demonstrated a significant success, as the blood brain barrier (BBB) still restricts its uptake, thus limiting the therapeutic options. Therefore, enormous efforts are being devoted to the development of novel nanomedicines with the ability to cross the BBB and specifically target the cancer cells. In this context, the use of nanoparticles represents a promising non-invasive route, allowing to evade BBB and reducing systemic concentration of drugs and, hence, side effects. In this review, we revise with a critical view the different families of nanoparticles and approaches followed so far with this aim.
Collapse
Affiliation(s)
- Daniel Ruiz-Molina
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain
| | - Xiaoman Mao
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain
| | - Paula Alfonso-Triguero
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain
- Institut de Biotecnologia i de Biomedicina, Departament de Bioquimica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
| | - Julia Lorenzo
- Institut de Biotecnologia i de Biomedicina, Departament de Bioquimica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
| | - Jordi Bruna
- Neuro-Oncology Unit, Bellvitge University Hospital-ICO (IDIBELL), Avinguda de la Gran Via de l’Hospitalet, 199-203, L’Hospitalet de Llobregat, 08908 Barcelona, Spain
| | - Victor J. Yuste
- Instituto de Neurociencias. Universitat Autònoma de Barcelona (UAB), Campus UAB, 08193 Cerdanyola del Vallès, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Campus UAB, 08193 Cerdanyola del Vallès, Spain
| | - Ana Paula Candiota
- Institut de Biotecnologia i de Biomedicina, Departament de Bioquimica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
- Centro de Investigación Biomédica en Red: Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 08193 Cerdanyola del Vallès, Spain
| | - Fernando Novio
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain
- Departament de Química, Universitat Autònoma de Barcelona (UAB), Campus UAB, 08193 Cerdanyola del Vallès, Spain
| |
Collapse
|
32
|
Walker DA. Childhood brain tumors: It is the child’s brain that really matters. Front Oncol 2022; 12:982914. [PMID: 36267979 PMCID: PMC9576866 DOI: 10.3389/fonc.2022.982914] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/19/2022] [Indexed: 11/13/2022] Open
|
33
|
Brain targeted delivery of carmustine using chitosan coated nanoparticles via nasal route for glioblastoma treatment. Int J Biol Macromol 2022; 221:435-445. [PMID: 36067850 DOI: 10.1016/j.ijbiomac.2022.08.210] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 08/25/2022] [Accepted: 08/31/2022] [Indexed: 11/24/2022]
Abstract
This study aims to develop chitosan-coated PLGA nanoparticles intended for nose-to-brain delivery of carmustine. Formulations were prepared by the double emulsion solvent evaporation method and optimized by using Box-Behnken Design. The optimized nanoparticles were obtained to satisfactory levels in terms of particle size, PDI, entrapment efficiency, and drug loading. In vitro drug release and ex-vivo permeation showed sustained release and enhanced permeability (approx. 2 fold) of carmustine compared to drug suspension. The AUC0-t of brain obtained with carmustine-loaded nanoparticles via nasal administration in Albino Wistar rats was 2.8 and 14.7 times that of intranasal carmustine suspension and intravenous carmustine, respectively. The MTT assay on U87 MG cell line showed a significant decrease (P < 0.05) in the IC50 value of the formulation (71.23 μg ml-1) as compared to drug suspension (90.02 μg ml-1).These findings suggest chitosan coated nanoparticles could be used to deliver carmustine via intranasal administration to treat Glioblastoma multiforme.
Collapse
|
34
|
Novel rapid intraoperative qualitative tumor detection by a residual convolutional neural network using label-free stimulated Raman scattering microscopy. Acta Neuropathol Commun 2022; 10:109. [PMID: 35933416 PMCID: PMC9356422 DOI: 10.1186/s40478-022-01411-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/17/2022] [Indexed: 12/03/2022] Open
Abstract
Determining the presence of tumor in biopsies and the decision-making during resections is often dependent on intraoperative rapid frozen-section histopathology. Recently, stimulated Raman scattering microscopy has been introduced to rapidly generate digital hematoxylin-and-eosin-stained-like images (stimulated Raman histology) for intraoperative analysis. To enable intraoperative prediction of tumor presence, we aimed to develop a new deep residual convolutional neural network in an automated pipeline and tested its validity. In a monocentric prospective clinical study with 94 patients undergoing biopsy, brain or spinal tumor resection, Stimulated Raman histology images of intraoperative tissue samples were obtained using a fiber-laser-based stimulated Raman scattering microscope. A residual network was established and trained in ResNetV50 to predict three classes for each image: (1) tumor, (2) non-tumor, and (3) low-quality. The residual network was validated on images obtained in three small random areas within the tissue samples and were blindly independently reviewed by a neuropathologist as ground truth. 402 images derived from 132 tissue samples were analyzed representing the entire spectrum of neurooncological surgery. The automated workflow took in a mean of 240 s per case, and the residual network correctly classified tumor (305/326), non-tumorous tissue (49/67), and low-quality (6/9) images with an inter-rater agreement of 89.6% (κ = 0.671). An excellent internal consistency was found among the random areas with 90.2% (Cα = 0.942) accuracy. In conclusion, the novel stimulated Raman histology-based residual network can reliably detect the microscopic presence of tumor and differentiate from non-tumorous brain tissue in resection and biopsy samples within 4 min and may pave a promising way for an alternative rapid intraoperative histopathological decision-making tool.
Collapse
|
35
|
Efficient spatially targeted gene editing using a near-infrared activatable protein-conjugated nanoparticle for brain applications. Nat Commun 2022; 13:4135. [PMID: 35840564 PMCID: PMC9287341 DOI: 10.1038/s41467-022-31791-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 07/05/2022] [Indexed: 12/27/2022] Open
Abstract
Spatial control of gene expression is critical to modulate cellular functions and deconstruct the function of individual genes in biological processes. Light-responsive gene-editing formulations have been recently developed; however, they have shown limited applicability in vivo due to poor tissue penetration, limited cellular transfection and the difficulty in evaluating the activity of the edited cells. Here, we report a formulation composed of upconversion nanoparticles conjugated with Cre recombinase enzyme through a photocleavable linker, and a lysosomotropic agent that facilitates endolysosomal escape. This formulation allows in vitro spatial control in gene editing after activation with near-infrared light. We further demonstrate the potential of this formulation in vivo through three different paradigms: (i) gene editing in neurogenic niches, (ii) gene editing in the ventral tegmental area to facilitate monitoring of edited cells by precise optogenetic control of reward and reinforcement, and (iii) gene editing in a localized brain region via a noninvasive administration route (i.e., intranasal). Spatial control of gene expression allows precise control over biological processes. Here, the authors develop an efficient light-responsive formulation based on upconversion nanoparticles, and demonstrate on-demand genetic manipulation in deep brain tissue.
Collapse
|
36
|
Boccellato C, Rehm M. Glioblastoma, from disease understanding towards optimal cell-based in vitro models. Cell Oncol (Dordr) 2022; 45:527-541. [PMID: 35763242 PMCID: PMC9424171 DOI: 10.1007/s13402-022-00684-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2022] [Indexed: 11/24/2022] Open
Abstract
Abstract
Background
Glioblastoma (GBM) patients are notoriously difficult to treat and ultimately all succumb to disease. This unfortunate scenario motivates research into better characterizing and understanding this disease, and into developing novel research tools by which potential novel therapeutics and treatment options initially can be evaluated pre-clinically. Here, we provide a concise overview of glioblastoma epidemiology, disease classification, the challenges faced in the treatment of glioblastoma and current novel treatment strategies. From this, we lead into a description and assessment of advanced cell-based models that aim to narrow the gap between pre-clinical and clinical studies. Such invitro models are required to deliver reliable and meaningful data for the development and pre-validation of novel therapeutics and treatments.
Conclusions
The toolbox for GBM cell-based models has expanded substantially, with the possibility of 3D printing tumour tissues and thereby replicating invivo tissue architectures now looming on the horizon. A comparison of experimental cell-based model systems and techniques highlights advantages and drawbacks of the various tools available, based on which cell-based models and experimental approaches best suited to address a diversity of research questions in the glioblastoma research field can be selected.
Collapse
Affiliation(s)
- Chiara Boccellato
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany.
| | - Markus Rehm
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany.
- Stuttgart Research Center Systems Biology, University of Stuttgart, 70569, Stuttgart, Germany.
| |
Collapse
|
37
|
Alajangi HK, Kaur M, Sharma A, Rana S, Thakur S, Chatterjee M, Singla N, Jaiswal PK, Singh G, Barnwal RP. Blood-brain barrier: emerging trends on transport models and new-age strategies for therapeutics intervention against neurological disorders. Mol Brain 2022; 15:49. [PMID: 35650613 PMCID: PMC9158215 DOI: 10.1186/s13041-022-00937-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/24/2022] [Indexed: 12/12/2022] Open
Abstract
The integrity of the blood–brain barrier (BBB) is essential for normal central nervous system (CNS) functioning. Considering the significance of BBB in maintaining homeostasis and the neural environment, we aim to provide an overview of significant aspects of BBB. Worldwide, the treatment of neurological diseases caused by BBB disruption has been a major challenge. BBB also restricts entry of neuro-therapeutic drugs and hinders treatment modalities. Hence, currently nanotechnology-based approaches are being explored on large scale as alternatives to conventional methodologies. It is necessary to investigate the in-depth characteristic features of BBB to facilitate the discovery of novel drugs that can successfully cross the barrier and target the disease effectively. It is imperative to discover novel strategies to treat life-threatening CNS diseases in humans. Therefore, insights regarding building blocks of BBB, activation of immune response on breach of this barrier, and various autoimmune neurological disorders caused due to BBB dysfunction are discussed. Further, special emphasis is given on delineating BBB disruption leading to CNS disorders. Moreover, various mechanisms of transport pathways across BBB, several novel strategies, and alternative routes by which drugs can be properly delivered into CNS are also discussed.
Collapse
Affiliation(s)
- Hema Kumari Alajangi
- Department of Biophysics, Panjab University, Chandigarh, 160014, India.,University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, 160014, India
| | - Mandeep Kaur
- Department of Biophysics, Panjab University, Chandigarh, 160014, India
| | - Akanksha Sharma
- Department of Biophysics, Panjab University, Chandigarh, 160014, India.,University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, 160014, India
| | - Sumedh Rana
- Department of Biophysics, Panjab University, Chandigarh, 160014, India
| | - Shipali Thakur
- Department of Biophysics, Panjab University, Chandigarh, 160014, India
| | - Mary Chatterjee
- Department of Biotechnology, UIET, Panjab University, Chandigarh, 160014, India
| | - Neha Singla
- Department of Biophysics, Panjab University, Chandigarh, 160014, India
| | - Pradeep Kumar Jaiswal
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, 77843, USA.
| | - Gurpal Singh
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, 160014, India.
| | | |
Collapse
|
38
|
Targeting the Axl and mTOR Pathway Synergizes Immunotherapy and Chemotherapy to Butylidenephthalide in a Recurrent GBM. JOURNAL OF ONCOLOGY 2022; 2022:3236058. [PMID: 35646111 PMCID: PMC9132698 DOI: 10.1155/2022/3236058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/26/2022] [Indexed: 12/13/2022]
Abstract
Background. The role of inherent tumor heterogeneity and an immunosuppressive microenvironment in therapeutic resistance has been determined to be of importance for the better management of glioblastoma multiforme (GBM). Some studies have suggested that combined drugs with divergent mechanisms may be promising in treating recurrent GBM. Methods. Intracranial sustained (Z)-n-butylidenephthalide [(Z)-BP] delivery through Cerebraca Wafers (CWs) to eliminate unresectable brain tumors was combined with the administration of temozolomide (TMZ), pembrolizumab, and cytokine-induced killer (CIK) cells for treating a patient with recurrent glioblastoma. Neurological adverse events and wound healing delay were monitored for estimating tolerance and efficacy. Response Assessment in Neuro-Oncology criteria were applied to evaluate progression-free survival (PFS); further, the molecular characteristics of GBM tissues were analyzed, and the underlying mechanism was investigated using primary culture. Results. Intracerebral (Z)-BP in residual tumors could not only inhibit cancer stem cells but also increase interferon gamma levels in serum, which then led to the regression of GBM and an immune-responsive microenvironment. Targeting receptor tyrosine kinases, including Axl and epidermal growth factor receptor (EGFR), and inhibiting the mechanistic target of rapamycin (mTOR) through (Z)-BP were determined to synergize CIK cells in the presence of pembrolizumab and TMZ in recurrent GBM. Therefore, this well-tolerated regimen could simultaneously block multiple cancer pathways, which allowed extended PFS and improved quality of life for 22 months. Conclusion. Given the several unique functions of (Z)-BP, greater sensitivity of chemotherapy and the synergism of pembrolizumab and CIK cells could have affected the excellent prognosis seen in this patient with recurrent GBM.
Collapse
|
39
|
Hauck M, Hellmold D, Kubelt C, Synowitz M, Adelung R, Schütt F, Held‐Feindt J. Localized Drug Delivery Systems in High‐Grade Glioma Therapy – From Construction to Application. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202200013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Margarethe Hauck
- Functional Nanomaterials, Institute for Materials Science Kiel University Kiel 24143 Germany
| | - Dana Hellmold
- Department of Neurosurgery University Medical Center Schleswig‐Holstein UKSH Campus Kiel Kiel 24105 Germany
| | - Carolin Kubelt
- Department of Neurosurgery University Medical Center Schleswig‐Holstein UKSH Campus Kiel Kiel 24105 Germany
| | - Michael Synowitz
- Department of Neurosurgery University Medical Center Schleswig‐Holstein UKSH Campus Kiel Kiel 24105 Germany
| | - Rainer Adelung
- Functional Nanomaterials, Institute for Materials Science Kiel University Kiel 24143 Germany
| | - Fabian Schütt
- Functional Nanomaterials, Institute for Materials Science Kiel University Kiel 24143 Germany
| | - Janka Held‐Feindt
- Department of Neurosurgery University Medical Center Schleswig‐Holstein UKSH Campus Kiel Kiel 24105 Germany
| |
Collapse
|
40
|
Lam FC, Salehi F, Kasper EM. Integrating Nanotechnology in Neurosurgery, Neuroradiology, and Neuro-Oncology Practice-The Clinicians' Perspective. Front Bioeng Biotechnol 2022; 10:801822. [PMID: 35223783 PMCID: PMC8864069 DOI: 10.3389/fbioe.2022.801822] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/11/2022] [Indexed: 12/11/2022] Open
Affiliation(s)
- Fred C Lam
- Division of Neurosurgery, Hamilton Health Sciences, McMaster University Faculty of Health Sciences, Hamilton, ON, Canada
| | - Fateme Salehi
- Department of Radiology, Hamilton Health Sciences, McMaster University Faculty of Health Sciences, Hamilton, ON, Canada
| | - Ekkehard M Kasper
- Division of Neurosurgery, Hamilton Health Sciences, McMaster University Faculty of Health Sciences, Hamilton, ON, Canada
| |
Collapse
|
41
|
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
|
42
|
Parakh S, Nicolazzo J, Scott AM, Gan HK. Antibody Drug Conjugates in Glioblastoma - Is There a Future for Them? Front Oncol 2021; 11:718590. [PMID: 34926242 PMCID: PMC8678283 DOI: 10.3389/fonc.2021.718590] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 11/15/2021] [Indexed: 12/25/2022] Open
Abstract
Glioblastoma (GBM) is an aggressive and fatal malignancy that despite decades of trials has limited therapeutic options. Antibody drug conjugates (ADCs) are composed of a monoclonal antibody which specifically recognizes a cellular surface antigen linked to a cytotoxic payload. ADCs have demonstrated superior efficacy and/or reduced toxicity in a range of haematological and solid tumors resulting in nine ADCs receiving regulatory approval. ADCs have also been explored in patients with brain tumours but with limited success to date. While earlier generations ADCs in glioma patients have had limited success and high toxicity, newer and improved ADCs characterised by low immunogenicity and more effective payloads have shown promise in a range of tumour types. These newer ADCs have also been tested in glioma patients, however, with mixed results. Factors affecting the effectiveness of ADCs to target the CNS include the blood brain barrier which acts as a physical and biochemical barrier, the pro-cancerogenic and immunosuppressive tumor microenvironment and tumour characteristics like tumour volume and antigen expression. In this paper we review the data regarding the ongoing the development of ADCs in glioma patients as well as potential strategies to overcome these barriers to maximise their therapeutic potential.
Collapse
Affiliation(s)
- Sagun Parakh
- Department of Medical Oncology, Austin Hospital, Heidelberg, VIC, Australia
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia
| | - Joseph Nicolazzo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Andrew M Scott
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia
- Department of Medicine, University of Melbourne, Heidelberg, VIC, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC, Australia
| | - Hui Kong Gan
- Department of Medical Oncology, Austin Hospital, Heidelberg, VIC, Australia
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia
- Department of Medicine, University of Melbourne, Heidelberg, VIC, Australia
| |
Collapse
|
43
|
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
|
44
|
Hazra RS, Dutta D, Mamnoon B, Nair G, Knight A, Mallik S, Ganai S, Reindl K, Jiang L, Quadir M. Polymeric Composite Matrix with High Biobased Content as Pharmaceutically Relevant Molecular Encapsulation and Release Platform. ACS APPLIED MATERIALS & INTERFACES 2021; 13:40229-40248. [PMID: 34423963 DOI: 10.1021/acsami.1c03805] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Drug delivery systems (DDS) that can temporally control the rate and extent of release of therapeutically active molecules find applications in many clinical settings, ranging from infection control to cancer therapy. With an aim to design a locally implantable, controlled-release DDS, we demonstrated the feasibility of using cellulose nanocrystal (CNC)-reinforced poly (l-lactic acid) (PLA) composite beads. The performance of the platform was evaluated using doxorubicin (DOX) as a model drug for applications in triple-negative breast cancer. A facile, nonsolvent-induced phase separation (NIPS) method was adopted to form composite beads. We observed that CNC loading within these beads played a critical role in the mechanical stability, porosity, water uptake, diffusion, release, and pharmacological activity of the drug from the delivery system. When loaded with DOX, composite beads significantly controlled the release of the drug in a pH-dependent pattern. For example, PLA/CNC beads containing 37.5 wt % of CNCs showed a biphasic release of DOX, where 41 and 82% of the loaded drug were released at pH 7.4 and pH 5.5, respectively, over 7 days. Drug release followed Korsmeyer's kinetics, indicating that the release mechanism was mostly diffusion and swelling-controlled. We showed that DOX released from drug-loaded PLA/CNC composite beads locally suppressed the growth and proliferation of triple-negative breast cancer cells, MBA-MB-231, via the apoptotic pathway. The efficacy of the DDS was evaluated in human tissue explants. We envision that such systems will find applications for designing biobased platforms with programmed stability and drug delivery functions.
Collapse
Affiliation(s)
- Raj Shankar Hazra
- Department of Mechanical Engineering, North Dakota State University, Fargo, North Dakota 58108, United States
- Materials and Nanotechnology Program, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Debasmita Dutta
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Babak Mamnoon
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Gauthami Nair
- Department of Biological Sciences, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Austin Knight
- Department of Mechanical Engineering, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Sanku Mallik
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Sabha Ganai
- Division of Surgical Oncology, Sanford Research, Fargo, North Dakota 58122, United States
| | - Katie Reindl
- Department of Biological Sciences, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Long Jiang
- Department of Mechanical Engineering, North Dakota State University, Fargo, North Dakota 58108, United States
- Materials and Nanotechnology Program, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Mohiuddin Quadir
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, North Dakota 58108, United States
- Materials and Nanotechnology Program, North Dakota State University, Fargo, North Dakota 58108, United States
| |
Collapse
|
45
|
Emerging Nano-Carrier Strategies for Brain Tumor Drug Delivery and Considerations for Clinical Translation. Pharmaceutics 2021; 13:pharmaceutics13081193. [PMID: 34452156 PMCID: PMC8399364 DOI: 10.3390/pharmaceutics13081193] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 12/13/2022] Open
Abstract
Treatment of brain tumors is challenging since the blood–brain tumor barrier prevents chemotherapy drugs from reaching the tumor site in sufficient concentrations. Nanomedicines have great potential for therapy of brain disorders but are still uncommon in clinical use despite decades of research and development. Here, we provide an update on nano-carrier strategies for improving brain drug delivery for treatment of brain tumors, focusing on liposomes, extracellular vesicles and biomimetic strategies as the most clinically feasible strategies. Finally, we describe the obstacles in translation of these technologies including pre-clinical models, analytical methods and regulatory issues.
Collapse
|
46
|
G-protein-coupled receptors as therapeutic targets for glioblastoma. Drug Discov Today 2021; 26:2858-2870. [PMID: 34271165 DOI: 10.1016/j.drudis.2021.07.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/03/2021] [Accepted: 07/05/2021] [Indexed: 12/29/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive primary brain tumour in adults. Treatments include surgical resection, radiotherapy, and chemotherapy. Despite this, the prognosis remains poor, with an impacted quality of life during treatment coupled with brain tumour recurrence; thus, new treatments are desperately needed. In this review, we focus on recent advances in G-protein-coupled receptor (GPCR) targets. To date, the most promising targets are the chemokine, cannabinoid, and dopamine receptors, but future work should further examine the melanocortin receptor-4 (MC4R), adhesion, lysophosphatidic acid (LPA) and smoothened (Smo) receptors to initiate new drug-screening strategies and targeted delivery of safe and effective GBM therapies.
Collapse
|
47
|
Johann P, Lenz D, Ries M. The drug development pipeline for glioblastoma-A cross sectional assessment of the FDA Orphan Drug Product designation database. PLoS One 2021; 16:e0252924. [PMID: 34234357 PMCID: PMC8263276 DOI: 10.1371/journal.pone.0252924] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 05/25/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most common malignant brain tumour among adult patients and represents an almost universally fatal disease. Novel therapies for GBM are being developed under the orphan drug legislation and the knowledge on the molecular makeup of this disease has been increasing rapidly. However, the clinical outcomes in GBM patients with currently available therapies are still dismal. An insight into the current drug development pipeline for GBM is therefore of particular interest. OBJECTIVES To provide a quantitative clinical-regulatory insight into the status of FDA orphan drug designations for compounds intended to treat GBM. METHODS Quantitative cross-sectional analysis of the U.S. Food and Drug Administration Orphan Drug Product database between 1983 and 2020. STROBE criteria were respected. RESULTS Four orphan drugs out of 161 (2,4%) orphan drug designations were approved for the treatment for GBM by the FDA between 1983 and 2020. Fourteen orphan drug designations were subsequently withdrawn for unknown reasons. The number of orphan drug designations per year shows a growing trend. In the last decade, the therapeutic mechanism of action of designated compounds intended to treat glioblastoma shifted from cytotoxic drugs (median year of designation 2008) to immunotherapeutic approaches and small molecules (median year of designation 2014 and 2015 respectively) suggesting an increased focus on precision in the therapeutic mechanism of action for compounds the development pipeline. CONCLUSION Despite the fact that current pharmacological treatment options in GBM are sparse, the drug development pipeline is steadily growing. In particular, the surge of designated immunotherapies detected in the last years raises the hope that elaborate combination possibilities between classical therapeutic backbones (radiotherapy and chemotherapy) and novel, currently experimental therapeutics may help to provide better therapies for this deadly disease in the future.
Collapse
Affiliation(s)
- Pascal Johann
- German Cancer Research Center (DKFZ), Division of Paediatric Neurooncology, Heidelberg, Germany
- Paediatric and Adolescent Medicine, Swabian Children’s Cancer Center Augsburg, Augsburg, Germany
| | - Dominic Lenz
- Paediatric Neurology and Metabolic Medicine, Center for Paediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Markus Ries
- Paediatric Neurology and Metabolic Medicine, Center for Paediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
- Center for Rare Disorders, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
48
|
Marizomib sensitizes primary glioma cells to apoptosis induced by a latest-generation TRAIL receptor agonist. Cell Death Dis 2021; 12:647. [PMID: 34168123 PMCID: PMC8225658 DOI: 10.1038/s41419-021-03927-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/28/2021] [Accepted: 06/07/2021] [Indexed: 12/26/2022]
Abstract
Due to the absence of curative treatments for glioblastoma (GBM), we assessed the efficacy of single and combination treatments with a translationally relevant 2nd generation TRAIL-receptor agonist (IZI1551) and the blood–brain barrier (BBB) permeant proteasome inhibitor marizomib in a panel of patient-derived glioblastoma cell lines. These cells were cultured using protocols that maintain the characteristics of primary tumor cells. IZI1551+marizomib combination treatments synergistically induced apoptotic cell death in the majority of cases, both in 2D, as well as in 3D spheroid cultures. In contrast, single-drug treatments largely failed to induce noticeable amounts of cell death. Kinetic analyses suggested that time-shifted drug exposure might further increase responsiveness, with marizomib pre-treatments indeed strongly enhancing cell death. Cell death responses upon the addition of IZI1551 could also be observed in GBM cells that were kept in a medium collected from the basolateral side of a human hCMEC/D3 BBB model that had been exposed to marizomib. Interestingly, the subset of GBM cell lines resistant to IZI1551+marizomib treatments expressed lower surface amounts of TRAIL death receptors, substantially lower amounts of procaspase-8, and increased amounts of cFLIP, suggesting that apoptosis initiation was likely too weak to initiate downstream apoptosis execution. Indeed, experiments in which the mitochondrial apoptosis threshold was lowered by antagonizing Mcl-1 re-established sensitivity to IZI1551+marizomib in otherwise resistant cells. Overall, our study demonstrates a high efficacy of combination treatments with a latest-generation TRAIL receptor agonist and the BBB permeant proteasome inhibitor marizomib in relevant GBM cell models, as well as strategies to further enhance responsiveness and to sensitize subgroups of otherwise resistant GBM cases.
Collapse
|
49
|
Pinheiro L, Perdomo-Pantoja A, Casaos J, Huq S, Paldor I, Vigilar V, Mangraviti A, Wang Y, Witham TF, Brem H, Tyler B. Captopril inhibits Matrix Metalloproteinase-2 and extends survival as a temozolomide adjuvant in an intracranial gliosarcoma model. Clin Neurol Neurosurg 2021; 207:106771. [PMID: 34198223 DOI: 10.1016/j.clineuro.2021.106771] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/22/2021] [Accepted: 06/17/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Captopril is a well-characterized, FDA-approved drug that has demonstrated promise as a repurposed oncology therapeutic. Captopril's known anti-cancer effects include inhibition of Matrix Metalloproteinase-2 (MMP-2), an endopeptidase which selectively breaks down the extracellular matrix to promote cell migration. MMP-2 is a known therapeutic target in gliomas, tumors with significant clinical need. Using an aggressive gliosarcoma model, we assessed captopril's effects on MMP-2 expression in vitro and in vivo as well as its efficacy as an adjuvant in combination therapy regimens in vivo. METHODS Following captopril treatment, MMP-2 protein expression and migratory capabilities of 9 L gliosarcoma cells were assessed in vitro via western blots and scratch wound assays, respectively. Rats were intracranially implanted with 9 L gliosarcoma tumors, and survival was assessed in the following groups: control; captopril (30 mg/kg/day); temozolomide (TMZ) (50 mg/kg/day), and captopril+TMZ. In vivo experiments were accompanied by immunohistochemistry for MMP-2 from brain tissue. RESULTS In vitro, captopril decreased MMP-2 protein expression and reduced migratory capacity in 9 L gliosarcoma cells. In a gliosarcoma animal model, captopril decreased MMP-2 protein expression and extended survival as a TMZ adjuvant relative to untreated controls, captopril monotherapy, and TMZ monotherapy groups (27.5 versus 14 (p < 0.001), 16 (p < 0.001), and 23 (p = 0.018) days, respectively). CONCLUSIONS Captopril decreases gliosarcoma cell migration, which may be mediated by reduction in MMP-2 protein expression. Captopril provided a survival advantage as a TMZ adjuvant in a rat intracranial gliosarcoma model. Captopril may represent a promising potential adjuvant to TMZ therapy in gliosarcoma as a modulator of the MMP-2 pathway.
Collapse
Affiliation(s)
- Leon Pinheiro
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Joshua Casaos
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sakibul Huq
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Iddo Paldor
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Veronica Vigilar
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Antonella Mangraviti
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yuan Wang
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Timothy F Witham
- 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
| | - Betty Tyler
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
50
|
Patel JP, Spiller SE, Barker ED. Drug penetration in pediatric brain tumors: Challenges and opportunities. Pediatr Blood Cancer 2021; 68:e28983. [PMID: 33719183 DOI: 10.1002/pbc.28983] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 12/23/2022]
Abstract
Larger clinical trial enrollments and a greater understanding of biological heterogeneity have led to improved survival rates for children diagnosed with brain tumors in the last 50 years. However, reducing long-term morbidities and improving survival rates of high-risk tumors remain major challenges. Chemotherapy can reduce tumor burden, but effective drug penetration at the tumor site is limited by barriers in the route of drug administration and within the tumor microenvironment. Bioavailability of drugs is impeded by the blood-brain barrier, plasma protein binding, and structural components by the tumor including the matrix and vasculature contributing to increased interstitial fluid pressure, hypoxia, and acidity. Designing drug delivery systems to circumvent these barriers could lead to improved drug penetration at the tumor site and reduce adverse systemic side effects. In this review, we expand on how systemic and local barriers limit drug penetration and present potential methods to enhance drug penetration in pediatric brain tumors.
Collapse
Affiliation(s)
- Jenny P Patel
- Department of Mechanical, Aerospace, and Biomedical Engineering, The University of Tennessee at Knoxville, Knoxville, Tennessee
| | - Susan E Spiller
- Pediatric Hematology/Oncology, East Tennessee Children's Hospital, Knoxville, Tennessee
| | - Elizabeth D Barker
- Department of Mechanical, Aerospace, and Biomedical Engineering, The University of Tennessee at Knoxville, Knoxville, Tennessee
| |
Collapse
|