51
|
Nair SK, Botros D, Chakravarti S, Mao Y, Wu E, Lu B, Liu S, Elshareif M, Jackson CM, Gallia GL, Bettegowda C, Weingart J, Brem H, Mukherjee D. Predictors of surgical site infection in glioblastoma patients undergoing craniotomy for tumor resection. J Neurosurg 2022; 138:1227-1234. [PMID: 36208433 DOI: 10.3171/2022.8.jns212799] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 08/03/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE
Surgical site infections (SSIs) burden patients and healthcare systems, often requiring additional intervention. The objective of this study was to identify the relationship between preoperative predictors inclusive of scalp incision type and postoperative SSI following glioblastoma resection.
METHODS
The authors retrospectively reviewed cases of glioblastoma resection performed at their institution from December 2006 to December 2019 and noted preoperative demographic and clinical presentations, excluding patients missing these data. Preoperative nutritional indices were available for a subset of cases. Scalp incisions were categorized as linear/curvilinear, reverse question mark, trapdoor, or frontotemporal. Patients were dichotomized by SSI incidence. Multivariable logistic regression was used to determine predictors of SSI.
RESULTS
A total of 911 cases of glioblastoma resection were identified, 30 (3.3%) of which demonstrated postoperative SSI. There were no significant differences in preoperative malnutrition or number of surgeries between SSI and non-SSI cases. The SSI cases had a significantly lower preoperative Karnofsky Performance Status (KPS) than the non-SSI cases (63.0 vs 75.1, p < 0.0001), were more likely to have prior radiation history (43.3% vs 26.4%, p = 0.042), and were more likely to have received steroids both preoperatively and postoperatively (83.3% vs 54.5%, p = 0.002). Linear/curvilinear incisions were more common in non-SSI than in SSI cases (56.9% vs 30.0%, p = 0.004). Trapdoor scalp incisions were more frequent in SSI than non-SSI cases (43.3% vs 24.2%, p = 0.012). On multivariable analysis, a lower preoperative KPS (OR 1.04, 95% CI 1.02–1.06), a trapdoor scalp incision (OR 3.34, 95% CI 1.37–8.49), and combined preoperative and postoperative steroid administration (OR 3.52, 95% CI 1.41–10.7) were independently associated with an elevated risk of postoperative SSI.
CONCLUSIONS
The study findings indicated that SSI risk following craniotomy for glioblastoma resection may be elevated in patients with a low preoperative KPS, a trapdoor scalp incision during surgery, and steroid treatment both preoperatively and postoperatively. These data may help guide future operative decision-making for these patients.
Collapse
Affiliation(s)
- Sumil K. Nair
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - David Botros
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sachiv Chakravarti
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yuncong Mao
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Esther Wu
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Brian Lu
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sophie Liu
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mazin Elshareif
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christopher M. Jackson
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Gary L. Gallia
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Chetan Bettegowda
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jon Weingart
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Henry Brem
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Debraj Mukherjee
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
52
|
Śledzińska P, Bebyn M, Furtak J, Koper A, Koper K. Current and promising treatment strategies in glioma. Rev Neurosci 2022:revneuro-2022-0060. [PMID: 36062548 DOI: 10.1515/revneuro-2022-0060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/30/2022] [Indexed: 12/14/2022]
Abstract
Gliomas are the most common primary central nervous system tumors; despite recent advances in diagnosis and treatment, glioma patients generally have a poor prognosis. Hence there is a clear need for improved therapeutic options. In recent years, significant effort has been made to investigate immunotherapy and precision oncology approaches. The review covers well-established strategies such as surgery, temozolomide, PCV, and mTOR inhibitors. Furthermore, it summarizes promising therapies: tumor treating fields, immune therapies, tyrosine kinases inhibitors, IDH(Isocitrate dehydrogenase)-targeted approaches, and others. While there are many promising treatment strategies, none fundamentally changed the management of glioma patients. However, we are still awaiting the outcome of ongoing trials, which have the potential to revolutionize the treatment of glioma.
Collapse
Affiliation(s)
- Paulina Śledzińska
- Molecular Oncology and Genetics Department, Innovative Medical Forum, The F. Lukaszczyk Oncology Center, 85-796 Bydgoszcz, Poland
| | - Marek Bebyn
- Molecular Oncology and Genetics Department, Innovative Medical Forum, The F. Lukaszczyk Oncology Center, 85-796 Bydgoszcz, Poland
| | - Jacek Furtak
- Department of Neurosurgery, 10th Military Research Hospital and Polyclinic, 85-681 Bydgoszcz, Poland.,Department of Neurooncology and Radiosurgery, The F. Lukaszczyk Oncology Center, 85-796 Bydgoszcz, Poland
| | - Agnieszka Koper
- Department of Oncology, Nicolaus Copernicus University in Torun, Ludwik Rydygier Collegium Medicum, 85-067 Bydgoszcz, Poland.,Department of Oncology, Franciszek Lukaszczyk Oncology Centre, 85-796 Bydgoszcz, Poland
| | - Krzysztof Koper
- Department of Oncology, Franciszek Lukaszczyk Oncology Centre, 85-796 Bydgoszcz, Poland.,Department of Clinical Oncology, and Nursing, Departament of Oncological Surgery, Nicolaus Copernicus University in Torun, Ludwik Rydygier Collegium Medicum, 85-067 Bydgoszcz, Poland
| |
Collapse
|
53
|
Shibahara I, Shibahara Y, Hagiwara H, Watanabe T, Orihashi Y, Handa H, Inukai M, Hide T, Yasui Y, Kumabe T. Ventricular opening and cerebrospinal fluid circulation accelerate the biodegradation process of carmustine wafers suggesting their immunomodulation potential in the human brain. J Neurooncol 2022; 159:425-435. [PMID: 35802230 DOI: 10.1007/s11060-022-04078-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/25/2022] [Indexed: 11/28/2022]
Abstract
PURPOSE Opening the ventricular system during glioblastoma surgery is often necessary, but the consequent effect on the tumor microenvironment of glioblastoma remains unknown. Implantation of carmustine wafer enables direct drug delivery to the tumor site; however, the exact mechanism of the wafer's biodegradation process is unclear, and the available data is limited to in vivo non-human mammalian studies. We hypothesized that the ventricular opening affects the degradation process of the wafer and the glioblastoma tumor microenvironment. METHODS This study included 30 glioblastoma patients. 21 patients underwent carmustine wafer implantation during initial surgery. All patients underwent repeated surgical resection upon recurrence, allowing for pathological comparison of changes associated with wafer implantation. Immunohistochemical analyses were performed using CD68, TMEM119, CD163, IBA1, BIN1, and CD31 antibodies to highlight microglia, macrophages, and tumor vascularity, and the quantitative scoring results were correlated with clinical, molecular, and surgical variables, including the effect of the ventricular opening. RESULTS The carmustine wafer implanted group presented significantly less TMEM119-positive microglia within the tumor (P = 0.0002). Simple and multiple regression analyses revealed that the decrease in TMEM119-positive microglia was correlated with longer intervals between surgeries and opened ventricular systems. No correlation was observed between age, methylated O6-methylguanine DNA methyltransferase promoter expression, and the extent of surgical resection. CONCLUSIONS Our study findings strongly suggest that biomaterials may possess immunomodulation capacity, which is significantly impacted by the ventricular opening procedure. Furthermore, our data highlights the pathophysiological effects of the ventricular opening within the surrounding human brain, especially after the wafer implantation.
Collapse
Affiliation(s)
- Ichiyo Shibahara
- Department of Neurosurgery, Kitasato University School of Medicine, 1-15-1 Kitasato Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan.
| | - Yukiko Shibahara
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Hiroyuki Hagiwara
- Department of Neurosurgery, Yamato Municipal Hospital, Yamato, Kanagawa, Japan
| | - Takashi Watanabe
- Department of General Internal Medicine, JCHO Sendai Hospital, Sendai, Miyagi, Japan
| | - Yasushi Orihashi
- Division of Clinical Research, Kitasato University Hospital, Sagamihara, Kanagawa, Japan
| | - Hajime Handa
- Department of Neurosurgery, Kitasato University School of Medicine, 1-15-1 Kitasato Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Madoka Inukai
- Department of Neurosurgery, Kitasato University School of Medicine, 1-15-1 Kitasato Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan.,Department of Pathology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Takuichiro Hide
- Department of Neurosurgery, Kitasato University School of Medicine, 1-15-1 Kitasato Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Yoshie Yasui
- Department of Neurosurgery, Kitasato University School of Medicine, 1-15-1 Kitasato Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Toshihiro Kumabe
- Department of Neurosurgery, Kitasato University School of Medicine, 1-15-1 Kitasato Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| |
Collapse
|
54
|
Poot E, Maguregui A, Brunton VG, Sieger D, Hulme AN. Targeting Glioblastoma through Nano- and Micro-particle-Mediated Immune Modulation. Bioorg Med Chem 2022; 72:116913. [DOI: 10.1016/j.bmc.2022.116913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 11/02/2022]
|
55
|
Lowe SR, Kunigelis K, Vogelbaum MA. Leveraging the neurosurgical operating room for therapeutic development in NeuroOncology. Adv Drug Deliv Rev 2022; 186:114337. [PMID: 35561836 DOI: 10.1016/j.addr.2022.114337] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 04/22/2022] [Accepted: 05/06/2022] [Indexed: 11/25/2022]
Abstract
Glioblastoma (GBM) remains a disease with a dismal prognosis. For all the hope and promise immunotherapies and molecular targeted therapies have shown for systemic malignancies, these treatments have failed to show any promise in GBM. In this context, the paradigm of investigation of therapeutics for this disease itself must be examined and modifications considered. The unique challenge of the presence of blood-brain and blood-tumor barriers (BBB/BTB) raises questions about both the true levels of systemic drug delivery to the affected tissues. Window-of-opportunity (WoO) trials in neuro-oncology allow for proof-of-concept at the start of a classic phase I-II-III clinical trial progression. For therapeutics that do not have the ability to cross the BBB/BTB, direct delivery into tumor and/or tumor-infiltrated brain in the setting of a surgical procedure can provide a novel route of therapeutic access. These approaches permit neurosurgeons to play a greater role in therapeutic development for brain tumors.
Collapse
|
56
|
Bota DA, Taylor TH, Lomeli N, Kong XT, Fu BD, Schönthal AH, Singer S, Blumenthal DT, Senecal FM, Linardou H, Rokas E, Antoniou DG, Schijns VEJC, Chen TC, Elliot J, Stathopoulos A. A Prospective, Cohort Study of SITOIGANAP to Treat Glioblastoma When Given in Combination With Granulocyte-Macrophage Colony-Stimulating Factor/Cyclophosphamide/Bevacizumab/Nivolumab or Granulocyte-Macrophage Colony-Stimulating Factor/Cyclophosphamide/Bevacizumab/Pembrolizumab in Patients Who Failed Prior Treatment With Surgical Resection, Radiation, and Temozolomide. Front Oncol 2022; 12:934638. [PMID: 35837107 PMCID: PMC9273968 DOI: 10.3389/fonc.2022.934638] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundGlioblastoma (GBM) is the most common primary, malignant brain tumor in adults and has a poor prognosis. The median progression-free survival (mPFS) of newly diagnosed GBM is approximately 6 months. The recurrence rate approaches 100%, and the case-fatality ratio approaches one. Half the patients die within 8 months of recurrence, and 5-year survival is less than 10%. Advances in treatment options are urgently needed. We report on the efficacy and safety of a therapeutic vaccine (SITOIGANAP: Epitopoietic Research Corporation) administered to 21 patients with recurrent GBM (rGBM) under a Right-to-Try/Expanded Access program. SITOIGANAP is composed of both autologous and allogeneic tumor cells and lysates.MethodsTwenty-one patients with rGBM received SITOIGANAP on 28-day cycles in combination with granulocyte-macrophage colony-stimulating factor (GM-CSF), cyclophosphamide, bevacizumab, and an anti-programmed cell death protein-1 (anti-PD-1) monoclonal antibody (either nivolumab or pembrolizumab).ResultsThe mPFS was 9.14 months, and the median overall survival (mOS) was 19.63 months from protocol entry. Currently, 14 patients (67%) are at least 6 months past their first SITOIGANAP cycle; 10 patients (48%) have received at least six cycles and have a mOS of 30.64 months and 1-year survival of 90%. The enrollment and end-of-study CD3+/CD4+ T-lymphocyte counts strongly correlate with OS.ConclusionsThe addition of SITOIGANAP/GM-CSF/cyclophosphamide to bevacizumab and an anti-PD-1 monoclonal antibody resulted in a significant survival benefit compared to historic control values in rGBM with minimal toxicity compared to current therapy.
Collapse
Affiliation(s)
- Daniela A. Bota
- Department of Neurology, University of California Irvine, Irvine, CA, United States
- Department of Neurological Surgery, University of California Irvine, Irvine, CA, United States
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, United States
- *Correspondence: Daniela A. Bota,
| | - Thomas H. Taylor
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, United States
- Department of Epidemiology and Biostatistics, University of California Irvine, Irvine, CA, United States
| | - Naomi Lomeli
- Department of Neurology, University of California Irvine, Irvine, CA, United States
| | - Xiao-Tang Kong
- Department of Neurology, University of California Irvine, Irvine, CA, United States
- Department of Neurological Surgery, University of California Irvine, Irvine, CA, United States
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, United States
| | - Beverly D. Fu
- Department of Neurology, University of California Irvine, Irvine, CA, United States
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, United States
| | - Axel H. Schönthal
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Samuel Singer
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, United States
| | - Deborah T. Blumenthal
- Neuro-oncology Division, Tel-Aviv Sourasky Medical Center, Tel-Aviv University, Tel-Aviv, Israel
| | - Frank M. Senecal
- Department of Hematology and Oncology, Northwest Medical Specialties, Tacoma, WA, United States
| | - Helena Linardou
- Fourth Oncology Department and Comprehensive Clinical Trials Center, Metropolitan Hospital, Athens, Greece
| | - Evangelos Rokas
- Department of Neurosurgery, Henry Dunant Hospital Center, Athens, Greece
| | | | | | - Thomas C. Chen
- Epitopoietic Research Corporation (ERC), Pasadena, CA, United States
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Joseph Elliot
- Epitopoietic Research Corporation (ERC), Pasadena, CA, United States
| | - Apostolos Stathopoulos
- Department of Neurosurgery, Henry Dunant Hospital Center, Athens, Greece
- Epitopoietic Research Corporation (ERC), Gembloux, Belgium
- Epitopoietic Research Corporation (ERC), Pasadena, CA, United States
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
57
|
McMahon DJ, Gleeson JP, O'Reilly S, Bambury RM. Management of newly diagnosed glioblastoma multiforme: current state of the art and emerging therapeutic approaches. Med Oncol 2022; 39:129. [PMID: 35716200 DOI: 10.1007/s12032-022-01708-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/14/2022] [Indexed: 12/21/2022]
Abstract
Glioblastoma multiforme represent > 50% of primary gliomas and have five year survival rates of ~ 5%. Maximal safe surgical resection followed by radiotherapy with concurrent and adjuvant temozolomide remains the standard treatment since published by Stupp et al. (in N Engl J Med 352:987-996, 2005), with additional benefit for patients with MGMT-methylated tumors. We review the current treatment landscape and ongoing efforts to improve these outcomes. An extensive literature search of Pubmed and Google Scholar involving the search terms "glioblastoma," "glioblastoma multiforme," or "GBM" for papers published to July 2021 was conducted and papers evaluated for relevance. As well as current data that informs clinical practice, we review ongoing clinical research in both newly diagnosed and recurrent settings that provides hope for a breakthrough. The Stupp protocol remains standard of care in 2021. Addition of tumor treating fields improved mOS modestly, with benefit seen in MGMT-methylated and unmethylated cohorts and also improved time to cognitive decline but has not been widely adopted. The addition of lomustine to temozolomide, in MGMT-methylated patients, also showed a mOS benefit but further investigation is required. Other promising therapeutic strategies including anti-angiogenic therapy, targeted therapy, and immunotherapy have yet to show a survival advantage. Improvements in the multidisciplinary management, surgical techniques and equipment, early palliative care, carrier support, and psychological support may be responsible for improving survival over time. Despite promising preclinical rationale, immunotherapy and targeted therapy are struggling to impact survival. A number of ongoing clinical trials provide hope for a breakthrough.
Collapse
Affiliation(s)
- D J McMahon
- Cork University Hospital, Cork, Ireland, UK.
| | | | - S O'Reilly
- Cork University Hospital, Cork, Ireland, UK
| | | |
Collapse
|
58
|
Pardridge WM. A Historical Review of Brain Drug Delivery. Pharmaceutics 2022; 14:1283. [PMID: 35745855 PMCID: PMC9229021 DOI: 10.3390/pharmaceutics14061283] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022] Open
Abstract
The history of brain drug delivery is reviewed beginning with the first demonstration, in 1914, that a drug for syphilis, salvarsan, did not enter the brain, due to the presence of a blood-brain barrier (BBB). Owing to restricted transport across the BBB, FDA-approved drugs for the CNS have been generally limited to lipid-soluble small molecules. Drugs that do not cross the BBB can be re-engineered for transport on endogenous BBB carrier-mediated transport and receptor-mediated transport systems, which were identified during the 1970s-1980s. By the 1990s, a multitude of brain drug delivery technologies emerged, including trans-cranial delivery, CSF delivery, BBB disruption, lipid carriers, prodrugs, stem cells, exosomes, nanoparticles, gene therapy, and biologics. The advantages and limitations of each of these brain drug delivery technologies are critically reviewed.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| |
Collapse
|
59
|
Germano IM, Ziu M, Wen P, Ormond DR, Olson JJ. Congress of Neurological Surgeons systematic review and evidence-based guidelines update on the role of cytotoxic chemotherapy and other cytotoxic therapies in the management of progressive glioblastoma in adults. J Neurooncol 2022; 158:225-253. [PMID: 35195819 DOI: 10.1007/s11060-021-03900-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/12/2021] [Indexed: 11/29/2022]
Abstract
TARGET POPULATION These recommendations apply to adult patients diagnosed with progressive glioblastoma (pGBM). QUESTION (Q1): In adult patients with pGBM does the use of temozolomide (TMZ) with alternative dosing or the use of TMZ in combination with other cytotoxic treatments result in increased overall survival compared to other chemotherapy? RECOMMENDATION Level III: Adult patients with pGBM might derive benefit in treatment with TMZ, especially those who progress after more than 5 months of TMZ-treatment free interval. LEVEL III Combination of TMZ with other cytotoxic agents such as nitrosourea, cisplatin, electrohyperthermia, or tamoxifen is not suggested in adult patients with pGBM as a stand-alone therapy. There is insufficient data to make a recommendation about which alternative TMZ dosing provides the best benefits. QUESTION (Q2): In adult patients with pGBM does the use of systemic or in situ nitrosourea result in increased overall survival compared to other chemotherapy? RECOMMENDATION Level III: In the setting of pGBM, fotemustine is suggested in elderly patients with methylated MGMT promoter status. There is insufficient evidence to compare fotemustine to other nitrosoureas. There is insufficient evidence to make a recommendation about the use of in situ nitrosourea in patients with pGBM who underwent the Stupp regimen. QUESTION (Q3): In adult patients with pGBM does the use of platinum compounds and topoisomerase result in increased survival compared to other chemotherapy? RECOMMENDATION Level III: Other chemotherapy including platinum compounds and topoisomerase inhibitors are not suggested to be used in adult patients with pGBM. LEVEL III Other cytotoxic therapies like perillyl acohol or ketogenic diet are not suggested for use in adult patients with pGBM as a stand-alone therapy. QUESTION (Q4): In adult patients with pGBM does the use of tumor treating field (TTF) result in increased overall survival compared to chemotherapy? RECOMMENDATION Level III: The use of TTF with other chemotherapy may be considered when treating adult patients with pGBM. There is insufficient evidence to recommend TTF to increase overall survival in adult patients with pGBM. QUESTION (Q5): In adult patients with pGBM does the use of oncolytic virotherapy result in increased survival compared to chemotherapy? RECOMMENDATION Level III: Oncolytic virotherapy is not suggested in patients with pGBM.
Collapse
Affiliation(s)
- Isabelle M Germano
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Mateo Ziu
- Department of Neurosurgery, Inova Neurosciences, Fairfax, VA, USA
| | - Patrick Wen
- Center For Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - D Ryan Ormond
- Department of Neurosurgery, University of Colorado School of Medicine, Aurora, CO, USA
| | - Jeffrey J Olson
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
60
|
Chromatin structure predicts survival in glioma patients. Sci Rep 2022; 12:8221. [PMID: 35581287 PMCID: PMC9114333 DOI: 10.1038/s41598-022-11019-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/15/2022] [Indexed: 11/08/2022] Open
Abstract
The pathological changes in epigenetics and gene regulation that accompany the progression of low-grade to high-grade gliomas are under-studied. The authors use a large set of paired atac-seq and RNA-seq data from surgically resected glioma specimens to infer gene regulatory relationships in glioma. Thirty-eight glioma patient samples underwent atac-seq sequencing and 16 samples underwent additional RNA-seq analysis. Using an atac-seq/RNA-seq correlation matrix, atac-seq peaks were paired with genes based on high correlation values (|r2| > 0.6). Samples clustered by IDH1 status but not by grade. Surprisingly there was a trend for IDH1 mutant samples to have more peaks. The majority of peaks are positively correlated with survival and positively correlated with gene expression. Constructing a model of the top six atac-seq peaks created a highly accurate survival prediction model (r2 = 0.68). Four of these peaks were still significant after controlling for age, grade, pathology, IDH1 status and gender. Grade II, III, and IV (primary) samples have similar transcription factors and gene modules. However, grade IV (recurrent) samples have strikingly few peaks. Patient-derived glioma cultures showed decreased peak counts following radiation indicating that this may be radiation-induced. This study supports the notion that IDH1 mutant and IDH1 wildtype gliomas have different epigenetic landscapes and that accessible chromatin sites mapped by atac-seq peaks tend to be positively correlated with expression. The data in this study leads to a new model of treatment response wherein glioma cells respond to radiation therapy by closing open regions of DNA.
Collapse
|
61
|
Brachytherapy for central nervous system tumors. J Neurooncol 2022; 158:393-403. [PMID: 35546384 DOI: 10.1007/s11060-022-04026-3] [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: 03/14/2022] [Accepted: 04/28/2022] [Indexed: 10/18/2022]
Abstract
Radiation is a mainstay of treatment for central nervous system (CNS) tumors. Brachytherapy involves the placement of a localized/interstitial radiation source into a tumor or resection bed and has distinct advantages that can make it an attractive form of radiation when used in the appropriate setting. However, the data supporting use of brachytherapy is clouded by variability in radiation sources, techniques, delivered doses, and trial designs. The goal of this manuscript is to identify consistent themes, review the highest-level evidence and potential indications for brachytherapy in CNS tumors, as well as highlight avenues for future work. Improved understanding of the underlying biology, indications, complications, and evolving industry-academic collaborations, place brachytherapy on the brink of a resurgence.
Collapse
|
62
|
Germano IM, Johnson DR, Patrick HH, Goodman AL, Ziu M, Ormond DR, Olson JJ. Congress of Neurological Surgeons Systematic Review and Evidence-Based Guidelines on the Management of Progressive Glioblastoma in Adults: Update of the 2014 Guidelines. Neurosurgery 2022; 90:e112-e115. [PMID: 35426875 DOI: 10.1227/neu.0000000000001903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 12/16/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The Institute of Medicine best practice recommendation to review guidelines every 5 years is followed by the Congress of Neurological Surgeons Guidelines Committee. The aim of this work was to provide an updated literature review and evidence-based recommendations on the topic of diagnosis and treatment of patients with progressive glioblastoma (pGBM). OBJECTIVE To review the literature published since the last guidelines on pGBM dated 2014, with literature search ending in June 2012. METHODS PubMed, Embase, and Cochrane were searched for the period July 1, 2012, to March 31, 2019, using search terms and search strategies to identify pertinent abstracts. These were then screened using published exclusion/inclusion criteria to identify full-text review articles. Evidence tables were constructed using data derived from full-text reviews and recommendations made from the evidence derived. RESULTS From the total 8786 abstracts identified by the search, 237 full-text articles met inclusion/exclusion criteria and were included in this update. Two new level II recommendations derived from this work. For the diagnosis of patients with GBM, the use of diffusion-weighted images is recommended to be included in the magnetic resonance images with and without contrast used for surveillance to detect pGBM. For the treatment of patients with pGBM, repeat cytoreductive surgery is recommended to improve overall survival. An additional 21 level III recommendations were provided. CONCLUSION Recent published literature provides new recommendations for the diagnosis and treatment of pGBM. The Central Nervous System Guidelines Committee will continue to pursue timely updates to further improve the care of patients with diagnosis.https://www.cns.org/guidelines/browse-guidelines-detail/guidelines-management-of-progressive-glioblastoma.
Collapse
Affiliation(s)
- Isabelle M Germano
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Derek R Johnson
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Hayes H Patrick
- Department of Neurological Surgery, George Washington University, Washington, District of Columbia, USA
| | - Abigail L Goodman
- Department of Pathology and Laboratory Medicine, Emory University Hospital, Atlanta, Georgia, USA
| | - Mateo Ziu
- Department of Neurosurgery, Inova Neuroscience and Spine Institute Fairfax, Virginia, USA
| | - D Ryan Ormond
- Department of Neurosurgery, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Jeffrey J Olson
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
63
|
Wanjale MV, Sunil Jaikumar V, Sivakumar KC, Ann Paul R, James J, Kumar GSV. Supramolecular Hydrogel Based Post-Surgical Implant System for Hydrophobic Drug Delivery Against Glioma Recurrence. Int J Nanomedicine 2022; 17:2203-2224. [PMID: 35599751 PMCID: PMC9122075 DOI: 10.2147/ijn.s348559] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/15/2022] [Indexed: 01/04/2023] Open
Abstract
Purpose The brain, protected by the cranium externally and the blood–brain barrier (BBB) internally, poses challenges in chemotherapy of aggressive brain tumors. Maximal tumor resection followed by radiation and chemotherapy is the standard treatment protocol; however, a substantial number of patients suffer from recurrence. Systemic circulation of drugs causes myelodysplasia and other side effects. To address these caveats, we report facile synthesis of a polyester-based supramolecular hydrogel as a brain biocompatible implant for in situ delivery of hydrophobic drugs. Methods Polycaprolactone-diol (PCL) was linked to polyethyleneglycol-diacid (PEG) via an ester bond. In silico modeling indicated micelle-based aggregation of PCL-PEG co-polymer to form a supramolecular hydrogel. Brain biocompatibility was checked in Sprague Dawley rat brain cortex with MRI, motor function test, and histology. Model hydrophobic drugs carmustine and curcumin entrapment propelled glioma cells into apoptosis-based death evaluated by in vitro cytotoxicity assays and Western blot. In vivo post-surgical xenograft glioma model was developed in NOD-SCID mice and evaluated for efficacy to restrict aggressive regrowth of tumors. Results 20% (w/v) PCL-PEG forms a soft hydrogel that can cover the uneven and large surface area of a tumor resection cavity and maintain brain density. The PCL-PEG hydrogel was biocompatible, and well-tolerated upon implantation in rat brain cortex, for a study period of 12 weeks. We report for the first time the combination of carmustine and curcumin entrapped as model hydrophobic drugs, increasing their bioavailability and yielding synergistic apoptotic effect on glioma cells. Further in vivo study indicated PCL-PEG hydrogel with a dual cargo of carmustine and curcumin restricted aggressive regrowth post-resection significantly compared with control and animals with intravenous drug treatment. Conclusion PCL-PEG soft gel-based implant is malleable compared with rigid wafers used as implants, thus providing larger surface area contact. This stable, biocompatible, supramolecular gel without external crosslinking can find wide applications by interchanging formulation of various hydrophobic drugs to ensure and increase site-specific delivery, avoiding systemic circulation.
Collapse
Affiliation(s)
- Mrunal Vitthal Wanjale
- Nano Drug Delivery Systems (NDDS), Cancer Biology Division, Rajiv Gandhi Centre for Biotechnology, Thycaud P.O, Thiruvananthapuram, Kerala, 695014, India
- Research Scholar, Department of Biotechnology, Faculty of Applied Sciences & Technology, University of Kerala, Thiruvananthapuram, Kerala, 695581, India
| | - Vishnu Sunil Jaikumar
- Animal Research Facility, Rajiv Gandhi Centre for Biotechnology, Thycaud P.O, Thiruvananthapuram, Kerala, 695014, India
| | - K C Sivakumar
- Distributed Information Sub-Centre (Bioinformatics Centre), Bio-Innovation Center (BIC), Rajiv Gandhi Centre for Biotechnology, Poojappura, Thiruvananthapuram, Kerala, 695014, India
| | - Riya Ann Paul
- Research Scholar, Department of Biotechnology, Faculty of Applied Sciences & Technology, University of Kerala, Thiruvananthapuram, Kerala, 695581, India
- Neuro-Stem Cell Biology Lab, Rajiv Gandhi Centre for Biotechnology, Thycaud P.O, Thiruvananthapuram, Kerala, 695014, India
| | - Jackson James
- Neuro-Stem Cell Biology Lab, Rajiv Gandhi Centre for Biotechnology, Thycaud P.O, Thiruvananthapuram, Kerala, 695014, India
| | - G S Vinod Kumar
- Nano Drug Delivery Systems (NDDS), Cancer Biology Division, Rajiv Gandhi Centre for Biotechnology, Thycaud P.O, Thiruvananthapuram, Kerala, 695014, India
- Correspondence: GS Vinod Kumar, Tel +91 471 2781217, Fax +91 471 2348096, Email
| |
Collapse
|
64
|
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
|
65
|
3D printed drug-loaded implantable devices for intraoperative treatment of cancer. J Control Release 2022; 344:147-156. [PMID: 35217100 PMCID: PMC9373975 DOI: 10.1016/j.jconrel.2022.02.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 02/14/2022] [Accepted: 02/17/2022] [Indexed: 12/23/2022]
Abstract
Surgery is an important treatment for cancer; however, local recurrence following macroscopically-complete resection is common and a significant cause of morbidity and mortality. Systemic chemotherapy is often employed as an adjuvant therapy to prevent recurrence of residual disease, but has limited efficacy due to poor penetration and dose-limiting off-target toxicities. Selective delivery of chemotherapeutics to the surgical bed may eliminate residual tumor cells while avoiding systemic toxicity. While this is challenging for traditional drug delivery technologies, we utilized advances in 3D printing and drug delivery science to engineer a drug-loaded arrowhead array device (AAD) to overcome these challenges. We demonstrated that such a device can be designed, fabricated, and implanted intraoperatively and provide extended release of chemotherapeutics directly to the resection area. Using paclitaxel and cisplatin as model drugs and murine models of cancer, we showed AADs significantly decreased local recurrence post-surgery and improved survival. We further demonstrated the potential for fabricating personalized AADs for intraoperative application in the clinical setting.
Collapse
|
66
|
Immunotherapeutic Approaches for Glioblastoma Treatment. Biomedicines 2022; 10:biomedicines10020427. [PMID: 35203636 PMCID: PMC8962267 DOI: 10.3390/biomedicines10020427] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 02/05/2022] [Accepted: 02/08/2022] [Indexed: 11/17/2022] Open
Abstract
Glioblastoma remains a challenging disease to treat, despite well-established standard-of-care treatments, with a median survival consistently of less than 2 years. In this review, we delineate the unique disease-specific challenges for immunotherapies, both brain-related and non-brain-related, which will need to be adequately overcome for the development of effective treatments. We also review current immunotherapy treatments, with a focus on clinical applications, and propose future directions for the field of GBM immunotherapy.
Collapse
|
67
|
Gazaille C, Sicot M, Saulnier P, Eyer J, Bastiat G. Local Delivery and Glioblastoma: Why Not Combining Sustained Release and Targeting? FRONTIERS IN MEDICAL TECHNOLOGY 2022; 3:791596. [PMID: 35047971 PMCID: PMC8757870 DOI: 10.3389/fmedt.2021.791596] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 10/28/2021] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma is one of the most aggressive brain tumors and is associated with a very low overall median survival despite the current treatment. The standard of care used in clinic is the Stupp's protocol which consists of a maximal resection of the tumor when possible, followed by radio and chemotherapy using temozolomide. However, in most cases, glioblastoma cells infiltrate healthy tissues and lead to fatal recurrences. There are a lot of hurdles to overcome in the development of new therapeutic strategies such as tumor heterogeneity, cell infiltration, alkylating agent resistance, physiological barriers, etc., and few treatments are on the market today. One of them is particularly appealing because it is a local therapy, which does not bring additional invasiveness since tumor resection is included in the gold standard treatment. They are implants: the Gliadel® wafers, which are deposited post-surgery. Nevertheless, in addition to presenting important undesirable effects, it does not bring any major benefit in the therapy despite the strategy being particularly attractive. The purpose of this review is to provide an overview of recent advances in the development of innovative therapeutic strategies for glioblastoma using an implant-type approach. The combination of this local strategy with effective targeting of the tumor microenvironment as a whole, also developed in this review, may be of interest to alleviate some of the obstacles encountered in the treatment of glioblastoma.
Collapse
Affiliation(s)
| | - Marion Sicot
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, Angers, France
| | | | - Joël Eyer
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, Angers, France
| | | |
Collapse
|
68
|
Xu Y, Scharfstein D, Müller P, Daniels M. A Bayesian nonparametric approach for evaluating the causal effect of treatment in randomized trials with semi-competing risks. Biostatistics 2022; 23:34-49. [PMID: 32247284 PMCID: PMC10448950 DOI: 10.1093/biostatistics/kxaa008] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 01/30/2020] [Accepted: 02/03/2020] [Indexed: 11/12/2022] Open
Abstract
We develop a Bayesian nonparametric (BNP) approach to evaluate the causal effect of treatment in a randomized trial where a nonterminal event may be censored by a terminal event, but not vice versa (i.e., semi-competing risks). Based on the idea of principal stratification, we define a novel estimand for the causal effect of treatment on the nonterminal event. We introduce identification assumptions, indexed by a sensitivity parameter, and show how to draw inference using our BNP approach. We conduct simulation studies and illustrate our methodology using data from a brain cancer trial. The R code implementing our model and algorithm is available for download at https://github.com/YanxunXu/BaySemiCompeting.
Collapse
Affiliation(s)
- Yanxun Xu
- Department of Applied Mathematics and Statistics, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
| | - Daniel Scharfstein
- Department of Biostatistics, Johns Hopkins University, 615 N Wolfe St, Baltimore, MD 21205, USA
| | - Peter Müller
- Department of Mathematics, The University of Texas at Austin, 2515 Speedway, RLM 8.100, Austin, TX 78712, USA
| | - Michael Daniels
- Department of Statistics, University of Florida, Union Rd, Gainesville, FL 32603, USA
| |
Collapse
|
69
|
Iuchi T, Inoue A, Hirose Y, Morioka M, Horiguchi K, Natsume A, Arakawa Y, Iwasaki K, Fujiki M, Kumabe T, Sakata Y. Long-term effectiveness of Gliadel implant for malignant glioma and prognostic factors for survival: 3-year results of a postmarketing surveillance in Japan. Neurooncol Adv 2022; 4:vdab189. [PMID: 35118382 PMCID: PMC8807118 DOI: 10.1093/noajnl/vdab189] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Adjuvant treatment with Gliadel wafers may prolong overall survival (OS) for malignant glioma patients without increasing toxicity. In Japan, the long-term OS of these patients treated with Gliadel 7.7 mg implants has not been studied. We evaluated OS and prognostic factors that might affect OS in Japanese patients with malignant glioma who received the Gliadel 7.7 mg implant. METHODS This observational, long-term, postmarketing surveillance was an extension of a previous surveillance. Data were collected through case report forms at 2 and 3 years after Gliadel implant. Up to 8 Gliadel wafers (61.6 mg of carmustine) were placed over the tumor resection site. Primary endpoints were OS and prognostic factors that may influence OS. RESULTS Among the 506 patients analyzed, 62.6% had newly diagnosed disease, and 37.4% had recurrent disease; 79.1% had glioblastoma histological type and 79.6% had World Health Organization Grade IV disease. Patients received a median of 8 wafers. The median OS was 18.0 months; OS rates were 39.8% and 31.5% at 2 and 3 years, respectively. Age ≥65 years (hazard ratio [HR]: 1.456; P = .002), lower resection rate (HR: 1.206; P < .001), recurrence (HR: 2.418; P < .001), and concomitant radiotherapy (HR: 0.588; P < .001) were identified as significant prognostic factors. CONCLUSIONS This study confirmed the 2- and 3-year OS of Japanese malignant glioma patients with varied backgrounds after Gliadel implant. With a careful interpretation of indirect comparisons with previously reported data, the results suggest that prognosis could be improved with Gliadel implants. CLINICAL TRIAL REGISTRATION NCT02300506.
Collapse
Affiliation(s)
- Toshihiko Iuchi
- Division of Neurological Surgery, Chiba Cancer Center, Chiba, Japan
| | - Akihiro Inoue
- Department of Neurosurgery, Ehime University School of Medicine, Ehime, Japan
| | - Yuichi Hirose
- Department of Neurosurgery, Fujita Health University, Aichi, Japan
| | - Motohiro Morioka
- Department of Neurosurgery, Kurume University School of Medicine, Fukuoka, Japan
| | - Keishi Horiguchi
- Department of Neurosurgery, Gunma University Hospital, Gunma, Japan
| | - Atsushi Natsume
- Department of Neurosurgery, Nagoya University Hospital, Aichi, Japan
| | - Yoshiki Arakawa
- Department of Neurosurgery, Kyoto University Hospital, Kyoto, Japan
| | - Koichi Iwasaki
- Department of Neurosurgery, Tazuke Kofukai Medical Research Institute, Kitano Hospital, Osaka, Japan
| | - Minoru Fujiki
- Department of Neurosurgery, Oita University Hospital, Oita, Japan
| | - Toshihiro Kumabe
- Department of Neurosurgery, Kitasato University Hospital, Kanagawa, Japan
| | - Yukinori Sakata
- Clinical Planning Department, Medical Headquarters, Eisai Co., Ltd, Tokyo, Japan
| |
Collapse
|
70
|
WAN J, HUANG M. Apigenin inhibits proliferation, migration, invasion and epithelial mesenchymal transition of glioma cells by regulating miR-103a-3p/NEED9/AKT axis. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.23022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Jing WAN
- Renmin Hospital of Wuhan University, China
| | - Min HUANG
- Renmin Hospital of Wuhan University, China
| |
Collapse
|
71
|
Serra R, Mangraviti A, Gorelick NL, Shapira-Furman T, Alomari S, Cecia A, Darjee N, Brem H, Rottenberg Y, Domb AJ, Tyler B. Combined Intracranial Acriflavine, Temozolomide and Radiation Extends Survival in a Rat Glioma Model. Eur J Pharm Biopharm 2021; 170:179-186. [PMID: 34968646 DOI: 10.1016/j.ejpb.2021.12.011] [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: 11/04/2021] [Accepted: 12/23/2021] [Indexed: 11/04/2022]
Abstract
Glioblastomas have been historically difficult to treat with poor long-term survival. With novel strategies focused on targeting hypoxia-inducible factor (HIF) regulatory pathways, recent evidence has shown that Acriflavine (ACF) can effectively target glioma invasiveness and recurrence. However, local delivery of ACF and its combinatory effects with Temozolomide (TMZ) and radiation therapy (XRT) have not yet been optimized. In this study we test a novel polymeric matrix that can gradually release ACF at the tumor bed site in combination with systemic TMZ and XRT. In vitro cytotoxicity assays of ACF in combination with TMZ and XRT were performed on rodent and human cell lines with CCK-8 and flow cytometry. In vitro drug release was measured and intracranial safety was assessed in tumor-free animals. Finally, efficacy was assessed in an intracranial gliosarcoma model and combination therapy with TMZ and XRT evaluated. Combination therapy of ACF, TMZ, and XRT was able to reduce cell viability and induce apoptosis in glioma cells. In vitro and in vivo release of ACF was measured in benchtop and animal models. Efficacy was established in an in vivo gliosarcoma model in which intracranial ACF (p<0.01) significantly improved median survival and the combination therapy of ACF, TMZ and XRT (p<0.01) significantly improved median survival and led to long-term survival (LTS). We provide evidence that ACF, combined with TMZ and XRT, led to LTS in an intracranial model of rat gliosarcoma. These findings, in combination with the use of a novel polymeric matrix that allows more gradual drug delivery, constitute a first step in the translation of this novel strategy to human use.
Collapse
Affiliation(s)
- Riccardo Serra
- Department of Neurosurgery, Johns Hopkins University, Baltimore, MD, United States; Department of Neurosurgery, University of Maryland, Baltimore, MD, United States
| | - Antonella Mangraviti
- Department of Neurosurgery, Johns Hopkins University, Baltimore, MD, United States; Department of Neurosurgery, School of Medicine - Catholic University of the Sacred Heart, Rome, Italy
| | - Noah L Gorelick
- Department of Neurosurgery, Johns Hopkins University, Baltimore, MD, United States
| | - Tovi Shapira-Furman
- Institute of Drug Research, School of Pharmacy-Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Safwan Alomari
- Department of Neurosurgery, Johns Hopkins University, Baltimore, MD, United States
| | - Arba Cecia
- Department of Neurosurgery, Johns Hopkins University, Baltimore, MD, United States
| | - Namrata Darjee
- Department of Neurosurgery, Johns Hopkins University, Baltimore, MD, United States
| | - Henry Brem
- Department of Neurosurgery, Johns Hopkins University, Baltimore, MD, United States; Department of Oncology, Johns Hopkins University, Baltimore, MD, United States; Department of Ophthalmology, Johns Hopkins University, Baltimore, MD, United States; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Yakir Rottenberg
- Department of Oncology, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Abraham J Domb
- Institute of Drug Research, School of Pharmacy-Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Betty Tyler
- Department of Neurosurgery, Johns Hopkins University, Baltimore, MD, United States.
| |
Collapse
|
72
|
Mohile NA, Messersmith H, Gatson NTN, Hottinger AF, Lassman AB, Morton J, Ney D, Nghiemphu PL, Olar A, Olson J, Perry J, Portnow J, Schiff D, Shannon A, Shih HA, Strowd R, van den Bent M, Ziu M, Blakeley J. Therapy for Diffuse Astrocytic and Oligodendroglial Tumors in Adults: ASCO-SNO Guideline. Neuro Oncol 2021. [DOI: 10.1093/neuonc/noab279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Abstract
Purpose
To provide guidance to clinicians regarding therapy for diffuse astrocytic and oligodendroglial tumors in adults.
Methods
ASCO and the Society for Neuro-Oncology convened an Expert Panel and conducted a systematic review of the literature.
Results
Fifty-nine randomized trials focusing on therapeutic management were identified.
Recommendations
Adults with newly diagnosed oligodendroglioma, isocitrate dehydrogenase (IDH)–mutant, 1p19q codeleted CNS WHO grade 2 and 3 should be offered radiation therapy (RT) and procarbazine, lomustine, and vincristine (PCV). Temozolomide (TMZ) is a reasonable alternative for patients who may not tolerate PCV, but no high-level evidence supports upfront TMZ in this setting. People with newly diagnosed astrocytoma, IDH-mutant, 1p19q non-codeleted CNS WHO grade 2 should be offered RT with adjuvant chemotherapy (TMZ or PCV). People with astrocytoma, IDH-mutant, 1p19q non-codeleted CNS WHO grade 3 should be offered RT and adjuvant TMZ. People with astrocytoma, IDH-mutant, CNS WHO grade 4 may follow recommendations for either astrocytoma, IDH-mutant, 1p19q non-codeleted CNS WHO grade 3 or glioblastoma, IDH-wildtype, CNS WHO grade 4. Concurrent TMZ and RT should be offered to patients with newly diagnosed glioblastoma, IDH-wildtype, CNS WHO grade 4 followed by 6 months of adjuvant TMZ. Alternating electric field therapy, approved by the US Food and Drug Administration, should be considered for these patients. Bevacizumab is not recommended. In situations in which the benefits of 6-week RT plus TMZ may not outweigh the harms, hypofractionated RT plus TMZ is reasonable. In patients age ≥ 60 to ≥ 70 years, with poor performance status or for whom toxicity or prognosis are concerns, best supportive care alone, RT alone (for MGMTpromoter unmethylated tumors), or TMZ alone (for MGMT promoter methylated tumors) are reasonable treatment options. Additional information is available at www.asco.org/neurooncology-guidelines.
Collapse
Affiliation(s)
- Nimish A Mohile
- Department of Neurology and Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | | | - Na Tosha N Gatson
- Banner MD Anderson Cancer Center, Phoenix, AZ, USA
- Geisinger Neuroscience Institute, Danville, PA, USA
| | - Andreas F Hottinger
- Department of Clinical Neurosciences and Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | | | - Jordan Morton
- University of Oklahoma Health Sciences, Oklahoma City, OK, USA
| | - Douglas Ney
- University of Colorado School of Medicine, Aurora, CO, USA
| | | | | | - Jeffery Olson
- Emory University, Atlanta, GA, USA
- Sunnybrook Health Sciences Center, Toronto, Ontario, Canada
| | - James Perry
- City of Hope National Medical Center, Duarte, CA, USA
| | - Jana Portnow
- City of Hope National Medical Center, Duarte, CA, USA
| | - David Schiff
- University of Virginia Medical Center, Charlottesville, VA, USA
| | | | | | - Roy Strowd
- Wake Forest Baptist Health Medical Center, Winston-Salem, NC, USA
| | - Martin van den Bent
- The Brain Tumor Center at Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Mateo Ziu
- INOVA Neurosciences and Inova Schar Cancer Institute, Falls Church, VA, USA
| | | |
Collapse
|
73
|
Nie W, Wang B, Mi X, Chen J, Yu T, Miao J, Lin Y, Yang T, Ran M, Hong Z, Liu X, Liang X, Qian Z, Gao X. Co-Delivery of Paclitaxel and shMCL-1 by Folic Acid-Modified Nonviral Vector to Overcome Cancer Chemotherapy Resistance. SMALL METHODS 2021; 5:e2001132. [PMID: 34928100 DOI: 10.1002/smtd.202001132] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/05/2021] [Indexed: 02/05/2023]
Abstract
Acquired chemoresistance presents a major clinical impediment, which is an urgent problem to be solved. Interestingly, myeloma cell leukemia-1 (MCL-1) and folate receptor expression levels are higher in chemotherapy-resistant patients than in pretreatment patients. In this study, a multifunctional folic acid (FA)-targeting core-shell structure is presented for simultaneous delivery of shMCL-1 and paclitaxel (PTX). The transfection efficiency of shMCL-1 with the FA-targeting delivery system is higher than with a nontargeting delivery system in Skov3 and A2780T cells. The FA-targeting system significantly inhibits cell growth, blocks cell cycles, and promotes apoptosis of cancer cells in vitro. The mechanisms involved in inhibiting growth are related to Bcl-2/Bax and cdc2/Cyclin B1 pathways. An analysis of RNA sequencing suggests that shMCL-1 reverses chemoresistance through regulating genes such as regulator of chromosome condensation 2 (RCC2). The synergetic effect of shMCL-1 and PTX effectively inhibits tumor growth in both PTX-resistant and normal cancer models by inducing tumor apoptosis, inhibiting proliferation, and limiting tumor angiogenesis. The study results indicate that a FA-targeting delivery system combining shMCL-1 with PTX can simultaneously target tumor sites and restore the sensitivity of chemotherapy-resistant cancer to PTX. These findings have important implications for patients with normal or PTX-resistant cancer.
Collapse
Affiliation(s)
- Wen Nie
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Bilan Wang
- Department of Pharmacy, West China Second University Hospital of Sichuan University, Chengdu, 610041, P. R. China
| | - Xue Mi
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Jing Chen
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Ting Yu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Junming Miao
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, P. R. China
| | - Yunzhu Lin
- Department of Pharmacy, West China Second University Hospital of Sichuan University, Chengdu, 610041, P. R. China
| | - Tingting Yang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Mengni Ran
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Zehuo Hong
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Xiaoxiao Liu
- Department of Radiation Oncology, Cancer Center, Affiliated Hospital of Xuzhou Medical University, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, 221000, P. R. China
| | - Xiao Liang
- Department of Pharmacy, West China Second University Hospital of Sichuan University, Chengdu, 610041, P. R. China
| | - Zhiyong Qian
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| |
Collapse
|
74
|
Drug Repurposing for Glioblastoma and Current Advances in Drug Delivery-A Comprehensive Review of the Literature. Biomolecules 2021; 11:biom11121870. [PMID: 34944514 PMCID: PMC8699739 DOI: 10.3390/biom11121870] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/19/2021] [Accepted: 12/03/2021] [Indexed: 12/22/2022] Open
Abstract
Glioblastoma (GBM) is the most common primary malignant brain tumor in adults with an extremely poor prognosis. There is a dire need to develop effective therapeutics to overcome the intrinsic and acquired resistance of GBM to current therapies. The process of developing novel anti-neoplastic drugs from bench to bedside can incur significant time and cost implications. Drug repurposing may help overcome that obstacle. A wide range of drugs that are already approved for clinical use for the treatment of other diseases have been found to target GBM-associated signaling pathways and are being repurposed for the treatment of GBM. While many of these drugs are undergoing pre-clinical testing, others are in the clinical trial phase. Since GBM stem cells (GSCs) have been found to be a main source of tumor recurrence after surgery, recent studies have also investigated whether repurposed drugs that target these pathways can be used to counteract tumor recurrence. While several repurposed drugs have shown significant efficacy against GBM cell lines, the blood–brain barrier (BBB) can limit the ability of many of these drugs to reach intratumoral therapeutic concentrations. Localized intracranial delivery may help to achieve therapeutic drug concentration at the site of tumor resection while simultaneously minimizing toxicity and side effects. These strategies can be considered while repurposing drugs for GBM.
Collapse
|
75
|
Mohile NA, Messersmith H, Gatson NT, Hottinger AF, Lassman A, Morton J, Ney D, Nghiemphu PL, Olar A, Olson J, Perry J, Portnow J, Schiff D, Shannon A, Shih HA, Strowd R, van den Bent M, Ziu M, Blakeley J. Therapy for Diffuse Astrocytic and Oligodendroglial Tumors in Adults: ASCO-SNO Guideline. J Clin Oncol 2021; 40:403-426. [PMID: 34898238 DOI: 10.1200/jco.21.02036] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
PURPOSE To provide guidance to clinicians regarding therapy for diffuse astrocytic and oligodendroglial tumors in adults. METHODS ASCO and the Society for Neuro-Oncology convened an Expert Panel and conducted a systematic review of the literature. RESULTS Fifty-nine randomized trials focusing on therapeutic management were identified. RECOMMENDATIONS Adults with newly diagnosed oligodendroglioma, isocitrate dehydrogenase (IDH)-mutant, 1p19q codeleted CNS WHO grade 2 and 3 should be offered radiation therapy (RT) and procarbazine, lomustine, and vincristine (PCV). Temozolomide (TMZ) is a reasonable alternative for patients who may not tolerate PCV, but no high-level evidence supports upfront TMZ in this setting. People with newly diagnosed astrocytoma, IDH-mutant, 1p19q non-codeleted CNS WHO grade 2 should be offered RT with adjuvant chemotherapy (TMZ or PCV). People with astrocytoma, IDH-mutant, 1p19q non-codeleted CNS WHO grade 3 should be offered RT and adjuvant TMZ. People with astrocytoma, IDH-mutant, CNS WHO grade 4 may follow recommendations for either astrocytoma, IDH-mutant, 1p19q non-codeleted CNS WHO grade 3 or glioblastoma, IDH-wildtype, CNS WHO grade 4. Concurrent TMZ and RT should be offered to patients with newly diagnosed glioblastoma, IDH-wildtype, CNS WHO grade 4 followed by 6 months of adjuvant TMZ. Alternating electric field therapy, approved by the US Food and Drug Administration, should be considered for these patients. Bevacizumab is not recommended. In situations in which the benefits of 6-week RT plus TMZ may not outweigh the harms, hypofractionated RT plus TMZ is reasonable. In patients age ≥ 60 to ≥ 70 years, with poor performance status or for whom toxicity or prognosis are concerns, best supportive care alone, RT alone (for MGMT promoter unmethylated tumors), or TMZ alone (for MGMT promoter methylated tumors) are reasonable treatment options. Additional information is available at www.asco.org/neurooncology-guidelines.
Collapse
Affiliation(s)
- Nimish A Mohile
- Department of Neurology and Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY
| | | | - Na Tosha Gatson
- Banner MD Anderson Cancer Center, Phoenix, AZ.,Geisinger Neuroscience Institute. Danville, PA
| | - Andreas F Hottinger
- Departments of Clinical Neurosciences and Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | | | - Jordan Morton
- University of Oklahoma Health Sciences, Oklahoma City, OK
| | - Douglas Ney
- University of Colorado School of Medicine, Aurora, CO
| | | | | | | | - James Perry
- Sunnybrook Health Sciences Center, Toronto, Ontario, Canada
| | - Jana Portnow
- City of Hope National Medical Center, Duarte, CA
| | - David Schiff
- University of Virginia Medical Center, Charlottesville, VA
| | | | | | - Roy Strowd
- Wake Forest Baptist Health Medical Center, Winston-Salem, NC
| | - Martin van den Bent
- The Brain Tumor Center at Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Mateo Ziu
- INOVA Neurosciences and Inova Schar Cancer Institute, Falls Church, VA
| | | |
Collapse
|
76
|
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
|
77
|
Lehnert S. Targeting of radio-enhancing drugs. Int J Radiat Biol 2021; 98:461-465. [PMID: 34747680 DOI: 10.1080/09553002.2021.2003465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
OBJECTIVE Toxicity to normal tissue is frequently the dose-limiting factor in the chemotherapy and mixed modality treatments of cancer. If the radio-enhancing drug could be localized at the disease site and released slowly over time, then systemic drug toxicities could be decreased while simultaneously maintaining high drug concentrations in the tumor. These considerations support a role for a sustained release intra-tumoral delivery systems for the delivery of radio-enhancing drugs. METHODS Two approaches aimed at achieving the end of localizing the radio-enhancing drug to the tumor are described. First, nanoparticles, which have a prolonged circulation time and facility for enhanced tumor targeting. Structural defects in the walls of the tumor vasculature allow the passage of particles too large to pass through the walls of normal blood vessels. This characteristic of tumor blood vessels, referred to as the enhanced permeability and retention (EPR) effect, allows relatively large entities (typically liposomes, nanoparticles, and macromolecular drugs) to pass from the blood vessels to tumor tissue and as a result nanoparticles accumulate in the tumor while being excluded from normal tissue. Second, biodegradable implanted polymers. In these devices, the radio-enhancing drug is physically trapped within the polymer matrix which is implanted in the tumor. The drug is released as the polymer degrades in response to its local environment. The degradation rate of the polymer device can be adjusted to control the rate of drug release. By this means, the level of radio-enhancing drug can be maintained at the tumor site for the duration of radiation treatment. RESULTS AND CONCLUSIONS Results of experiments indicate that for both methods tumor control could be optimized by maintaining the radio-enhancing drug at a useful concentration in the tumor over a period of time compatible with the duration of fractionated radiation treatment. These studies have provided proof of principle support for the further development of this approach. To date, while some of the methods and devices for drug delivery described in this paper have been involved in clinical trials, none have so far been developed for routine clinical application.
Collapse
|
78
|
Karlsson J, Luly KM, Tzeng SY, Green JJ. Nanoparticle designs for delivery of nucleic acid therapeutics as brain cancer therapies. Adv Drug Deliv Rev 2021; 179:113999. [PMID: 34715258 PMCID: PMC8720292 DOI: 10.1016/j.addr.2021.113999] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/06/2021] [Accepted: 10/05/2021] [Indexed: 12/18/2022]
Abstract
Glioblastoma (GBM) is an aggressive central nervous system cancer with a dismal prognosis. The standard of care involves surgical resection followed by radiotherapy and chemotherapy, but five-year survival is only 5.6% despite these measures. Novel therapeutic approaches, such as immunotherapies, targeted therapies, and gene therapies, have been explored to attempt to extend survival for patients. Nanoparticles have been receiving increasing attention as promising vehicles for non-viral nucleic acid delivery in the context of GBM, though delivery is often limited by low blood-brain barrier permeability, particle instability, and low trafficking to target brain structures and cells. In this review, nanoparticle design considerations and new advances to overcome nucleic acid delivery challenges to treat brain cancer are summarized and discussed.
Collapse
Affiliation(s)
- Johan Karlsson
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Kathryn M. Luly
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Stephany Y. Tzeng
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Jordan J. Green
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Departments of Ophthalmology, Oncology, Neurosurgery, Materials Science & Engineering, and Chemical & Biomolecular Engineering, and the Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| |
Collapse
|
79
|
Abstract
Glioblastoma is one of the deadliest forms of primary adult tumors, with median survival of 14.6 months post-diagnosis despite aggressive standard of care treatment. This grim prognosis for glioblastoma patients has changed little in the past two decades, necessitating novel treatment modalities. One potential treatment modality is cancer immunotherapy, which has shown remarkable progress in slowing disease progression or even potentially curing certain solid tumors. However, the transport barriers posed by the blood-brain barrier and the immune privileged status of the central nervous system pose drug delivery obstacles that are unique to brain tumors. In this review, we provide an overview of the various physiological, immunological, and drug delivery barriers that must be overcome for effective glioblastoma treatment. We discuss chemical modification strategies to enable nanomedicines to bypass the blood-brain barrier and reach intracranial tumors. Finally, we highlight recent advances in biomaterial-based strategies for cancer immunotherapy that can be adapted to glioblastoma treatment.
Collapse
Affiliation(s)
- Yuan Rui
- Department of Biomedical Engineering, the Institute for Nanobiotechnology and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jordan J Green
- Department of Biomedical Engineering, the Institute for Nanobiotechnology and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Departments of Neurosurgery, Ophthalmology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Departments of Materials Science & Engineering and Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Johns Hopkins University School of Medicine, Bloomberg~Kimmel Institute for Cancer Immunotherapy, Baltimore, MD, USA.
| |
Collapse
|
80
|
Kang JH, Desjardins A. Convection-enhanced delivery for high-grade glioma. Neurooncol Pract 2021; 9:24-34. [DOI: 10.1093/nop/npab065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Abstract
Glioblastoma (GBM) is the most common adult primary malignant brain tumor and is associated with a dire prognosis. Despite multi-modality therapies of surgery, radiation, and chemotherapy, its 5-year survival rate is 6.8%. The presence of the blood-brain barrier (BBB) is one factor that has made GBM difficult to treat. Convection-enhanced delivery (CED) is a modality that bypasses the BBB, which allows the intracranial delivery of therapies that would not otherwise cross the BBB and avoids systemic toxicities. This review will summarize prior and ongoing studies and highlights practical considerations related to clinical care to aid providers caring for a high-grade glioma patient being treated with CED. Although not the main scope of this paper, this review also touches upon relevant technical considerations of using CED, an area still under much development.
Collapse
Affiliation(s)
- Jennifer H Kang
- Department of Neurology, Duke University Medical Center, Durham, North Carolina, USA
| | - Annick Desjardins
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
81
|
Discriminating surgical bed cysts from bacterial brain abscesses after Carmustine wafer implantation in newly diagnosed IDH-wildtype glioblastomas. Neurosurg Rev 2021; 45:1501-1511. [PMID: 34651215 DOI: 10.1007/s10143-021-01670-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/16/2021] [Accepted: 10/04/2021] [Indexed: 10/20/2022]
Abstract
Carmustine wafers can be implanted in the surgical bed of high-grade gliomas, which can induce surgical bed cyst formation, leading to clinically relevant mass effect. An observational retrospective monocentric study was conducted including 122 consecutive adult patients with a newly diagnosed supratentorial glioblastoma who underwent a surgical resection with Carmustine wafer implantation as first line treatment (2005-2018). Twenty-two patients (18.0%) developed a postoperative contrast-enhancing cyst within the surgical bed: 16 surgical bed cysts and six bacterial abscesses. All patients with a surgical bed cyst were managed conservatively, all resolved on imaging follow-up, and no patient stopped the radiochemotherapy. Independent risk factors of formation of a postoperative surgical bed cyst were age ≥ 60 years (p = 0.019), number of Carmustine wafers implanted ≥ 8 (p = 0.040), and partial resection (p = 0.025). Compared to surgical bed cysts, the occurrence of a postoperative bacterial abscess requiring surgical management was associated more frequently with a shorter time to diagnosis from surgery (p = 0.009), new neurological deficit (p < 0.001), fever (p < 0.001), residual air in the cyst (p = 0.018), a cyst diameter greater than that of the initial tumor (p = 0.027), and increased mass effect and brain edema compared to early postoperative MRI (p = 0.024). Contrast enhancement (p = 0.473) and diffusion signal abnormalities (p = 0.471) did not differ between postoperative bacterial abscesses and surgical bed cysts. Clinical and imaging findings help discriminate between surgical bed cysts and bacterial abscesses following Carmustine wafer implantation. Surgical bed cysts can be managed conservatively. Individual risk factors will help tailor their steroid therapy and imaging follow-up.
Collapse
|
82
|
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
|
83
|
NISHIKAWA R, IWATA H, SAKATA Y, MURAMOTO K, MATSUOKA T. Safety of Gliadel Implant for Malignant Glioma: Report of Postmarketing Surveillance in Japan. Neurol Med Chir (Tokyo) 2021; 61:536-548. [PMID: 34092748 PMCID: PMC8443968 DOI: 10.2176/nmc.oa.2021-0024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/26/2021] [Indexed: 11/20/2022] Open
Abstract
Clinical trial data of Carmustine implant (Gliadel Wafer) in Japanese patients with malignant glioma are limited; thus, we conducted a postmarketing surveillance study to evaluate the safety of Gliadel in real-world clinical practice in Japan. In this postmarketing surveillance study, all patients who received Gliadel placement for malignant glioma surgeries from its market launch (January 9, 2013) to July 10, 2013 were enrolled from 229 institutions using a central registration system. Up to eight wafers of Gliadel (containing 61.6 mg of carmustine) were used to cover the site of brain tumor resection intraoperatively according to the size and shape of the tumor resection cavity. The observation period lasted 3 months after Gliadel placement. Patients were followed up for 1 year postoperatively. Safety was assessed by the incidence of adverse events (AEs) and adverse drug reactions (ADRs). In total, 558 patients were included. Most patients (66.7%) received eight Gliadel wafers. The percentage of patients with ADRs was 35.7% (365 ADR episodes in 199 patients). Of the AEs of special interest, the most common were cerebral edema (22.2%, 124/558 patients), convulsion (9.9%, 55/558 patients), impaired healing (4.8%, 27/558 patients), and infection (3.4%, 19/558 patients). This first all-case postmarketing surveillance report of the safety of Gliadel in real-world clinical practice in Japan suggests that the risk of toxicity with Gliadel placement is relatively tolerable. The survival benefits of Gliadel placement should be evaluated and considered carefully by the clinician taking into account possible toxicities.
Collapse
Affiliation(s)
- Ryo NISHIKAWA
- Department of Neuro-Oncology and Neurosurgery, Saitama Medical University
International Medical Center, Hidaka, Saitama, Japan
| | | | | | | | | |
Collapse
|
84
|
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
|
85
|
Alghamdi M, Gumbleton M, Newland B. Local delivery to malignant brain tumors: potential biomaterial-based therapeutic/adjuvant strategies. Biomater Sci 2021; 9:6037-6051. [PMID: 34357362 DOI: 10.1039/d1bm00896j] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Glioblastoma (GBM) is the most aggressive malignant brain tumor and is associated with a very poor prognosis. The standard treatment for newly diagnosed patients involves total tumor surgical resection (if possible), plus irradiation and adjuvant chemotherapy. Despite treatment, the prognosis is still poor, and the tumor often recurs within two centimeters of the original tumor. A promising approach to improving the efficacy of GBM therapeutics is to utilize biomaterials to deliver them locally at the tumor site. Local delivery to GBM offers several advantages over systemic administration, such as bypassing the blood-brain barrier and increasing the bioavailability of the therapeutic at the tumor site without causing systemic toxicity. Local delivery may also combat tumor recurrence by maintaining sufficient drug concentrations at and surrounding the original tumor area. Herein, we critically appraised the literature on local delivery systems based within the following categories: polymer-based implantable devices, polymeric injectable systems, and hydrogel drug delivery systems. We also discussed the negative effect of hypoxia on treatment strategies and how one might utilize local implantation of oxygen-generating biomaterials as an adjuvant to enhance current therapeutic strategies.
Collapse
Affiliation(s)
- Majed Alghamdi
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK. and Faculty of Pharmacy, King Abdulaziz University, Jeddah, 22522, Kingdom of Saudi Arabia
| | - Mark Gumbleton
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK.
| | - Ben Newland
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK. and Leibniz-Institut für Polymerforschung Dresden, Max Bergmann Center of Biomaterials Dresden, Hohe Straße 6, D-01069 Dresden, Germany
| |
Collapse
|
86
|
Darbasizadeh B, Mortazavi SA, Kobarfard F, Jaafari MR, Hashemi A, Farhadnejad H, Feyzi-barnaji B. Electrospun Doxorubicin-loaded PEO/PCL core/sheath nanofibers for chemopreventive action against breast cancer cells. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102576] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
87
|
Bouché M, Dong YC, Sheikh S, Taing K, Saxena D, Hsu JC, Chen MH, Salinas RD, Song H, Burdick JA, Dorsey J, Cormode DP. Novel Treatment for Glioblastoma Delivered by a Radiation Responsive and Radiopaque Hydrogel. ACS Biomater Sci Eng 2021; 7:3209-3220. [PMID: 34160196 PMCID: PMC8411482 DOI: 10.1021/acsbiomaterials.1c00385] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Successful treatment of glioblastoma (GBM) is hampered by primary tumor recurrence after surgical resection and poor prognosis, despite adjuvant radiotherapy and chemotherapy. In search of improved outcomes for this disease, quisinostat appeared as a lead compound in drug screening. A delivery system was devised for this drug and to exploit current clinical methodology: an injectable hydrogel, loaded with both the quisinostat drug and radiopaque gold nanoparticles (AuNP) as contrast agent, that can release these payloads as a response to radiation. This hydrogel grants high local drug concentrations, overcoming issues with current standards of care. Significant hydrogel degradation and quisinostat release were observed due to the radiation trigger, providing high in vitro anticancer activity. In vivo, the combination of radiotherapy and the radiation-induced delivery of quisinostat from the hydrogel, successfully inhibited tumor growth in a mice model bearing xenografted human GBM tumors with a total response rate of 67%. Long-term tolerability was observed after intratumoral injection of the quisinostat loaded hydrogel. The AuNP payload enabled precise image-guided radiation delivery and the monitoring of hydrogel degradation using computed tomography (CT). These exciting results highlight this hydrogel as a versatile imageable drug delivery platform that can be activated simultaneously to radiation therapy and potentially offers improved treatment for GBM.
Collapse
Affiliation(s)
- Mathilde Bouché
- Department of Radiology, University of Pennsylvania, 3400 Spruce Street, 1 Silverstein Philadelphia, Pennsylvania 19104, United States
| | - Yuxi C Dong
- Department of Radiology, University of Pennsylvania, 3400 Spruce Street, 1 Silverstein Philadelphia, Pennsylvania 19104, United States
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, Pennsylvania 19104, United States
| | - Saad Sheikh
- Department of Radiation Oncology, University of Pennsylvania, 3400 Civic Center Boulevard Atrium, Philadelphia, Pennsylvania 19104, United States
| | - Kimberly Taing
- Department of Radiology, University of Pennsylvania, 3400 Spruce Street, 1 Silverstein Philadelphia, Pennsylvania 19104, United States
| | - Deeksha Saxena
- Department of Radiation Oncology, University of Pennsylvania, 3400 Civic Center Boulevard Atrium, Philadelphia, Pennsylvania 19104, United States
| | - Jessica C Hsu
- Department of Radiology, University of Pennsylvania, 3400 Spruce Street, 1 Silverstein Philadelphia, Pennsylvania 19104, United States
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, Pennsylvania 19104, United States
| | - Minna H Chen
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, Pennsylvania 19104, United States
| | - Ryan D Salinas
- Department of Neurosurgery, University of Pennsylvania, 3400 Spruce Street, Philadelphia, Pennsylvania 19104, United States
| | - Hongjun Song
- Department of Neuroscience, University of Pennsylvania, 3400 Spruce Street, Philadelphia, Pennsylvania 19104, United States
| | - Jason A Burdick
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, Pennsylvania 19104, United States
| | - Jay Dorsey
- Department of Radiation Oncology, University of Pennsylvania, 3400 Civic Center Boulevard Atrium, Philadelphia, Pennsylvania 19104, United States
| | - David P Cormode
- Department of Radiology, University of Pennsylvania, 3400 Spruce Street, 1 Silverstein Philadelphia, Pennsylvania 19104, United States
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
88
|
Kumthekar P, Ko CH, Paunesku T, Dixit K, Sonabend AM, Bloch O, Tate M, Schwartz M, Zuckerman L, Lezon R, Lukas RV, Jovanovic B, McCortney K, Colman H, Chen S, Lai B, Antipova O, Deng J, Li L, Tommasini-Ghelfi S, Hurley LA, Unruh D, Sharma NV, Kandpal M, Kouri FM, Davuluri RV, Brat DJ, Muzzio M, Glass M, Vijayakumar V, Heidel J, Giles FJ, Adams AK, James CD, Woloschak GE, Horbinski C, Stegh AH. A first-in-human phase 0 clinical study of RNA interference-based spherical nucleic acids in patients with recurrent glioblastoma. Sci Transl Med 2021; 13:13/584/eabb3945. [PMID: 33692132 DOI: 10.1126/scitranslmed.abb3945] [Citation(s) in RCA: 194] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 02/19/2021] [Indexed: 12/13/2022]
Abstract
Glioblastoma (GBM) is one of the most difficult cancers to effectively treat, in part because of the lack of precision therapies and limited therapeutic access to intracranial tumor sites due to the presence of the blood-brain and blood-tumor barriers. We have developed a precision medicine approach for GBM treatment that involves the use of brain-penetrant RNA interference-based spherical nucleic acids (SNAs), which consist of gold nanoparticle cores covalently conjugated with radially oriented and densely packed small interfering RNA (siRNA) oligonucleotides. On the basis of previous preclinical evaluation, we conducted toxicology and toxicokinetic studies in nonhuman primates and a single-arm, open-label phase 0 first-in-human trial (NCT03020017) to determine safety, pharmacokinetics, intratumoral accumulation and gene-suppressive activity of systemically administered SNAs carrying siRNA specific for the GBM oncogene Bcl2Like12 (Bcl2L12). Patients with recurrent GBM were treated with intravenous administration of siBcl2L12-SNAs (drug moniker: NU-0129), at a dose corresponding to 1/50th of the no-observed-adverse-event level, followed by tumor resection. Safety assessment revealed no grade 4 or 5 treatment-related toxicities. Inductively coupled plasma mass spectrometry, x-ray fluorescence microscopy, and silver staining of resected GBM tissue demonstrated that intravenously administered SNAs reached patient tumors, with gold enrichment observed in the tumor-associated endothelium, macrophages, and tumor cells. NU-0129 uptake into glioma cells correlated with a reduction in tumor-associated Bcl2L12 protein expression, as indicated by comparison of matched primary tumor and NU-0129-treated recurrent tumor. Our results establish SNA nanoconjugates as a potential brain-penetrant precision medicine approach for the systemic treatment of GBM.
Collapse
Affiliation(s)
- Priya Kumthekar
- Ken and Ruth Davee Department of Neurology, The Northwestern Malnati Brain Tumor Institute, Feinberg School of Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA.
| | - Caroline H Ko
- International Institute for Nanotechnology, Northwestern University, Evanston, IL 60208, USA
| | - Tatjana Paunesku
- Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Karan Dixit
- Ken and Ruth Davee Department of Neurology, The Northwestern Malnati Brain Tumor Institute, Feinberg School of Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | - Adam M Sonabend
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Orin Bloch
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Matthew Tate
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Margaret Schwartz
- Ken and Ruth Davee Department of Neurology, The Northwestern Malnati Brain Tumor Institute, Feinberg School of Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | - Laura Zuckerman
- Ken and Ruth Davee Department of Neurology, The Northwestern Malnati Brain Tumor Institute, Feinberg School of Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | - Ray Lezon
- Ken and Ruth Davee Department of Neurology, The Northwestern Malnati Brain Tumor Institute, Feinberg School of Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | - Rimas V Lukas
- Ken and Ruth Davee Department of Neurology, The Northwestern Malnati Brain Tumor Institute, Feinberg School of Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | - Borko Jovanovic
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Kathleen McCortney
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Howard Colman
- Huntsman Cancer Institute and Department of Neurosurgery, University of Utah, Salt Lake City, UT 84112, USA
| | - Si Chen
- X-ray Science Division, Advanced Photon Source, Argonne National Laboratory, Argonne, IL 60439, USA
| | - Barry Lai
- X-ray Science Division, Advanced Photon Source, Argonne National Laboratory, Argonne, IL 60439, USA
| | - Olga Antipova
- X-ray Science Division, Advanced Photon Source, Argonne National Laboratory, Argonne, IL 60439, USA
| | - Junjing Deng
- X-ray Science Division, Advanced Photon Source, Argonne National Laboratory, Argonne, IL 60439, USA
| | - Luxi Li
- X-ray Science Division, Advanced Photon Source, Argonne National Laboratory, Argonne, IL 60439, USA
| | - Serena Tommasini-Ghelfi
- Ken and Ruth Davee Department of Neurology, The Northwestern Malnati Brain Tumor Institute, Feinberg School of Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | - Lisa A Hurley
- Ken and Ruth Davee Department of Neurology, The Northwestern Malnati Brain Tumor Institute, Feinberg School of Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | - Dusten Unruh
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Nitya V Sharma
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Manoj Kandpal
- Preventive Medicine, Health and Biomedical Informatics, Feinberg School of Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | - Fotini M Kouri
- Ken and Ruth Davee Department of Neurology, The Northwestern Malnati Brain Tumor Institute, Feinberg School of Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | - Ramana V Davuluri
- Preventive Medicine, Health and Biomedical Informatics, Feinberg School of Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | - Daniel J Brat
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Miguel Muzzio
- Life Sciences Group, IIT Research Institute, Chicago, IL 60616, USA
| | | | | | | | - Francis J Giles
- Developmental Therapeutics Program of the Division of Hematology Oncology, Feinberg School of Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | - Ann K Adams
- Office for Research, Northwestern University, Evanston, IL 60208, USA
| | - C David James
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Gayle E Woloschak
- Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Craig Horbinski
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.,Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Alexander H Stegh
- Ken and Ruth Davee Department of Neurology, The Northwestern Malnati Brain Tumor Institute, Feinberg School of Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA. .,International Institute for Nanotechnology, Northwestern University, Evanston, IL 60208, USA
| |
Collapse
|
89
|
Tseng YY, Chen TY, Liu SJ. Role of Polymeric Local Drug Delivery in Multimodal Treatment of Malignant Glioma: A Review. Int J Nanomedicine 2021; 16:4597-4614. [PMID: 34267515 PMCID: PMC8275179 DOI: 10.2147/ijn.s309937] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/21/2021] [Indexed: 12/29/2022] Open
Abstract
Malignant gliomas (MGs) are the most common and devastating primary brain tumor. At present, surgical interventions, radiotherapy, and chemotherapy are only marginally effective in prolonging the life expectancy of patients with MGs. Inherent heterogeneity, aggressive invasion and infiltration, intact physical barriers, and the numerous mechanisms underlying chemotherapy and radiotherapy resistance contribute to the poor prognosis for patients with MGs. Various studies have investigated methods to overcome these obstacles in MG treatment. In this review, we address difficulties in MG treatment and focus on promising polymeric local drug delivery systems. In contrast to most local delivery systems, which are directly implanted into the residual cavity after intratumoral injection or the surgical removal of a tumor, some rapidly developing and promising nanotechnological methods—including surface-decorated nanoparticles, magnetic nanoparticles, and focused ultrasound assist transport—are administered through (systemic) intravascular injection. We also discuss further synergistic and multimodal strategies for heightening therapeutic efficacy. Finally, we outline the challenges and therapeutic potential of these polymeric drug delivery systems.
Collapse
Affiliation(s)
- Yuan-Yun Tseng
- Department of Neurosurgery, New Taipei Municipal Tu-Cheng Hospital (Built and Operated by Chang Gung Medical Foundation), New Taipei City, Taiwan
| | - Tai-Yuan Chen
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shih-Jung Liu
- Department of Mechanical Engineering, Chang Gung University, Tao-Yuan, Taiwan.,Department of Orthopedic Surgery, Chang Gung Memorial Hospital-Linkuo, Tao-Yuan, Taiwan
| |
Collapse
|
90
|
Di Mascolo D, Palange AL, Primavera R, Macchi F, Catelani T, Piccardi F, Spanò R, Ferreira M, Marotta R, Armirotti A, Gallotti AL, Galli R, Wilson C, Grant GA, Decuzzi P. Conformable hierarchically engineered polymeric micromeshes enabling combinatorial therapies in brain tumours. NATURE NANOTECHNOLOGY 2021; 16:820-829. [PMID: 33795849 DOI: 10.1038/s41565-021-00879-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 02/17/2021] [Indexed: 06/12/2023]
Abstract
The poor transport of molecular and nanoscale agents through the blood-brain barrier together with tumour heterogeneity contribute to the dismal prognosis in patients with glioblastoma multiforme. Here, a biodegradable implant (μMESH) is engineered in the form of a micrometre-sized poly(lactic-co-glycolic acid) mesh laid over a water-soluble poly(vinyl alcohol) layer. Upon poly(vinyl alcohol) dissolution, the flexible poly(lactic-co-glycolic acid) mesh conforms to the resected tumour cavity as docetaxel-loaded nanomedicines and diclofenac molecules are continuously and directly released into the adjacent tumour bed. In orthotopic brain cancer models, generated with a conventional, reference cell line and patient-derived cells, a single μMESH application, carrying 0.75 mg kg-1 of docetaxel and diclofenac, abrogates disease recurrence up to eight months after tumour resection, with no appreciable adverse effects. Without tumour resection, the μMESH increases the median overall survival (∼30 d) as compared with the one-time intracranial deposition of docetaxel-loaded nanomedicines (15 d) or 10 cycles of systemically administered temozolomide (12 d). The μMESH modular structure, for the independent coloading of different molecules and nanomedicines, together with its mechanical flexibility, can be exploited to treat a variety of cancers, realizing patient-specific dosing and interventions.
Collapse
Affiliation(s)
- Daniele Di Mascolo
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Anna Lisa Palange
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Rosita Primavera
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University, Palo Alto, CA, USA
| | - Francesca Macchi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Tiziano Catelani
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
- Microscopy Facility, University of Milano-Bicocca, Milan, Italy
| | - Federica Piccardi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Raffaele Spanò
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Miguel Ferreira
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Roberto Marotta
- Electron Microscopy Facility, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Andrea Armirotti
- Analytical Chemistry Lab, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Alberto L Gallotti
- Neural Stem Cell Biology Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Rossella Galli
- Neural Stem Cell Biology Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Christy Wilson
- Department of Neurosurgery, Stanford University, Palo Alto, CA, USA
| | - Gerald A Grant
- Department of Neurosurgery, Stanford University, Palo Alto, CA, USA
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy.
| |
Collapse
|
91
|
Bettag C, Hussein A, Sachkova A, Bock HC, Mielke D, Rohde V, Abboud T. Implantation of Carmustine wafers after resection of malignant glioma with and without opening of the ventricular system. J Neurooncol 2021; 153:519-525. [PMID: 34148163 DOI: 10.1007/s11060-021-03792-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/18/2021] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Implantation of biodegradable Carmustine wafers in patients with malignant glioma is not generally recommended when the ventricular system is opened during tumor resection. Thrombin/fibrinogenn-covered collagen fleeces showed promising results in sufficiently closing ventricular defects. The aim of this study was to evaluate the postoperative morbidity in patients with implanted Carmustine wafers either with opened or intact ventricular system. METHODS A consecutive series of patients who underwent resection of malignant glioma with implantation of Carmustine wafers was analyzed. In case of opening of the ventricular system, the defect in the ventricle wall was sealed using a collagen sponge coated with fibrinogen and thrombin prior to the implantation of the wafers. Postoperative adverse events (AE) and Karnofsky performance status scale (KPS) at follow up were compared between both groups. RESULTS Fifty-four patients were included. The ventricular system was opened in 33 patients and remained intact in 21 patients. Both groups were comparable in terms of age, rate of primary and recurrent glioma, preoperative KPS, rate of gross total resection and number of implanted wafers. Postoperative AEs occurred in 9/33 patients (27.3%) with opened and in 5/21 patients (23.8%) with intact ventricular system (p = 0.13). At follow-up assessments, KPS was not significantly different between both groups (p = 0.18). Opened ventricular system was not associated with a higher incidence of postoperative AEs (p = 0.98). CONCLUSION Appropriate closure of opened ventricular system during resection of malignant glioma allows for a safe implantation of Carmustine wafers and is not associated with a higher incidence of postoperative AEs.
Collapse
Affiliation(s)
- Christoph Bettag
- Department of Neurosurgery, University Medical Center Goettingen, University Hospital Göttingen, Georg-August-University, Goettingen, Germany
| | - Abdelhalim Hussein
- Department of Neurosurgery, University Medical Center Goettingen, University Hospital Göttingen, Georg-August-University, Goettingen, Germany
| | - Alexandra Sachkova
- Department of Neurosurgery, University Medical Center Goettingen, University Hospital Göttingen, Georg-August-University, Goettingen, Germany
| | - Hans Christoph Bock
- Department of Neurosurgery, University Medical Center Goettingen, University Hospital Göttingen, Georg-August-University, Goettingen, Germany
| | - Dorothee Mielke
- Department of Neurosurgery, University Medical Center Goettingen, University Hospital Göttingen, Georg-August-University, Goettingen, Germany
| | - Veit Rohde
- Department of Neurosurgery, University Medical Center Goettingen, University Hospital Göttingen, Georg-August-University, Goettingen, Germany
| | - Tammam Abboud
- Department of Neurosurgery, University Medical Center Goettingen, University Hospital Göttingen, Georg-August-University, Goettingen, Germany.
| |
Collapse
|
92
|
Keutgen XM, Ornell KJ, Vogle A, Lakiza O, Williams J, Miller P, Mistretta KS, Setia N, Weichselbaum RR, Coburn JM. Sunitinib-Loaded Chondroitin Sulfate Hydrogels as a Novel Drug-Delivery Mechanism for the Treatment of Pancreatic Neuroendocrine Tumors. Ann Surg Oncol 2021; 28:8532-8543. [PMID: 34091777 DOI: 10.1245/s10434-021-10245-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 05/20/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Pancreatic neuroendocrine tumors (PanNETs) are increasingly common. Experts debate whether small tumors should be resected. Tumor destruction via injection of cytotoxic agents could offer a minimal invasive approach to this controversy. We hypothesize that a new drug delivery system comprising chondroitin sulfate (CS) hydrogels loaded with sunitinib (SUN) suppresses tumor growth in PanNET cells. METHODS Injectable hydrogels composed of CS modified with methacrylate groups (MA) were fabricated and loaded with SUN. Loading target was either 200 µg (SUN200-G) or 500 µg (SUN500-G) as well as sham hydrogel with no drug loading (SUN0-G). SUN release from hydrogels was monitored in vitro over time and cytotoxicity induced by the released SUN was evaluated using QGP-1 and BON1 PanNET cell lines. QGP-1 xenografts were developed in 35 mice and directly injected with 25 µL of either SUN200-G, SUN500-G, SUN0-G, 100 µL of Sunitinib Malate (SUN-inj), or given 40 mg/kg/day oral sunitinib (SUN-oral). RESULTS SUN-loaded CSMA hydrogel retained complete in vitro cytotoxicity toward the QGP-1 PanNET and BON-1 PanNET cell lines for 21 days. Mouse xenograft models with QGP-1 PanNETs showed a significant delay in tumor growth in the SUN200/500-G, SUN-inj and SUN-oral groups compared with SUN0-G (p = 0.0014). SUN500-G hydrogels induced significantly more tumor necrosis than SUN0-G (p = 0.04). There was no difference in tumor growth delay between SUN200/500G, SUN-inj, and SUN-oral. CONCLUSIONS This study demonstrates that CSMA hydrogels loaded with SUN suppress PanNETs growth. This drug delivery could approach represents a novel way to treat PanNETs and other neoplasms via intratumoral injection.
Collapse
Affiliation(s)
- Xavier M Keutgen
- Endocrine Surgery Research Program, Division of General Surgery and Surgical Oncology, Department of Surgery, The University of Chicago Medicine, Chicago, IL, USA.
| | - Kimberly J Ornell
- Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Alyx Vogle
- Endocrine Surgery Research Program, Division of General Surgery and Surgical Oncology, Department of Surgery, The University of Chicago Medicine, Chicago, IL, USA
| | - Olga Lakiza
- Endocrine Surgery Research Program, Division of General Surgery and Surgical Oncology, Department of Surgery, The University of Chicago Medicine, Chicago, IL, USA
| | - Jelani Williams
- Endocrine Surgery Research Program, Division of General Surgery and Surgical Oncology, Department of Surgery, The University of Chicago Medicine, Chicago, IL, USA
| | - Paul Miller
- Endocrine Surgery Research Program, Division of General Surgery and Surgical Oncology, Department of Surgery, The University of Chicago Medicine, Chicago, IL, USA
| | | | - Namrata Setia
- Department of Pathology, The University of Chicago Medicine, Chicago, IL, USA
| | - Ralph R Weichselbaum
- Department of Radiation Oncology and Cellular Biology, The University of Chicago Medicine, Chicago, IL, USA
| | - Jeannine M Coburn
- Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, USA.
| |
Collapse
|
93
|
Phipps MD, Sanders VA, Deri MA. Current State of Targeted Radiometal-Based Constructs for the Detection and Treatment of Disease in the Brain. Bioconjug Chem 2021; 32:1331-1347. [PMID: 34015928 DOI: 10.1021/acs.bioconjchem.1c00180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The continual development of radiopharmaceutical agents for the field of nuclear medicine is integral to promoting the necessity of personalized medicine. One way to greatly expand the selection of radiopharmaceuticals available is to broaden the range of radionuclides employed in such agents. Widening the scope of development to include radiometals with their variety of physical decay characteristics and chemical properties opens up a myriad of possibilities for new actively targeted molecules and bioconjugates. This is especially true to further advance the imaging and treatment of disease in the brain. Over the past few decades, imaging of disease in the brain has heavily relied on agents which exploit metabolic uptake. However, through utilizing the broad range of physical characteristics that radiometals offer, the ability to target other processes has become more available. The varied chemistries of radiometals also allows for them to incorporated into specifically designed diverse constructs. A major limitation to efficient treatment of disease in the brain is the ability for relevant agents to penetrate the blood-brain barrier. Thus, along with efficient disease targeting, there must be intentional thought put into overcoming this challenge. Here, we review the current field of radiometal-based agents aimed at either imaging or therapy of brain disease that have been evaluated through at least in vivo studies.
Collapse
Affiliation(s)
- Michael D Phipps
- Ph.D. Program in Chemistry, Graduate Center of the City University of New York, New York, New York 10016, United States.,Department of Chemistry, Lehman College of the City University of New York, New York, New York 10468, United States.,Department of Chemistry, Hunter College of the City University of New York, New York, New York 10065, United States.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Vanessa A Sanders
- Collider Accelerator Department, Brookhaven National Laboratory, Upton, New York 11973, United States
| | - Melissa A Deri
- Ph.D. Program in Chemistry, Graduate Center of the City University of New York, New York, New York 10016, United States.,Department of Chemistry, Lehman College of the City University of New York, New York, New York 10468, United States
| |
Collapse
|
94
|
Marcucci F, Corti A, Ferreri AJM. Breaching the Blood-Brain Tumor Barrier for Tumor Therapy. Cancers (Basel) 2021; 13:cancers13102391. [PMID: 34063335 PMCID: PMC8156088 DOI: 10.3390/cancers13102391] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/06/2021] [Accepted: 05/11/2021] [Indexed: 12/26/2022] Open
Abstract
Tumors affecting the central nervous system (CNS), either primary or secondary, are highly prevalent and represent an unmet medical need. Prognosis of these tumors remains poor, mostly due to the low intrinsic chemo/radio-sensitivity of tumor cells, a meagerly known role of the microenvironment and the poor CNS bioavailability of most used anti-cancer agents. The BBTB is the main obstacle for anticancer drugs to achieve therapeutic concentrations in the tumor tissues. During the last decades, many efforts have been devoted to the identification of modalities allowing to increase drug delivery into brain tumors. Until recently, success has been modest, as few of these approaches reached clinical testing and even less gained regulatory approval. In recent years, the scenario has changed, as various conjugates and drug delivery technologies have advanced into clinical testing, with encouraging results and without being burdened by a heavy adverse event profile. In this article, we review the different approaches aimed at increasing drug delivery to brain tumors, with particular attention to new, promising approaches that increase the permeability of the BBTB or exploit physiological transport mechanisms.
Collapse
Affiliation(s)
- Fabrizio Marcucci
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20132 Milan, Italy
- Correspondence: (F.M.); (A.C.)
| | - Angelo Corti
- Division of Experimental Oncology, Tumor Biology and Vascular Targeting Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Faculty of Medicine and Surgery, Università Vita-Salute San Raffaele, 20132 Milan, Italy
- Correspondence: (F.M.); (A.C.)
| | - Andrés J. M. Ferreri
- Lymphoma Unit, Department of Onco-Hematology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy;
| |
Collapse
|
95
|
Mansouri A, Beyn ME, Pancholi A, Chow CT, Wang R, Boutet A, Elias GJB, Germann J, Loh A, Voisin MR, Lozano AM, Chiocca EA, Vogelbaum MA, Zadeh G. Evolution of the Neurosurgeon's Role in Clinical Trials for Glioblastoma: A Systematic Overview of the Clinicaltrials.Gov Database. Neurosurgery 2021; 89:196-203. [PMID: 33989408 DOI: 10.1093/neuros/nyab169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 03/12/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The therapeutic challenge of glioblastoma (GBM) has catalyzed the development of clinical trials to evaluate novel interventions. With increased understanding of GBM biology and technological advances, the neurosurgeon's role in neuro-oncology has evolved. OBJECTIVE To evaluate the current landscape of procedure-based clinical trials for GBM to characterize this evolution, gain insight into past failures, and accordingly outline implications for future research and practice that may inform future studies. METHODS The ClinicalTrials.gov database was searched for surgical/procedural trials in individuals with GBM. Demographics, specific intervention, trial phase, and main outcome measures were abstracted. RESULTS A total of 224 of 2311 GBM trials (9.7%) were identified as procedural, with the majority being based in the United States (155/224, 69.2%), single-center (155/224, 69.2%), and not randomized (176/224, 78.6%). Primary and recurrent GBMs were evenly addressed. The leading interventions were local delivery of therapeutics (50.0%), surgical techniques (33.9%), such as image-guided surgery, and novel device applications (14.3%). Phase I designs predominated (82/224, 36.6%). The top primary outcome was safety/tolerability/feasibility (88/224, 39.3%), followed by survival (46/224, 20.5%). Approximately 17% of studies were terminated, withdrawn, or suspended. Fifty-two linked publications were identified, among which 42 were classified as having a positive result. CONCLUSION Procedural interventions comprised ∼10% of all registered GBM trials. Local delivery of therapeutics, use of surgical imaging techniques and novel device applications, predominantly through phase I designs, represent the evolved role of the neurosurgeon in neuro-oncology. Improved reporting of trial designs, outcomes, and results are needed to better inform the field and increase efficiency.
Collapse
Affiliation(s)
- Alireza Mansouri
- Department of Neurosurgery, Penn State Hershey Medical Center, Hershey, Pennsylvania, USA.,Penn State Cancer Institute, Hershey, Pennsylvania, USA
| | | | | | | | - Ryan Wang
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Alexandre Boutet
- University Health Network, Toronto, Ontario, Canada.,Joint Department of Medical Imaging, University of Toronto, Toronto, Ontario, Canada
| | | | | | - Aaron Loh
- University Health Network, Toronto, Ontario, Canada
| | - Mathew R Voisin
- Division of Neurosurgery, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Andres M Lozano
- Division of Neurosurgery, Toronto Western Hospital, Toronto, Ontario, Canada.,Krembil Research Institute, Toronto, Ontario, Canada
| | - E Antonio Chiocca
- Harvey W. Cushing Neuro-Oncology Laboratories (HCNL), Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Gelareh Zadeh
- Division of Neurosurgery, Toronto Western Hospital, Toronto, Ontario, Canada.,MacFeeters Hamilton Centre for Neuro-Oncology Research, Princess Margaret Cancer Centre, Toronto, Ontario, Canada.,Institute of Medical Science, Toronto, Ontario, Canada
| |
Collapse
|
96
|
Yu MW, Quail DF. Immunotherapy for Glioblastoma: Current Progress and Challenge. Front Immunol 2021; 12:676301. [PMID: 34054867 PMCID: PMC8158294 DOI: 10.3389/fimmu.2021.676301] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 04/21/2021] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma is a highly lethal brain cancer with a median survival rate of less than 15 months when treated with the current standard of care, which consists of surgery, radiotherapy and chemotherapy. With the recent success of immunotherapy in other aggressive cancers such as advanced melanoma and advanced non-small cell lung cancer, glioblastoma has been brought to the forefront of immunotherapy research. Resistance to therapy has been a major challenge across a multitude of experimental candidates and no immunotherapies have been approved for glioblastoma to-date. Intra- and inter-tumoral heterogeneity, an inherently immunosuppressive environment and tumor plasticity remain barriers to be overcome. Moreover, the unique tissue-specific interactions between the central nervous system and the peripheral immune system present an additional challenge for immune-based therapies. Nevertheless, there is sufficient evidence that these challenges may be overcome, and immunotherapy continues to be actively pursued in glioblastoma. Herein, we review the primary ongoing immunotherapy candidates for glioblastoma with a focus on immune checkpoint inhibitors, myeloid-targeted therapies, vaccines and chimeric antigen receptor (CAR) immunotherapies. We further provide insight on mechanisms of resistance and how our understanding of these mechanisms may pave the way for more effective immunotherapeutics against glioblastoma.
Collapse
Affiliation(s)
- Miranda W Yu
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada.,Department of Physiology, McGill University, Montreal, QC, Canada
| | - Daniela F Quail
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada.,Department of Physiology, McGill University, Montreal, QC, Canada.,Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
97
|
Waldherr L, Seitanidou M, Jakešová M, Handl V, Honeder S, Nowakowska M, Tomin T, Karami Rad M, Schmidt T, Distl J, Birner‐Gruenberger R, von Campe G, Schäfer U, Berggren M, Rinner B, Asslaber M, Ghaffari‐Tabrizi‐Wizsy N, Patz S, Simon DT, Schindl R. Targeted Chemotherapy of Glioblastoma Spheroids with an Iontronic Pump. ADVANCED MATERIALS TECHNOLOGIES 2021; 6:2001302. [PMID: 34195355 PMCID: PMC8218220 DOI: 10.1002/admt.202001302] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/15/2021] [Indexed: 05/13/2023]
Abstract
Successful treatment of glioblastoma multiforme (GBM), the most lethal tumor of the brain, is presently hampered by (i) the limits of safe surgical resection and (ii) "shielding" of residual tumor cells from promising chemotherapeutic drugs such as Gemcitabine (Gem) by the blood brain barrier (BBB). Here, the vastly greater GBM cell-killing potency of Gem compared to the gold standard temozolomide is confirmed, moreover, it shows neuronal cells to be at least 104-fold less sensitive to Gem than GBM cells. The study also demonstrates the potential of an electronically-driven organic ion pump ("GemIP") to achieve controlled, targeted Gem delivery to GBM cells. Thus, GemIP-mediated Gem delivery is confirmed to be temporally and electrically controllable with pmol min-1 precision and electric addressing is linked to the efficient killing of GBM cell monolayers. Most strikingly, GemIP-mediated GEM delivery leads to the overt disintegration of targeted GBM tumor spheroids. Electrically-driven chemotherapy, here exemplified, has the potential to radically improve the efficacy of GBM adjuvant chemotherapy by enabling exquisitely-targeted and controllable delivery of drugs irrespective of whether these can cross the BBB.
Collapse
Affiliation(s)
- Linda Waldherr
- Gottfried Schatz Research Center – BiophysicsMedical University of GrazGraz8010Austria
| | - Maria Seitanidou
- Laboratory of Organic ElectronicsDepartment of Science and TechnologyLinköping UniversityNorrköping60174Sweden
| | - Marie Jakešová
- Laboratory of Organic ElectronicsDepartment of Science and TechnologyLinköping UniversityNorrköping60174Sweden
| | - Verena Handl
- Department of NeurosurgeryMedical University of GrazGraz8010Austria
| | - Sophie Honeder
- Diagnostic and Research Institute of PathologyMedical University of GrazGraz8010Austria
| | - Marta Nowakowska
- Department of NeurosurgeryMedical University of GrazGraz8010Austria
| | - Tamara Tomin
- Diagnostic and Research Institute of PathologyMedical University of GrazGraz8010Austria
- Institute of Chemical Technologies and AnalyticsTechnische Universität WienVienna1060Austria
| | - Meysam Karami Rad
- Laboratory of Organic ElectronicsDepartment of Science and TechnologyLinköping UniversityNorrköping60174Sweden
| | - Tony Schmidt
- Gottfried Schatz Research Center – BiophysicsMedical University of GrazGraz8010Austria
| | - Joachim Distl
- Gottfried Schatz Research Center – BiophysicsMedical University of GrazGraz8010Austria
| | - Ruth Birner‐Gruenberger
- Diagnostic and Research Institute of PathologyMedical University of GrazGraz8010Austria
- Institute of Chemical Technologies and AnalyticsTechnische Universität WienVienna1060Austria
| | - Gord von Campe
- Department of NeurosurgeryMedical University of GrazGraz8010Austria
| | - Ute Schäfer
- Department of NeurosurgeryMedical University of GrazGraz8010Austria
| | - Magnus Berggren
- Laboratory of Organic ElectronicsDepartment of Science and TechnologyLinköping UniversityNorrköping60174Sweden
| | - Beate Rinner
- Division of Biomedical ResearchMedical University of GrazGraz8036Austria
| | - Martin Asslaber
- Diagnostic and Research Institute of PathologyMedical University of GrazGraz8010Austria
| | | | - Silke Patz
- Department of NeurosurgeryMedical University of GrazGraz8010Austria
| | - Daniel T. Simon
- Laboratory of Organic ElectronicsDepartment of Science and TechnologyLinköping UniversityNorrköping60174Sweden
| | - Rainer Schindl
- Gottfried Schatz Research Center – BiophysicsMedical University of GrazGraz8010Austria
| |
Collapse
|
98
|
Janjua TI, Rewatkar P, Ahmed-Cox A, Saeed I, Mansfeld FM, Kulshreshtha R, Kumeria T, Ziegler DS, Kavallaris M, Mazzieri R, Popat A. Frontiers in the treatment of glioblastoma: Past, present and emerging. Adv Drug Deliv Rev 2021; 171:108-138. [PMID: 33486006 DOI: 10.1016/j.addr.2021.01.012] [Citation(s) in RCA: 158] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/13/2020] [Accepted: 01/09/2021] [Indexed: 12/13/2022]
Abstract
Glioblastoma (GBM) is one of the most aggressive cancers of the brain. Despite extensive research over the last several decades, the survival rates for GBM have not improved and prognosis remains poor. To date, only a few therapies are approved for the treatment of GBM with the main reasons being: 1) significant tumour heterogeneity which promotes the selection of resistant subpopulations 2) GBM induced immunosuppression and 3) fortified location of the tumour in the brain which hinders the delivery of therapeutics. Existing therapies for GBM such as radiotherapy, surgery and chemotherapy have been unable to reach the clinical efficacy necessary to prolong patient survival more than a few months. This comprehensive review evaluates the current and emerging therapies including those in clinical trials that may potentially improve both targeted delivery of therapeutics directly to the tumour site and the development of agents that may specifically target GBM. Particular focus has also been given to emerging delivery technologies such as focused ultrasound, cellular delivery systems nanomedicines and immunotherapy. Finally, we discuss the importance of developing novel materials for improved delivery efficacy of nanoparticles and therapeutics to reduce the suffering of GBM patients.
Collapse
|
99
|
The biomedical significance of multifunctional nanobiomaterials: The key components for site-specific delivery of therapeutics. Life Sci 2021; 277:119400. [PMID: 33794255 DOI: 10.1016/j.lfs.2021.119400] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/08/2021] [Accepted: 03/13/2021] [Indexed: 01/07/2023]
Abstract
The emergence of nanotechnology has provided the possibilities to overcome the potential problems associated with the development of pharmaceuticals including the low solubility, non-specific cellular uptake or action, and rapid clearance. Regarding the biomaterials (BMs), huge efforts have been made for improving their multi-functionalities via incorporation of various nanomaterials (NMs). Nanocomposite hydrogels with suitable properties could exhibit a variety of beneficial effects in biomedicine particularly in the delivery of therapeutics or tissue engineering. NMs including the silica- or carbon-based ones are capable of integration into various BMs that might be due to their special compositions or properties such as the hydrophilicity, hydrophobicity, magnetic or electrical characteristics, and responsiveness to various stimuli. This might provide multi-functional nanobiomaterials against a wide variety of disorders. Meanwhile, inappropriate distribution or penetration into the cells or tissues, bio-nano interface complexity, targeting ability loss, or any other unpredicted phenomena are the serious challenging issues. Computational simulations and models enable development of NMs with optimal characteristics and provide a deeper knowledge of NM interaction with biosystems. This review highlights the biomedical significance of the multifunctional NMs particularly those applied for the development of 2-D or 3-D BMs for a variety of applications including the site-specific delivery of therapeutics. The powerful impacts of the computational techniques on the design process of NMs, quantitation and prediction of protein corona formation, risk assessment, and individualized therapy for improved therapeutic outcomes have also been discussed.
Collapse
|
100
|
Vermandel M, Dupont C, Lecomte F, Leroy HA, Tuleasca C, Mordon S, Hadjipanayis CG, Reyns N. Standardized intraoperative 5-ALA photodynamic therapy for newly diagnosed glioblastoma patients: a preliminary analysis of the INDYGO clinical trial. J Neurooncol 2021; 152:501-514. [PMID: 33743128 DOI: 10.1007/s11060-021-03718-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/13/2021] [Indexed: 12/11/2022]
Abstract
PURPOSE Glioblastoma (GBM) is the most aggressive malignant primary brain tumor. The unfavorable prognosis despite maximal therapy relates to high propensity for recurrence. Thus, overall survival (OS) is quite limited and local failure remains the fundamental problem. Here, we present a safety and feasibility trial after treating GBM intraoperatively by photodynamic therapy (PDT) after 5-aminolevulinic acid (5-ALA) administration and maximal resection. METHODS Ten patients with newly diagnosed GBM were enrolled and treated between May 2017 and June 2018. The standardized therapeutic approach included maximal resection (near total or gross total tumor resection (GTR)) guided by 5-ALA fluorescence-guided surgery (FGS), followed by intraoperative PDT. Postoperatively, patients underwent adjuvant therapy (Stupp protocol). Follow-up included clinical examinations and brain MR imaging was performed every 3 months until tumor progression and/or death. RESULTS There were no unacceptable or unexpected toxicities or serious adverse effects. At the time of the interim analysis, the actuarial 12-months progression-free survival (PFS) rate was 60% (median 17.1 months), and the actuarial 12-months OS rate was 80% (median 23.1 months). CONCLUSIONS This trial assessed the feasibility and the safety of intraoperative 5-ALA PDT as a novel approach for treating GBM after maximal tumor resection. The current standard of care remains microsurgical resection whenever feasible, followed by adjuvant therapy (Stupp protocol). We postulate that PDT delivered immediately after resection as an add-on therapy of this primary brain cancer is safe and may help to decrease the recurrence risk by targeting residual tumor cells in the resection cavity. Trial registration NCT number: NCT03048240. EudraCT number: 2016-002706-39.
Collapse
Affiliation(s)
- Maximilien Vermandel
- Univ. Lille, Inserm, CHU Lille, U1189 - ONCO-THAI -Laser Assisted Therapies and Immunotherapies for Oncology, 59000, Lille, France
- Neurosurgery Department, CHU Lille, 59000, Lille, France
| | - Clément Dupont
- Univ. Lille, Inserm, CHU Lille, U1189 - ONCO-THAI -Laser Assisted Therapies and Immunotherapies for Oncology, 59000, Lille, France
| | - Fabienne Lecomte
- Univ. Lille, Inserm, CHU Lille, U1189 - ONCO-THAI -Laser Assisted Therapies and Immunotherapies for Oncology, 59000, Lille, France
| | - Henri-Arthur Leroy
- Univ. Lille, Inserm, CHU Lille, U1189 - ONCO-THAI -Laser Assisted Therapies and Immunotherapies for Oncology, 59000, Lille, France
- Neurosurgery Department, CHU Lille, 59000, Lille, France
| | - Constantin Tuleasca
- Faculty of Biology and Medicine (FBM) and Centre Hospitalier Universitaire Vaudois (CHUV), Clinical Neurosciences Department, Neurosurgery Service and Gamma Knife Center, University of Lausanne (Unil), Lausanne, Switzerland
- Signal Processing Laboratory (LTS 5), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Serge Mordon
- Univ. Lille, Inserm, CHU Lille, U1189 - ONCO-THAI -Laser Assisted Therapies and Immunotherapies for Oncology, 59000, Lille, France
| | - Constantinos G Hadjipanayis
- Department of Neurosurgery, Icahn School of Medicine At Mount Sinai, Mount Sinai Health System, New York, NY, USA
- Department of Neurosurgery, Mount Sinai Beth Israel, New York, NY, USA
| | - Nicolas Reyns
- Univ. Lille, Inserm, CHU Lille, U1189 - ONCO-THAI -Laser Assisted Therapies and Immunotherapies for Oncology, 59000, Lille, France.
- Neurosurgery Department, CHU Lille, 59000, Lille, France.
| |
Collapse
|