Commentary: Current Status of Intratumoral Therapy for Glioblastoma Gregory D

Total Page:16

File Type:pdf, Size:1020Kb

Commentary: Current Status of Intratumoral Therapy for Glioblastoma Gregory D Arnone GD, Wonais M, Linninger A, Mehta AI. J Neurol Neuromedicine (2016) 1(6): 27-31 Neuromedicine www.jneurology.com www.jneurology.com Journal of Neurology & Neuromedicine Commentary Open Access Commentary: Current status of intratumoral therapy for glioblastoma Gregory D. Arnone1, Matt Wonais2, Andreas Linninger1,3, Ankit I. Mehta1* 1Department of Neurosurgery, University of Illinois at Chicago, Chicago, IL 60612, USA 2University of Illinois College of Medicine at Chicago, Chicago, IL 60612, USA 3Department of Bioengineering, The University of Illinois at Chicago, Chicago, IL, USA Glial tumors, particularly high grade glioma and glioblastoma Article Info Article Notes neuro-oncologists and neurosurgeons. Despite vigorous research Received: August 04, 2016 multiforme (GBM), continue to pose a significant challenge to Accepted: September 12, 2016 efforts over the past half-century, survival of GBM after maximal surgery, chemotherapy, and radiation therapy remains poor, *Correspondence: 1 Department of Neurosurgery reaching a median of 14-16 months after diagnosis . New therapies University of Illinois at Chicago have shown modest increases in median survival, but the majority of 912 S. Wood Street (MC-799) patients in these studies still die within two years after diagnosis2,3. Chicago, IL 60612 Telephone: 312-996-4842 Drug delivery to the central nervous system (CNS) poses unique Fax: 312-996-9018 challenges. Availability of drugs to cross the blood-brain-barrier Email: [email protected] (BBB), or more accurately termed blood-CNS barrier, have limited © 2016 Mehta AI. This article is distributed under the terms of the number, type, and doses of effective therapies against GBM. the Creative Commons Attribution 4.0 International License Various agents, including chemotherapeutics4, antiangiogenic 5,6 7,8 9 Keywords drugs , immunotoxins , viral vectors and gene therapy , among convection-enhanced delivery others, have been tried, all with suboptimal results. Nanoparticles Glioblastoma Strategies to circumvent the blood-CNS barrier have long been Localized drug delivery under investigation. Transient osmotic or mechanical disruption of Intratumoral therapy the blood-CNS barrier has been used in order to permit passage of chemotherapeutics into the CNS, though questions regarding lack of 10. Localized drug delivery into a postoperative tumor bed also dodges the blood-CNS issue without disruptionspecific targeting of normal and brainsafety homeostasis, persist and therefore remains an attractive option for delivery of therapeutics. While ability to cross the blood-CNS barrier has traditionally been critical in designing drugs for use in GBM, the opposite may prove to be most useful for a local delivery strategy. For example, platinum drugs were previously abandoned for treatment of glioma due to their poor passage across the blood-CNS barrier, but are now being re-explored for localized delivery. Adding to the fact that they are not a substrate for principle cross the blood-CNS barrier may now prove to be an advantage in prolongingefflux transporters the half-life in theand therapeuticbrain, the inability effect of theof these drug 11-13agents. to Gliadel wafers, a carmustine-imbedded polyanhydride co- polymer matrix implant, have perhaps been the most successful localized therapy and are FDA approved for newly diagnosed and recurrent glioblastoma, modestly extending overall survival of both groups14. Wafer implantation has drawbacks, however, with increased risk of complications such as surgical site breakdown, and cyst formation reported in some studies15, though not all16,17. Nonetheless,infection, cerebrospinal tumor recurrence fluid leak, is common brain edema, even after hydrocephalus, carmustine Page 27 of 31 Arnone GD, Wonais M, Linninger A, Mehta AI. J Neurol Neuromedicine (2016) 1(6): 27-31 Journal of Neurology & Neuromedicine wafer use, with median survival times of 16.4 and the ECS27. Consequently, CED has become among the most 9.7 months for newly diagnosed and recurrent GBM, popular vehicles for localized delivery in current time, with respectively. Histological analysis reveals that most tumor 52 studies regarding CED for gliomas published in the past recurrence in high-grade glioma is within 2-3 cm of the year (pubmed) and 5 ongoing clinical trials of convection tumor margin18,19, while drug penetration from surface enhanced delivery for glioma treatment (clinicaltrials.gov). erosion of the wafer is only within millimeters of the Unfortunately, most randomized trials related to CED implant20. Recent efforts in localized drug delivery have for glioma have been underwhelming28. Perhaps the most therefore focused on increasing local drug penetration notable failure of CED was the PRECISE trial, which did to surrounding brain parenchyma. To that effect, much work has been done regarding understanding the brain of cintredekin besudotox (CB) immunotoxin to gliadel extracellular matrix/space (ECS). not find any difference in survival when comparing29. A closeCED In addition to size of molecules, diffusion through the examination of that study reveals that inconsistent catheter ECS is governed by multiple factors, including ECS local placement,wafers in patients as well withas spread GBM ofat drugfirst recurrenceinto unwanted areas of low resistance, such as subarachnoid space and the to receptors, variable degree of dead space, extracellular matrixgeometry, molecules a net fixed negative(hyaluronan), charge, bindingand intratumoral of antigens of currently available image-guidance technology, these pressure gradients21. In order to promote diffusion of issuesventricles may may be havemitigated reduced in thefuture efficacy studies. of CED. Additionally, With use chemotherapeutics, nanocarriers such as liposomes and novel methods to track CED infusion of drugs may allow PEGylated nanoparticles have been developed. These real-time feedback of drug distribution. For example, surfaces both protect drugs from being prematurely superparamagnetic iron oxide nanoparticles detectable metabolized, as well as provide charge-shielding from the on T2 MRI imaging have recently been incorporated into ECS. In fact, studies on rat and human brain have shown brain penetrating nanoparticles, providing ability to that substances of 114 nm were able to diffuse through track therapeutic in the brain for up to one month after administration30. Radiolabeled therapeutics are also being local environment, as opposed to the previous size of 64 used to study the volume of distribution of drug after nmthe ECSwhen when unshielded protected22. Another from the important charge influence consideration of the CED, utilizing interval high-resolution positron emission when studying drug distribution in the brain parenchyma tomography (PET) scanning to evaluate biologically and interstitial space is the rate and mechanism of drug clearance from the brain. Recently, a novel, ubiquitous distributions31. Still, treatment failure of CED in the PRECISE pathway in the brain has been described that facilitates trialabsorbed may notdoses be fullyof radiationexplained inby suboptimalspecific anatomical catheter placement alone. In fact, a retrospective review of catheter (ISF), and the clearance of solutes from the interstitium. placement on patient outcomes in the PRECISE trial showed exchange of cerebrospinal fluid (CSF), interstitial fluid Designated the “glymphatic system”, this pathway consists that overall catheter placement scores did not correlate with local tumor control, progression free survival, or pathway that is dependent upon the astrocytic aquaporin-4 overall survival32 of a para-arterial CSF influx route, a trans-parenchymal (AQP4) water channel for astroglial water transport, also exists in CED, though catheter design has recently been 23. As . The additional issue of infusion backflow understanding of this pathway increases, researchers may technology, as well as variable injection rates33-36. Overall, and finally a para-venous ISF clearance route be able to better predict the clearance of drug from the theimproved pattern with of usedrug of dispersalmultiple ports,during backflow-reducing CED, especially interstitial space and therefore tailor dosing of local drug around inhomogeneous peritumoral tissue, has proven to depots. Additionally, knowledge of the glymphatic system be unpredictable37, but is steadily improving. Technology and its transport channels may ultimately help to develop development and mathematical modeling may play a novel methods to prolong the half-life of chemotherapeutics future role in determining optimal stereotactic catheter and nanocarriers. Another way to promote local drug penetration through environment38-40. Aside from the infusion and dispersal the ECS is by convection instead of relying only on bulk issues,placement, CED specific has an to additional each given disadvantage tumor and peritumoral related to diffusion alone. Indeed, convection-enhanced delivery connection of the catheter to an outside system, potentially (CED) of a variety of therapeutics has been a popular topic increasing chance of infection with prolonged or repeated of drug development and delivery for recurrent glioma in infusions. Additionally, in-hospital monitoring is required the past decade24-26. The CED technique utilizes a catheter during infusion, drastically limiting the effective time that that is connected to a pump delivering a pressure head drugs can be administered. Subcutaneous reservoirs of behind the infusion of drug. Studies
Recommended publications
  • Central Nervous System Tumors General ~1% of Tumors in Adults, but ~25% of Malignancies in Children (Only 2Nd to Leukemia)
    Last updated: 3/4/2021 Prepared by Kurt Schaberg Central Nervous System Tumors General ~1% of tumors in adults, but ~25% of malignancies in children (only 2nd to leukemia). Significant increase in incidence in primary brain tumors in elderly. Metastases to the brain far outnumber primary CNS tumors→ multiple cerebral tumors. One can develop a very good DDX by just location, age, and imaging. Differential Diagnosis by clinical information: Location Pediatric/Young Adult Older Adult Cerebral/ Ganglioglioma, DNET, PXA, Glioblastoma Multiforme (GBM) Supratentorial Ependymoma, AT/RT Infiltrating Astrocytoma (grades II-III), CNS Embryonal Neoplasms Oligodendroglioma, Metastases, Lymphoma, Infection Cerebellar/ PA, Medulloblastoma, Ependymoma, Metastases, Hemangioblastoma, Infratentorial/ Choroid plexus papilloma, AT/RT Choroid plexus papilloma, Subependymoma Fourth ventricle Brainstem PA, DMG Astrocytoma, Glioblastoma, DMG, Metastases Spinal cord Ependymoma, PA, DMG, MPE, Drop Ependymoma, Astrocytoma, DMG, MPE (filum), (intramedullary) metastases Paraganglioma (filum), Spinal cord Meningioma, Schwannoma, Schwannoma, Meningioma, (extramedullary) Metastases, Melanocytoma/melanoma Melanocytoma/melanoma, MPNST Spinal cord Bone tumor, Meningioma, Abscess, Herniated disk, Lymphoma, Abscess, (extradural) Vascular malformation, Metastases, Extra-axial/Dural/ Leukemia/lymphoma, Ewing Sarcoma, Meningioma, SFT, Metastases, Lymphoma, Leptomeningeal Rhabdomyosarcoma, Disseminated medulloblastoma, DLGNT, Sellar/infundibular Pituitary adenoma, Pituitary adenoma,
    [Show full text]
  • Inhibition of Mir-1193 Leads to Synthetic Lethality in Glioblastoma
    Zhang et al. Cell Death and Disease (2020) 11:602 https://doi.org/10.1038/s41419-020-02812-3 Cell Death & Disease ARTICLE Open Access Inhibition of miR-1193 leads to synthetic lethality in glioblastoma multiforme cells deficient of DNA-PKcs Jing Zhang1,LiJing1,SubeeTan2, Er-Ming Zeng3, Yingbo Lin4, Lingfeng He 1, Zhigang Hu 1, Jianping Liu1 and Zhigang Guo1 Abstract Glioblastoma multiforme (GBM) is the most malignant primary brain tumor and has the highest mortality rate among cancers and high resistance to radiation and cytotoxic chemotherapy. Although some targeted therapies can partially inhibit oncogenic mutation-driven proliferation of GBM cells, therapies harnessing synthetic lethality are ‘coincidental’ treatments with high effectiveness in cancers with gene mutations, such as GBM, which frequently exhibits DNA-PKcs mutation. By implementing a highly efficient high-throughput screening (HTS) platform using an in-house-constructed genome-wide human microRNA inhibitor library, we demonstrated that miR-1193 inhibition sensitized GBM tumor cells with DNA-PKcs deficiency. Furthermore, we found that miR-1193 directly targets YY1AP1, leading to subsequent inhibition of FEN1, an important factor in DNA damage repair. Inhibition of miR-1193 resulted in accumulation of DNA double-strand breaks and thus increased genomic instability. RPA-coated ssDNA structures enhanced ATR checkpoint kinase activity, subsequently activating the CHK1/p53/apoptosis axis. These data provide a preclinical theory for the application of miR-1193 inhibition as a potential synthetic lethal approach targeting GBM cancer cells with DNA-PKcs fi 1234567890():,; 1234567890():,; 1234567890():,; 1234567890():,; de ciency. Introduction In response to DNA damage, cells activate the DNA Glioblastoma multiforme (GBM), exhibits highly damage response (DDR) network, allowing DNA repair aggressive invasion, a high mortality rate, and high resis- through the regulation of cell-cycle progression, DNA tance to radiation and cytotoxic chemotherapy, and thus damage repair or apoptosis4.
    [Show full text]
  • Meningioma ACKNOWLEDGEMENTS
    AMERICAN BRAIN TUMOR ASSOCIATION Meningioma ACKNOWLEDGEMENTS ABOUT THE AMERICAN BRAIN TUMOR ASSOCIATION Meningioma Founded in 1973, the American Brain Tumor Association (ABTA) was the first national nonprofit advocacy organization dedicated solely to brain tumor research. For nearly 45 years, the ABTA has been providing comprehensive resources that support the complex needs of brain tumor patients and caregivers, as well as the critical funding of research in the pursuit of breakthroughs in brain tumor diagnosis, treatment and care. To learn more about the ABTA, visit www.abta.org. We gratefully acknowledge Santosh Kesari, MD, PhD, FANA, FAAN chair of department of translational neuro- oncology and neurotherapeutics, and Marlon Saria, MSN, RN, AOCNS®, FAAN clinical nurse specialist, John Wayne Cancer Institute at Providence Saint John’s Health Center, Santa Monica, CA; and Albert Lai, MD, PhD, assistant clinical professor, Adult Brain Tumors, UCLA Neuro-Oncology Program, for their review of this edition of this publication. This publication is not intended as a substitute for professional medical advice and does not provide advice on treatments or conditions for individual patients. All health and treatment decisions must be made in consultation with your physician(s), utilizing your specific medical information. Inclusion in this publication is not a recommendation of any product, treatment, physician or hospital. COPYRIGHT © 2017 ABTA REPRODUCTION WITHOUT PRIOR WRITTEN PERMISSION IS PROHIBITED AMERICAN BRAIN TUMOR ASSOCIATION Meningioma INTRODUCTION Although meningiomas are considered a type of primary brain tumor, they do not grow from brain tissue itself, but instead arise from the meninges, three thin layers of tissue covering the brain and spinal cord.
    [Show full text]
  • The CNS and the Brain Tumor Microenvironment: Implications for Glioblastoma Immunotherapy
    International Journal of Molecular Sciences Review The CNS and the Brain Tumor Microenvironment: Implications for Glioblastoma Immunotherapy Fiona A. Desland and Adília Hormigo * Icahn School of Medicine at Mount Sinai, The Tisch Cancer Institute, Department of Neurology, Box 1137, 1 Gustave L. Levy Pl, New York, NY 10029-6574, USA; fi[email protected] * Correspondence: [email protected] Received: 28 August 2020; Accepted: 29 September 2020; Published: 5 October 2020 Abstract: Glioblastoma (GBM) is the most common and aggressive malignant primary brain tumor in adults. Its aggressive nature is attributed partly to its deeply invasive margins, its molecular and cellular heterogeneity, and uniquely tolerant site of origin—the brain. The immunosuppressive central nervous system (CNS) and GBM microenvironments are significant obstacles to generating an effective and long-lasting anti-tumoral response, as evidenced by this tumor’s reduced rate of treatment response and high probability of recurrence. Immunotherapy has revolutionized patients’ outcomes across many cancers and may open new avenues for patients with GBM. There is now a range of immunotherapeutic strategies being tested in patients with GBM that target both the innate and adaptive immune compartment. These strategies include antibodies that re-educate tumor macrophages, vaccines that introduce tumor-specific dendritic cells, checkpoint molecule inhibition, engineered T cells, and proteins that help T cells engage directly with tumor cells. Despite this, there is still much ground to be gained in improving the response rates of the various immunotherapies currently being trialed. Through historical and contemporary studies, we examine the fundamentals of CNS immunity that shape how to approach immune modulation in GBM, including the now revamped concept of CNS privilege.
    [Show full text]
  • Rehabilitation of Adult Patients with Primary Brain Tumors: a Narrative Review
    brain sciences Review Rehabilitation of Adult Patients with Primary Brain Tumors: A Narrative Review Parth Thakkar 1, Brian D. Greenwald 2,* and Palak Patel 1 1 Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA; [email protected] (P.T.); [email protected] (P.P.) 2 JFK Johnson Rehabilitation Institute, Edison, NJ 08820, USA * Correspondence: [email protected] Received: 22 May 2020; Accepted: 25 July 2020; Published: 29 July 2020 Abstract: Rehabilitative measures have been shown to benefit patients with primary brain tumors (PBT). To provide a high quality of care, clinicians should be aware of common challenges in this population including a variety of medical complications, symptoms, and impairments, such as headaches, seizures, cognitive deficits, fatigue, and mood changes. By taking communication and family training into consideration, clinicians can provide integrated and patient-centered care to this population. This article looks to review the current literature in outpatient and inpatient rehabilitation options for adult patients with PBTs as well as explore the role of the interdisciplinary team in providing survivorship care. Keywords: primary brain tumor; neuro-oncology; brain cancer; rehabilitation 1. Introduction About 80,000 people in the United States are diagnosed with a primary brain tumor (PBT) every year [1]. PBTs vary widely in their clinical presentations and can carry devastating prognoses for patients. The average survival rate for patients with malignant brain tumors is 35%, and the five-year survival rate for glioblastoma multiforme, the most common form of primary malignant brain tumor in adults, is 5.6%. There are an estimated 700,000 Americans living with a PBT, of which approximately 70% are benign and 30% are malignant [2].
    [Show full text]
  • Information Sheet 4: High-Grade Glioma Brain Tumours and Treatment Options
    Information Sheet 4: High-Grade Glioma brain tumours and treatment options This information document on high grade brain tumours simply provides a basis for discussion with the healthcare professionals with whom you are in contact. It should not in any way be used as a substitute for professional care. What is a Glioma? There are nearly 100 different types of brain tumours. Most primary brain tumours develop from the cells that support the nerve cells of the brain. These are called glial cells. So a tumour of glial cells is given the collective name of a glioma. About half of all primary brain tumours are gliomas. Low-grade or high-grade? Brain tumours are put into groups according to how fast they are likely to grow. There are four groups, called Grades 1–4. The cells are examined under a microscope, and the more normal they look, the more slowly the brain tumour is likely to develop and the lower the grade. The more abnormal the cells look, the more quickly the brain tumour is likely to grow and the higher the grade. You may have been told you have a benign tumour or a malignant tumour. Low-grade tumours are generally regarded as benign and high-grade as malignant. Malignant So, generally speaking, malignant means that: The tumour is relatively fast growing It may come back after surgery, even if completely removed It may spread to other parts of the brain or spinal cord It cannot just be treated with surgery and will need further treatments, such as radiotherapy or chemotherapy, to try to stop it from returning.
    [Show full text]
  • Neurofibromatosis Type 1 and Type 2 Associated Tumours: Current Trends in Diagnosis and Management with a Focus on Novel Medical Therapies
    Neurofibromatosis Type 1 and Type 2 Associated Tumours: Current trends in Diagnosis and Management with a focus on Novel Medical Therapies Simone Lisa Ardern-Holmes MBChB, MSc, FRACP A thesis submitted in fulfilment of the requirements for the degree of Doctor of Philosophy Faculty of Health Sciences, The University of Sydney February 2018 1 STATEMENT OF ORIGINALITY This is to certify that this submission is my own work and that, to the best of my knowledge, it contains no material previously published or written by another person, or material which to a substantial extent has been accepted for the award of any other degree or diploma of the university or other institute of higher learning, except where due acknowledgement has been made in the text. Simone L. Ardern-Holmes 2 SUMMARY Neurofibromatosis type 1 (NF1) and Neurofibromatosis type 2 (NF2) are distinct single gene disorders, which share a predisposition to formation of benign nervous system tumours due to loss of tumour suppressor function. Since identification of the genes encoding NF1 and NF2 in the early 1990s, significant progress has been made in understanding the biological processes and molecular pathways underlying tumour formation. As a result, identifying safe and effective medical approaches to treating NF1 and NF2-associated tumours has become a focus of clinical research and patient care in recent years. This thesis presents a comprehensive discussion of the complications of NF1 and NF2 and approaches to treatment, with a focus on key tumours in each condition. The significant functional impact of these disorders in children and young adults is illustrated, demonstrating the need for coordinated care from experienced multidisciplinary teams.
    [Show full text]
  • Proceedings from the 2009 NF Conference: New Frontiers Joseph L
    CONFERENCE REPORT What’s New in Neurofibromatosis? Proceedings From the 2009 NF Conference: New Frontiers Joseph L. Kissil,1 Jaishri O. Blakeley,2 Rosalie E. Ferner,3 Susan M. Huson,4 Michel Kalamarides,5 Victor-Felix Mautner,6 Frank McCormick,7 Helen Morrison,8 Roger Packer,9 Vijaya Ramesh,10 Nancy Ratner,11 Katherine A. Rauen,7 David A. Stevenson,12 Kim Hunter-Schaedle,13* and Kathryn North14 1The Wistar Institute, Philadelphia, Pennsylvania 2Johns Hopkins University, Baltimore, Maryland 3Guy’s and St Thomas’!NHS Trust, London, United Kingdom 4St. Mary’s Hospital, University of Manchester, Manchester, United Kingdom 5Hopital Beaujon, APHP, Clichy, France and Inserm U674 6University Eppendorf, Hamburg, Germany 7University of California, San Francisco, California 8Leibniz Institute for Age Research, Jena, Germany 9Children’s National Medical Center, Washington, District of Columbia 10Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 11Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio 12University of Utah, Salt Lake City, Utah 13Children’s Tumor Foundation, New York, New York 14University of Sydney and The Children’s Hospital at Westmead, Sydney, Australia Received 30 September 2009; Accepted 17 October 2009 The NF Conference is the largest annual gathering of researchers and clinicians focused on neurofibromatosis and has been con- How to Cite this Article: vened by the Children’s Tumor Foundation for over 20 years. The Kissil JL, Blakeley JO, Ferner RE, Huson SM, 2009 NF Conference was held in Portland, Oregon from June 13 to Kalamarides M, Mautner V-F, McCormick F, June 16, 2009 andco-chaired by Kathryn North from the University Morrison H, Packer R, Ramesh V, Ratner N, of Sydney and The Children’s Hospital at Westmead, Sydney, Rauen KA, Stevenson DA, Hunter-Schaedle Australia; and Joseph Kissil from the Wistar Institute, Philadel- K, North K.
    [Show full text]
  • Clinical and Genetic Patterns Ofneurofibromatosis 1 and 2
    662 BritishJournalofOphthalmology 1993; 77: 662-672 PERSPECTIVE Br J Ophthalmol: first published as 10.1136/bjo.77.10.662 on 1 October 1993. Downloaded from Clinical and genetic patterns of neurofibromatosis 1 and 2 Nicola K Ragge General introduction to the neurofibromatoses Neurofibromatosis type 1 The diseases traditionally known as neurofibromatosis have now been formally separated into two types: neurofibromato- CLINICAL ASPECTS sis type 1 or NFl (the type described by von Recklinghausen) Neurofibromatosis type 1 (NFI) is the commonest form of and neurofibromatosis type 2 or NF2 (a much rarer form).' It neurofibromatosis and has a frequency of about 1 in 3000 to is now recognised that although they have overlapping 1 in 3500.192° Although the gene is almost 100% penetrant, features, including an inherited propensity to neurofibromas the disease itselfhas extremely variable expressivity.20 About and tumours of the central nervous system, they are indeed 50% ofindex cases and 30% ofall NFl cases are considered to separate diseases and map to different chromosomes - 17 for be new mutations. The high mutation rate may be due in part NFl and 22 for NF2. Furthermore, developmental abnor- to the large size of the gene and its transcript, or possibly to malities such as hamartomas occur in both types of neuro- the presence of sequences within the gene highly susceptible fibromatosis, illustrating a need to define the role of the to mutation. normal NF genes in development. There may also be further forms of neurofibromatosis, including NF3, NF4, multiple meningiomatosis, and spinal Clinicalfeatures schwannomatosis, that do not fit precisely into current Particular clinical features are critical for establishing the diagnostic classifications.
    [Show full text]
  • Current FDA-Approved Therapies for High-Grade Malignant Gliomas
    biomedicines Review Current FDA-Approved Therapies for High-Grade Malignant Gliomas Jacob P. Fisher 1,* and David C. Adamson 2,3 1 Division of Biochemistry, Southern Virginia University, Buena Vista, VA 24416, USA 2 Department of Neurosurgery, School of Medicine, Emory University, Atlanta, GA 30322, USA; [email protected] 3 Atlanta VA Healthcare System, Decatur, GA 30033, USA * Correspondence: Jacob.fi[email protected] Abstract: The standard of care (SOC) for high-grade gliomas (HGG) is maximally safe surgical resection, followed by concurrent radiation therapy (RT) and temozolomide (TMZ) for 6 weeks, then adjuvant TMZ for 6 months. Before this SOC was established, glioblastoma (GBM) patients typically lived for less than one year after diagnosis, and no adjuvant chemotherapy had demonstrated significant survival benefits compared with radiation alone. In 2005, the Stupp et al. randomized controlled trial (RCT) on newly diagnosed GBM patients concluded that RT plus TMZ compared to RT alone significantly improved overall survival (OS) (14.6 vs. 12.1 months) and progression-free survival (PFS) at 6 months (PFS6) (53.9% vs. 36.4%). Outside of TMZ, there are four drugs and one device FDA-approved for the treatment of HGGs: lomustine, intravenous carmustine, carmustine wafer implants, bevacizumab (BVZ), and tumor treatment fields (TTFields). These treatments are now mainly used to treat recurrent HGGs and symptoms. TTFields is the only treatment that has been shown to improve OS (20.5 vs. 15.6 months) and PFS6 (56% vs. 37%) in comparison to the current SOC. TTFields is the newest addition to this list of FDA-approved treatments, but has not been universally accepted yet as part of SOC.
    [Show full text]
  • Current Understanding of Neurofibromatosis Type 1, 2, And
    International Journal of Molecular Sciences Review Current Understanding of Neurofibromatosis Type 1, 2, and Schwannomatosis Ryota Tamura Department of Neurosurgery, Kawasaki Municipal Hospital, Shinkawadori, Kanagawa, Kawasaki-ku 210-0013, Japan; [email protected] Abstract: Neurofibromatosis (NF) is a neurocutaneous syndrome characterized by the development of tumors of the central or peripheral nervous system including the brain, spinal cord, organs, skin, and bones. There are three types of NF: NF1 accounting for 96% of all cases, NF2 in 3%, and schwannomatosis (SWN) in <1%. The NF1 gene is located on chromosome 17q11.2, which encodes for a tumor suppressor protein, neurofibromin, that functions as a negative regulator of Ras/MAPK and PI3K/mTOR signaling pathways. The NF2 gene is identified on chromosome 22q12, which encodes for merlin, a tumor suppressor protein related to ezrin-radixin-moesin that modulates the activity of PI3K/AKT, Raf/MEK/ERK, and mTOR signaling pathways. In contrast, molecular insights on the different forms of SWN remain unclear. Inactivating mutations in the tumor suppressor genes SMARCB1 and LZTR1 are considered responsible for a majority of cases. Recently, treatment strategies to target specific genetic or molecular events involved in their tumorigenesis are developed. This study discusses molecular pathways and related targeted therapies for NF1, NF2, and SWN and reviews recent clinical trials which involve NF patients. Keywords: neurofibromatosis type 1; neurofibromatosis type 2; schwannomatosis; molecular tar- geted therapy; clinical trial Citation: Tamura, R. Current Understanding of Neurofibromatosis Int. Type 1, 2, and Schwannomatosis. 1. Introduction J. Mol. Sci. 2021, 22, 5850. https:// doi.org/10.3390/ijms22115850 Neurofibromatosis (NF) is a genetic disorder that causes multiple tumors on nerve tissues, including brain, spinal cord, and peripheral nerves [1–3].
    [Show full text]
  • Genetic Testing for Neurofibromatosis and Related Disorders
    Corporate Medical Policy Genetic Testing for Neurofibromatosis and Related Disorders AHS – M2134 File Name: genetic_testing_for_neurofibromatosis_and_related_disorders Origination: 01/2019 Last CAPReview: 07/2021 Next CAP Review: 07/2022 Last Review: 07/2021 Description of Procedure or Service Neurofibromatoses are a group of three clinically and genetically distinct disorders that cause tumors to form on nerve tissue. Neurofibromatosis type 1 (NF1) is caused by autosomal dominant mutations in the neurofibromin (NF1) gene and is characterized by multiple café-au-lait macules and neurofibromas (Korf, 2021). Neurofibromatosis type (NF2) is caused by autosomal dominant mutations in the merlin, also known as schwannomin, (NF2) gene, and is characterized by multiple tumors of the nervous system, including the more common bilateral vestibular schwannomas as well as intracranial and spinal meningiomas, intrinsic ependymomas, and other spine tumors (Evans, 2020). Schwannomatosis is caused by inactivating mutations in SMARCB1 and LZTR and is characterized by multiple schwannomas and pain arising in adulthood (Yohay & Bergner, 2019). Legius syndrome is an NF1-like disorder caused by autosomal dominant mutations in the sprout-related EVH1 [enabled/vasodilator-stimulated phosphoprotein homology 1] domain-containing protein 1 (SPRED1) gene, resulting in café-au-lait macules. Constitutional mismatch repair-deficiency syndrome (CMMR-D), caused by mutations in mismatch repair genes, can also result in café-au-lait macules, axillary freckling, and Lisch nodules similar to NF1; however, unlike NF1, CMMR-D can also result in a variety of different malignancies, including glioblastoma and colorectal cancer (Korf, 2021). Related Policies Prenatal Screening AHS-G2035 ***Note: This Medical Policy is complex and technical. For questions concerning the technical language and/or specific clinical indications for its use, please consult your physician.
    [Show full text]