1
|
Vittay O, Christopher J, Mehta SG, Toms AP. Genetic basis and imaging findings of neurofibromatosis 1 and other somatic overgrowth disorders. Skeletal Radiol 2025; 54:915-923. [PMID: 39254838 PMCID: PMC11953187 DOI: 10.1007/s00256-024-04772-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 08/02/2024] [Accepted: 08/02/2024] [Indexed: 09/11/2024]
Abstract
Somatic overgrowth disorders comprise a wide range of rare conditions that present with focal enlargement of one or more tissue types. The PI3K-AKT-mTOR pathway is a signalling pathway that induces angiogenesis and cell proliferation, and is one of the most commonly overactivated signalling pathways in cancer. The PI3K-AKT-mTOR pathway can be up-regulated by genetic variants that code for proteins in this pathway, or down-regulated by proteins that inhibit the pathway. Mosaic genetic variations can result in cells that proliferate excessively in specific anatomical locations. The PIK3CA-related overgrowth spectrum (PROS) disorders include CLOVES syndrome, macrodystrophia lipomatosa, and Klippel-Trenaunay syndrome among many. The neurofibromatosis type 1 (NF1) gene encodes neurofibromin which down-regulates the PI3K-AKT-mTOR pathway. Thousands of pathological variants in the NF1 gene have been described which can result in lower-than-normal levels of neurofibromin and therefore up-regulation of the PI3K-AKT-mTOR pathway promoting cellular overgrowth. Somatic overgrowth is a rare presentation in NF1 with a wide range of clinical and radiological presentations. Hypertrophy of all ectodermal and mesodermal elements has been described in NF1 including bone, muscle, fat, nerve, lymphatics, arteries and veins, and skin. The shared signalling pathway for cellular overgrowth means that these radiological appearances can overlap with other conditions in the PIK3CA-related overgrowth spectrum. The aim of this review is to describe the genetic basis for the radiological features of NF1 and in particular compare the appearances of the somatic overgrowth disorders in NF1 with other conditions in the PIK3CA-related overgrowth spectrum.
Collapse
Affiliation(s)
- Orsolya Vittay
- Department of Radiology, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 2QQ, UK
| | - Joseph Christopher
- Department of Clinical Genetics, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 2QQ, UK
- Academic Department of Medical Genetics, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Sarju G Mehta
- Department of Clinical Genetics, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 2QQ, UK
| | - Andoni P Toms
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norfolk, NR4 7TJ, UK.
- Department of Radiology, Norfolk & Norwich University Hospital, Colney Lane, Norwich, NR4 7UB, UK.
| |
Collapse
|
2
|
Weber CAM, Krönke N, Volk V, Auber B, Förster A, Trost D, Geffers R, Esmaeilzadeh M, Lalk M, Nabavi A, Samii A, Krauss JK, Feuerhake F, Hartmann C, Wiese B, Brand F, Weber RG. Rare germline variants in POLE and POLD1 encoding the catalytic subunits of DNA polymerases ε and δ in glioma families. Acta Neuropathol Commun 2023; 11:184. [PMID: 37990341 PMCID: PMC10664377 DOI: 10.1186/s40478-023-01689-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/11/2023] [Indexed: 11/23/2023] Open
Abstract
Pathogenic germline variants in the DNA polymerase genes POLE and POLD1 cause polymerase proofreading-associated polyposis, a dominantly inherited disorder with increased risk of colorectal carcinomas and other tumors. POLE/POLD1 variants may result in high somatic mutation and neoantigen loads that confer susceptibility to immune checkpoint inhibitors (ICIs). To explore the role of POLE/POLD1 germline variants in glioma predisposition, whole-exome sequencing was applied to leukocyte DNA of glioma patients from 61 tumor families with at least one glioma case each. Rare heterozygous POLE/POLD1 missense variants predicted to be deleterious were identified in glioma patients from 10 (16%) families, co-segregating with the tumor phenotype in families with available DNA from several tumor patients. Glioblastoma patients carrying rare POLE variants had a mean overall survival of 21 months. Additionally, germline variants in POLD1, located at 19q13.33, were detected in 2/34 (6%) patients with 1p/19q-codeleted oligodendrogliomas, while POLE variants were identified in 2/4 (50%) glioblastoma patients with a spinal metastasis. In 13/15 (87%) gliomas from patients carrying POLE/POLD1 variants, features of defective polymerase proofreading, e.g. hypermutation, POLE/POLD1-associated mutational signatures, multinucleated cells, and increased intratumoral T cell response, were observed. In a CRISPR/Cas9-derived POLE-deficient LN-229 glioblastoma cell clone, a mutator phenotype and delayed S phase progression were detected compared to wildtype POLE cells. Our data provide evidence that rare POLE/POLD1 germline variants predispose to gliomas that may be susceptible to ICIs. Data compiled here suggest that glioma patients carrying POLE/POLD1 variants may be recognized by cutaneous manifestations, e.g. café-au-lait macules, and benefit from surveillance colonoscopy.
Collapse
Affiliation(s)
- Christine A M Weber
- Department of Human Genetics, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Nicole Krönke
- Department of Neuropathology, Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Valery Volk
- Department of Neuropathology, Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Bernd Auber
- Department of Human Genetics, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Alisa Förster
- Department of Human Genetics, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | | | - Robert Geffers
- Genome Analytics Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | - Michael Lalk
- Department of Neurosurgery, KRH Klinikum Nordstadt, Hannover, Germany
| | - Arya Nabavi
- Department of Neurosurgery, KRH Klinikum Nordstadt, Hannover, Germany
| | - Amir Samii
- Department of Neurosurgery, International Neuroscience Institute, Hannover, Germany
| | - Joachim K Krauss
- Department of Neurosurgery, Hannover Medical School, Hannover, Germany
| | - Friedrich Feuerhake
- Department of Neuropathology, Institute of Pathology, Hannover Medical School, Hannover, Germany
- Institute for Neuropathology, University Clinic Freiburg, Freiburg, Germany
| | - Christian Hartmann
- Department of Neuropathology, Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Bettina Wiese
- Department of Neurosurgery, Hannover Medical School, Hannover, Germany
- Department of Neurology, Henriettenstift, Diakovere Krankenhaus gGmbH, Hannover, Germany
| | - Frank Brand
- Department of Human Genetics, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Ruthild G Weber
- Department of Human Genetics, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
3
|
Novel treatments for autism spectrum disorder based on genomics and systems biology. Pharmacol Ther 2021; 230:107939. [PMID: 34174273 DOI: 10.1016/j.pharmthera.2021.107939] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 06/03/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a highly heterogeneous neurodevelopmental disorder with a complex underlying genetic architecture. There are currently no known pharmacologic treatments for the core ASD symptoms of social deficits and restricted/ repetitive behavior. However, there are dozens of clinical trials currently underway that are testing the impact of novel and existing agents on core and associated symptoms in ASD. METHODS We present a narrative synthesis of the historical and contemporary challenges to drug discovery in ASD. We then provide an overview of novel treatments currently under investigation from a genomics and systems biology perspective. RESULTS Data driven network and cluster analyses suggest alterations in transcriptional regulation, chromatin remodelling, synaptic transmission, neuropeptide signalling, and/or immunological mechanisms may contribute to or underlie the development of ASD. Agents and upcoming trials targeting each of the above listed systems are reviewed. CONCLUSION Identifying effective pharmacologic treatments for the core and associated symptom domains in ASD will require further collaboration and innovation in the areas of outcome measurement, biomarker research, and genomics, as well as systematic efforts to identify and treat subgroups of individuals with ASD who may be differentially responsive to specific treatments.
Collapse
|
4
|
Gropman AL. Epigenetics and pervasive developmental disorders. EPIGENETICS IN PSYCHIATRY 2021:519-552. [DOI: 10.1016/b978-0-12-823577-5.00011-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
5
|
Löscher W, Klein P. The Pharmacology and Clinical Efficacy of Antiseizure Medications: From Bromide Salts to Cenobamate and Beyond. CNS Drugs 2021; 35:935-963. [PMID: 34145528 PMCID: PMC8408078 DOI: 10.1007/s40263-021-00827-8] [Citation(s) in RCA: 155] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/04/2021] [Indexed: 12/16/2022]
Abstract
Epilepsy is one of the most common and disabling chronic neurological disorders. Antiseizure medications (ASMs), previously referred to as anticonvulsant or antiepileptic drugs, are the mainstay of symptomatic epilepsy treatment. Epilepsy is a multifaceted complex disease and so is its treatment. Currently, about 30 ASMs are available for epilepsy therapy. Furthermore, several ASMs are approved therapies in nonepileptic conditions, including neuropathic pain, migraine, bipolar disorder, and generalized anxiety disorder. Because of this wide spectrum of therapeutic activity, ASMs are among the most often prescribed centrally active agents. Most ASMs act by modulation of voltage-gated ion channels; by enhancement of gamma aminobutyric acid-mediated inhibition; through interactions with elements of the synaptic release machinery; by blockade of ionotropic glutamate receptors; or by combinations of these mechanisms. Because of differences in their mechanisms of action, most ASMs do not suppress all types of seizures, so appropriate treatment choices are important. The goal of epilepsy therapy is the complete elimination of seizures; however, this is not achievable in about one-third of patients. Both in vivo and in vitro models of seizures and epilepsy are used to discover ASMs that are more effective in patients with continued drug-resistant seizures. Furthermore, therapies that are specific to epilepsy etiology are being developed. Currently, ~ 30 new compounds with diverse antiseizure mechanisms are in the preclinical or clinical drug development pipeline. Moreover, therapies with potential antiepileptogenic or disease-modifying effects are in preclinical and clinical development. Overall, the world of epilepsy therapy development is changing and evolving in many exciting and important ways. However, while new epilepsy therapies are developed, knowledge of the pharmacokinetics, antiseizure efficacy and spectrum, and adverse effect profiles of currently used ASMs is an essential component of treating epilepsy successfully and maintaining a high quality of life for every patient, particularly those receiving polypharmacy for drug-resistant seizures.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Bünteweg 17, 30559, Hannover, Germany. .,Center for Systems Neuroscience, Hannover, Germany.
| | - Pavel Klein
- grid.429576.bMid-Atlantic Epilepsy and Sleep Center, Bethesda, MD USA
| |
Collapse
|
6
|
Sabetghadam A, Wu C, Liu J, Zhang L, Reid AY. Increased epileptogenicity in a mouse model of neurofibromatosis type 1. Exp Neurol 2020; 331:113373. [PMID: 32502580 DOI: 10.1016/j.expneurol.2020.113373] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 05/22/2020] [Accepted: 06/01/2020] [Indexed: 11/19/2022]
Abstract
RATIONALE Neurofibromatosis type 1 (NF1) is associated with higher rates of epilepsy compared to the general population. Some NF1 patients with epilepsy do not have intracranial lesions, suggesting the genetic mutation itself may contribute to higher rates of epilepsy in these patients. We have recently demonstrated increased seizure susceptibility in the Nf1+/- mouse, but it is unknown whether this model displays altered epileptogenicity, as has been reported in patients with NF1. The aim of this study was to determine whether the Nf1+/- mouse is more susceptible to electrical kindling-induced epileptogenesis. METHODS Young male or female adult Nf1+/- or Nf1+/+ (wild-type; WT) mice were implanted with electrodes for neocortical or hippocampal kindling paradigms. Neocortical kindling was performed for 40 stimulation sessions followed by baseline EEG monitoring to detect possible SRSs. Hippocampal kindling was performed with a modified extended kindling paradigm, completed to a maximum of 80 sessions to try to induce spontaneous repetitive seizures (SRSs). Western blot assays were performed in naïve and kindled mice to compare levels of Akt and MAPK (ERK1/2), proteins downstream of the NF1 mutation. RESULTS The average initial neocortical after-discharge threshold (ADT) was significantly lower in the Nf1+/- group, which also required fewer stimulations to reach stage 5 seizure, had greater average seizure severity across all kindling sessions, had a greater number of convulsive seizures, and had a faster progression of after-discharge duration and Racine score during kindling. No WT mice exhibited SRS after neocortical kindling, versus 33% of Nf1+/- mice. The average initial hippocampal ADT was not significantly different between the WT and Nf1+/- groups, nor was there a difference in the number of stimulations required to reach the kindled state. The WT group had a significantly higher average seizure severity across all kindling sessions as compared with the Nf1+/- mice. The WT group also had faster progression of the Racine seizure score over the kindling sessions, mainly due to a faster increase in seizures severity early during the kindling process. However, SRSs were seen in 50% of Nf1+/- mice after modified extended kindling and in no WT mice. Western blots showed hippocampal kindling increased the ratio of phosphorylated/total Akt in both the WT and Nf1+/- mice, while neocortical kindling led to increased ratios of phosphorylated/total Akt and MAPK in Nf1+/- mice only. CONCLUSIONS We have demonstrated for the first time an increased rate of epileptogenesis in an animal model of NF1 with no known macroscopic/neoplastic brain lesions. This work provides evidence for the genetic mutation itself playing a role in seizures and epilepsy in patients with NF1, and supports the use of the Nf1+/- mouse model in future mechanistic studies.
Collapse
Affiliation(s)
- A Sabetghadam
- Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, Ontario M5T 0S8, Canada.
| | - C Wu
- Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, Ontario M5T 0S8, Canada
| | - J Liu
- Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, Ontario M5T 0S8, Canada
| | - L Zhang
- Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, Ontario M5T 0S8, Canada; Department of Medicine (Neurology), University of Toronto, Toronto, Ontario, Canada
| | - A Y Reid
- Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, Ontario M5T 0S8, Canada; Department of Medicine (Neurology), University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Slopis JM, Arevalo O, Bell CS, Hebert AA, Northrup H, Riascos RF, Samuels JA, Smith KC, Tate P, Koenig MK. Treatment of Disfiguring Cutaneous Lesions in Neurofibromatosis-1 with Everolimus: A Phase II, Open-Label, Single-Arm Trial. Drugs R D 2019; 18:295-302. [PMID: 30284154 PMCID: PMC6277319 DOI: 10.1007/s40268-018-0248-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Cutaneous neurofibromas cause disfigurement and discomfort in individuals with neurofibromatosis type 1 (NF-1). Methods The primary objective of this phase II, open-label, single-arm trial was to assess whether orally administered everolimus reduced the surface volume of cutaneous neurofibromas in patients with NF-1. Results Of 22 patients who took the study drug, 17 completed the trial; 5 patients withdrew due to adverse events. Sixteen patients had photographs of sufficient quality for assessment of the primary outcome. A significant reduction in lesion surface volume, defined as an end of trial volume > 2 standard errors (SE) less than baseline volume, was observed for 4/31 lesions (13%) from 3/16 patients (19%). Additionally, a statistically significant absolute change in average height for paired lesions was observed (p = 0.048). Although not a prespecified outcome measure, a dramatic reduction in the size of 3 large plexiform neurofibromas with a cutaneous component was also noted and documented by measurement of maximum circumference or magnetic resonance imaging-based volumetric analysis. Adverse events were common in this trial, but no serious adverse events occurred. Conclusions Although this was a small, exploratory trial that was not powered for significance, the reduction in surface volume observed in this study is noteworthy assuming that the natural course for untreated lesions is to maintain or increase in volume. Future studies are needed with larger study populations that incorporate longer durations of treatment and better standardization of volumetric measurements. Trial Registration ClinicalTrials.gov Identifier: NCT02332902 Electronic supplementary material The online version of this article (10.1007/s40268-018-0248-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- John M Slopis
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Octavio Arevalo
- Department of Diagnostic and Interventional Imaging, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Cynthia S Bell
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Adelaide A Hebert
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.,Department of Dermatology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Hope Northrup
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Roy F Riascos
- Department of Diagnostic and Interventional Imaging, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Joshua A Samuels
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Keri C Smith
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Patti Tate
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Mary Kay Koenig
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA. .,Department of Pediatrics, Division of Child and Adolescent Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, 6410 Fannin Street, UTPB 732, Houston, TX, 77030, USA.
| |
Collapse
|
8
|
Abstract
Neurofibromatosis type 1 and type 2, affecting both children and adults, often results in devastating complications. The rapid unravelling of the genetic underpinnings of these unique disorders has led to the development of novel therapies, especially molecular-targeted therapies. To facilitate clinical trial development, the Neurofibromatosis Clinical Trial Consortium (NFCTC) was established in 2006 by the Department of Defense. Over the past decade, the Consortium has successfully completed studies for children and adults with neurofibromatosis type 1 and plexiform neurofibromas, neurocognitive challenges, low-grade gliomas, and malignant peripheral nerve sheath tumors. In addition, a study for children and adults with neurofibromatosis type 2 and acoustic schwannomas is near completion. The NFCTC has now been expanded to 19 sites in the United Stated and Australia. Mechanisms have been put in place to work closely with other consortia, foundations, and industry to expeditiously translate preclinical discoveries into clinical trials.
Collapse
Affiliation(s)
- Roger J Packer
- 1 From the Center for Neuroscience and Behavioral Medicine, Children's National Health System, Washington, DC, USA.,2 Gilbert Neurofibromatosis Family Institute, Children's National Health System, Washington, DC, USA.,3 Brain Tumor Institute, Children's National Health System, Washington, DC, USA
| | - Michael J Fisher
- 4 Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Gary Cutter
- 5 University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Bruce R Korf
- 5 University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
9
|
Archer TC, Sengupta S, Pomeroy SL. Brain cancer genomics and epigenomics. HANDBOOK OF CLINICAL NEUROLOGY 2018; 148:785-797. [PMID: 29478614 DOI: 10.1016/b978-0-444-64076-5.00050-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Classically, brain cancers have been graded and diagnosed based on histology and risk stratified by clinical criteria. Recent advances in genomics and epigenomics have ushered in an era of defining cancers based on molecular criteria. These advances have increased our precision of identifying oncogenic driving events and, most importantly, increased our precision at predicting clinical outcome. For the first time in its history, the 2016 revision of the WHO Classification of Tumors of the Central Nervous System included molecular features as tumor classification criteria. Brain tumors can develop in the context of genetic cancer predisposition syndromes, such as Li-Fraumeni or Gorlin syndrome, but by far most commonly arise through the acquisition of somatic mutations and chromosome changes in the malignant cells. By taking a survey across this cancer landscape, certain themes emerge as being common events to drive cancer: DNA damage repair, genomic instability, mechanistic target of rapamycin pathway, sonic hedgehog pathway, hypoxia, and epigenetic dysfunction. Understanding these mechanisms is of paramount importance for improving targeted therapies, and for identifying the right patients for those therapies.
Collapse
Affiliation(s)
- Tenley C Archer
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States; Broad Institute of Harvard and MIT, Cambridge, MA, United States
| | - Soma Sengupta
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, United States
| | - Scott L Pomeroy
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States; Broad Institute of Harvard and MIT, Cambridge, MA, United States.
| |
Collapse
|
10
|
Poopal AC, Schroeder LM, Horn PS, Bassell GJ, Gross C. Increased expression of the PI3K catalytic subunit p110δ underlies elevated S6 phosphorylation and protein synthesis in an individual with autism from a multiplex family. Mol Autism 2016; 7:3. [PMID: 26770665 PMCID: PMC4712554 DOI: 10.1186/s13229-015-0066-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/28/2015] [Indexed: 12/26/2022] Open
Abstract
Background Dysfunctions in the PI3K/mTOR pathway have gained a lot of attention in autism research. This was initially based on the discovery of several monogenic autism spectrum disorders with mutations or defects in PI3K/mTOR signaling components. Recent genetic studies corroborate that defective PI3K/mTOR signaling might be a shared pathomechanism in autism disorders of so far unknown etiology, but functional molecular analyses in human cells are rare. The goals of this study were to perform a functional screen of cell lines from patients with idiopathic autism for defects in PI3K/mTOR signaling, to test if further functional analyses are suitable to detect underlying molecular mechanisms, and to evaluate this approach as a biomarker tool to identify therapeutic targets. Methods We performed phospho-S6- and S6-specific ELISA experiments on 21 lymphoblastoid cell lines from the AGRE collection and on 37 lymphoblastoid cell lines from the Simons Simplex Collection and their healthy siblings. Cell lines from one individual with increased S6 phosphorylation and his multiplex family were analyzed in further detail to identify upstream defects in PI3K signaling associated with autism diagnosis. Results We detected significantly increased S6 phosphorylation in 3 of the 21 lymphoblastoid cell lines from AGRE compared to a healthy control and in 1 of the 37 lymphoblastoid cell lines from the Simons Simplex Collection compared to the healthy sibling. Further analysis of cells from one individual with elevated S6 phosphorylation showed increased expression of the PI3K catalytic subunit p110δ, which was also observed in lymphoblastoid cells from other autistic siblings but not unaffected members in his multiplex family. The p110δ-selective inhibitor IC87114 reduced elevated S6 phosphorylation and protein synthesis in this cell line. Conclusions Our results suggest that functional analysis of PI3K/mTOR signaling is a biomarker tool to identify disease-associated molecular defects that could serve as therapeutic targets in autism. Using this approach, we discovered impaired signaling and protein synthesis through the PI3K catalytic subunit p110δ as an underlying molecular defect and potential treatment target in select autism spectrum disorders. Increased p110δ activity was recently associated with schizophrenia, and our results suggest that p110δ may also be implicated in autism. Electronic supplementary material The online version of this article (doi:10.1186/s13229-015-0066-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ashwini C Poopal
- Department of Cell Biology, Emory University Medical School, 615 Michael Street, Atlanta, GA 30322 USA
| | - Lindsay M Schroeder
- Division of Neurology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229 USA
| | - Paul S Horn
- Division of Neurology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229 USA ; Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229 USA
| | - Gary J Bassell
- Department of Cell Biology, Emory University Medical School, 615 Michael Street, Atlanta, GA 30322 USA
| | - Christina Gross
- Department of Cell Biology, Emory University Medical School, 615 Michael Street, Atlanta, GA 30322 USA ; Division of Neurology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229 USA
| |
Collapse
|
11
|
Gierut JJ, Wood LB, Lau KS, Lin YJ, Genetti C, Samatar AA, Lauffenburger DA, Haigis KM. Network-level effects of kinase inhibitors modulate TNF-α-induced apoptosis in the intestinal epithelium. Sci Signal 2015; 8:ra129. [PMID: 26671150 DOI: 10.1126/scisignal.aac7235] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Individual signaling pathways operate in the context of the broader signaling network. Thus, the response of a cell to signals from the environment is affected by the state of the signaling network, such as the clinically relevant example of whether some components in the network are inhibited. The cytokine tumor necrosis factor-α (TNF-α) promotes opposing cellular behaviors under different conditions; the outcome is influenced by the state of the network. For example, in the mouse intestinal epithelium, inhibition of the mitogen-activated protein kinase (MAPK) kinase MEK alters the timing of TNF-α-induced apoptosis. We investigated whether MAPK signaling directly influences TNF-α-induced apoptosis or whether network-level effects secondary to inhibition of the MAPK pathway alter the cellular response. We found that inhibitors of the MAPK kinase kinase Raf, MEK, or extracellular signal-regulated kinase (ERK) exerted distinct effects on the timing and magnitude of TNF-α-induced apoptosis in the mouse intestine. Furthermore, even different MEK inhibitors exerted distinct effects; one, CH5126766, potentiated TNF-α-induced apoptosis, and the others reduced cell death. Computational modeling and experimental perturbation identified the kinase Akt as the primary signaling node that enhanced apoptosis in the context of TNF-α signaling in the presence of CH5126766. Our work emphasizes the importance of integrated network signaling in specifying cellular behavior in response to experimental or therapeutic manipulation. More broadly, this study highlighted the importance of considering the network-level effects of pathway inhibitors and showed the distinct effects of inhibitors that share the same target.
Collapse
Affiliation(s)
- Jessica J Gierut
- Cancer Research Institute, Beth Israel Deaconess Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Levi B Wood
- Cancer Research Institute, Beth Israel Deaconess Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA 02215, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ken S Lau
- Department of Chemical and Physical Biology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.,Epithelial Biology Center and Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Yi-Jang Lin
- Cancer Research Institute, Beth Israel Deaconess Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Casie Genetti
- Cancer Research Institute, Beth Israel Deaconess Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | | | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kevin M Haigis
- Cancer Research Institute, Beth Israel Deaconess Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
12
|
A Potential Role for Felbamate in TSC- and NF1-Related Epilepsy: A Case Report and Review of the Literature. Case Rep Neurol Med 2015; 2015:960746. [PMID: 26579319 PMCID: PMC4633543 DOI: 10.1155/2015/960746] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 09/23/2015] [Accepted: 10/05/2015] [Indexed: 12/31/2022] Open
Abstract
A 15-year-old girl with maternal inheritance of neurofibromatosis type 1 (NF1) and paternal inheritance of tuberous sclerosis complex (TSC) developed intractable epilepsy at age 5. Her seizures were refractory to adequate doses of four antiepileptic medications until felbamate was initiated at age 7. She has since remained seizure-free on felbamate monotherapy. Although felbamate has multiple mechanisms of action, it is thought to have its most potent antiepileptic effects through inhibition of the N-methyl-D-aspartate receptor (NMDAR). Previous studies have shown that the NMDAR is altered in varying epilepsy syndromes and notably in the cortical tubers found in TSC. The aim of this paper is to examine how felbamate monotherapy was able to achieve such robust antiepileptic effects in a unique patient and possibly offer a novel therapeutic approach to patients suffering from TSC- and NF-related epilepsy.
Collapse
|
13
|
Mistry KA, Sood D, Bhoil R, Chadha V, Ahluwalia AK, Sood S, Suthar PP. A Classic Case of Tuberous Sclerosis with Multisystem Involvement Including Giant Bilateral Renal Angiomyolipomas Presenting as Massive Hematuria. Pol J Radiol 2015; 80:435-41. [PMID: 26491490 PMCID: PMC4588676 DOI: 10.12659/pjr.894741] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Accepted: 06/19/2015] [Indexed: 12/19/2022] Open
Abstract
Background Tuberous Sclerosis (TSC) also known as Bourneville disease is a neurocutaneous syndrome having an autosomal dominant inheritance pattern, though the condition has a high rate of spontaneous mutation. It is the second most common neurocutaneous syndrome after neurofibromatosis. This disease demonstrates a widespread potential for hamartomatous growths in multiple organ systems. Case Report We report a case of a 36-year-old female with TSC presenting as massive hematuria with underlying giant bilateral renal angiomyolipomas (AML) with estimated total tumor burden of more than 8 kg which is to the best of our knowledge the highest ever reported. The patient also had lymphangioleiomyomatosis and lesions in the brain, skin, teeth and bones. Conclusions TSC has a wide variety of clinical and radiologic manifestations. It should be suspected when some of the common radiological manifestations are found, including CNS involvement, renal and hepatic AMLs and LAM, even if clinical signs are not obvious. Renal AMLs in setting of TSC may reach giant proportions and may present with massive hematuria.
Collapse
Affiliation(s)
- Kewal A Mistry
- Department of Radiology, Dr. Rajendra Prasad Government Medical College, Kangra, India
| | - Dinesh Sood
- Department of Radiology, Dr. Rajendra Prasad Government Medical College, Kangra, India
| | - Rohit Bhoil
- Department of Radiology, Indira Gandhi Medical College, Shimla, India
| | - Veenal Chadha
- Department of Community Medicine, Dr. Rajendra Prasad Government Medical College, Kangra, India
| | - Ajay K Ahluwalia
- Department of Radiology, Dr. Rajendra Prasad Government Medical College, Kangra, India
| | - Saurabh Sood
- Department of Radiology, Dr. Rajendra Prasad Government Medical College, Kangra, India
| | - Pokhraj P Suthar
- Department of Radiology, Baroda Medical College, Vadodara, India
| |
Collapse
|
14
|
Immune mediators in the brain and peripheral tissues in autism spectrum disorder. Nat Rev Neurosci 2015; 16:469-86. [PMID: 26189694 DOI: 10.1038/nrn3978] [Citation(s) in RCA: 343] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Increasing evidence points to a central role for immune dysregulation in autism spectrum disorder (ASD). Several ASD risk genes encode components of the immune system and many maternal immune system-related risk factors--including autoimmunity, infection and fetal reactive antibodies--are associated with ASD. In addition, there is evidence of ongoing immune dysregulation in individuals with ASD and in animal models of this disorder. Recently, several molecular signalling pathways--including pathways downstream of cytokines, the receptor MET, major histocompatibility complex class I molecules, microglia and complement factors--have been identified that link immune activation to ASD phenotypes. Together, these findings indicate that the immune system is a point of convergence for multiple ASD-related genetic and environmental risk factors.
Collapse
|
15
|
Sasongko TH, Ismail NFD, Nik Abdul Malik NMA, Zabidi-Hussin ZAMH. Rapamycin and its analogues (rapalogs) for Tuberous Sclerosis Complex-associated tumors: a systematic review on non-randomized studies using meta-analysis. Orphanet J Rare Dis 2015; 10:95. [PMID: 26259610 PMCID: PMC4531483 DOI: 10.1186/s13023-015-0317-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 08/03/2015] [Indexed: 12/26/2022] Open
Abstract
Background Rapamycin has gained significant attention for its potential activity in reducing the size of TSC-associated tumors, thus providing alternative to surgery. This study aimed at determining the efficacy of rapamycin and rapalogs for reducing the size of TSC-associated solid tumors in patients with Tuberous Sclerosis Complex (TSC). Methods Our data sources included electronic searches of the PubMed. We included into our meta-analysis any type of non-randomized study that reported the use of rapamycin and rapalogs for reducing the size of TSC-associated solid tumors in patients with TSC. Data was entered into Cochrane Review Manager Version 5.3 and analyzed. Results Four case reports and 4 clinical trials were included. Five patients from the case reports (all with SEGA) and 91 patients from the clinical trials (41 with SEGA, 63 with kidney angiomyolipoma and 5 with liver angiomyolipoma) were included into the analysis. Volume and diameter of SEGAs were significantly reduced by mean difference of 1.23 cc (95 % CI −2.32 to −0.13; p = 0.03) and 7.91 mm (95 % CI −11.82 to −4.01; p < 0.0001), respectively. Volume and mean of sum of longest diameter of kidney angiomyolipomas were significantly reduced by mean difference of 39.5 cc (95 % CI −48.85 to −30.15; p <0.00001) and 69.03 mm (95 % CI −158.05 to 12.65; p = 0.008), respectively. In liver angiomyolipomas, however, reduction in tumor size was not evident. Sum of longest diameter of liver angiomyolipomas in 4 patients were enlarged by 2.7 mm (95 % CI 28.42 to −23.02) by the end of treatment, though not significant (p = 0.84). Conclusions Rapamycin and rapalogs showed efficacy towards reducing the size of SEGA and kidney angiomyolipoma but not liver angiomyolipomas. This finding is strengthening the conclusion of our Cochrane systematic review on the randomized trials.
Collapse
Affiliation(s)
- Teguh Haryo Sasongko
- Human Genome Center, School of Medical Sciences, Universiti Sains Malaysia, USM Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia. .,Center for Neuroscience Services and Research, Universiti Sains Malaysia, USM Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia.
| | - Nur Farrah Dila Ismail
- Human Genome Center, School of Medical Sciences, Universiti Sains Malaysia, USM Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia.,Center for Neuroscience Services and Research, Universiti Sains Malaysia, USM Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Nik Mohamad Ariff Nik Abdul Malik
- Human Genome Center, School of Medical Sciences, Universiti Sains Malaysia, USM Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia.,Center for Neuroscience Services and Research, Universiti Sains Malaysia, USM Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Z A M H Zabidi-Hussin
- Department of Pediatrics, School of Medical Sciences, Universiti Sains Malaysia, USM Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia.,Center for Neuroscience Services and Research, Universiti Sains Malaysia, USM Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
16
|
Okumura A, Ozaki M, Niida Y. Development of a practical NF1 genetic testing method through the pilot analysis of five Japanese families with neurofibromatosis type 1. Brain Dev 2015; 37:677-89. [PMID: 25480383 DOI: 10.1016/j.braindev.2014.11.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 11/08/2014] [Accepted: 11/10/2014] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Mutation analysis of NF1, the responsible gene for neurofibromatosis type 1 (NF1), is still difficult due to its large size, lack of mutational hotspots, the presence of many pseudogenes, and its wide spectrum of mutations. To develop a simple and inexpensive NF1 genetic testing for clinical use, we analyzed five Japanese families with NF1 as a pilot study. METHODS Our original method, CEL endonuclease mediated heteroduplex incision with polyacrylamide gel electrophoresis and silver staining (CHIPS) was optimized for NF1 mutation screening, and reverse transcription polymerase chain reaction (RT-PCR) was performed to determine the effect of transcription. Also, we employed DNA microarray analysis to evaluate the break points of the large deletion. RESULTS A new nonsense mutation, p.Gln209(∗), was detected in family 1 and the splicing donor site mutation, c.2850+1G>T, was detected in family 2. In family 3, c.4402A>G was detected in exon 34 and the p.Ser1468Gly missense mutation was predicted. However mRNA analysis revealed that this substitution created an aberrant splicing acceptor site, thereby causing the p.Phe1457(∗) nonsense mutation. In the other two families, type-1 and unique NF1 microdeletions were detected by DNA microarray analysis. CONCLUSIONS Our results show that the combination of CHIPS and RT-PCR effectively screen and characterize NF1 point mutations, and both DNA and RNA level analysis are required to understand the nature of the NF1 mutation. Our results also suggest the possibility of a higher incidence and unique profile of NF1 large deletions in the Japanese population as compared to previous studies performed in Europe.
Collapse
Affiliation(s)
- Akiko Okumura
- Department of Pediatrics, Kanazawa University Graduate School of Medical Science, Japan
| | - Mamoru Ozaki
- Division of Genomic Medicine, Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Ishikawa, Japan; Center for Medical Genetics, Kanazawa Medical University Hospital, Japan
| | - Yo Niida
- Division of Genomic Medicine, Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Ishikawa, Japan; Center for Medical Genetics, Kanazawa Medical University Hospital, Japan.
| |
Collapse
|
17
|
Curatolo P. Mechanistic target of rapamycin (mTOR) in tuberous sclerosis complex-associated epilepsy. Pediatr Neurol 2015; 52:281-9. [PMID: 25591831 DOI: 10.1016/j.pediatrneurol.2014.10.028] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 09/29/2014] [Accepted: 10/29/2014] [Indexed: 01/12/2023]
Abstract
BACKGROUND Tuberous sclerosis complex is a multiorgan disease resulting from a mutation of one of two TSC genes. The two gene products form a functional complex that regulates the mTOR signaling pathway (mTOR initially represented mammalian target of rapamycin, but increasingly the term mechanistic target of rapamycin is used to reflect the ubiquitous occurrence of mTOR). Epilepsy is the most common neurological symptom of tuberous sclerosis complex, occurring in 80% to 90% of affected individuals over the course of their lifetimes and causing significant morbidity and mortality. The mechanistic target of rapamycin (mTOR) signaling pathway is intricately involved in multiple cellular functions--including protein synthesis, cell growth and proliferation, and synaptic plasticity--which may influence neuronal excitability and precipitate epileptogenesis. Recent preclinical and clinical studies have increased interest in the potential role of mTOR inhibitors for the treatment of tuberous sclerosis complex-related epilepsy. METHODS Medline and PubMed database searches were used to identify relevant studies and other information on tuberous sclerosis complex-related epilepsies, the mTOR pathway, and current advances in treatment approaches. RESULTS Although current management strategies that provide symptomatic relief are effective at reducing the frequency of seizures in individuals with tuberous sclerosis complex, there is further room for the exploration of therapies that directly address hyperactive mTOR signaling--the underlying etiology of the disease. The role of the antiepileptic effect of mTOR inhibition was first demonstrated in knockout TSC1 mouse models. Additionally, several case studies demonstrated a positive effect on seizure frequency and severity in patients with pharmacoresistant epilepsy. In a phase 1/2 clinical trial with 28 patients, clinically relevant reduction in overall seizure frequency was documented in individuals treated with the mTOR inhibitor everolimus. In a phase 3 trial evaluating the role of everolimus in subependymal giant cell astrocytoma, seizures were a secondary end point. Because the median seizure frequency was zero in this study, the analysis was inconclusive. CONCLUSION Various preclinical models provide substantial evidence for the role of mTOR inhibition in the treatment of epilepsy in individuals with tuberous sclerosis complex. Preliminary clinical studies provide supportive evidence for a role of mTOR inhibition in the management of tuberous sclerosis complex-associated epilepsy and pave the way for new randomized placebo-controlled studies. This article reviews current treatment recommendations for the management of tuberous sclerosis complex-associated epilepsy as well as the rationale and evidence to support the use of mTOR inhibitors.
Collapse
Affiliation(s)
- Paolo Curatolo
- Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University of Rome, Rome, Italy.
| |
Collapse
|
18
|
Ma DJ, Galanis E, Anderson SK, Schiff D, Kaufmann TJ, Peller PJ, Giannini C, Brown PD, Uhm JH, McGraw S, Jaeckle KA, Flynn PJ, Ligon KL, Buckner JC, Sarkaria JN. A phase II trial of everolimus, temozolomide, and radiotherapy in patients with newly diagnosed glioblastoma: NCCTG N057K. Neuro Oncol 2014; 17:1261-9. [PMID: 25526733 DOI: 10.1093/neuonc/nou328] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2007] [Accepted: 10/31/2014] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The mammalian target of rapamycin (mTOR) functions within the phosphatidylinositol-3 kinase (PI3K)/Akt pathway as a critical modulator of cell survival. This clinical trial evaluated the combination of the mTOR inhibitor everolimus with conventional temozolomide (TMZ)-based chemoradiotherapy. METHODS Newly diagnosed patients with glioblastoma multiforme were eligible for this single arm, phase II study. Everolimus (70 mg/wk) was started 1 week prior to radiation and TMZ, followed by adjuvant TMZ, and continued until disease progression. The primary endpoint was overall survival at 12 months, and secondary endpoints were toxicity and time to progression. Eleven patients were imaged with 3'-deoxy-3'-(18)F-fluorothymidine ((18)FLT)-PET/CT before and after the initial 2 doses of everolimus before initiating radiation/TMZ. Imaged patients with sufficient tumor samples also underwent immunohistochemical and focused exon sequencing analysis. RESULTS This study accrued 100 evaluable patients. Fourteen percent of patients had grade 4 hematologic toxicities. Twelve percent had at least one grade 4 nonhematologic toxicity, and there was one treatment-related death. Overall survival at 12 months was 64% and median time to progression was 6.4 months. Of the patients who had (18)FLT-PET data, 4/9 had a partial response after 2 doses of everolimus. Focused exon sequencing demonstrated that (18)FLT-PET responders were less likely to have alterations within the PI3K/Akt/mTOR or tuberous sclerosis complex/neurofibromatosis type 1 pathway compared with nonresponders. CONCLUSION Combining everolimus with conventional chemoradiation had moderate toxicity. (18)FLT-PET studies suggested an initial antiproliferative effect in a genetically distinct subset of tumors, but this did not translate into an appreciable survival benefit compared with historical controls treated with conventional therapy.
Collapse
Affiliation(s)
- Daniel J Ma
- Mayo Clinic, Rochester, Minnesota (D.J.M., E.G., T.J.K., P.J.P., C.G., P.D.B., J.H.U., J.C.B., J.N.S.); Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota (S.K.A.); University of Virginia, Charlottesville, Virginia (D.S.); MD Anderson Cancer Center, Houston, Texas (P.D.B.); Sioux Community Cancer Consortium, Sioux Falls, South Dakota (S.M.); Mayo Clinic, Jacksonville, Florida (K.A.J.); Metro-Minnesota Community Clinical Oncology Program, St. Louis Park, Minnesota (P.J.F.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Medical Oncology, Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts (K.L.L.)
| | - Evanthia Galanis
- Mayo Clinic, Rochester, Minnesota (D.J.M., E.G., T.J.K., P.J.P., C.G., P.D.B., J.H.U., J.C.B., J.N.S.); Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota (S.K.A.); University of Virginia, Charlottesville, Virginia (D.S.); MD Anderson Cancer Center, Houston, Texas (P.D.B.); Sioux Community Cancer Consortium, Sioux Falls, South Dakota (S.M.); Mayo Clinic, Jacksonville, Florida (K.A.J.); Metro-Minnesota Community Clinical Oncology Program, St. Louis Park, Minnesota (P.J.F.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Medical Oncology, Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts (K.L.L.)
| | - S Keith Anderson
- Mayo Clinic, Rochester, Minnesota (D.J.M., E.G., T.J.K., P.J.P., C.G., P.D.B., J.H.U., J.C.B., J.N.S.); Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota (S.K.A.); University of Virginia, Charlottesville, Virginia (D.S.); MD Anderson Cancer Center, Houston, Texas (P.D.B.); Sioux Community Cancer Consortium, Sioux Falls, South Dakota (S.M.); Mayo Clinic, Jacksonville, Florida (K.A.J.); Metro-Minnesota Community Clinical Oncology Program, St. Louis Park, Minnesota (P.J.F.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Medical Oncology, Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts (K.L.L.)
| | - David Schiff
- Mayo Clinic, Rochester, Minnesota (D.J.M., E.G., T.J.K., P.J.P., C.G., P.D.B., J.H.U., J.C.B., J.N.S.); Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota (S.K.A.); University of Virginia, Charlottesville, Virginia (D.S.); MD Anderson Cancer Center, Houston, Texas (P.D.B.); Sioux Community Cancer Consortium, Sioux Falls, South Dakota (S.M.); Mayo Clinic, Jacksonville, Florida (K.A.J.); Metro-Minnesota Community Clinical Oncology Program, St. Louis Park, Minnesota (P.J.F.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Medical Oncology, Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts (K.L.L.)
| | - Timothy J Kaufmann
- Mayo Clinic, Rochester, Minnesota (D.J.M., E.G., T.J.K., P.J.P., C.G., P.D.B., J.H.U., J.C.B., J.N.S.); Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota (S.K.A.); University of Virginia, Charlottesville, Virginia (D.S.); MD Anderson Cancer Center, Houston, Texas (P.D.B.); Sioux Community Cancer Consortium, Sioux Falls, South Dakota (S.M.); Mayo Clinic, Jacksonville, Florida (K.A.J.); Metro-Minnesota Community Clinical Oncology Program, St. Louis Park, Minnesota (P.J.F.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Medical Oncology, Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts (K.L.L.)
| | - Patrick J Peller
- Mayo Clinic, Rochester, Minnesota (D.J.M., E.G., T.J.K., P.J.P., C.G., P.D.B., J.H.U., J.C.B., J.N.S.); Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota (S.K.A.); University of Virginia, Charlottesville, Virginia (D.S.); MD Anderson Cancer Center, Houston, Texas (P.D.B.); Sioux Community Cancer Consortium, Sioux Falls, South Dakota (S.M.); Mayo Clinic, Jacksonville, Florida (K.A.J.); Metro-Minnesota Community Clinical Oncology Program, St. Louis Park, Minnesota (P.J.F.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Medical Oncology, Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts (K.L.L.)
| | - Caterina Giannini
- Mayo Clinic, Rochester, Minnesota (D.J.M., E.G., T.J.K., P.J.P., C.G., P.D.B., J.H.U., J.C.B., J.N.S.); Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota (S.K.A.); University of Virginia, Charlottesville, Virginia (D.S.); MD Anderson Cancer Center, Houston, Texas (P.D.B.); Sioux Community Cancer Consortium, Sioux Falls, South Dakota (S.M.); Mayo Clinic, Jacksonville, Florida (K.A.J.); Metro-Minnesota Community Clinical Oncology Program, St. Louis Park, Minnesota (P.J.F.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Medical Oncology, Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts (K.L.L.)
| | - Paul D Brown
- Mayo Clinic, Rochester, Minnesota (D.J.M., E.G., T.J.K., P.J.P., C.G., P.D.B., J.H.U., J.C.B., J.N.S.); Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota (S.K.A.); University of Virginia, Charlottesville, Virginia (D.S.); MD Anderson Cancer Center, Houston, Texas (P.D.B.); Sioux Community Cancer Consortium, Sioux Falls, South Dakota (S.M.); Mayo Clinic, Jacksonville, Florida (K.A.J.); Metro-Minnesota Community Clinical Oncology Program, St. Louis Park, Minnesota (P.J.F.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Medical Oncology, Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts (K.L.L.)
| | - Joon H Uhm
- Mayo Clinic, Rochester, Minnesota (D.J.M., E.G., T.J.K., P.J.P., C.G., P.D.B., J.H.U., J.C.B., J.N.S.); Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota (S.K.A.); University of Virginia, Charlottesville, Virginia (D.S.); MD Anderson Cancer Center, Houston, Texas (P.D.B.); Sioux Community Cancer Consortium, Sioux Falls, South Dakota (S.M.); Mayo Clinic, Jacksonville, Florida (K.A.J.); Metro-Minnesota Community Clinical Oncology Program, St. Louis Park, Minnesota (P.J.F.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Medical Oncology, Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts (K.L.L.)
| | - Steven McGraw
- Mayo Clinic, Rochester, Minnesota (D.J.M., E.G., T.J.K., P.J.P., C.G., P.D.B., J.H.U., J.C.B., J.N.S.); Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota (S.K.A.); University of Virginia, Charlottesville, Virginia (D.S.); MD Anderson Cancer Center, Houston, Texas (P.D.B.); Sioux Community Cancer Consortium, Sioux Falls, South Dakota (S.M.); Mayo Clinic, Jacksonville, Florida (K.A.J.); Metro-Minnesota Community Clinical Oncology Program, St. Louis Park, Minnesota (P.J.F.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Medical Oncology, Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts (K.L.L.)
| | - Kurt A Jaeckle
- Mayo Clinic, Rochester, Minnesota (D.J.M., E.G., T.J.K., P.J.P., C.G., P.D.B., J.H.U., J.C.B., J.N.S.); Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota (S.K.A.); University of Virginia, Charlottesville, Virginia (D.S.); MD Anderson Cancer Center, Houston, Texas (P.D.B.); Sioux Community Cancer Consortium, Sioux Falls, South Dakota (S.M.); Mayo Clinic, Jacksonville, Florida (K.A.J.); Metro-Minnesota Community Clinical Oncology Program, St. Louis Park, Minnesota (P.J.F.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Medical Oncology, Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts (K.L.L.)
| | - Patrick J Flynn
- Mayo Clinic, Rochester, Minnesota (D.J.M., E.G., T.J.K., P.J.P., C.G., P.D.B., J.H.U., J.C.B., J.N.S.); Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota (S.K.A.); University of Virginia, Charlottesville, Virginia (D.S.); MD Anderson Cancer Center, Houston, Texas (P.D.B.); Sioux Community Cancer Consortium, Sioux Falls, South Dakota (S.M.); Mayo Clinic, Jacksonville, Florida (K.A.J.); Metro-Minnesota Community Clinical Oncology Program, St. Louis Park, Minnesota (P.J.F.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Medical Oncology, Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts (K.L.L.)
| | - Keith L Ligon
- Mayo Clinic, Rochester, Minnesota (D.J.M., E.G., T.J.K., P.J.P., C.G., P.D.B., J.H.U., J.C.B., J.N.S.); Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota (S.K.A.); University of Virginia, Charlottesville, Virginia (D.S.); MD Anderson Cancer Center, Houston, Texas (P.D.B.); Sioux Community Cancer Consortium, Sioux Falls, South Dakota (S.M.); Mayo Clinic, Jacksonville, Florida (K.A.J.); Metro-Minnesota Community Clinical Oncology Program, St. Louis Park, Minnesota (P.J.F.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Medical Oncology, Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts (K.L.L.)
| | - Jan C Buckner
- Mayo Clinic, Rochester, Minnesota (D.J.M., E.G., T.J.K., P.J.P., C.G., P.D.B., J.H.U., J.C.B., J.N.S.); Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota (S.K.A.); University of Virginia, Charlottesville, Virginia (D.S.); MD Anderson Cancer Center, Houston, Texas (P.D.B.); Sioux Community Cancer Consortium, Sioux Falls, South Dakota (S.M.); Mayo Clinic, Jacksonville, Florida (K.A.J.); Metro-Minnesota Community Clinical Oncology Program, St. Louis Park, Minnesota (P.J.F.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Medical Oncology, Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts (K.L.L.)
| | - Jann N Sarkaria
- Mayo Clinic, Rochester, Minnesota (D.J.M., E.G., T.J.K., P.J.P., C.G., P.D.B., J.H.U., J.C.B., J.N.S.); Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota (S.K.A.); University of Virginia, Charlottesville, Virginia (D.S.); MD Anderson Cancer Center, Houston, Texas (P.D.B.); Sioux Community Cancer Consortium, Sioux Falls, South Dakota (S.M.); Mayo Clinic, Jacksonville, Florida (K.A.J.); Metro-Minnesota Community Clinical Oncology Program, St. Louis Park, Minnesota (P.J.F.); Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts (K.L.L.); Department of Medical Oncology, Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts (K.L.L.)
| |
Collapse
|
19
|
Chiarini F, Evangelisti C, McCubrey JA, Martelli AM. Current treatment strategies for inhibiting mTOR in cancer. Trends Pharmacol Sci 2014; 36:124-35. [PMID: 25497227 DOI: 10.1016/j.tips.2014.11.004] [Citation(s) in RCA: 213] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 11/07/2014] [Accepted: 11/12/2014] [Indexed: 02/07/2023]
Abstract
Mammalian target of rapamycin (mTOR) is a Ser/Thr kinase that regulates a wide range of functions, including cell growth, proliferation, survival, autophagy, metabolism, and cytoskeletal organization. mTOR activity is dysregulated in several human disorders, including cancer. The crucial role of mTOR in cancer cell biology has stimulated interest in mTOR inhibitors, placing mTOR on the radar of the pharmaceutical industry. Several mTOR inhibitors have already undergone clinical trials for treating tumors, without great success, although mTOR inhibitors are approved for the treatment of some types of cancer, including advanced renal cell carcinoma. However, the role of mTOR inhibitors in cancer treatment continues to evolve as new compounds are continuously being disclosed. Here we review the three classes of mTOR inhibitors currently available for treating cancer patients. Moreover, we highlight efforts to identify markers of resistance and sensitivity to mTOR inhibition that could prove useful in the emerging field of personalized medicine.
Collapse
Affiliation(s)
- Francesca Chiarini
- Institute of Molecular Genetics, National Research Council, Bologna, Italy; Rizzoli Orthopedic Institute, Bologna, Italy
| | - Camilla Evangelisti
- Institute of Molecular Genetics, National Research Council, Bologna, Italy; Rizzoli Orthopedic Institute, Bologna, Italy
| | - James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Alberto M Martelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.
| |
Collapse
|
20
|
Yang G, Yang L, Yang X, Shi X, Wang J, Liu Y, Ju J, Zou L. Efficacy and safety of a mammalian target of rapamycin inhibitor in pediatric patients with tuberous sclerosis complex: A systematic review and meta-analysis. Exp Ther Med 2014; 9:626-630. [PMID: 25574245 PMCID: PMC4280930 DOI: 10.3892/etm.2014.2093] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 11/25/2014] [Indexed: 12/27/2022] Open
Abstract
Inhibitors of mammalian target of rapamycin (mTOR) are increasingly used as therapy for pediatric patients with tuberous sclerosis complex (TSC). The uncertainty over the efficacy and safety of mTOR inhibitor therapy for the treatment of pediatric patients with TSC emphasizes the necessity for a synthesis of existing evidence. The aim of this study was to assess the efficacy and safety of mTOR inhibitor therapy for the treatment of pediatric patients with TSC. The PubMed, EmBase and Cochrane Library electronic databases were searched, and studies of mTOR inhibitor therapy and non-mTOR inhibitor therapy in pediatric patients with TSC (<18 years old) were selected. Eleven studies met the inclusion criteria. There was evidence of a significantly increased response rate in pediatric patients with TSC treated with mTOR inhibitor therapy compared with those treated with non-mTOR inhibitor therapy (odds ratio, 24.71; 95% confidence interval, 7.46–81.72; P<0.001). The majority of studies reported few adverse events. There was an increased incidence of mouth ulceration, stomatitis, convulsion and pyrexia in pediatric patients with TSC treated with mTOR inhibitor therapy. In conclusion, mTOR inhibitor therapy is an efficacious and safe treatment for pediatric patients with TSC.
Collapse
Affiliation(s)
- Guang Yang
- Department of Pediatrics, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Lu Yang
- Special Care Medical Center, Navy General Hospital of PLA, Beijing 100048, P.R. China
| | - Xiaofan Yang
- Department of Pediatrics, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Xiuyu Shi
- Department of Pediatrics, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Jing Wang
- Department of Pediatrics, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Yujie Liu
- Department of Pediatrics, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Jun Ju
- Department of Pediatrics, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Liping Zou
- Department of Pediatrics, Chinese PLA General Hospital, Beijing 100853, P.R. China ; Beijing Institute for Brain Disorders, Beijing 100069, P.R. China
| |
Collapse
|
21
|
Johnson CE, Hunt DK, Wiltshire M, Herbert TP, Sampson JR, Errington RJ, Davies DM, Tee AR. Endoplasmic reticulum stress and cell death in mTORC1-overactive cells is induced by nelfinavir and enhanced by chloroquine. Mol Oncol 2014; 9:675-88. [PMID: 25498902 DOI: 10.1016/j.molonc.2014.11.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 11/17/2014] [Accepted: 11/18/2014] [Indexed: 01/29/2023] Open
Abstract
Inappropriate activation of mammalian/mechanistic target of rapamycin complex 1 (mTORC1) is common in cancer and has many cellular consequences including elevated endoplasmic reticulum (ER) stress. Cells employ autophagy as a critical compensatory survival mechanism during ER stress. This study utilised drug-induced ER stress through nelfinavir in order to examine ER stress tolerance in cell lines with hyper-active mTORC1 signalling. Our initial findings in wild type cells showed nelfinavir inhibited mTORC1 signalling and upregulated autophagy, as determined by decreased rpS6 and S6K1 phosphorylation, and SQTSM1 protein expression, respectively. Contrastingly, cells with hyper-active mTORC1 displayed basally elevated levels of ER stress which was greatly exaggerated following nelfinavir treatment, seen through increased CHOP mRNA and XBP1 splicing. To further enhance the effects of nelfinavir, we introduced chloroquine as an autophagy inhibitor. Combination of nelfinavir and chloroquine significantly increased ER stress and caused selective cell death in multiple cell line models with hyper-active mTORC1, whilst control cells with normalised mTORC1 signalling tolerated treatment. By comparing chloroquine to other autophagy inhibitors, we uncovered that selective toxicity invoked by chloroquine was independent of autophagy inhibition yet entrapment of chloroquine to acidified lysosomal/endosomal compartments was necessary for cytotoxicity. Our research demonstrates that combination of nelfinavir and chloroquine has therapeutic potential for treatment of mTORC1-driven tumours.
Collapse
Affiliation(s)
- Charlotte E Johnson
- Institute of Cancer and Genetics, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - David K Hunt
- Institute of Cancer and Genetics, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - Marie Wiltshire
- Institute of Cancer and Genetics, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - Terry P Herbert
- Department of Cell Physiology and Pharmacology, University of Leicester, The Henry Wellcome Building, University Road, Leicester LE1 9HN, UK
| | - Julian R Sampson
- Institute of Cancer and Genetics, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - Rachel J Errington
- Institute of Cancer and Genetics, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - D Mark Davies
- Institute of Cancer and Genetics, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - Andrew R Tee
- Institute of Cancer and Genetics, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.
| |
Collapse
|
22
|
Knöpfel N, Martín-Santiago A, Bauza A, Hervás J. Topical 0.2% Rapamycin to Treat Facial Angiofibromas and Hypomelanotic Macules in Tuberous Sclerosis. ACTAS DERMO-SIFILIOGRAFICAS 2014. [DOI: 10.1016/j.adengl.2013.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
23
|
Rapamicina tópica al 0,2% para el tratamiento de angiofibromas faciales y máculas hipomelanóticas en la esclerosis tuberosa. ACTAS DERMO-SIFILIOGRAFICAS 2014; 105:802-3. [DOI: 10.1016/j.ad.2013.09.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 09/03/2013] [Accepted: 09/08/2013] [Indexed: 01/09/2023] Open
|
24
|
Kurschat CE, Müller RU, Franke M, Maintz D, Schermer B, Benzing T. An approach to cystic kidney diseases: the clinician's view. Nat Rev Nephrol 2014; 10:687-99. [DOI: 10.1038/nrneph.2014.173] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
25
|
Prato G, Mancardi MM, Baglietto MG, Janis S, Vercellino N, Rossi A, Consales A, Raso A, Garrè ML. Congenital segmental lymphedema in tuberous sclerosis complex with associated subependymal giant cell astrocytomas treated with Mammalian target of rapamycin inhibitors. J Child Neurol 2014; 29:NP54-7. [PMID: 24056156 DOI: 10.1177/0883073813499969] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Tuberous sclerosis complex is a genetic, multisystemic disorder characterized by circumscribed benign lesions (hamartomas) in several organs, including brain. This is the result of defects in the TSC1 and/or TSC2 tumor suppressor genes, encoding the hamartin-tuberin complex that inhibits the mammalian target of rapamycin pathway. Specific inhibitors of this pathway have been shown to reduce the volume of subependymal giant cell astrocytomas associated with tuberous sclerosis. Congenital lymphedema is rarely seen in association with tuberous sclerosis, with only a few reported cases. Although this association can be coincidental, the dysgenetic lymphatic system can represent a hamartia as a consequence of gene mutation. We describe a child with congenital lymphedema in tuberous sclerosis and associated subependymal giant cell astrocytoma who experienced lymphangitis under treatment with mammalian target of rapamycin inhibitors. Because our patient did not show worsening of lymphedema, congenital lymphedema does not seem to be a contraindication for this therapy.
Collapse
Affiliation(s)
- Giulia Prato
- Child Neuropsychiatry Unit, G. Gaslini Institute, Genova, Italy
| | | | | | - Sara Janis
- Child Neuropsychiatry Unit, G. Gaslini Institute, Genova, Italy
| | | | - Andrea Rossi
- Neuroradiology Unit, G. Gaslini Institute, Genova, Italy
| | | | | | | |
Collapse
|
26
|
Navarrete A, Armitage EG, Musteanu M, García A, Mastrangelo A, Bujak R, López-Casas PP, Hidalgo M, Barbas C. Metabolomic evaluation of Mitomycin C and rapamycin in a personalized treatment of pancreatic cancer. Pharmacol Res Perspect 2014; 2:e00067. [PMID: 25505613 PMCID: PMC4186447 DOI: 10.1002/prp2.67] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 07/08/2014] [Accepted: 07/10/2014] [Indexed: 01/05/2023] Open
Abstract
In a personalized treatment designed for a patient with pancreatic cancer resistant to other treatments, the success of Mitomycin C (MMC) has been highlighted. This was revealed in a murine xenograft tumor model encompassing pancreatic adenocarcinoma cells extracted from the patient. The patient was found to exhibit a biallelic inactivation of the PALB2 gene, involved in DNA repair in addition to another mutation in the TSC2 gene that induces susceptibility of the tumor to therapeutic targets of the PI3K-mTOR pathway. The aim of the study was to apply metabolomics to elucidate the modes of action of each therapy, suggesting why MMC was so successful in this patient and why it could be a more popular choice in future pancreatic cancer treatment. The effectiveness of MMC compared to rapamycin (RM), another relevant therapeutic agent has been evaluated through liquid- and gas-chromatography mass spectrometry-based metabolomic analyses of the xenograft tumors. The relative concentrations of many metabolites in the xenograft tumors were found to be increased by MMC relative to other treatments (RM and a combination of both), including a number that are involved in central carbon metabolism (CCM). Metabolic fingerprinting revealed statistically significantly altered pathways including, but not restricted to, the pentose phosphate pathway, glycolysis, TCA cycle, purine metabolism, fatty acid biosynthesis, in addition to many significant lipid and amino acid alterations. Given the genetic background of the patient, it was expected that the combined therapy would be most effective; however, the most effective was MMC alone. It is proposed that the effectiveness of MMC is owed to its direct effect on CCM, a vital region of tumor metabolism.
Collapse
Affiliation(s)
- Alicia Navarrete
- CEMBIO (Center for Metabolomics and Bioanalysis), Facultad de Farmacia, Universidad CEU San Pablo Campus Montepríncipe, Boadilla del Monte, 28668, Madrid, Spain
| | - Emily G Armitage
- CEMBIO (Center for Metabolomics and Bioanalysis), Facultad de Farmacia, Universidad CEU San Pablo Campus Montepríncipe, Boadilla del Monte, 28668, Madrid, Spain
| | - Monica Musteanu
- CNIO (Spanish National Cancer Research Centre) E-28029, Madrid, Madrid, Spain
| | - Antonia García
- CEMBIO (Center for Metabolomics and Bioanalysis), Facultad de Farmacia, Universidad CEU San Pablo Campus Montepríncipe, Boadilla del Monte, 28668, Madrid, Spain
| | - Annalaura Mastrangelo
- CEMBIO (Center for Metabolomics and Bioanalysis), Facultad de Farmacia, Universidad CEU San Pablo Campus Montepríncipe, Boadilla del Monte, 28668, Madrid, Spain
| | - Renata Bujak
- CEMBIO (Center for Metabolomics and Bioanalysis), Facultad de Farmacia, Universidad CEU San Pablo Campus Montepríncipe, Boadilla del Monte, 28668, Madrid, Spain ; Department of Biopharmacy and Pharmacodynamics, Medical University of Gdansk Al. Hallera 107, Gdansk, 80-416, Poland
| | - Pedro P López-Casas
- CNIO (Spanish National Cancer Research Centre) E-28029, Madrid, Madrid, Spain
| | - Manuel Hidalgo
- CNIO (Spanish National Cancer Research Centre) E-28029, Madrid, Madrid, Spain
| | - Coral Barbas
- CEMBIO (Center for Metabolomics and Bioanalysis), Facultad de Farmacia, Universidad CEU San Pablo Campus Montepríncipe, Boadilla del Monte, 28668, Madrid, Spain
| |
Collapse
|
27
|
Khunger N. The dynamics and power of change: changing practices in cutaneous and aesthetic surgery. J Cutan Aesthet Surg 2014; 7:73-4. [PMID: 25136205 PMCID: PMC4134655 DOI: 10.4103/0974-2077.138323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Niti Khunger
- Department of Dermatology and Sexually Transmitted Diseases, Vardhman Mahavir Medical College and Safdarjang Hospital, New Delhi, India
| |
Collapse
|
28
|
Natural history and CT scan follow-up of subependymal giant cell tumors in tuberous sclerosis complex patients. J Clin Neurosci 2014; 21:939-41. [DOI: 10.1016/j.jocn.2013.08.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 08/19/2013] [Accepted: 08/21/2013] [Indexed: 11/17/2022]
|
29
|
Co-targeting the MAPK and PI3K/AKT/mTOR pathways in two genetically engineered mouse models of schwann cell tumors reduces tumor grade and multiplicity. Oncotarget 2014; 5:1502-14. [PMID: 24681606 PMCID: PMC4039227 DOI: 10.18632/oncotarget.1609] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are soft tissue sarcomas that occur spontaneously, or from benign plexiform neurofibromas, in the context of the genetic disorder Neurofibromatosis Type 1 (NF1). The current standard treatment includes surgical resection, high-dose chemotherapy, and/or radiation. To date, most targeted therapies have failed to demonstrate effectiveness against plexiform neurofibromas and MPNSTs. Recently, several studies suggested that the mTOR and MAPK pathways are involved in the formation and progression of MPNSTs. Everolimus (RAD001) inhibits the mTOR and is currently FDA approved for several types of solid tumors. PD-0325901 (PD-901) inhibits MEK, a component of the MAPK pathway, and is currently in clinical trials. Here, we show in vitro than MPNST cell lines are more sensitive to inhibition of cellular growth by Everolimus and PD-901 than immortalized human Schwann cells. In combination, these drugs synergistically inhibit cell growth and induce apoptosis. In two genetically engineered mouse models of MPNST formation, modeling both sporadic and NF1-associated MPNSTs, Everolimus, or PD-901 treatment alone each transiently reduced tumor burden and size, and extended lifespan. However, prolonged treatment of each single agent resulted in the development of resistance and reactivation of target pathways. Combination therapy using Everolimus and PD-901 had synergistic effects on reducing tumor burden and size, and increased lifespan. Combination therapy allowed persistent and prolonged reduction in signaling through both pathways. These data suggest that co-targeting mTOR and MEK may be effective in patients with sporadic or NF1-associated MPNSTs.
Collapse
|
30
|
Gropman AL. Epigenetics and Pervasive Developmental Disorders. EPIGENETICS IN PSYCHIATRY 2014:395-424. [DOI: 10.1016/b978-0-12-417114-5.00019-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
31
|
Scaringi C, Enrici RM, Minniti G. Combining molecular targeted agents with radiation therapy for malignant gliomas. Onco Targets Ther 2013; 6:1079-95. [PMID: 23966794 PMCID: PMC3745290 DOI: 10.2147/ott.s48224] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The expansion in understanding the molecular biology that characterizes cancer cells has led to the rapid development of new agents to target important molecular pathways associated with aberrant activation or suppression of cellular signal transduction pathways involved in gliomagenesis, including epidermal growth factor receptor, vascular endothelial growth factor receptor, mammalian target of rapamycin, and integrins signaling pathways. The use of antiangiogenic agent bevacizumab, epidermal growth factor receptor tyrosine kinase inhibitors gefitinib and erlotinib, mammalian target of rapamycin inhibitors temsirolimus and everolimus, and integrin inhibitor cilengitide, in combination with radiation therapy, has been supported by encouraging preclinical data, resulting in a rapid translation into clinical trials. Currently, the majority of published clinical studies on the use of these agents in combination with radiation and cytotoxic therapies have shown only modest survival benefits at best. Tumor heterogeneity and genetic instability may, at least in part, explain the poor results observed with a single-target approach. Much remains to be learned regarding the optimal combination of targeted agents with conventional chemoradiation, including the use of multipathways-targeted therapies, the selection of patients who may benefit from combined treatments based on molecular biomarkers, and the verification of effective blockade of signaling pathways.
Collapse
Affiliation(s)
- Claudia Scaringi
- Department of Radiation Oncology, Sant'Andrea Hospital, University Sapienza, Rome, Italy
| | | | | |
Collapse
|
32
|
Bartmann AP, Sander JW. Epilepsy treatment: a paradigm shift is urgently need. ARQUIVOS DE NEURO-PSIQUIATRIA 2013; 71:180-2. [DOI: 10.1590/s0004-282x2013000300010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 12/20/2012] [Indexed: 01/01/2023]
Abstract
Epilepsy affects between 5 and 10 people in a 1,000 and carries considerable morbidity and premature mortality. The complex inheritance pattern of a lowered seizure threshold is not fully understood but is likely to be polygenic. In the majority of people with epilepsy, we do not understand the pathophysiology, how a seizure is triggered, and how it can be prevented. In the centennial year of the discovery of the antiepileptic properties of phenobarbital, we have over 20 antiepileptic drugs; however, none have dramatically changed the long-term prognosis of the condition. The cascade of events triggering epilepsy is likely to vary greatly among individuals. The hope for the future is a shift of paradigm away from the symptomatic approach that currently exists. Indeed, once epileptogenesis is fully understood, treatment can be targeted at specific mechanisms, and then we will have truly disease-modifying therapies.
Collapse
Affiliation(s)
- Ana Paula Bartmann
- UCL Institute of Neurology; Chalfont Centre for Epilepsy, United Kingdom
| | - Josemir W. Sander
- UCL Institute of Neurology; Chalfont Centre for Epilepsy, United Kingdom
| |
Collapse
|
33
|
Millan MJ. An epigenetic framework for neurodevelopmental disorders: from pathogenesis to potential therapy. Neuropharmacology 2012; 68:2-82. [PMID: 23246909 DOI: 10.1016/j.neuropharm.2012.11.015] [Citation(s) in RCA: 151] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2012] [Revised: 11/11/2012] [Accepted: 11/22/2012] [Indexed: 12/12/2022]
Abstract
Neurodevelopmental disorders (NDDs) are characterized by aberrant and delayed early-life development of the brain, leading to deficits in language, cognition, motor behaviour and other functional domains, often accompanied by somatic symptoms. Environmental factors like perinatal infection, malnutrition and trauma can increase the risk of the heterogeneous, multifactorial and polygenic disorders, autism and schizophrenia. Conversely, discrete genetic anomalies are involved in Down, Rett and Fragile X syndromes, tuberous sclerosis and neurofibromatosis, the less familiar Phelan-McDermid, Sotos, Kleefstra, Coffin-Lowry and "ATRX" syndromes, and the disorders of imprinting, Angelman and Prader-Willi syndromes. NDDs have been termed "synaptopathies" in reference to structural and functional disturbance of synaptic plasticity, several involve abnormal Ras-Kinase signalling ("rasopathies"), and many are characterized by disrupted cerebral connectivity and an imbalance between excitatory and inhibitory transmission. However, at a different level of integration, NDDs are accompanied by aberrant "epigenetic" regulation of processes critical for normal and orderly development of the brain. Epigenetics refers to potentially-heritable (by mitosis and/or meiosis) mechanisms controlling gene expression without changes in DNA sequence. In certain NDDs, prototypical epigenetic processes of DNA methylation and covalent histone marking are impacted. Conversely, others involve anomalies in chromatin-modelling, mRNA splicing/editing, mRNA translation, ribosome biogenesis and/or the regulatory actions of small nucleolar RNAs and micro-RNAs. Since epigenetic mechanisms are modifiable, this raises the hope of novel therapy, though questions remain concerning efficacy and safety. The above issues are critically surveyed in this review, which advocates a broad-based epigenetic framework for understanding and ultimately treating a diverse assemblage of NDDs ("epigenopathies") lying at the interface of genetic, developmental and environmental processes. This article is part of the Special Issue entitled 'Neurodevelopmental Disorders'.
Collapse
Affiliation(s)
- Mark J Millan
- Unit for Research and Discovery in Neuroscience, IDR Servier, 125 chemin de ronde, 78290 Croissy sur Seine, Paris, France.
| |
Collapse
|
34
|
Stefanaki K, Alexiou GA, Stefanaki C, Prodromou N. Tumors of central and peripheral nervous system associated with inherited genetic syndromes. Pediatr Neurosurg 2012; 48:271-85. [PMID: 23796843 DOI: 10.1159/000351546] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Accepted: 04/18/2013] [Indexed: 11/19/2022]
Abstract
There are several genetic syndromes that predispose to the development of tumors of the nervous system. In the present study, we provide a review of the tumors that are associated with neurofibromatosis type 1, neurofibromatosis type 2, tuberous sclerosis complex, von Hippel-Lindau disease, Li-Fraumeni syndrome, Cowden disease, Turcot syndrome, nevoid basal cell carcinoma syndrome (Gorlin syndrome) and rhabdoid predisposition syndrome, which are the most common.
Collapse
|