51
|
Zhang G, Li Y, Reuss JL, Liu N, Wu C, Li J, Xu S, Wang F, Hazel TG, Cunningham M, Zhang H, Dai Y, Hong P, Zhang P, He J, Feng H, Lu X, Ulmer JL, Johe KK, Xu R. Stable Intracerebral Transplantation of Neural Stem Cells for the Treatment of Paralysis Due to Ischemic Stroke. Stem Cells Transl Med 2019; 8:999-1007. [PMID: 31241246 PMCID: PMC6766600 DOI: 10.1002/sctm.18-0220] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 05/10/2019] [Indexed: 12/14/2022] Open
Abstract
NSI‐566 is a stable, primary adherent neural stem cell line derived from a single human fetal spinal cord and expanded epigenetically with no genetic modification. This cell line is being tested in clinical trials in the U.S. for treatment of amyotrophic lateral sclerosis and spinal cord injury. In a single‐site, phase I study, we evaluated the feasibility and safety of NSI‐566 transplantation for the treatment of hemiparesis due to chronic motor stroke and determined the maximum tolerated dose for future trials. Three cohorts (n = 3 per cohort) were transplanted with one‐time intracerebral injections of 1.2 × 107, 2.4 × 107, or 7.2 × 107 cells. Immunosuppression therapy with tacrolimus was maintained for 28 days. All subjects had sustained chronic motor strokes, verified by magnetic resonance imaging (MRI), initiated between 5 and 24 months prior to surgery with modified Rankin Scores [MRSs] of 2, 3, or 4 and Fugl‐Meyer Motor Scores of 55 or less. At the 12‐month visit, the mean Fugl‐Meyer Motor Score (FMMS, total score of 100) for the nine participants showed 16 points of improvement (p = .0078), the mean MRS showed 0.8 points of improvement (p = .031), and the mean National Institutes of Health Stroke Scale showed 3.1 points of improvement (p = .020). For six participants who were followed up for 24 months, these mean changes remained stable. The treatment was well tolerated at all doses. Longitudinal MRI studies showed evidence indicating cavity‐filling by new neural tissue formation in all nine patients. Although this was a small, one‐arm study of feasibility, the results are encouraging to warrant further studies. stem cells translational medicine2019;8:999–1007
Collapse
Affiliation(s)
- Guangzhu Zhang
- Affiliated BaYi Brain Hospital, Army General Hospital of PLA, Beijing, People's Republic of China
| | - Ying Li
- Neurology Department, Army General Hospital of PLA, Beijing, People's Republic of China
| | - James L Reuss
- Prism Clinical Imaging, Inc., Milwaukee, Wisconsin, USA
| | - Nan Liu
- Neurology Department, Army General Hospital of PLA, Beijing, People's Republic of China
| | - Cuiying Wu
- Affiliated BaYi Brain Hospital, Army General Hospital of PLA, Beijing, People's Republic of China
| | - Jingpo Li
- Suzhou Neuralstem Biopharmaceutical Co., Ltd., Suzhou, People's Republic of China
| | - Shuangshuang Xu
- Suzhou Neuralstem Biopharmaceutical Co., Ltd., Suzhou, People's Republic of China
| | - Feng Wang
- Suzhou Neuralstem Biopharmaceutical Co., Ltd., Suzhou, People's Republic of China
| | | | - Miles Cunningham
- Laboratory for Neural Reconstruction, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
| | - Hongtian Zhang
- Affiliated BaYi Brain Hospital, Army General Hospital of PLA, Beijing, People's Republic of China
| | - Yiwu Dai
- Affiliated BaYi Brain Hospital, Army General Hospital of PLA, Beijing, People's Republic of China
| | - Peng Hong
- Affiliated BaYi Brain Hospital, Army General Hospital of PLA, Beijing, People's Republic of China
| | - Ping Zhang
- Affiliated BaYi Brain Hospital, Army General Hospital of PLA, Beijing, People's Republic of China
| | - Jianghong He
- Affiliated BaYi Brain Hospital, Army General Hospital of PLA, Beijing, People's Republic of China
| | - Huiru Feng
- Department of Nuclear Medicine, Army General Hospital of PLA, Beijing, People's Republic of China
| | - Xiangdong Lu
- Department of Nuclear Medicine, Army General Hospital of PLA, Beijing, People's Republic of China
| | - John L Ulmer
- Department of Neuroradiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | | - Ruxiang Xu
- Affiliated BaYi Brain Hospital, Army General Hospital of PLA, Beijing, People's Republic of China
| |
Collapse
|
52
|
Goutman SA, Savelieff MG, Sakowski SA, Feldman EL. Stem cell treatments for amyotrophic lateral sclerosis: a critical overview of early phase trials. Expert Opin Investig Drugs 2019; 28:525-543. [PMID: 31189354 DOI: 10.1080/13543784.2019.1627324] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease of cortical, brainstem, and spinal motor neurons; it causes progressive muscle weakness and atrophy, respiratory failure, and death. No currently available treatment either stops or reverses this disease. Therapeutics to slow, stop, and reverse ALS are needed. Stem cells may be a viable solution to sustain and nurture diseased motor neurons. Several early-stage clinical trials have been launched to assess the potential of stem cells for ALS treatment. Areas covered: Expert opinion: AREAS COVERED This review covers the key advances from early phase clinical trials of stem cell therapy for ALS and identifies promising avenues and key challenges. EXPERT OPINION Clinical trials in humans are still in the nascent stages of development. It will be critical to ensure that powered, well-controlled trials are conducted, that optimal treatment windows are identified, and that the ideal cell type, cell dose, and delivery site and method are determined. Several trials have used more invasive procedures, and ethical concerns of sham procedures on patients in the control arm and on their safety should be considered.
Collapse
Affiliation(s)
- Stephen A Goutman
- a Department of Neurology , University of Michigan , Ann Arbor , MI , USA.,b Program for Neurology Research & Discovery , University of Michigan , Ann Arbor , MI , USA
| | - Masha G Savelieff
- a Department of Neurology , University of Michigan , Ann Arbor , MI , USA.,b Program for Neurology Research & Discovery , University of Michigan , Ann Arbor , MI , USA
| | - Stacey A Sakowski
- a Department of Neurology , University of Michigan , Ann Arbor , MI , USA.,b Program for Neurology Research & Discovery , University of Michigan , Ann Arbor , MI , USA
| | - Eva L Feldman
- a Department of Neurology , University of Michigan , Ann Arbor , MI , USA.,b Program for Neurology Research & Discovery , University of Michigan , Ann Arbor , MI , USA
| |
Collapse
|
53
|
Mazzini L, Gelati M, Profico DC, Sorarù G, Ferrari D, Copetti M, Muzi G, Ricciolini C, Carletti S, Giorgi C, Spera C, Frondizi D, Masiero S, Stecco A, Cisari C, Bersano E, De Marchi F, Sarnelli MF, Querin G, Cantello R, Petruzzelli F, Maglione A, Zalfa C, Binda E, Visioli A, Trombetta D, Torres B, Bernardini L, Gaiani A, Massara M, Paolucci S, Boulis NM, Vescovi AL. Results from Phase I Clinical Trial with Intraspinal Injection of Neural Stem Cells in Amyotrophic Lateral Sclerosis: A Long-Term Outcome. Stem Cells Transl Med 2019; 8:887-897. [PMID: 31104357 PMCID: PMC6708070 DOI: 10.1002/sctm.18-0154] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 02/19/2019] [Indexed: 12/13/2022] Open
Abstract
The main objective of this phase I trial was to assess the feasibility and safety of microtransplanting human neural stem cell (hNSC) lines into the spinal cord of patients with amyotrophic lateral sclerosis (ALS). Eighteen patients with a definite diagnosis of ALS received microinjections of hNSCs into the gray matter tracts of the lumbar or cervical spinal cord. Patients were monitored before and after transplantation by clinical, psychological, neuroradiological, and neurophysiological assessment. For up to 60 months after surgery, none of the patients manifested severe adverse effects or increased disease progression because of the treatment. Eleven patients died, and two underwent tracheotomy as a result of the natural history of the disease. We detected a transitory decrease in progression of ALS Functional Rating Scale Revised, starting within the first month after surgery and up to 4 months after transplantation. Our results show that transplantation of hNSC is a safe procedure that causes no major deleterious effects over the short or long term. This study is the first example of medical transplantation of a highly standardized cell drug product, which can be reproducibly and stably expanded ex vivo, comprising hNSC that are not immortalized, and are derived from the forebrain of the same two donors throughout this entire study as well as across future trials. Our experimental design provides benefits in terms of enhancing both intra‐ and interstudy reproducibility and homogeneity. Given the potential therapeutic effects of the hNSCs, our observations support undertaking future phase II clinical studies in which increased cell dosages are studied in larger cohorts of patients. stem cells translational medicine2019;8:887&897
Collapse
Affiliation(s)
- Letizia Mazzini
- Eastern Piedmont University, "Maggiore della Carità" Hospital, Dipartimento di Neurologia, Novara
| | - Maurizio Gelati
- Laboratorio Cellule Staminali, Cell Factory e Biobanca, Terni Hospital, Italy.,Fondazione IRCCS Casa Sollievo della Sofferenza, Advanced Therapies Production Unit, San Giovanni Rotondo, Foggia, Italy
| | - Daniela Celeste Profico
- Fondazione IRCCS Casa Sollievo della Sofferenza, Advanced Therapies Production Unit, San Giovanni Rotondo, Foggia, Italy
| | - Gianni Sorarù
- Department of Neuroscience, University of Padua, Padua, Italy
| | - Daniela Ferrari
- Biotechnology and Bioscience Department Bicocca University, Milan, Italy
| | - Massimiliano Copetti
- Fondazione IRCCS Casa Sollievo della Sofferenza, Biostatistic Unit, San Giovanni Rotondo, Foggia, Italy
| | - Gianmarco Muzi
- Laboratorio Cellule Staminali, Cell Factory e Biobanca, Terni Hospital, Italy
| | - Claudia Ricciolini
- Laboratorio Cellule Staminali, Cell Factory e Biobanca, Terni Hospital, Italy
| | - Sandro Carletti
- Department of Neurosurgery and Neuroscience, "Santa Maria" Hospital, Terni, Italy
| | - Cesare Giorgi
- Department of Neurosurgery and Neuroscience, "Santa Maria" Hospital, Terni, Italy
| | - Cristina Spera
- Department of Neurosurgery and Neuroscience, "Santa Maria" Hospital, Terni, Italy
| | - Domenico Frondizi
- Department of Neurosurgery and Neuroscience, "Santa Maria" Hospital, Terni, Italy
| | - Stefano Masiero
- Department of Neuroscience, University of Padua, Padua, Italy
| | - Alessandro Stecco
- Department of Diagnostic and Interventional Radiology, "Eastern Piedmont" University, "Maggiore della Carità" Hospital, Novara
| | - Carlo Cisari
- Department of Physical Therapy, "Eastern Piedmont" University, "Maggiore della Carità" Hospital, Novara
| | - Enrica Bersano
- Eastern Piedmont University, "Maggiore della Carità" Hospital, Dipartimento di Neurologia, Novara
| | - Fabiola De Marchi
- Eastern Piedmont University, "Maggiore della Carità" Hospital, Dipartimento di Neurologia, Novara
| | - Maria Francesca Sarnelli
- Eastern Piedmont University, "Maggiore della Carità" Hospital, Dipartimento di Neurologia, Novara
| | - Giorgia Querin
- Department of Neuroscience, University of Padua, Padua, Italy
| | - Roberto Cantello
- Eastern Piedmont University, "Maggiore della Carità" Hospital, Dipartimento di Neurologia, Novara
| | - Francesco Petruzzelli
- Fondazione IRCCS Casa Sollievo della Sofferenza, Obstetrics and Gynaecology Department, San Giovanni Rotondo, Foggia, Italy
| | - Annamaria Maglione
- Fondazione IRCCS Casa Sollievo della Sofferenza, Obstetrics and Gynaecology Department, San Giovanni Rotondo, Foggia, Italy
| | - Cristina Zalfa
- Biotechnology and Bioscience Department Bicocca University, Milan, Italy
| | - Elena Binda
- Fondazione IRCCS Casa Sollievo della Sofferenza, Cancer Stem Cells Unit, San Giovanni Rotondo, Foggia, Italy
| | | | - Domenico Trombetta
- Fondazione IRCCS Casa Sollievo della Sofferenza, Department of Oncology, San Giovanni Rotondo, Foggia, Italy
| | - Barbara Torres
- Fondazione IRCCS Casa Sollievo della Sofferenza, Cytogenetics Unit, San Giovanni Rotondo, Foggia, Italy
| | - Laura Bernardini
- Fondazione IRCCS Casa Sollievo della Sofferenza, Cytogenetics Unit, San Giovanni Rotondo, Foggia, Italy
| | | | - Maurilio Massara
- Eastern Piedmont University, "Maggiore della Carità" Hospital, Dipartimento di Neurologia, Novara
| | - Silvia Paolucci
- Eastern Piedmont University, "Maggiore della Carità" Hospital, Dipartimento di Neurologia, Novara
| | | | - Angelo L Vescovi
- Laboratorio Cellule Staminali, Cell Factory e Biobanca, Terni Hospital, Italy.,Fondazione IRCCS Casa Sollievo della Sofferenza, Advanced Therapies Production Unit, San Giovanni Rotondo, Foggia, Italy.,Biotechnology and Bioscience Department Bicocca University, Milan, Italy
| | | |
Collapse
|
54
|
Neuroimaging and clinical trials with stem cells in amyotrophic lateral sclerosis: Present and future perspectives. RADIOLOGIA 2019. [DOI: 10.1016/j.rxeng.2019.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
55
|
Zalfa C, Rota Nodari L, Vacchi E, Gelati M, Profico D, Boido M, Binda E, De Filippis L, Copetti M, Garlatti V, Daniele P, Rosati J, De Luca A, Pinos F, Cajola L, Visioli A, Mazzini L, Vercelli A, Svelto M, Vescovi AL, Ferrari D. Transplantation of clinical-grade human neural stem cells reduces neuroinflammation, prolongs survival and delays disease progression in the SOD1 rats. Cell Death Dis 2019; 10:345. [PMID: 31024007 PMCID: PMC6484011 DOI: 10.1038/s41419-019-1582-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 03/26/2019] [Accepted: 03/29/2019] [Indexed: 12/13/2022]
Abstract
Stem cells are emerging as a therapeutic option for incurable diseases, such as Amyotrophic Lateral Sclerosis (ALS). However, critical issues are related to their origin as well as to the need to deepen our knowledge of the therapeutic actions exerted by these cells. Here, we investigate the therapeutic potential of clinical-grade human neural stem cells (hNSCs) that have been successfully used in a recently concluded phase I clinical trial for ALS patients (NCT01640067). The hNSCs were transplanted bilaterally into the anterior horns of the lumbar spinal cord (four grafts each, segments L3–L4) of superoxide dismutase 1 G93A transgenic rats (SOD1 rats) at the symptomatic stage. Controls included untreated SOD1 rats (CTRL) and those treated with HBSS (HBSS). Motor symptoms and histological hallmarks of the disease were evaluated at three progressive time points: 15 and 40 days after transplant (DAT), and end stage. Animals were treated by transient immunosuppression (for 15 days, starting at time of transplantation). Under these conditions, hNSCs integrated extensively within the cord, differentiated into neural phenotypes and migrated rostro-caudally, up to 3.77 ± 0.63 cm from the injection site. The transplanted cells delayed decreases in body weight and deterioration of motor performance in the SOD1 rats. At 40DAT, the anterior horns at L3–L4 revealed a higher density of motoneurons and fewer activated astroglial and microglial cells. Accordingly, the overall survival of transplanted rats was significantly enhanced with no rejection of hNSCs observed. We demonstrated that the beneficial effects observed after stem cell transplantation arises from multiple events that counteract several aspects of the disease, a crucial feature for multifactorial diseases, such as ALS. The combination of therapeutic approaches that target different pathogenic mechanisms of the disorder, including pharmacology, molecular therapy and cell transplantation, will increase the chances of a clinically successful therapy for ALS.
Collapse
Affiliation(s)
- Cristina Zalfa
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2, 20126, Milan, Italy
| | - Laura Rota Nodari
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2, 20126, Milan, Italy
| | - Elena Vacchi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2, 20126, Milan, Italy
| | - Maurizio Gelati
- Fondazione IRCCS Casa Sollievo della Sofferenza, Production Unit of Advanced Therapies (UPTA), Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), 71013, San Giovanni Rotondo, Foggia, Italy
| | - Daniela Profico
- Fondazione IRCCS Casa Sollievo della Sofferenza, Production Unit of Advanced Therapies (UPTA), Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), 71013, San Giovanni Rotondo, Foggia, Italy
| | - Marina Boido
- Neuroscience Institute Cavalieri Ottolenghi, Department of Neuroscience "Rita Levi Montalcini", University of Torino, Torino, Italy
| | - Elena Binda
- Fondazione IRCCS Casa Sollievo della Sofferenza, Cancer Stem Cells Unit, Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), 71013, San Giovanni Rotondo, (FG), Italy
| | - Lidia De Filippis
- Fondazione IRCCS Casa Sollievo della Sofferenza, Regenerative Medicine and Innovative Therapies (ISBReMIT), 71013, San Giovanni Rotondo, (FG), Italy
| | - Massimiliano Copetti
- Fondazione IRCCS Casa Sollievo della Sofferenza, Bioinformatics Unit, Viale dei Cappuccini, 71013, San Giovanni Rotondo, (FG), Italy
| | - Valentina Garlatti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2, 20126, Milan, Italy
| | - Paola Daniele
- Fondazione IRCCS Casa Sollievo della Sofferenza, Molecular Genetics Unit, Viale dei Cappuccini, 71013, San Giovanni Rotondo, (FG), Italy
| | - Jessica Rosati
- Fondazione IRCCS Casa Sollievo della Sofferenza, Cellular Reprogramming Unit, San Giovanni Rotondo, (FG), Italy
| | - Alessandro De Luca
- Fondazione IRCCS Casa Sollievo della Sofferenza, Molecular Genetics Unit, Viale dei Cappuccini, 71013, San Giovanni Rotondo, (FG), Italy
| | - Francesca Pinos
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2, 20126, Milan, Italy
| | - Laura Cajola
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2, 20126, Milan, Italy
| | | | - Letizia Mazzini
- Centro Regionale Esperto SLA Azienda Ospedaliero-Universitaria "Maggiore della Carità", Novara, Italy
| | - Alessandro Vercelli
- Neuroscience Institute Cavalieri Ottolenghi, Department of Neuroscience "Rita Levi Montalcini", University of Torino, Torino, Italy
| | - Maria Svelto
- Department of Bioscience, Biotechnology and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
| | - Angelo Luigi Vescovi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2, 20126, Milan, Italy. .,Fondazione IRCCS Casa Sollievo della Sofferenza, Production Unit of Advanced Therapies (UPTA), Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), 71013, San Giovanni Rotondo, Foggia, Italy. .,Department of Bioscience, Biotechnology and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy.
| | - Daniela Ferrari
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2, 20126, Milan, Italy.
| |
Collapse
|
56
|
Abati E, Bresolin N, Comi G, Corti S. Advances, Challenges, and Perspectives in Translational Stem Cell Therapy for Amyotrophic Lateral Sclerosis. Mol Neurobiol 2019; 56:6703-6715. [DOI: 10.1007/s12035-019-1554-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 03/13/2019] [Indexed: 12/13/2022]
|
57
|
Mazzini L, Ferrari D, Andjus PR, Buzanska L, Cantello R, De Marchi F, Gelati M, Giniatullin R, Glover JC, Grilli M, Kozlova EN, Maioli M, Mitrečić D, Pivoriunas A, Sanchez-Pernaute R, Sarnowska A, Vescovi AL. Advances in stem cell therapy for amyotrophic lateral sclerosis. Expert Opin Biol Ther 2019; 18:865-881. [PMID: 30025485 DOI: 10.1080/14712598.2018.1503248] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Amyotrophic Lateral Sclerosis (ALS) is a progressive, incurable neurodegenerative disease that targets motoneurons. Cell-based therapies have generated widespread interest as a potential therapeutic approach but no conclusive results have yet been reported either from pre-clinical or clinical studies. AREAS COVERED This is an integrated review of pre-clinical and clinical studies focused on the development of cell-based therapies for ALS. We analyze the biology of stem cell treatments and results obtained from pre-clinical models of ALS and examine the methods and the results obtained to date from clinical trials. We discuss scientific, clinical, and ethical issues and propose some directions for future studies. EXPERT OPINION While data from individual studies are encouraging, stem-cell-based therapies do not yet represent a satisfactory, reliable clinical option. The field will critically benefit from the introduction of well-designed, randomized and reproducible, powered clinical trials. Comparative studies addressing key issues such as the nature, properties, and number of donor cells, the delivery mode and the selection of proper patient populations that may benefit the most from cell-based therapies are now of the essence. Multidisciplinary networks of experts should be established to empower effective translation of research into the clinic.
Collapse
Affiliation(s)
- Letizia Mazzini
- a ALS Centre Department of Neurology , "Maggiore della Carità" University Hospital Novara , Novara , Italy
| | - Daniela Ferrari
- b Department of Biotechnology and Biosciences , University Milano Bicocca , Milano , Italy
| | - Pavle R Andjus
- c Center for laser microscopy, Faculty of Biology , University of Belgrade , Belgrade , Serbia
| | - Leonora Buzanska
- d Stem Cell Bioengineering Unit , Mossakowski Medical Research Center, Polish Academy of Sciences , Warsaw , Poland
| | - Roberto Cantello
- a ALS Centre Department of Neurology , "Maggiore della Carità" University Hospital Novara , Novara , Italy
| | - Fabiola De Marchi
- a ALS Centre Department of Neurology , "Maggiore della Carità" University Hospital Novara , Novara , Italy
| | - Maurizio Gelati
- e Scientific Direction , IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo , Foggia , Italy.,f Cell Factory e biobanca, Fondazione Cellule Staminali , Terni , Italy
| | - Rashid Giniatullin
- g A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland , Neulaniementie 2, Kuopio , FINLAND
| | - Joel C Glover
- h Department of Molecular Medicine , Institute of Basic Medical Sciences, University of Oslo and Norwegian Center for Stem Cell Research, Oslo University Hospital , Oslo , Norway
| | - Mariagrazia Grilli
- i Department Pharmaceutical Sciences , Laboratory of Neuroplasticity, University of Piemonte Orientale , Novara , Italy
| | - Elena N Kozlova
- j Department of Neuroscience , Uppsala University Biomedical Centre , Uppsala , Sweden
| | - Margherita Maioli
- k Department of Biomedical Sciences and Center for Developmental Biology and Reprogramming (CEDEBIOR) , University of Sassari, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR) , Sassari , Italy
| | - Dinko Mitrečić
- l Laboratory for Stem Cells, Croatian Institute for Brain Research , University of Zagreb School of Medicine , Zagreb , Croatia
| | - Augustas Pivoriunas
- m Department of Stem Cell Biology , State Research Institute Centre for Innovative Medicine , Vilnius , Lithuania
| | - Rosario Sanchez-Pernaute
- n Preclinical Research , Andalusian Initiative for Advanced Therapies, Andalusian Health Ministry , Sevilla , Spain
| | - Anna Sarnowska
- d Stem Cell Bioengineering Unit , Mossakowski Medical Research Center, Polish Academy of Sciences , Warsaw , Poland
| | - Angelo L Vescovi
- b Department of Biotechnology and Biosciences , University Milano Bicocca , Milano , Italy.,f Cell Factory e biobanca, Fondazione Cellule Staminali , Terni , Italy
| | | |
Collapse
|
58
|
Yokobori S, Sasaki K, Kanaya T, Igarashi Y, Nakae R, Onda H, Masuno T, Suda S, Sowa K, Nakajima M, Spurlock MS, Onn Chieng L, Hazel TG, Johe K, Gajavelli S, Fuse A, Bullock MR, Yokota H. Feasibility of Human Neural Stem Cell Transplantation for the Treatment of Acute Subdural Hematoma in a Rat Model: A Pilot Study. Front Neurol 2019; 10:82. [PMID: 30809187 PMCID: PMC6379455 DOI: 10.3389/fneur.2019.00082] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 01/21/2019] [Indexed: 12/20/2022] Open
Abstract
Human neural stem cells (hNSCs) transplantation in several brain injury models has established their therapeutic potential. However, the feasibility of hNSCs transplantation is still not clear for acute subdural hematoma (ASDH) brain injury that needs external decompression. Thus, the aim of this pilot study was to test feasibility using a rat ASDH decompression model with two clinically relevant transplantation methods. Two different methods, in situ stereotactic injection and hNSC-embedded matrix seating on the brain surface, were attempted. Athymic rats were randomized to uninjured or ASDH groups (F344/NJcl-rnu/rnu, n = 7-10/group). Animals in injury group were subjected to ASDH, and received decompressive craniectomy and 1-week after decompression surgery were transplanted with green fluorescent protein (GFP)-transduced hNSCs using one of two approaches. Histopathological examinations at 4 and 8 weeks showed that the GFP-positive hNSCs survived in injured brain tissue, extended neurite-like projections resembling neural dendrites. The in situ transplantation group had greater engraftment of hNSCs than matrix embedding approach. Immunohistochemistry with doublecortin, NeuN, and GFAP at 8 weeks after transplantation showed that transplanted hNSCs remained as immature neurons and did not differentiate toward to glial cell lines. Motor function was assessed with rotarod, compared to control group (n = 10). The latency to fall from the rotarod in hNSC in situ transplanted rats was significantly higher than in control rats (median, 113 s in hNSC vs. 69 s in control, P = 0.02). This study first demonstrates the robust engraftment of in situ transplanted hNSCs in a clinically-relevant ASDH decompression rat model. Further preclinical studies with longer study duration are warranted to verify the effectiveness of hNSC transplantation in amelioration of TBI induced deficits.
Collapse
Affiliation(s)
- Shoji Yokobori
- Department of Emergency and Critical Care Medicine, Nippon Medical School, Tokyo, Japan
| | - Kazuma Sasaki
- Department of Emergency and Critical Care Medicine, Nippon Medical School, Tokyo, Japan
| | - Takahiro Kanaya
- Department of Emergency and Critical Care Medicine, Nippon Medical School, Tokyo, Japan
| | - Yutaka Igarashi
- Department of Emergency and Critical Care Medicine, Nippon Medical School, Tokyo, Japan
| | - Ryuta Nakae
- Department of Emergency and Critical Care Medicine, Nippon Medical School, Tokyo, Japan
| | - Hidetaka Onda
- Department of Emergency and Critical Care Medicine, Nippon Medical School, Tokyo, Japan
| | - Tomohiko Masuno
- Department of Emergency and Critical Care Medicine, Nippon Medical School, Tokyo, Japan
| | - Satoshi Suda
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Kota Sowa
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Masataka Nakajima
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Markus S. Spurlock
- Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Lee Onn Chieng
- Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | | | - Karl Johe
- Neuralstem, Inc., Germantown, MD, United States
| | - Shyam Gajavelli
- Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Akira Fuse
- Department of Emergency and Critical Care Medicine, Nippon Medical School, Tokyo, Japan
| | - M. Ross Bullock
- Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Hiroyuki Yokota
- Department of Emergency and Critical Care Medicine, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
59
|
Dwivedi N, Shah J, Mishra V, Tambuwala M, Kesharwani P. Nanoneuromedicine for management of neurodegenerative disorder. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2018.12.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
60
|
Chen KS, McGinley LM, Kashlan ON, Hayes JM, Bruno ES, Chang JS, Mendelson FE, Tabbey MA, Johe K, Sakowski SA, Feldman EL. Targeted intraspinal injections to assess therapies in rodent models of neurological disorders. Nat Protoc 2019; 14:331-349. [PMID: 30610242 DOI: 10.1038/s41596-018-0095-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Despite decades of research, pharmacological therapies for spinal cord motor pathologies are limited. Alternatives using macromolecular, viral, or cell-based therapies show early promise. However, introducing these substances into the spinal cord, past the blood-brain barrier, without causing injury is challenging. We describe a technique for intraspinal injection targeting the lumbar ventral horn in rodents. This technique preserves motor performance and has a proven track record of translation into phase 1 and 2 clinical trials in amyotrophic lateral sclerosis (ALS) patients. The procedure, in brief, involves exposure of the thoracolumbar spine and dissection of paraspinous muscles over the target vertebrae. Following laminectomy, the spine is affixed to a stereotactic frame, permitting precise and reproducible injection throughout the lumbar spine. We have used this protocol to inject various stem cell types, primarily human spinal stem cells (HSSCs); however, the injection is adaptable to any candidate therapeutic cell, virus, or macromolecule product. In addition to a detailed procedure, we provide stereotactic coordinates that assist in targeting of the lumbar spine and instructional videos. The protocol takes ~2 h per animal.
Collapse
Affiliation(s)
- Kevin S Chen
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Lisa M McGinley
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Osama N Kashlan
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - John M Hayes
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | | | - Josh S Chang
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Faye E Mendelson
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Maegan A Tabbey
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | | | | | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
61
|
Kassi AAY, Mahavadi AK, Clavijo A, Caliz D, Lee SW, Ahmed AI, Yokobori S, Hu Z, Spurlock MS, Wasserman JM, Rivera KN, Nodal S, Powell HR, Di L, Torres R, Leung LY, Rubiano AM, Bullock RM, Gajavelli S. Enduring Neuroprotective Effect of Subacute Neural Stem Cell Transplantation After Penetrating TBI. Front Neurol 2019; 9:1097. [PMID: 30719019 PMCID: PMC6348935 DOI: 10.3389/fneur.2018.01097] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 12/03/2018] [Indexed: 12/13/2022] Open
Abstract
Traumatic brain injury (TBI) is the largest cause of death and disability of persons under 45 years old, worldwide. Independent of the distribution, outcomes such as disability are associated with huge societal costs. The heterogeneity of TBI and its complicated biological response have helped clarify the limitations of current pharmacological approaches to TBI management. Five decades of effort have made some strides in reducing TBI mortality but little progress has been made to mitigate TBI-induced disability. Lessons learned from the failure of numerous randomized clinical trials and the inability to scale up results from single center clinical trials with neuroprotective agents led to the formation of organizations such as the Neurological Emergencies Treatment Trials (NETT) Network, and international collaborative comparative effectiveness research (CER) to re-orient TBI clinical research. With initiatives such as TRACK-TBI, generating rich and comprehensive human datasets with demographic, clinical, genomic, proteomic, imaging, and detailed outcome data across multiple time points has become the focus of the field in the United States (US). In addition, government institutions such as the US Department of Defense are investing in groups such as Operation Brain Trauma Therapy (OBTT), a multicenter, pre-clinical drug-screening consortium to address the barriers in translation. The consensus from such efforts including "The Lancet Neurology Commission" and current literature is that unmitigated cell death processes, incomplete debris clearance, aberrant neurotoxic immune, and glia cell response induce progressive tissue loss and spatiotemporal magnification of primary TBI. Our analysis suggests that the focus of neuroprotection research needs to shift from protecting dying and injured neurons at acute time points to modulating the aberrant glial response in sub-acute and chronic time points. One unexpected agent with neuroprotective properties that shows promise is transplantation of neural stem cells. In this review we present (i) a short survey of TBI epidemiology and summary of current care, (ii) findings of past neuroprotective clinical trials and possible reasons for failure based upon insights from human and preclinical TBI pathophysiology studies, including our group's inflammation-centered approach, (iii) the unmet need of TBI and unproven treatments and lastly, (iv) present evidence to support the rationale for sub-acute neural stem cell therapy to mediate enduring neuroprotection.
Collapse
Affiliation(s)
- Anelia A. Y. Kassi
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Anil K. Mahavadi
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Angelica Clavijo
- Neurosurgery Service, INUB-MEDITECH Research Group, El Bosque University, Bogotá, CO, United States
| | - Daniela Caliz
- Neurosurgery Service, INUB-MEDITECH Research Group, El Bosque University, Bogotá, CO, United States
| | - Stephanie W. Lee
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Aminul I. Ahmed
- Wessex Neurological Centre, University Hospitals Southampton, Southampton, United Kingdom
| | - Shoji Yokobori
- Department of Emergency and Critical Care Medicine, Nippon Medical School, Tokyo, Japan
| | - Zhen Hu
- Department of Neurosurgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Markus S. Spurlock
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Joseph M Wasserman
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Karla N. Rivera
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Samuel Nodal
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Henry R. Powell
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Long Di
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Rolando Torres
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Lai Yee Leung
- Branch of Brain Trauma Neuroprotection and Neurorestoration, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Andres Mariano Rubiano
- Neurosurgery Service, INUB-MEDITECH Research Group, El Bosque University, Bogotá, CO, United States
| | - Ross M. Bullock
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Shyam Gajavelli
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
62
|
Richard JP, Hussain U, Gross S, Taga A, Kouser M, Almad A, Campanelli JT, Bulte JWM, Maragakis NJ. Perfluorocarbon Labeling of Human Glial-Restricted Progenitors for 19 F Magnetic Resonance Imaging. Stem Cells Transl Med 2019; 8:355-365. [PMID: 30618148 PMCID: PMC6431733 DOI: 10.1002/sctm.18-0094] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 11/21/2018] [Indexed: 12/20/2022] Open
Abstract
One of the fundamental limitations in assessing potential efficacy in Central Nervous System (CNS) transplantation of stem cells is the capacity for monitoring cell survival and migration noninvasively and longitudinally. Human glial‐restricted progenitor (hGRP) cells (Q‐Cells) have been investigated for their utility in providing neuroprotection following transplantation into models of amyotrophic lateral sclerosis (ALS) and have been granted a Food and Drug Administration (FDA) Investigational New Drug (IND) for intraspinal transplantation in ALS patients. Furthermore, clinical development of these cells for therapeutic use will rely on the ability to track the cells using noninvasive imaging methodologies as well as the verification that the transplanted GRPs have disease‐relevant activity. As a first step in development, we investigated the use of a perfluorocarbon (PFC) dual‐modal (19F magnetic resonance imaging [MRI] and fluorescence) tracer agent to label Q‐Cells in culture and following spinal cord transplantation. PFCs have a number of potential benefits that make them appealing for clinical use. They are quantitative, noninvasive, biologically inert, and highly specific. In this study, we developed optimized PFC labeling protocols for Q‐Cells and demonstrate that PFCs do not significantly alter the glial identity of Q‐Cells. We also show that PFCs do not interfere with the capacity for differentiation into astrocytes either in vitro or following transplantation into the ventral horn of the mouse spinal cord, and can be visualized in vivo by hot spot 19F MRI. These studies provide a foundation for further preclinical development of PFCs within the context of evaluating Q‐Cell transplantation in the brain and spinal cord of future ALS patients using 19F MRI. stem cells translational medicine2019;8:355–365
Collapse
Affiliation(s)
- Jean-Philippe Richard
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Uzma Hussain
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sarah Gross
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Arens Taga
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mehreen Kouser
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Akshata Almad
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Jeff W M Bulte
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nicholas J Maragakis
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
63
|
Neuroimaging and clinical trials with stem cells in amyotrophic lateral sclerosis: present and future perspectives. RADIOLOGIA 2019; 61:183-190. [PMID: 30606510 DOI: 10.1016/j.rx.2018.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 11/01/2018] [Accepted: 11/17/2018] [Indexed: 11/20/2022]
Abstract
Amyotrophic lateral sclerosis is a rare neurodegenerative disease with a rapid fatal course. The absence of effective treatments has led to new lines of research, some of which are based on stem cells. Surgical injection into the spinal cord, the most common route of administration of stem cells, has proven safe in trials to test the safety of the procedure. Nevertheless, challenges remain, such as determining the best route of administration or the way of checking the survival of the cells and their interaction with the therapeutic target. To date, the mission of neuroimaging techniques has been to detect lesions and complications in the spine and spinal cord, but neuroimaging also has the potential to supplant histologic study in analyzing the relations between the implanted cells and the therapeutic target, and as biomarkers of the disease, by measuring morphological and functional changes after treatment. These developments would increase the role of radiologists in the clinical management of patients with amyotrophic lateral sclerosis.
Collapse
|
64
|
Goncalves K, Przyborski S. The utility of stem cells for neural regeneration. Brain Neurosci Adv 2018; 2:2398212818818071. [PMID: 32166173 PMCID: PMC7058206 DOI: 10.1177/2398212818818071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Indexed: 12/22/2022] Open
Abstract
The use of stem cells in biomedical research is an extremely active area of science. This is because they provide tools that can be used both in vivo and vitro to either replace cells lost in degenerative processes, or to model such diseases to elucidate their underlying mechanisms. This review aims to discuss the use of stem cells in terms of providing regeneration within the nervous system, which is particularly important as neurons of the central nervous system lack the ability to inherently regenerate and repair lost connections. As populations are ageing, incidence of neurodegenerative diseases are increasing, highlighting the need to better understand the regenerative capacity and many uses of stem cells in this field.
Collapse
Affiliation(s)
| | - Stefan Przyborski
- Department of Biosciences, Durham University, Durham, UK.,Reprocell Europe, Sedgefield, UK
| |
Collapse
|
65
|
Feng S, Xiao J, Han F, Chen L, Gao W, Mao G, Huang H. Neurorestorative clinical application standards for the culture and quality control of neural progenitor/precursor cells (version 2017). JOURNAL OF NEURORESTORATOLOGY 2018. [DOI: 10.2147/jn.s147917] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
66
|
Mikhailov A, Sankai Y. Donation of neural stem cells? Post mortal viability of spinal cord neuronal cells. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2018; 2018:5333-5337. [PMID: 30441541 DOI: 10.1109/embc.2018.8513487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Transplantation of cells into central nervous system (CNS) shows a potential for treatment of post-traumatic and neurodegenerative diseases. Cadaver-derived neural cells can help reducing deficit of allogeneic material ready for transplantation. In this study we analyze post-mortal survival of spinal cord neural cells. Maximal time when alive neuronal cells can be recovered form spinal cord of the animals was determined as 56hr for human-size animal and 18hr for rat. Cells with surface expression of ganglioside GD2 and antigen CD24 constituted up to one percent of all recovered alive cells in earlier samples with time dependent decline in percentage. GD2-positive cells from rat spinal cord demonstrated spontaneous and induced electrical activity, which reduces with time post mortem.
Collapse
|
67
|
Oskarsson B, Gendron TF, Staff NP. Amyotrophic Lateral Sclerosis: An Update for 2018. Mayo Clin Proc 2018; 93:1617-1628. [PMID: 30401437 DOI: 10.1016/j.mayocp.2018.04.007] [Citation(s) in RCA: 195] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 03/12/2018] [Accepted: 04/13/2018] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease affecting motor neurons and other neuronal cells, leading to severe disability and eventually death from ventilatory failure. It has a prevalence of 5 in 100,000, with an incidence of 1.7 per 100,000, reflecting short average survival. The pathogenesis is incompletely understood, but defects of RNA processing and protein clearance may be fundamental. Repeat expansions in the chromosome 9 open reading frame 72 gene (C9orf72) are the most common known genetic cause of ALS and are seen in approximately 40% of patients with a family history and approximately 10% of those without. No environmental risk factors are proved to be causative, but many have been proposed, including military service. The diagnosis of ALS rests on a history of painless progressive weakness coupled with examination findings of upper and lower motor dysfunction. No diagnostic test is yet available, but electromyography and genetic tests can support the diagnosis. Care for patients is best provided by a multidisciplinary team, and most interventions are directed at managing symptoms. Two medications with modest benefits have Food and Drug Administration approval for the treatment of ALS: riluzole, a glutamate receptor antagonist, and, new in 2017, edaravone, a free radical scavenger. Many other encouraging treatment strategies are being explored in clinical trials for ALS; herein we review stem cell and antisense oligonucleotide gene therapies.
Collapse
|
68
|
Abstract
ALS is a neurodegenerative disease in which the primary symptoms result in progressive neuromuscular weakness. Recent studies have highlighted that there is significant heterogeneity with regard to anatomical and temporal disease progression. Importantly, more recent advances in genetics have revealed new causative genes to the disease. New efforts have focused on the development of biomarkers that could aid in diagnosis, prognosis, and serve as pharmacodynamics markers. Although traditional pharmaceuticals continue to undergo trials for ALS, new therapeutic strategies including stem cell transplantation studies, gene therapies, and antisense therapies targeting some of the familial forms of ALS are gaining momentum.
Collapse
|
69
|
Yeon JY, Hwang JY, Lee HW, Pyeon HJ, Won JS, Noh YJ, Nam H, Joo KM. Optimized Clump Culture Methods for Adult Human Multipotent Neural Cells. Int J Mol Sci 2018; 19:ijms19113380. [PMID: 30380605 PMCID: PMC6274905 DOI: 10.3390/ijms19113380] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 10/24/2018] [Accepted: 10/25/2018] [Indexed: 12/30/2022] Open
Abstract
Adult human multipotent neural cell (ahMNC) is a candidate for regeneration therapy for neurodegenerative diseases. Here, we developed a primary clump culture method for ahMNCs to increase the efficiency of isolation and in vitro expansion. The same amount of human temporal lobe (1 g) was partially digested and then filtered through strainers with various pore sizes, resulting in four types of clumps: Clump I > 100 µm, 70 µm < Clump II < 100 µm, 40 µm < Clump III < 70 µm, and Clump IV < 40 µm. At 3 and 6 days after culture, Clump II showed significantly higher number of colonies than the other Clumps. Moreover, ahMNCs derived from Clump II (ahMNCs-Clump II) showed stable proliferation, and shortened the time to first passage from 19 to 15 days, and the time to 1 × 109 cells from 42 to 34 days compared with the previous single-cell method. ahMNCs-Clump II had neural differentiation and pro-angiogenic potentials, which are the characteristics of ahMNCs. In conclusion, the novel clump culture method for ahMNCs has significantly higher efficiency than previous techniques. Considering the small amount of available human brain tissue, the clump culture method would promote further clinical applications of ahMNCs.
Collapse
Affiliation(s)
- Je Young Yeon
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea.
- Stem Cell and Regenerative Medicine Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea.
| | - Ji-Yoon Hwang
- Stem Cell and Regenerative Medicine Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea.
- Department of Anatomy & Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea.
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16419, Korea.
| | - Hye Won Lee
- Department of Anatomy & Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea.
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16419, Korea.
| | - Hee-Jang Pyeon
- Stem Cell and Regenerative Medicine Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea.
- Department of Anatomy & Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea.
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16419, Korea.
| | - Jeong-Seob Won
- Stem Cell and Regenerative Medicine Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea.
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16419, Korea.
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea.
| | - Yoo-Jung Noh
- Stem Cell and Regenerative Medicine Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea.
- Department of Anatomy & Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea.
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16419, Korea.
| | - Hyun Nam
- Stem Cell and Regenerative Medicine Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea.
- Department of Anatomy & Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea.
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16419, Korea.
| | - Kyeung Min Joo
- Stem Cell and Regenerative Medicine Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea.
- Department of Anatomy & Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea.
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16419, Korea.
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea.
| |
Collapse
|
70
|
Donnelly EM, Kubelick KP, Dumani DS, Emelianov SY. Photoacoustic Image-Guided Delivery of Plasmonic-Nanoparticle-Labeled Mesenchymal Stem Cells to the Spinal Cord. NANO LETTERS 2018; 18:6625-6632. [PMID: 30160124 DOI: 10.1021/acs.nanolett.8b03305] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Regenerative therapies using stem cells have great potential for treating neurodegenerative diseases and traumatic injuries in the spinal cord. In spite of significant research efforts, many therapies fail at the clinical phase. As stem cell technologies advance toward clinical use, there is a need for a minimally invasive, safe, affordable, and real-time imaging technique that allows for the accurate and safe monitoring of stem cell delivery in the operating room. In this work, we present a combined ultrasound and photoacoustic imaging tool to provide image-guided needle placement and monitoring of nanoparticle-labeled stem cell delivery into the spinal cord. We successfully tagged stem cells using gold nanospheres and provided image-guided delivery of stem cells into the spinal cord in real-time, detecting as few as 1000 cells. Ultrasound and photoacoustic imaging was used to guide needle placement for direct stem cell injection to minimize the risk of needle shear and accidental injury and to improve therapeutic outcomes with accurate, localized stem cell delivery. Following injections of various volumes of cells, three-dimensional ultrasound and photoacoustic images allowed the visualization of stem cell distribution along the spinal cord, showing the potential to monitor the migration of the cells in the future. The feasibility of quantitative imaging was also shown by correlating the total photoacoustic signal over the imaging volume to the volume of cells injected. Overall, the presented method may allow clinicians to utilize imaged-guided delivery for more accurate and safer stem cell delivery to the spinal cord.
Collapse
Affiliation(s)
- Eleanor M Donnelly
- School of Electrical and Computer Engineering , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| | - Kelsey P Kubelick
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University School of Medicine , Atlanta , Georgia 30332 , United States
| | - Diego S Dumani
- School of Electrical and Computer Engineering , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University School of Medicine , Atlanta , Georgia 30332 , United States
| | - Stanislav Y Emelianov
- School of Electrical and Computer Engineering , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University School of Medicine , Atlanta , Georgia 30332 , United States
| |
Collapse
|
71
|
McGinley LM, Kashlan ON, Bruno ES, Chen KS, Hayes JM, Kashlan SR, Raykin J, Johe K, Murphy GG, Feldman EL. Human neural stem cell transplantation improves cognition in a murine model of Alzheimer's disease. Sci Rep 2018; 8:14776. [PMID: 30283042 PMCID: PMC6170460 DOI: 10.1038/s41598-018-33017-6] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 09/17/2018] [Indexed: 02/06/2023] Open
Abstract
Stem cell transplantation offers a potentially transformative approach to treating neurodegenerative disorders. The safety of cellular therapies is established in multiple clinical trials, including our own in amyotrophic lateral sclerosis. To initiate similar trials in Alzheimer's disease, efficacious cell lines must be identified. Here, we completed a preclinical proof-of-concept study in the APP/PS1 murine model of Alzheimer's disease. Human neural stem cell transplantation targeted to the fimbria fornix significantly improved cognition in two hippocampal-dependent memory tasks at 4 and 16 weeks post-transplantation. While levels of synapse-related proteins and cholinergic neurons were unaffected, amyloid plaque load was significantly reduced in stem cell transplanted mice and associated with increased recruitment of activated microglia. In vitro, these same neural stem cells induced microglial activation and amyloid phagocytosis, suggesting an immunomodulatory capacity. Although long-term transplantation resulted in significant functional and pathological improvements in APP/PS1 mice, stem cells were not identified by immunohistochemistry or PCR at the study endpoint. These data suggest integration into native tissue or the idea that transient engraftment may be adequate for therapeutic efficacy, reducing the need for continued immunosuppression. Overall, our results support further preclinical development of human neural stem cells as a safe and effective therapy for Alzheimer's disease.
Collapse
Affiliation(s)
- Lisa M McGinley
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Osama N Kashlan
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | | | - Kevin S Chen
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - John M Hayes
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Samy R Kashlan
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Julia Raykin
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Karl Johe
- Neuralstem, Inc, Germantown, MD, USA
| | - Geoffrey G Murphy
- Department of Molecular & Integrative Physiology, Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
72
|
|
73
|
Rosati J, Ferrari D, Altieri F, Tardivo S, Ricciolini C, Fusilli C, Zalfa C, Profico DC, Pinos F, Bernardini L, Torres B, Manni I, Piaggio G, Binda E, Copetti M, Lamorte G, Mazza T, Carella M, Gelati M, Valente EM, Simeone A, Vescovi AL. Establishment of stable iPS-derived human neural stem cell lines suitable for cell therapies. Cell Death Dis 2018; 9:937. [PMID: 30224709 PMCID: PMC6141489 DOI: 10.1038/s41419-018-0990-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 07/30/2018] [Accepted: 08/01/2018] [Indexed: 12/13/2022]
Abstract
Establishing specific cell lineages from human induced pluripotent stem cells (hiPSCs) is vital for cell therapy approaches in regenerative medicine, particularly for neurodegenerative disorders. While neural precursors have been induced from hiPSCs, the establishment of hiPSC-derived human neural stem cells (hiNSCs), with characteristics that match foetal hNSCs and abide by cGMP standards, thus allowing clinical applications, has not been described. We generated hiNSCs by a virus-free technique, whose properties recapitulate those of the clinical-grade hNSCs successfully used in an Amyotrophic Lateral Sclerosis (ALS) phase I clinical trial. Ex vivo, hiNSCs critically depend on exogenous mitogens for stable self-renewal and amplification and spontaneously differentiate into astrocytes, oligodendrocytes and neurons upon their removal. In the brain of immunodeficient mice, hiNSCs engraft and differentiate into neurons and glia, without tumour formation. These findings now warrant the establishment of clinical-grade, autologous and continuous hiNSC lines for clinical trials in neurological diseases such as Huntington’s, Parkinson’s and Alzheimer’s, among others.
Collapse
Affiliation(s)
- Jessica Rosati
- Cellular Reprogramming Unit, IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini, 71013, San Giovanni Rotondo, Foggia, Italy.
| | - Daniela Ferrari
- Department of Biotechnology and Biosciences, University of Milan Bicocca, Piazza della Scienza, 220126, Milan, Italy
| | - Filomena Altieri
- Cellular Reprogramming Unit, IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Silvia Tardivo
- Neurogenetics Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Claudia Ricciolini
- Stem Cell Laboratory, Cell Factory e Biobank, Terni Hospital, Via Tristano di Joannuccio 1, 05100, Terni, Italy
| | - Caterina Fusilli
- Bioinformatics Unit, IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Cristina Zalfa
- Department of Biotechnology and Biosciences, University of Milan Bicocca, Piazza della Scienza, 220126, Milan, Italy
| | - Daniela C Profico
- Production Unit of Advanced Therapies (UPTA), Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Francesca Pinos
- Department of Biotechnology and Biosciences, University of Milan Bicocca, Piazza della Scienza, 220126, Milan, Italy
| | - Laura Bernardini
- Medical Genetics Unit, IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Barbara Torres
- Medical Genetics Unit, IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Isabella Manni
- Department of Research, Diagnosis and Innovative Technologies, Regina Elena National Cancer Institute, Rome, Italy
| | - Giulia Piaggio
- Department of Research, Diagnosis and Innovative Technologies, Regina Elena National Cancer Institute, Rome, Italy
| | - Elena Binda
- Cancer Stem Cells Unit (ICS), Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Massimiliano Copetti
- Biostatistic Unit, IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Giuseppe Lamorte
- Medical Genetics Unit, IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Tommaso Mazza
- Bioinformatics Unit, IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Massimo Carella
- Medical Genetics Unit, IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Maurizio Gelati
- Department of Biotechnology and Biosciences, University of Milan Bicocca, Piazza della Scienza, 220126, Milan, Italy.,Stem Cell Laboratory, Cell Factory e Biobank, Terni Hospital, Via Tristano di Joannuccio 1, 05100, Terni, Italy.,Production Unit of Advanced Therapies (UPTA), Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Enza Maria Valente
- Neurogenetics Unit, IRCCS Santa Lucia Foundation, Rome, Italy.,Department of Molecular Medicine, University of Pavia, Via Forlanini 14, 27100, Pavia, Italy
| | - Antonio Simeone
- Institute of Genetics and Biophysics Adriano Buzzati Traverso, CNR, Via P. Castellino 111, 80131, Naples, Italy.,IRCSS Neuromed, 86077, Pozzilli, Isernia, Italy
| | - Angelo L Vescovi
- Cellular Reprogramming Unit, IRCCS Casa Sollievo della Sofferenza, Viale dei Cappuccini, 71013, San Giovanni Rotondo, Foggia, Italy. .,Department of Biotechnology and Biosciences, University of Milan Bicocca, Piazza della Scienza, 220126, Milan, Italy. .,Stem Cell Laboratory, Cell Factory e Biobank, Terni Hospital, Via Tristano di Joannuccio 1, 05100, Terni, Italy.
| |
Collapse
|
74
|
Gurusamy N, Alsayari A, Rajasingh S, Rajasingh J. Adult Stem Cells for Regenerative Therapy. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 160:1-22. [PMID: 30470288 DOI: 10.1016/bs.pmbts.2018.07.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cell therapy has been identified as an effective method to regenerate damaged tissue. Adult stem cells, also known as somatic stem cells or resident stem cells, are a rare population of undifferentiated cells, located within a differentiated organ, in a specialized structure, called a niche, which maintains the microenvironments that regulate the growth and development of adult stem cells. The adult stem cells are self-renewing, clonogenic, and multipotent in nature, and their main role is to maintain the tissue homeostasis. They can be activated to proliferate and differentiate into the required type of cells, upon the loss of cells or injury to the tissue. Adult stem cells have been identified in many tissues including blood, intestine, skin, muscle, brain, and heart. Extensive preclinical and clinical studies have demonstrated the structural and functional regeneration capabilities of these adult stem cells, such as bone marrow-derived mononuclear cells, hematopoietic stem cells, mesenchymal stromal/stem cells, resident adult stem cells, induced pluripotent stem cells, and umbilical cord stem cells. In this review, we focus on the human therapies, utilizing adult stem cells for their regenerative capabilities in the treatment of cardiac, brain, pancreatic, and eye disorders.
Collapse
Affiliation(s)
- Narasimman Gurusamy
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Abdulrhman Alsayari
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Sheeja Rajasingh
- Department of Internal Medicine, University of Kansas Medical Center, Kansas, KS, United States
| | - Johnson Rajasingh
- Department of Internal Medicine, University of Kansas Medical Center, Kansas, KS, United States.
| |
Collapse
|
75
|
Abati E, Bresolin N, Comi GP, Corti S. Preconditioning and Cellular Engineering to Increase the Survival of Transplanted Neural Stem Cells for Motor Neuron Disease Therapy. Mol Neurobiol 2018; 56:3356-3367. [PMID: 30120734 DOI: 10.1007/s12035-018-1305-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 08/07/2018] [Indexed: 12/11/2022]
Abstract
Despite the extensive research effort that has been made in the field, motor neuron diseases, namely, amyotrophic lateral sclerosis and spinal muscular atrophies, still represent an overwhelming cause of morbidity and mortality worldwide. Exogenous neural stem cell-based transplantation approaches have been investigated as multifaceted strategies to both protect and repair upper and lower motor neurons from degeneration and inflammation. Transplanted neural stem cells (NSCs) exert their beneficial effects not only through the replacement of damaged cells but also via bystander immunomodulatory and neurotrophic actions. Notwithstanding these promising findings, the clinical translatability of such techniques is jeopardized by the limited engraftment success and survival of transplanted cells within the hostile disease microenvironment. To overcome this obstacle, different methods to enhance graft survival, stability, and therapeutic potential have been developed, including environmental stress preconditioning, biopolymers scaffolds, and genetic engineering. In this review, we discuss current engineering techniques aimed at the exploitation of the migratory, proliferative, and secretive capacity of NSCs and their relevance for the therapeutic arsenal against motor neuron disorders and other neurological disorders.
Collapse
Affiliation(s)
- Elena Abati
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Neuroscience Section, University of Milan, Milan, Italy
| | - Nereo Bresolin
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Neuroscience Section, University of Milan, Milan, Italy.,Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, via F. Sforza 35, 20122, Milan, Italy
| | - Giacomo Pietro Comi
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Neuroscience Section, University of Milan, Milan, Italy.,Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, via F. Sforza 35, 20122, Milan, Italy
| | - Stefania Corti
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Neuroscience Section, University of Milan, Milan, Italy. .,Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, via F. Sforza 35, 20122, Milan, Italy.
| |
Collapse
|
76
|
Nabavi SM, Arab L, Jarooghi N, Bolurieh T, Abbasi F, Mardpour S, Azimyian V, Moeininia F, Maroufizadeh S, Sanjari L, Hosseini SE, Aghdami N. Safety, Feasibility of Intravenous and Intrathecal Injection of Autologous Bone Marrow Derived Mesenchymal Stromal Cells in Patients with Amyotrophic Lateral Sclerosis: An Open Label Phase I Clinical Trial. CELL JOURNAL 2018; 20:592-598. [PMID: 30124008 PMCID: PMC6099146 DOI: 10.22074/cellj.2019.5370] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 12/28/2018] [Indexed: 12/11/2022]
Abstract
Objective Amyotrophic lateral sclerosis (ALS) is the most severe disorder within the spectrum of motor neuron diseases
(MND) that has no effective treatment and a progressively fatal outcome. We have conducted two clinical trials to assess the
safety and feasibility of intravenous (IV) and intrathecal (IT) injections of bone marrow derived mesenchymal stromal cells
(BM-MSCs) in patients with ALS.
Materials and Methods This is an interventional/experimental study. We enrolled 14 patients that met the following inclusion
criteria: definitive diagnosis of sporadic ALS, ALS Functional Rating Scale (ALS-FRS) ≥24, and ≥40% predicted forced vital
capacity (FVC). All patients underwent bone marrow (BM) aspiration to obtain an adequate sample for cell isolation and
culture. Patients in group 1 (n=6) received an IV and patients in group 2 (n=8) received an IT injection of the cell suspension. All
patients in both groups were followed at 24 hours and 2, 4, 6, and 12 months after the injection with ALS-FRS, FVC, laboratory
tests, check list of side effects and brain/spinal cord magnetic resonance imaging (MRI). In each group, one patient was lost to
follow up one month after cell injection and one patient from IV group died due to severe respiratory insufficiency and infection.
Results During the follow up there were no reports of adverse events in terms of clinical and laboratory assessments.
In MRI, there was not any new abnormal finding. The ALS-FRS score and FVC percentage significantly reduced in all
patients from both groups.
Conclusion This study has shown that IV and IT transplantation of BM-derived stromal cells is safe and feasible (Registration
numbers: NCT01759797 and NCT01771640).
Collapse
Affiliation(s)
- Seyed Massood Nabavi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Leila Arab
- Department of Neuroscience, School of Advanced Technologies in Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Neda Jarooghi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Tina Bolurieh
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Fatemeh Abbasi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Soura Mardpour
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Vajihe Azimyian
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Fatemeh Moeininia
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Saman Maroufizadeh
- Department of Epidemiology and Reproductive Health, Reproductive Epidemiology Research Center, Royan Institute for Reproductive Medicine, ACECR, Tehran, Iran
| | - Leila Sanjari
- Intensive Care Unit, Mostafa Khomeini Hospital, Tehran, Iran
| | - Seyedeh Esmat Hosseini
- Student Research Committee, School of Nursing and Midwifery, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nasser Aghdami
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.Electronic Address:
| |
Collapse
|
77
|
Teng CF, Jeng LB, Shyu WC. Role of Insulin-like Growth Factor 1 Receptor Signaling in Stem Cell Stemness and Therapeutic Efficacy. Cell Transplant 2018; 27:1313-1319. [PMID: 29882416 PMCID: PMC6168993 DOI: 10.1177/0963689718779777] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Evidence has emerged that stem cells represent a promising therapeutic tool for tissue engineering and regenerative medicine. Thus, identifying functional markers for selecting stem cells capable of superior self-renewal and pluripotency (or multipotency) and maintaining stem cell identity under appropriate culture conditions are critical for guiding the use of stem cells toward clinical applications. Many investigations have implicated the insulin-like growth factor 1 receptor (IGF1R) signaling in maintenance of stem cell characteristics and enhancement of stem cell therapy efficacy. IGF1R-expressing stem cells display robust pluripotent or multipotent properties. In this review, we summarize the essential roles of IGF1R signaling in self-renewal, pluripotency (or multipotency), and therapeutic efficacy of stem cells, including human embryonic stem cells, neural stem cells, cardiac stem cells, bone marrow mesenchymal stem cells, placental mesenchymal stem cells, and dental pulp mesenchymal stem cells. Modifying IGF1R signaling may thus provide potential strategies for maintaining stem cell properties and improving stem-cell-based therapeutic applications.
Collapse
Affiliation(s)
- Chiao-Fang Teng
- 1 Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,2 Organ Transplantation Center, China Medical University Hospital, Taichung, Taiwan
| | - Long-Bin Jeng
- 2 Organ Transplantation Center, China Medical University Hospital, Taichung, Taiwan
| | - Woei-Cherng Shyu
- 1 Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,3 Translational Medicine Research Center and Department of Neurology, China Medical University Hospital, Taichung, Taiwan.,4 Department of Occupational Therapy, Asia University, Taichung, Taiwan
| |
Collapse
|
78
|
Goutman SA, Brown MB, Glass JD, Boulis NM, Johe K, Hazel T, Cudkowicz M, Atassi N, Borges L, Patil PG, Sakowski SA, Feldman EL. Long-term Phase 1/2 intraspinal stem cell transplantation outcomes in ALS. Ann Clin Transl Neurol 2018; 5:730-740. [PMID: 29928656 PMCID: PMC5989736 DOI: 10.1002/acn3.567] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 03/25/2018] [Indexed: 12/12/2022] Open
Abstract
Objective Intraspinal human spinal cord‐derived neural stem cell (HSSC) transplantation is a potential therapy for amyotrophic lateral sclerosis (ALS); however, previous trials lack controls. This post hoc analysis compared ambulatory limb‐onset ALS participants in Phase 1 and 2 (Ph1/2) open‐label intraspinal HSSC transplantation studies up to 3 years after transplant to matched participants in Pooled Resource Open‐Access ALS Clinical Trials (PRO‐ACT) and ceftriaxone datasets to provide required analyses to inform future clinical trial designs. Methods Survival, ALSFRS‐R, and a composite statistic (ALS/SURV) combining survival and ALS Functional Rating Scale revised (ALSFRS‐R) functional status were assessed for matched participant subsets: PRO‐ACT n = 1108, Ph1/2 n = 21 and ceftriaxone n = 177, Ph1/2 n = 20. Results Survival did not differ significantly between cohorts: Ph1/2 median survival 4.7 years, 95% CI (1.2, ∞) versus PRO‐ACT 2.3 years (1.9, 2.5), P = 1.0; Ph1/2 3.0 years (1.2, 5.6) versus ceftriaxone 2.3 years (1.8, 2.8), P = 0.88. Mean ALSFRS‐R at 24 months significantly differed between Ph1/2 and both comparison cohorts (Ph1/2 30.1 ± 8.6 vs. PRO‐ACT 24.0 ± 10.2, P = 0.048; Ph1/2 30.7 ± 8.8 vs. ceftriaxone 19.2 ± 9.5, P = 0.0023). Using ALS/SURV, median PRO‐ACT and ceftriaxone participants died by 24 months, whereas median Ph1/2 participant ALSFRS‐Rs were 23 (P = 0.0038) and 19 (P = 0.14) in PRO‐ACT and ceftriaxone comparisons at 24 months, respectively, supporting improved functional outcomes in the Ph1/2 study. Interpretation Comparison of Ph1/2 studies to historical datasets revealed significantly improved survival and function using ALS/SURV versus PRO‐ACT controls. While results are encouraging, comparison against historical populations demonstrate limitations in noncontrolled studies. These findings support continued evaluation of HSSC transplantation in ALS, support the benefit of control populations, and enable necessary power calculations to design a randomized, sham surgery‐controlled efficacy study.
Collapse
Affiliation(s)
- Stephen A Goutman
- Department of Neurology University of Michigan 109 Zina Pitcher Place 5017 AAT-BSRB Ann Arbor Michigan 48109
| | - Morton B Brown
- Department of Biostatistics University of Michigan 1415 Washington Heights M4039 SPH II Ann Arbor Michigan 48109
| | - Jonathan D Glass
- Department of Neurology Emory University School of Medicine 101 Woodruff Circle Atlanta Georgia 30322
| | - Nicholas M Boulis
- Department of Neurosurgery Emory University School of Medicine 101 Woodruff Circle WMB Room 6309 Atlanta Georgia
| | - Karl Johe
- Neuralstem, Inc. 20271 Goldenrod Lane Suite 2033 Germantown Maryland 20876
| | - Tom Hazel
- Neuralstem, Inc. 20271 Goldenrod Lane Suite 2033 Germantown Maryland 20876
| | - Merit Cudkowicz
- Department of Neurology Massachusetts General Hospital Harvard Medical School 165 Cambridge Street Boston Massachusetts 02114
| | - Nazem Atassi
- Department of Neurology Massachusetts General Hospital Harvard Medical School 165 Cambridge Street Boston Massachusetts 02114
| | - Lawrence Borges
- Department of Neurosurgery Massachusetts General Hospital Harvard Medical School 15 Parkman Street Wand ACC 745 Boston Massachusetts 02114
| | - Parag G Patil
- Department of Neurology University of Michigan 109 Zina Pitcher Place 5017 AAT-BSRB Ann Arbor Michigan 48109.,Department of Neurosurgery University of Michigan 1500 E. Medical Center Drive SPC 5338 Ann Arbor Michigan 48109
| | - Stacey A Sakowski
- Program for Neurology Research and Discovery University of Michigan 109 Zina Pitcher Place 5017 AAT-BSRB Ann Arbor Michigan 48109
| | - Eva L Feldman
- Department of Neurology University of Michigan 109 Zina Pitcher Place 5017 AAT-BSRB Ann Arbor Michigan 48109.,Program for Neurology Research and Discovery University of Michigan 109 Zina Pitcher Place 5017 AAT-BSRB Ann Arbor Michigan 48109
| |
Collapse
|
79
|
Lee KH, Nam H, Won JS, Hwang JY, Jang HW, Lee SH, Joo KM. In Vivo Spinal Distribution of Cy5.5 Fluorescent Dye after Injection via the Lateral Ventricle and Cisterna Magna in Rat Model. J Korean Neurosurg Soc 2018; 61:434-440. [PMID: 29660974 PMCID: PMC6046575 DOI: 10.3340/jkns.2017.0252] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 10/23/2017] [Indexed: 11/27/2022] Open
Abstract
Objective The purpose of this study was to find an optimal delivery route for clinical trials of intrathecal cell therapy for spinal cord injury in preclinical stage. Methods We compared in vivo distribution of Cy5.5 fluorescent dye in the spinal cord region at various time points utilizing in vivo optical imaging techniques, which was injected into the lateral ventricle (LV) or cisterna magna (CM) of rats. Results Although CM locates nearer to the spinal cord than the LV, significantly higher signal of Cy5.5 was detected in the thoracic and lumbar spinal cord region at all time points tested when Cy5.5 was injected into the LV. In the LV injection Cy5.5 signal in the thoracic and lumbar spinal cord was observed within 12 hours after injection, which was maintained until 72 hours after injection. In contrast, Cy5.5 signal was concentrated at the injection site in the CM injection at all time points. Conclusion These data suggested that the LV might be suitable for preclinical injection route of therapeutics targeting the spinal cord to test their treatment efficacy and biosafety for spinal cord diseases in small animal models.
Collapse
Affiliation(s)
- Kee-Hang Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea.,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, Korea.,Stem Cell and Regenerative Medicine Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Hyun Nam
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, Korea.,Stem Cell and Regenerative Medicine Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea.,Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jeong-Seob Won
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea.,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, Korea.,Stem Cell and Regenerative Medicine Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Ji-Yoon Hwang
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, Korea.,Stem Cell and Regenerative Medicine Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea.,Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hye Won Jang
- Department of Medical Education, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sun-Ho Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea.,Stem Cell and Regenerative Medicine Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea.,Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyeung Min Joo
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea.,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, Korea.,Stem Cell and Regenerative Medicine Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea.,Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
| |
Collapse
|
80
|
Rando A, Pastor D, Viso-León MC, Martínez A, Manzano R, Navarro X, Osta R, Martínez S. Intramuscular transplantation of bone marrow cells prolongs the lifespan of SOD1 G93A mice and modulates expression of prognosis biomarkers of the disease. Stem Cell Res Ther 2018; 9:90. [PMID: 29625589 PMCID: PMC5889612 DOI: 10.1186/s13287-018-0843-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 02/28/2018] [Accepted: 03/15/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder characterized by progressive muscle weakness, paralysis and death. There is no effective treatment for ALS and stem cell therapy has arisen as a potential therapeutic approach. METHODS SOD1 mutant mice were used to study the potential neurotrophic effect of bone marrow cells grafted into quadriceps femoris muscle. RESULTS Bone marrow intramuscular transplants resulted in increased longevity with improved motor function and decreased motoneuron degeneration in the spinal cord. Moreover, the increment of the glial-derived neurotrophic factor and neurotrophin 4 observed in the grafted muscles suggests that this partial neuroprotective effect is mediated by neurotrophic factor release at the neuromuscular junction level. Finally, certain neurodegeneration and muscle disease-specific markers, which are altered in the SOD1G93A mutant mouse and may serve as molecular biomarkers for the early detection of ALS in patients, have been studied with encouraging results. CONCLUSIONS This work demonstrates that stem cell transplantation in the muscle prolonged the lifespan, increased motoneuron survival and slowed disease progression, which was also assessed by genetic expression analysis.
Collapse
Affiliation(s)
- Amaya Rando
- LAGENBIO-I3A, Facultad de Veterinaria, IIS Aragón, Universidad de Zaragoza, Zaragoza, Spain
| | - Diego Pastor
- Centro de Investigación Deporte, Universidad Miguel Hernández de Elche, Alicante, Spain
- Instituto de Neurociencias de Alicante, UMH-CSIC, Universidad Miguel Hernández de Elche, Alicante, Spain
| | - Mari Carmen Viso-León
- Instituto de Neurociencias de Alicante, UMH-CSIC, Universidad Miguel Hernández de Elche, Alicante, Spain
| | - Anna Martínez
- Grupo de Neuroplasticidad y Regeneración, Instituto de Neurociencias y Departamento de Biología Celular, Fisiología e Inmunología, Universidad Autónoma de Barcelona, Barcelona, Spain
| | - Raquel Manzano
- LAGENBIO-I3A, Facultad de Veterinaria, IIS Aragón, Universidad de Zaragoza, Zaragoza, Spain
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Xavier Navarro
- Grupo de Neuroplasticidad y Regeneración, Instituto de Neurociencias y Departamento de Biología Celular, Fisiología e Inmunología, Universidad Autónoma de Barcelona, Barcelona, Spain
| | - Rosario Osta
- LAGENBIO-I3A, Facultad de Veterinaria, IIS Aragón, Universidad de Zaragoza, Zaragoza, Spain
| | - Salvador Martínez
- Instituto de Neurociencias de Alicante, UMH-CSIC, Universidad Miguel Hernández de Elche, Alicante, Spain
| |
Collapse
|
81
|
Huang H, Young W, Chen L, Feng S, Zoubi ZMA, Sharma HS, Saberi H, Moviglia GA, He X, Muresanu DF, Sharma A, Otom A, Andrews RJ, Al-Zoubi A, Bryukhovetskiy AS, Chernykh ER, Domańska-Janik K, Jafar E, Johnson WE, Li Y, Li D, Luan Z, Mao G, Shetty AK, Siniscalco D, Skaper S, Sun T, Wang Y, Wiklund L, Xue Q, You SW, Zheng Z, Dimitrijevic MR, Masri WSE, Sanberg PR, Xu Q, Luan G, Chopp M, Cho KS, Zhou XF, Wu P, Liu K, Mobasheri H, Ohtori S, Tanaka H, Han F, Feng Y, Zhang S, Lu Y, Zhang Z, Rao Y, Tang Z, Xi H, Wu L, Shen S, Xue M, Xiang G, Guo X, Yang X, Hao Y, Hu Y, Li J, AO Q, Wang B, Zhang Z, Lu M, Li T. Clinical Cell Therapy Guidelines for Neurorestoration (IANR/CANR 2017). Cell Transplant 2018; 27:310-324. [PMID: 29637817 PMCID: PMC5898693 DOI: 10.1177/0963689717746999] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 07/22/2017] [Accepted: 11/13/2017] [Indexed: 12/11/2022] Open
Abstract
Cell therapy has been shown to be a key clinical therapeutic option for central nervous system diseases or damage. Standardization of clinical cell therapy procedures is an important task for professional associations devoted to cell therapy. The Chinese Branch of the International Association of Neurorestoratology (IANR) completed the first set of guidelines governing the clinical application of neurorestoration in 2011. The IANR and the Chinese Association of Neurorestoratology (CANR) collaborated to propose the current version "Clinical Cell Therapy Guidelines for Neurorestoration (IANR/CANR 2017)". The IANR council board members and CANR committee members approved this proposal on September 1, 2016, and recommend it to clinical practitioners of cellular therapy. These guidelines include items of cell type nomenclature, cell quality control, minimal suggested cell doses, patient-informed consent, indications for undergoing cell therapy, contraindications for undergoing cell therapy, documentation of procedure and therapy, safety evaluation, efficacy evaluation, policy of repeated treatments, do not charge patients for unproven therapies, basic principles of cell therapy, and publishing responsibility.
Collapse
Affiliation(s)
- Hongyun Huang
- Institute of Neurorestoratology, General Hospital of Armed Police Forces, Beijing, People’s Republic of China
| | - Wise Young
- W. M. Keck Center for Collaborative Neuroscience, Rutgers, State University of New Jersey, Piscataway, NJ, USA
| | - Lin Chen
- Department of Neurosurgery, Tsinghua University Yuquan Hospital, Beijing, People’s Republic of China
| | - Shiqing Feng
- Department of Orthopaedics, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Ziad M. Al Zoubi
- Jordan Ortho and Spinal Centre, Al-Saif Medical Center, Amman, Jordan
| | - Hari Shanker Sharma
- Intensive Experimental CNS Injury and Repair, University Hospital, Uppsala University, Uppsala, Sweden
| | - Hooshang Saberi
- Department of Neurosurgery, Brain and Spinal Injury Research center, Tehran University of Medical Sciences, Tehran, Iran
| | - Gustavo A. Moviglia
- Center of Research and Engineer of Tissues and Cellular Therapy, Maimonides University, Buenos Aires, Argentina
| | - Xijing He
- Department of Orthopaedics, Second Affiliated Hospital of Xi’an Jiaotong University, Xian, People’s Republic of China
| | - Dafin F. Muresanu
- Department of Neurosciences “Iuliu Hatieganu,” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Alok Sharma
- Department of Neurosurgery, LTM Medical College, LTMG Hospital, Mumbai, Mumbai, India
| | - Ali Otom
- Royal Rehabilitation Center, King Hussein Medical Centre-RJRC Amman, Jordan
| | - Russell J. Andrews
- Nanotechnology & Smart Systems, NASA Ames Research Center, Silicon Valley, CA, USA
| | - Adeeb Al-Zoubi
- The University of Illinois College of Medicine in Peoria, Peoria, IL, USA
| | - Andrey S. Bryukhovetskiy
- NeuroVita Clinic of Interventional and Restorative Neurology and Therapy, Kashirskoye shosse, Moscow, Russia
| | - Elena R. Chernykh
- Lab of Cellular Immunotherapy, Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | | | - Emad Jafar
- Jordan Ortho and Spinal Centre, Al-Saif Medical Center, Amman, Jordan
| | - W. Eustace Johnson
- Stem Cells and Regenerative Biology, Faculty of Medicine Dentistry and Life Sciences, University of Chester, Chester, United Kingdom
| | - Ying Li
- Spinal Repair Unit, Department of Brain Repair and Rehabilitation, UCL Institute of Neurology, London, United Kingdom
| | - Daqing Li
- Spinal Repair Unit, Department of Brain Repair and Rehabilitation, UCL Institute of Neurology, London, United Kingdom
| | - Zuo Luan
- Department of Pediatrics, Navy General Hospital of PLA, Beijing, People’s Republic of China
| | - Gengsheng Mao
- Institute of Neurorestoratology, General Hospital of Armed Police Forces, Beijing, People’s Republic of China
| | - Ashok K. Shetty
- Department of Molecular and Cellular Medicine, Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA
| | - Dario Siniscalco
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli,” Naples, Italy
| | - Stephen Skaper
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Tiansheng Sun
- Department of orthopedics, PLA Army General Hospital, Beijing, People’s Republic of China
| | - Yunliang Wang
- Department of Neurology, 148th Hospital, Zibo, Shandong, People’s Republic of China
| | - Lars Wiklund
- Unit of Neurology, Department of Pharmacology and Clinical Neuroscience, Umea University, Ostersund, Sweden
| | - Qun Xue
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou Jiangsu, People’s Republic of China
| | - Si-Wei You
- Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Zuncheng Zheng
- Department of Rehabilitation Medicine, The Central Hospital of Taian, Taian, Shandong, People’s Republic of China
| | | | - W. S. El Masri
- Spinal Injuries Unit, Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, United Kingdom
| | - Paul R. Sanberg
- Center of Excellence for Aging & Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Qunyuan Xu
- Institute of Neuroscience, Capital Medical University, Beijing, People’s Republic of China
| | - Guoming Luan
- Department of Neurosurgery, Beijing Sanbo Brain Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Michael Chopp
- Henry Ford Hospital, Henry Ford Health System, Neurology Research, Detroit, MI, USA
| | - Kyoung-Suok Cho
- Department of Neurosurgery, Uijongbu St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Uijongbu, South Korea
| | - Xin-Fu Zhou
- Division of Health Sciences, School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia, Australia
| | - Ping Wu
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Kai Liu
- Division of Life Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong
| | - Hamid Mobasheri
- Biomaterials Research Center, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Seiji Ohtori
- Department of Orthopedic Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hiroyuki Tanaka
- Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Fabin Han
- Centre for Stem Cells and Regenerative Medicine, Liaocheng University/Liaocheng People’s Hospital, Liaocheng, Shandong, People’s Republic of China
| | - Yaping Feng
- Department of Neurosurgery, Kunming General Hospital of Chengdu Military Command of Chinese PLA, Kunming, Yunnan, People’s Republic of China
| | - Shaocheng Zhang
- Department of Orthopedics, Changhai Hospital, The Second Military Medical University, Shanghai, People’s Republic of China
| | - Yingjie Lu
- Department of Neurosurgery, Chengde Dadu Hospital, Weichang, Hebei, People’s Republic of China
| | - Zhicheng Zhang
- Department of orthopedics, PLA Army General Hospital, Beijing, People’s Republic of China
| | - Yaojian Rao
- Department of Spinal Surgery, Luoyang Orthopedic Hospital of Henan Province, Luoyang, Henan, People’s Republic of China
| | - Zhouping Tang
- Department of Neurology, Tongji Medical College of HUST, Tongji Hospital, Wuhan, People’s Republic of China
| | - Haitao Xi
- Department of Neurology, Beijing Rehabilitation Hospital of Capital Medical University, Beijing, People’s Republic of China
| | - Liang Wu
- Center of Rehabilitation, Beijing Xiaotangshan Rehabilitation Hospital, Beijing, People’s Republic of China
| | - Shunji Shen
- Department of Rehabilitation, Weihai Municipal Hospital, Weihai, Shandong, People’s Republic of China
| | - Mengzhou Xue
- Department of Neurorehabilitation, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
| | - Guanghong Xiang
- Brain Hospital of Hunan Province, Changsha, Hunan, People’s Republic of China
| | - Xiaoling Guo
- Department of Neurology, PLA Army 266 Hospital, Chengde, Hebei, People’s Republic of China
| | - Xiaofeng Yang
- Department of Neurosurgery, The First Affiliated Hospital of Zhejiang University Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Yujun Hao
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| | - Yong Hu
- Department of Orthopaedic and Traumatology, The University of Hong Kong, Pokfulam, Hong Kong
| | - Jinfeng Li
- Unit of Neurology, Department of Pharmacology and Clinical Neuroscience, Umea University, Ostersund, Sweden
| | - Qiang AO
- Department of tissue engineering, China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Bin Wang
- Department of Traumatology, The Second Affiliated Hospital of Guangzhou Medical University, Haizhu District, Guangzhou, People’s Republic of China
| | - Zhiwen Zhang
- Department of Neurosurgery, First Affiliated Hospital of Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Ming Lu
- Department of Neurosurgery, Second Affiliated Hospital of Hunan Normal University (163 Hospital of PLA), Changsha, Hunan, People’s Republic of China
| | - Tong Li
- Department of Neurology, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, People’s Republic of China
| |
Collapse
|
82
|
Ferrari D, Gelati M, Profico DC, Vescovi AL. Human Fetal Neural Stem Cells for Neurodegenerative Disease Treatment. Results Probl Cell Differ 2018; 66:307-329. [DOI: 10.1007/978-3-319-93485-3_14] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
|
83
|
Indications and prospects of neural transplantation for chronic neurological diseases. Curr Opin Organ Transplant 2017; 21:490-6. [PMID: 27517509 DOI: 10.1097/mot.0000000000000344] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW The replacement of damaged cells in the central nervous system (CNS) affected by degenerative disorders represents an attractive therapeutic strategy. The advent of stem cell technology may offer the possibility of generating a large number of renewable, specifically differentiated cells to potentially cure large cohorts of patients. In this review, we discuss current knowledge and issues involved in neural cell transplantation. The most important preclinical and clinical results of cellular transplantation applied to Parkinson's, Huntington's disease and amyotrophic lateral sclerosis will be summarized. RECENT FINDINGS Cellular transplantation is emerging as a possible therapy for a variety of incurable neurological disorders. The disorders that will primarily take advantage from neural stem cell grafting are those involving a well defined cell population in a restricted area of the CNS. Several clinical trials have been initiated to assess safety and efficacy of different stem cell-derived products, and promising results have been obtained for disorders such as Parkinson's disease. However, several scientific questions remain unanswered. Among these, the impact of the immunological interaction between host and graft in the particular environment of the CNS still requires additional investigations. SUMMARY Several chronic neurological disorders appear to be amenable to cell regenerative therapies. However, safety, efficacy and immunological issues will need to be carefully evaluated beforehand.
Collapse
|
84
|
Lamanna JJ, Gutierrez J, Espinosa JR, Wagner J, Urquia LN, Moreton C, Victor Hurtig C, Tora M, Kirk AD, Federici T, Boulis NM. Peripheral blood detection of systemic graft-specific xeno-antibodies following transplantation of human neural progenitor cells into the porcine spinal cord. J Clin Neurosci 2017; 48:173-180. [PMID: 29089163 DOI: 10.1016/j.jocn.2017.10.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 10/10/2017] [Indexed: 12/17/2022]
Abstract
Extensive pre-clinical and clinical studies have searched for therapeutic efficacy of cell-based therapeutics in diseases of the Central Nervous System (CNS) with no other viable options. Allogeneic cells represent the primary source of these therapies and immunosuppressive regimens have been empirically employed based on experience with solid organ transplantation, attempting to avoid immune mediated graft rejection. In this study, we aimed to 1) characterize the host immune response to stem cells transplanted into the CNS and 2) develop a non-invasive method for detecting immune response to transplanted cell grafts. Human neural progenitor cells were transplanted into the spinal cord of 10 Göttingen minipigs, of which 5 received no immunosuppression and 5 received Tacrolimus. Peripheral blood samples were collected longitudinally for flow cytometry cross match studies. Necropsy was performed at day 21 and spinal cord tissue analysis. We observed a transient increase in xeno-reactive antibodies was detected on post-operative day 7 and 14 in pigs that did not receive immunosuppression. This response was not detected in pigs that received Tacrolimus immunosuppression. No difference in graft survival was observed between the groups. Infiltration of numerous immune mediators including granulocytes, T lymphocytes, and activated microglia, and complement deposition were detected. In summary, a systemic immunologic response to stem cell grafts was detected for two weeks after transplantation using peripheral blood. This could be used as a non-invasive biomarker by investigators for detection of immunologic rejection. However, the absence of a detectable response in peripheral blood does not rule out a parenchymal immune response.
Collapse
Affiliation(s)
- Jason J Lamanna
- Department of Neurosurgery, School of Medicine, Emory University, 101 Woodruff Circle, Room 6339, Atlanta, GA 30322, USA; Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA 30322, USA.
| | - Juanmarco Gutierrez
- Department of Neurosurgery, School of Medicine, Emory University, 101 Woodruff Circle, Room 6339, Atlanta, GA 30322, USA.
| | - Jaclyn R Espinosa
- Department of Surgery, School of Medicine, Emory University, Atlanta, GA 30322, USA; Department of Surgery, Duke University, Durham, NC 27710, USA.
| | - Jacob Wagner
- Department of Neurosurgery, School of Medicine, Emory University, 101 Woodruff Circle, Room 6339, Atlanta, GA 30322, USA.
| | - Lindsey N Urquia
- Department of Neurosurgery, School of Medicine, Emory University, 101 Woodruff Circle, Room 6339, Atlanta, GA 30322, USA.
| | - Cheryl Moreton
- Department of Neurosurgery, School of Medicine, Emory University, 101 Woodruff Circle, Room 6339, Atlanta, GA 30322, USA.
| | - C Victor Hurtig
- Department of Neurosurgery, School of Medicine, Emory University, 101 Woodruff Circle, Room 6339, Atlanta, GA 30322, USA.
| | - Muhibullah Tora
- Department of Neurosurgery, School of Medicine, Emory University, 101 Woodruff Circle, Room 6339, Atlanta, GA 30322, USA; Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA 30322, USA.
| | - Allan D Kirk
- Department of Surgery, Duke University, Durham, NC 27710, USA.
| | - Thais Federici
- Department of Neurosurgery, School of Medicine, Emory University, 101 Woodruff Circle, Room 6339, Atlanta, GA 30322, USA.
| | - Nicholas M Boulis
- Department of Neurosurgery, School of Medicine, Emory University, 101 Woodruff Circle, Room 6339, Atlanta, GA 30322, USA; Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
85
|
Tang Y, Yu P, Cheng L. Current progress in the derivation and therapeutic application of neural stem cells. Cell Death Dis 2017; 8:e3108. [PMID: 29022921 PMCID: PMC5682670 DOI: 10.1038/cddis.2017.504] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 08/28/2017] [Accepted: 08/31/2017] [Indexed: 12/13/2022]
Abstract
Neural stem cells (NSCs) have a unique role in neural regeneration. Cell therapy based on NSC transplantation is a promising tool for the treatment of nervous system diseases. However, there are still many issues and controversies associated with the derivation and therapeutic application of these cells. In this review, we summarize the different sources of NSCs and their derivation methods, including direct isolation from primary tissues, differentiation from pluripotent stem cells and transdifferentiation from somatic cells. We also review the current progress in NSC implantation for the treatment of various neural defects and injuries in animal models and clinical trials. Finally, we discuss potential optimization strategies for NSC derivation and propose urgent challenges to the clinical translation of NSC-based therapies in the near future.
Collapse
Affiliation(s)
- Yuewen Tang
- National Research Center for Translational Medicine, State Key Laboratory of Medical Genomics, Shanghai Institute of Haematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pei Yu
- Department of Orthopaedics, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Cheng
- National Research Center for Translational Medicine, State Key Laboratory of Medical Genomics, Shanghai Institute of Haematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
86
|
Liu J, Wang F. Role of Neuroinflammation in Amyotrophic Lateral Sclerosis: Cellular Mechanisms and Therapeutic Implications. Front Immunol 2017; 8:1005. [PMID: 28871262 PMCID: PMC5567007 DOI: 10.3389/fimmu.2017.01005] [Citation(s) in RCA: 247] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 08/07/2017] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease that affects upper motor neurons (MNs) comprising the corticospinal tract and lower MNs arising from the brain stem nuclei and ventral roots of the spinal cord, leading to fatal paralysis. Currently, there are no effective therapies for ALS. Increasing evidence indicates that neuroinflammation plays an important role in ALS pathogenesis. The neuroinflammation in ALS is characterized by infiltration of lymphocytes and macrophages, activation of microglia and reactive astrocytes, as well as the involvement of complement. In this review, we focus on the key cellular players of neuroinflammation during the pathogenesis of ALS by discussing not only their detrimental roles but also their immunomodulatory actions. We will summarize the pharmacological therapies for ALS that target neuroinflammation, as well as recent advances in the field of stem cell therapy aimed at modulating the inflammatory environment to preserve the remaining MNs in ALS patients and animal models of the disease.
Collapse
Affiliation(s)
- Jia Liu
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Fei Wang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
87
|
Three-Dimensional Organoid System Transplantation Technologies in Future Treatment of Central Nervous System Diseases. Stem Cells Int 2017; 2017:5682354. [PMID: 28904534 PMCID: PMC5585580 DOI: 10.1155/2017/5682354] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 05/24/2017] [Accepted: 06/08/2017] [Indexed: 02/06/2023] Open
Abstract
In recent years, scientists have made great achievements in understanding the development of human brain and elucidating critical elements of stepwise spatiotemporal control strategies in neural stem cell specification lineage, which facilitates successful induction of neural organoid in vitro including the cerebral cortex, cerebellar, neural tube, hippocampus cortex, pituitary, and optic cup. Besides, emerging researches on neural organogenesis promote the application of 3D organoid system transplantation in treating central nervous system (CNS) diseases. Present review will categorize current researches on organogenesis into three approaches: (a) stepwise, direct organization of region-specific or population-enriched neural organoid; (b) assemble and direct distinct organ-specific progenitor cells or stem cells to form specific morphogenesis organoid; and (c) assemble embryoid bodies for induction of multilayer organoid. However, the majority of these researches focus on elucidating cellular and molecular mechanisms involving in brain organogenesis or disease development and only a few of them conducted for treating diseases. In this work, we will compare three approaches and also analyze their possible indications for diseases in future treatment on the basis of their distinct characteristics.
Collapse
|
88
|
Mohamed LA, Markandaiah S, Bonanno S, Pasinelli P, Trotti D. Blood-Brain Barrier Driven Pharmacoresistance in Amyotrophic Lateral Sclerosis and Challenges for Effective Drug Therapies. AAPS JOURNAL 2017; 19:1600-1614. [PMID: 28779378 DOI: 10.1208/s12248-017-0120-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 06/28/2017] [Indexed: 12/11/2022]
Abstract
The blood-brain barrier (BBB) is essential for proper neuronal function, homeostasis, and protection of the central nervous system (CNS) microenvironment from blood-borne pathogens and neurotoxins. The BBB is also an impediment for CNS penetration of drugs. In some neurologic conditions, such as epilepsy and brain tumors, overexpression of P-glycoprotein, an efflux transporter whose physiological function is to expel catabolites and xenobiotics from the CNS into the blood stream, has been reported. Recent studies reported that overexpression of P-glycoprotein and increase in its activity at the BBB drives a progressive resistance to CNS penetration and persistence of riluzole, the only drug approved thus far for treatment of amyotrophic lateral sclerosis (ALS), rapidly progressive and mostly fatal neurologic disease. This review will discuss the impact of transporter-mediated pharmacoresistance for ALS drug therapy and the potential therapeutic strategies to improve the outcome of ALS clinical trials and efficacy of current and future drug treatments.
Collapse
Affiliation(s)
- Loqman A Mohamed
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University Hospitals, 900 Walnut Street, Philadelphia, Pennsylvania, 19107, USA.
| | - Shashirekha Markandaiah
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University Hospitals, 900 Walnut Street, Philadelphia, Pennsylvania, 19107, USA
| | - Silvia Bonanno
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University Hospitals, 900 Walnut Street, Philadelphia, Pennsylvania, 19107, USA
| | - Piera Pasinelli
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University Hospitals, 900 Walnut Street, Philadelphia, Pennsylvania, 19107, USA
| | - Davide Trotti
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University Hospitals, 900 Walnut Street, Philadelphia, Pennsylvania, 19107, USA
| |
Collapse
|
89
|
Rushton DJ, Andres PL, Allred P, Baloh RH, Svendsen CN. Patients with ALS show highly correlated progression rates in left and right limb muscles. Neurology 2017; 89:196-206. [PMID: 28600459 PMCID: PMC5501935 DOI: 10.1212/wnl.0000000000004105] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 04/04/2017] [Indexed: 12/13/2022] Open
Abstract
Objective: Amyotrophic lateral sclerosis (ALS) progresses at different rates between patients, making clinical trial design difficult and dependent on large cohorts of patients. Currently, there are few data showing whether the left and right limbs progress at the same or different rates. This study addresses rates of decline in specific muscle groups of patients with ALS and assesses whether there is a relationship between left and right muscles in the same patient, regardless of overall progression. Methods: A large cohort of patients was used to assess decline in muscle strength in right and left limbs over time using 2 different methods: The Tufts Quantitative Neuromuscular Exam and Accurate Test of Limb Isometric Strength protocol. Then advanced linear regression statistical methods were applied to assess progression rates in each limb. Results: This report shows that linearized progression models can predict general slopes of decline with good accuracy. Critically, the data demonstrate that while overall decline is variable, there is a high degree of correlation between left and right muscle decline in ALS. This implies that irrespective of which muscle starts declining soonest or latest, their rates of decline following onset are more consistent. Conclusions: First, this study demonstrates a high degree of power when using unilateral treatment approaches to detect a slowing in disease progression in smaller groups of patients, thus allowing for paired statistical tests. These findings will be useful in transplantation trials that use muscle decline to track disease progression in ALS. Second, these findings discuss methods, such as tactical selection of muscle groups, which can improve the power efficiency of all ALS clinical trials.
Collapse
Affiliation(s)
- David J Rushton
- From the Board of Governors Regenerative Medicine Institute (D.J.R., P.A., R.H.B., C.N.S.), Department of Biomedical Sciences (D.J.R., C.N.S.), and Neurology (P.A., R.H.B.), Cedars-Sinai Medical Center, Los Angeles, CA; and Neurological Clinical Research Institute (P.L.A.), Massachusetts General Hospital, Boston
| | - Patricia L Andres
- From the Board of Governors Regenerative Medicine Institute (D.J.R., P.A., R.H.B., C.N.S.), Department of Biomedical Sciences (D.J.R., C.N.S.), and Neurology (P.A., R.H.B.), Cedars-Sinai Medical Center, Los Angeles, CA; and Neurological Clinical Research Institute (P.L.A.), Massachusetts General Hospital, Boston
| | - Peggy Allred
- From the Board of Governors Regenerative Medicine Institute (D.J.R., P.A., R.H.B., C.N.S.), Department of Biomedical Sciences (D.J.R., C.N.S.), and Neurology (P.A., R.H.B.), Cedars-Sinai Medical Center, Los Angeles, CA; and Neurological Clinical Research Institute (P.L.A.), Massachusetts General Hospital, Boston
| | - Robert H Baloh
- From the Board of Governors Regenerative Medicine Institute (D.J.R., P.A., R.H.B., C.N.S.), Department of Biomedical Sciences (D.J.R., C.N.S.), and Neurology (P.A., R.H.B.), Cedars-Sinai Medical Center, Los Angeles, CA; and Neurological Clinical Research Institute (P.L.A.), Massachusetts General Hospital, Boston
| | - Clive N Svendsen
- From the Board of Governors Regenerative Medicine Institute (D.J.R., P.A., R.H.B., C.N.S.), Department of Biomedical Sciences (D.J.R., C.N.S.), and Neurology (P.A., R.H.B.), Cedars-Sinai Medical Center, Los Angeles, CA; and Neurological Clinical Research Institute (P.L.A.), Massachusetts General Hospital, Boston.
| |
Collapse
|
90
|
Spurlock MS, Ahmed AI, Rivera KN, Yokobori S, Lee SW, Sam PN, Shear DA, Hefferan MP, Hazel TG, Johe KK, Gajavelli S, Tortella FC, Bullock RM. Amelioration of Penetrating Ballistic-Like Brain Injury Induced Cognitive Deficits after Neuronal Differentiation of Transplanted Human Neural Stem Cells. J Neurotrauma 2017; 34:1981-1995. [PMID: 28249550 PMCID: PMC6913783 DOI: 10.1089/neu.2016.4602] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Penetrating traumatic brain injury (PTBI) is one of the major cause of death and disability worldwide. Previous studies with penetrating ballistic-like brain injury (PBBI), a PTBI rat model revealed widespread perilesional neurodegeneration, similar to that seen in humans following gunshot wound to the head, which is unmitigated by any available therapies to date. Therefore, we evaluated human neural stem cell (hNSC) engraftment to putatively exploit the potential of cell therapy that has been seen in other central nervous system injury models. Toward this objective, green fluorescent protein (GFP) labeled hNSC (400,000 per animal) were transplanted in immunosuppressed Sprague-Dawley (SD), Fisher, and athymic (ATN) PBBI rats 1 week after injury. Tacrolimus (3 mg/kg 2 days prior to transplantation, then 1 mg/kg/day), methylprednisolone (10 mg/kg on the day of transplant, 1 mg/kg/week thereafter), and mycophenolate mofetil (30 mg/kg/day) for 7 days following transplantation were used to confer immunosuppression. Engraftment in SD and ATN was comparable at 8 weeks post-transplantation. Evaluation of hNSC differentiation and distribution revealed increased neuronal differentiation of transplanted cells with time. At 16 weeks post-transplantation, neither cell proliferation nor glial lineage markers were detected. Transplanted cell morphology was similar to that of neighboring host neurons, and there was relatively little migration of cells from the peritransplant site. By 16 weeks, GFP-positive processes extended both rostrocaudally and bilaterally into parenchyma, spreading along host white matter tracts, traversing the internal capsule, and extending ∼13 mm caudally from transplantation site reaching into the brainstem. In a Morris water maze test at 8 weeks post-transplantation, animals with transplants had shorter latency to platform than vehicle-treated animals. However, weak injury-induced cognitive deficits in the control group at the delayed time point confounded benefits of durable engraftment and neuronal differentiation. Therefore, these results justify further studies to progress towards clinical translation of hNSC therapy for PTBI.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Deborah A. Shear
- Branch of Brain Trauma Neuroprotection and Neurorestoration, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | | | | | | | | | - Frank C. Tortella
- Branch of Brain Trauma Neuroprotection and Neurorestoration, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | | |
Collapse
|
91
|
Abstract
INTRODUCTION Spinal cord injury (SCI) is a devastating condition, where regenerative failure and cell loss lead to paralysis. The heterogeneous and time-sensitive pathophysiology has made it difficult to target tissue repair. Despite many medical advances, there are no effective regenerative therapies. As stem cells offer multi-targeted and environmentally responsive benefits, cell therapy is a promising treatment approach. Areas covered: This review highlights the cell therapies being investigated for SCI, including Schwann cells, olfactory ensheathing cells, mensenchymal stem/stromal cells, neural precursors, oligodendrocyte progenitors, embryonic stem cells, and induced pluripotent stem cells. Through mechanisms of cell replacement, scaffolding, trophic support and immune modulation, each approach targets unique features of SCI pathology. However, as the injury is multifaceted, it is increasingly recognized that a combinatorial approach will be necessary to treat SCI. Expert opinion: Most preclinical studies, and an increasing number of clinical trials, are finding that single cell therapies have only modest benefits after SCI. These considerations, alongside issues of therapy cost-effectiveness, need to be addressed at the bench. In addition to exploring combinatorial strategies, researchers should consider cell reproducibility and storage parameters when designing animal experiments. Equally important, clinical trials must follow strict regulatory guidelines that will enable transparency of results.
Collapse
Affiliation(s)
- Anna Badner
- a Department of Genetics and Development , Krembil Research Institute, University Health Network , Toronto , ON , Canada.,b Institute of Medical Sciences , University of Toronto , Toronto , ON , Canada
| | - Ahad M Siddiqui
- a Department of Genetics and Development , Krembil Research Institute, University Health Network , Toronto , ON , Canada
| | - Michael G Fehlings
- a Department of Genetics and Development , Krembil Research Institute, University Health Network , Toronto , ON , Canada.,b Institute of Medical Sciences , University of Toronto , Toronto , ON , Canada.,c Canada Spinal Program , University Health Network, Toronto Western Hospital , Toronto , ON , Canada
| |
Collapse
|
92
|
Czarzasta J, Habich A, Siwek T, Czapliński A, Maksymowicz W, Wojtkiewicz J. Stem cells for ALS: An overview of possible therapeutic approaches. Int J Dev Neurosci 2017; 57:46-55. [PMID: 28088365 DOI: 10.1016/j.ijdevneu.2017.01.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 01/06/2017] [Accepted: 01/06/2017] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an unusual, fatal, neurodegenerative disorder leading to the loss of motor neurons. After diagnosis, the average lifespan ranges from 3 to 5 years, and death usually results from respiratory failure. Although the pathogenesis of ALS remains unclear, multiple factors are thought to contribute to the progression of ALS, such as network interactions between genes, environmental exposure, impaired molecular pathways and many others. The neuroprotective properties of neural stem cells (NSCs) and the paracrine signaling of mesenchymal stem cells (MSCs) have been examined in multiple pre-clinical trials of ALS with promising results. The data from these initial trials indicate a reduction in the rate of disease progression. The mechanism through which stem cells achieve this reduction is of major interest. Here, we review the to-date pre-clinical and clinical therapeutic approaches employing stem cells, and discuss the most promising ones.
Collapse
Affiliation(s)
- Joanna Czarzasta
- Department of Pathophysiology, Faculty of Medical Sciences, University of Warmia and Mazury, Olsztyn, Poland.
| | - Aleksandra Habich
- Department of Neurology and Neurosurgery, Faculty of Medical Sciences, University of Warmia and Mazury, Olsztyn, Poland
| | - Tomasz Siwek
- Department of Neurology and Neurosurgery, Faculty of Medical Sciences, University of Warmia and Mazury, Olsztyn, Poland
| | - Adam Czapliński
- Department of Neurology and Neurosurgery, Faculty of Medical Sciences, University of Warmia and Mazury, Olsztyn, Poland; Neurocentrum Bellevue, Neurology, Zurich, Switzerland
| | - Wojciech Maksymowicz
- Department of Neurology and Neurosurgery, Faculty of Medical Sciences, University of Warmia and Mazury, Olsztyn, Poland
| | - Joanna Wojtkiewicz
- Department of Pathophysiology, Faculty of Medical Sciences, University of Warmia and Mazury, Olsztyn, Poland; Laboratory of Regenerative Medicine, University of Warmia and Mazury, Olsztyn, Poland; Foundation for nerve cells regeneration, Olsztyn, Poland
| |
Collapse
|
93
|
Rauskolb S, Dombert B, Sendtner M. Insulin-like growth factor 1 in diabetic neuropathy and amyotrophic lateral sclerosis. Neurobiol Dis 2017; 97:103-113. [DOI: 10.1016/j.nbd.2016.04.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 03/29/2016] [Accepted: 04/29/2016] [Indexed: 12/12/2022] Open
|
94
|
Devine H, Patani R. The translational potential of human induced pluripotent stem cells for clinical neurology : The translational potential of hiPSCs in neurology. Cell Biol Toxicol 2016; 33:129-144. [PMID: 27915387 PMCID: PMC5325844 DOI: 10.1007/s10565-016-9372-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 11/18/2016] [Indexed: 12/14/2022]
Abstract
The induced pluripotent state represents a decade-old Nobel prize-winning discovery. Human-induced pluripotent stem cells (hiPSCs) are generated by the nuclear reprogramming of any somatic cell using a variety of established but evolving methods. This approach offers medical science unparalleled experimental opportunity to model an individual patient’s disease “in a dish.” HiPSCs permit developmentally rationalized directed differentiation into any cell type, which express donor cell mutation(s) at pathophysiological levels and thus hold considerable potential for disease modeling, drug discovery, and potentially cell-based therapies. This review will focus on the translational potential of hiPSCs in clinical neurology and the importance of integrating this approach with complementary model systems to increase the translational yield of preclinical testing for the benefit of patients. This strategy is particularly important given the expected increase in prevalence of neurodegenerative disease, which poses a major burden to global health over the coming decades.
Collapse
Affiliation(s)
- Helen Devine
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, WC1N3BG, UK.,Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London, UK
| | - Rickie Patani
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, WC1N3BG, UK. .,National Hospital for Neurology and Neurosurgery, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK. .,Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK. .,Euan MacDonald Centre for MND, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
95
|
Atassi N, Beghi E, Blanquer M, Boulis NM, Cantello R, Caponnetto C, Chiò A, Dunnett SB, Feldman EL, Vescovi A, Mazzini L, Bendotti C, Bersano E, Brajkovic S, Car P, De Marchi F, Fantozzi R, Follenzi A, Gelati M, Giorgi C, Grilli M, Guenzi P, La Bella V, Mancardi GL, Panzarasa G, Poloni M, Profico D, Silani V, Sorarù G, Spataro R, Stecco A, Vercelli A. Intraspinal stem cell transplantation for amyotrophic lateral sclerosis: Ready for efficacy clinical trials? Cytotherapy 2016; 18:1471-1475. [DOI: 10.1016/j.jcyt.2016.08.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 08/11/2016] [Accepted: 08/13/2016] [Indexed: 12/13/2022]
|
96
|
Abdul Wahid SF, Law ZK, Ismail NA, Azman Ali R, Lai NM. Cell-based therapies for amyotrophic lateral sclerosis/motor neuron disease. Cochrane Database Syst Rev 2016; 11:CD011742. [PMID: 27822919 PMCID: PMC6464737 DOI: 10.1002/14651858.cd011742.pub2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS), which is also known as motor neuron disease (MND) is a fatal disease associated with rapidly progressive disability, for which no definitive treatment as yet exists. Current treatment regimens largely focus on relieving symptoms to improve the quality of life of those affected. Based on data from preclinical studies, cell-based therapy is a promising treatment for ALS/MND. OBJECTIVES To assess the effects of cell-based therapy for people with ALS/MND, compared with placebo or no additional treatment. SEARCH METHODS On 21 June 2016, we searched the Cochrane Neuromuscular Specialised Register, CENTRAL, MEDLINE, and Embase. We also searched two clinical trials' registries for ongoing or unpublished studies. SELECTION CRITERIA We planned to include randomised controlled trials (RCTs), quasi-RCTs and cluster RCTs that assigned people with ALS/MND to receive cell-based therapy versus a placebo or no additional treatment. Co-interventions were allowable, provided that they were given to each group equally. DATA COLLECTION AND ANALYSIS We followed standard Cochrane methodology. MAIN RESULTS No studies were eligible for inclusion in the review. We identified four ongoing trials. AUTHORS' CONCLUSIONS Currently, there is a lack of high-quality evidence to guide practice on the use of cell-based therapy to treat ALS/MND.We need large, prospective RCTs to establish the efficacy of cellular therapy and to determine patient-, disease- and cell treatment-related factors that may influence the outcome of cell-based therapy. The major goals of future research should be to determine the appropriate cell source, phenotype, dose, and route of delivery, as these will be key elements in designing an optimal cell-based therapy programme for people with ALS/MND. Future research should also explore novel treatment strategies, including combinations of cellular therapy and standard or novel neuroprotective agents, to find the best possible approach to prevent or reverse the neurological deficit in ALS/MND, and to prolong survival in this debilitating and fatal condition.
Collapse
Affiliation(s)
| | - Zhe Kang Law
- Universiti Kebangsaan Malaysia Medical CentreDepartment of MedicineJalan Yaacob LatifBandar Tun RazakKuala LumpurMalaysia56000
| | - Nor Azimah Ismail
- Universiti Kebangsaan Malaysia Medical CentreCell Therapy CenterJalan Yaacob LatifKuala LumpurMalaysia56000
| | - Raymond Azman Ali
- Universiti Kebangsaan Malaysia Medical CentreNeurology Unit, Department of MedicineJalan Yaacob LatifBandar Tun RazakKuala LumpurMalaysia56000
| | - Nai Ming Lai
- Taylor's UniversitySchool of MedicineSubang JayaMalaysia
| |
Collapse
|
97
|
Syková E, Rychmach P, Drahorádová I, Konrádová Š, Růžičková K, Voříšek I, Forostyak S, Homola A, Bojar M. Transplantation of Mesenchymal Stromal Cells in Patients With Amyotrophic Lateral Sclerosis: Results of Phase I/IIa Clinical Trial. Cell Transplant 2016; 26:647-658. [PMID: 27938483 DOI: 10.3727/096368916x693716] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive untreatable neurodegenerative disorder, leading to the death of the cortical and spinal motoneurons (MNs). Bone marrow-derived mesenchymal stem/stromal cells (BM-MSCs) may represent a new approach to slowing down the progression of ALS by providing neurotrophic support to host MNs and by having an anti-inflammatory effect. We have designed a prospective, nonrandomized, open-label clinical trial (phase I/IIa, EudraCT No. 2011-000362-35) to assess the safety and efficacy of autologous multipotent BM-MSCs in ALS treatment. Autologous BM-MSCs were isolated and expanded under GMP conditions. Patients received 15 ± 4.5 × 106 of BM-MSCs via lumbar puncture into the cerebrospinal fluid. Patients were monitored for 6 months before treatment and then for an 18-month follow-up period. Potential adverse reactions were assessed, and the clinical outcome was evaluated by the ALS functional rating scale (ALSFRS), forced vital capacity (FVC), and weakness scales (WSs) to assess muscle strength on the lower and upper extremities. In total, 26 patients were enrolled in the study and were assessed for safety; 23 patients were suitable for efficacy evaluation. After intrathecal BM-MSC application, about 30% of the patients experienced a mild to moderate headache, resembling the headaches after a standard lumbar puncture. No suspected serious adverse reactions (SUSAR) were observed. We found a reduction in ALSFRS decline at 3 months after application (p < 0.02) that, in some cases, persisted for 6 months ( p < 0.05). In about 80% of the patients, FVC values remained stable or above 70% for a time period of 9 months. Values of WS were stable in 75% of patients at 3 months after application. Our results demonstrate that the intrathecal application of BM-MSCs in ALS patients is a safe procedure and that it can slow down progression of the disease.
Collapse
|
98
|
de Bie E, Oskarsson B, Joyce NC, Nicorici A, Kurillo G, Han JJ. Longitudinal evaluation of upper extremity reachable workspace in ALS by Kinect sensor. Amyotroph Lateral Scler Frontotemporal Degener 2016; 18:17-23. [DOI: 10.1080/21678421.2016.1241278] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Evan de Bie
- University of California at Davis, School of Medicine, Department of Public Health Sciences, Division of Biostatistics, Davis, California,
| | - Bjorn Oskarsson
- University of California at Davis, School of Medicine, Department of Neurology, Sacramento, California,
- Mayo Clinic, Jacksonville, Florida,
| | - Nanette C. Joyce
- University of California at Davis, School of Medicine, Department of Physical Medicine and Rehabilitation, Sacramento, California,
| | - Alina Nicorici
- University of California at Davis, School of Medicine, Department of Physical Medicine and Rehabilitation, Sacramento, California,
| | - Gregorij Kurillo
- University of California at Berkeley, College of Engineering, Department of Electrical Engineering and Computer Science, Berkeley, California, and
| | - Jay J. Han
- University of California at Irvine, School of Medicine, Department of Physical Medicine and Rehabilitation, Orange, California, USA
| |
Collapse
|
99
|
Goldman SA. Stem and Progenitor Cell-Based Therapy of the Central Nervous System: Hopes, Hype, and Wishful Thinking. Cell Stem Cell 2016; 18:174-88. [PMID: 26849304 DOI: 10.1016/j.stem.2016.01.012] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A variety of neurological disorders are attractive targets for stem and progenitor cell-based therapy. Yet many conditions are not, whether by virtue of an inhospitable disease environment, poorly understood pathophysiology, or poor alignment of donor cell capabilities with patient needs. Moreover, some disorders may be medically feasible targets but are not practicable, in light of already available treatments, poor risk-benefit and cost-benefit profiles, or resource limitations. This Perspective seeks to define those neurological conditions most appropriate for cell replacement therapy by considering its potential efficacy and clinical feasibility in those disorders, as well as potential impediments to its application.
Collapse
Affiliation(s)
- Steven A Goldman
- Center for Translational Neuromedicine and the Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA; Center for Basic and Translational Neuroscience, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen 2200, Denmark.
| |
Collapse
|
100
|
Abstract
Trauma is a leading cause of death in both military and civilian populations worldwide. Although medical advances have improved the overall morbidity and mortality often associated with trauma, additional research and innovative advancements in therapeutic interventions are needed to optimize patient outcomes. Cell-based therapies present a novel opportunity to improve trauma and critical care at both the acute and chronic phases that often follow injury. Although this field is still in its infancy, animal and human studies suggest that stem cells may hold great promise for the treatment of brain and spinal cord injuries, organ injuries, and extremity injuries such as those caused by orthopedic trauma, burns, and critical limb ischemia. However, barriers in the translation of cell therapies that include regulatory obstacles, challenges in manufacturing and clinical trial design, and a lack of funding are critical areas in need of development. In 2015, the Department of Defense Combat Casualty Care Research Program held a joint military–civilian meeting as part of its effort to inform the research community about this field and allow for effective planning and programmatic decisions regarding research and development. The objective of this article is to provide a “state of the science” review regarding cellular therapies in trauma and critical care, and to provide a foundation from which the potential of this emerging field can be harnessed to mitigate outcomes in critically ill trauma patients.
Collapse
|