Intranasal delivery of neural stem/progenitor cells: a noninvasive passage to target intracerebral glioma.

Standard

Intranasal delivery of neural stem/progenitor cells: a noninvasive passage to target intracerebral glioma. / Reitz, Matthias; Demestre, Maria; Sedlacik, Jan; Meissner, Hildegard; Fiehler, Jens; Kim, Seung U; Westphal, Manfred; Schmidt, Nils-Ole.

In: STEM CELL TRANSL MED, Vol. 1, No. 12, 12, 2012, p. 866-873.

Research output: SCORING: Contribution to journalSCORING: Journal articleResearchpeer-review

Harvard

Reitz, M, Demestre, M, Sedlacik, J, Meissner, H, Fiehler, J, Kim, SU, Westphal, M & Schmidt, N-O 2012, 'Intranasal delivery of neural stem/progenitor cells: a noninvasive passage to target intracerebral glioma.', STEM CELL TRANSL MED, vol. 1, no. 12, 12, pp. 866-873. https://doi.org/10.5966/sctm.2012-0045

APA

Reitz, M., Demestre, M., Sedlacik, J., Meissner, H., Fiehler, J., Kim, S. U., Westphal, M., & Schmidt, N-O. (2012). Intranasal delivery of neural stem/progenitor cells: a noninvasive passage to target intracerebral glioma. STEM CELL TRANSL MED, 1(12), 866-873. [12]. https://doi.org/10.5966/sctm.2012-0045

Vancouver

Bibtex

@article{e3bfbcc87d9d477ba8cd09e3d8c791e6,
title = "Intranasal delivery of neural stem/progenitor cells: a noninvasive passage to target intracerebral glioma.",
abstract = "Stem cell-based therapies for neurological disorders, including brain tumors, advance continuously toward clinical trials. Optimized cell delivery to the central nervous system remains a challenge since direct intracerebral injection is an invasive method with low transplantation efficiency. We investigated the feasibility of intranasal administration of neural stem/progenitor cells (NSPCs) as an alternative, noninvasive, and direct passage for the delivery of stem cells to target malignant gliomas. Tumor-targeting and migratory pathways of murine and human NSPCs were investigated by intravital magnetic resonance imaging and in histological time course analyses in the intracerebral U87, NCE-G55T2, and syngenic Gl261 glioblastoma models. Intranasally administered NSPCs displayed a rapid, targeted tumor tropism with significant numbers of NSPCs accumulating specifically at the intracerebral glioma site within 6 hours after intranasal delivery. Histological time series analysis revealed that NSPCs migrated within the first 24 hours mainly via olfactory pathways but also by systemic distribution via the microvasculature of the nasal mucosa. Intranasal application of NSPCs leads to a rapid, targeted migration of cells toward intracerebral gliomas. The directional distribution of cells accumulating intra- and peritumorally makes the intranasal delivery of NSPCs a promising noninvasive and convenient alternative delivery method for the treatment of malignant gliomas with the possibility of multiple dosing regimens.",
keywords = "Animals, Humans, Cells, Cultured, Disease Models, Animal, Mice, Mice, Inbred C57BL, Cell Movement, Magnetic Resonance Imaging, Cell Line, Tumor, Neoplasm Transplantation, Transplantation, Heterologous, Administration, Intranasal/*methods, Brain Neoplasms/pathology/*therapy, Fibroblasts/cytology, Frontal Lobe/cytology, Glioblastoma/pathology/*therapy, Neural Stem Cells/cytology/*transplantation, Parietal Lobe/cytology, Stem Cell Transplantation/*methods, Animals, Humans, Cells, Cultured, Disease Models, Animal, Mice, Mice, Inbred C57BL, Cell Movement, Magnetic Resonance Imaging, Cell Line, Tumor, Neoplasm Transplantation, Transplantation, Heterologous, Administration, Intranasal/*methods, Brain Neoplasms/pathology/*therapy, Fibroblasts/cytology, Frontal Lobe/cytology, Glioblastoma/pathology/*therapy, Neural Stem Cells/cytology/*transplantation, Parietal Lobe/cytology, Stem Cell Transplantation/*methods",
author = "Matthias Reitz and Maria Demestre and Jan Sedlacik and Hildegard Meissner and Jens Fiehler and Kim, {Seung U} and Manfred Westphal and Nils-Ole Schmidt",
year = "2012",
doi = "10.5966/sctm.2012-0045",
language = "English",
volume = "1",
pages = "866--873",
journal = "STEM CELL TRANSL MED",
issn = "2157-6564",
publisher = "ALPHAMED PRESS",
number = "12",

}

RIS

TY - JOUR

T1 - Intranasal delivery of neural stem/progenitor cells: a noninvasive passage to target intracerebral glioma.

AU - Reitz, Matthias

AU - Demestre, Maria

AU - Sedlacik, Jan

AU - Meissner, Hildegard

AU - Fiehler, Jens

AU - Kim, Seung U

AU - Westphal, Manfred

AU - Schmidt, Nils-Ole

PY - 2012

Y1 - 2012

N2 - Stem cell-based therapies for neurological disorders, including brain tumors, advance continuously toward clinical trials. Optimized cell delivery to the central nervous system remains a challenge since direct intracerebral injection is an invasive method with low transplantation efficiency. We investigated the feasibility of intranasal administration of neural stem/progenitor cells (NSPCs) as an alternative, noninvasive, and direct passage for the delivery of stem cells to target malignant gliomas. Tumor-targeting and migratory pathways of murine and human NSPCs were investigated by intravital magnetic resonance imaging and in histological time course analyses in the intracerebral U87, NCE-G55T2, and syngenic Gl261 glioblastoma models. Intranasally administered NSPCs displayed a rapid, targeted tumor tropism with significant numbers of NSPCs accumulating specifically at the intracerebral glioma site within 6 hours after intranasal delivery. Histological time series analysis revealed that NSPCs migrated within the first 24 hours mainly via olfactory pathways but also by systemic distribution via the microvasculature of the nasal mucosa. Intranasal application of NSPCs leads to a rapid, targeted migration of cells toward intracerebral gliomas. The directional distribution of cells accumulating intra- and peritumorally makes the intranasal delivery of NSPCs a promising noninvasive and convenient alternative delivery method for the treatment of malignant gliomas with the possibility of multiple dosing regimens.

AB - Stem cell-based therapies for neurological disorders, including brain tumors, advance continuously toward clinical trials. Optimized cell delivery to the central nervous system remains a challenge since direct intracerebral injection is an invasive method with low transplantation efficiency. We investigated the feasibility of intranasal administration of neural stem/progenitor cells (NSPCs) as an alternative, noninvasive, and direct passage for the delivery of stem cells to target malignant gliomas. Tumor-targeting and migratory pathways of murine and human NSPCs were investigated by intravital magnetic resonance imaging and in histological time course analyses in the intracerebral U87, NCE-G55T2, and syngenic Gl261 glioblastoma models. Intranasally administered NSPCs displayed a rapid, targeted tumor tropism with significant numbers of NSPCs accumulating specifically at the intracerebral glioma site within 6 hours after intranasal delivery. Histological time series analysis revealed that NSPCs migrated within the first 24 hours mainly via olfactory pathways but also by systemic distribution via the microvasculature of the nasal mucosa. Intranasal application of NSPCs leads to a rapid, targeted migration of cells toward intracerebral gliomas. The directional distribution of cells accumulating intra- and peritumorally makes the intranasal delivery of NSPCs a promising noninvasive and convenient alternative delivery method for the treatment of malignant gliomas with the possibility of multiple dosing regimens.

KW - Animals

KW - Humans

KW - Cells, Cultured

KW - Disease Models, Animal

KW - Mice

KW - Mice, Inbred C57BL

KW - Cell Movement

KW - Magnetic Resonance Imaging

KW - Cell Line, Tumor

KW - Neoplasm Transplantation

KW - Transplantation, Heterologous

KW - Administration, Intranasal/methods

KW - Brain Neoplasms/pathology/therapy

KW - Fibroblasts/cytology

KW - Frontal Lobe/cytology

KW - Glioblastoma/pathology/therapy

KW - Neural Stem Cells/cytology/transplantation

KW - Parietal Lobe/cytology

KW - Stem Cell Transplantation/methods

KW - Animals

KW - Humans

KW - Cells, Cultured

KW - Disease Models, Animal

KW - Mice

KW - Mice, Inbred C57BL

KW - Cell Movement

KW - Magnetic Resonance Imaging

KW - Cell Line, Tumor

KW - Neoplasm Transplantation

KW - Transplantation, Heterologous

KW - Administration, Intranasal/methods

KW - Brain Neoplasms/pathology/therapy

KW - Fibroblasts/cytology

KW - Frontal Lobe/cytology

KW - Glioblastoma/pathology/therapy

KW - Neural Stem Cells/cytology/transplantation

KW - Parietal Lobe/cytology

KW - Stem Cell Transplantation/methods

U2 - 10.5966/sctm.2012-0045

DO - 10.5966/sctm.2012-0045

M3 - SCORING: Journal article

VL - 1

SP - 866

EP - 873

JO - STEM CELL TRANSL MED

JF - STEM CELL TRANSL MED

SN - 2157-6564

IS - 12

M1 - 12

ER -