scholarly journals STMC-38. EYA1 IDENTIFIED BY IN VITRO AND IN VIVO PHAGE DISPLAY BIOPANNING DEMONSTRATES ROLE IN GLIOBLASTOMA STEM CELL PROLIFERATION THROUGH ONCOGENE INTERACTION

2016 ◽  
Vol 18 (suppl_6) ◽  
pp. vi190-vi191
Author(s):  
Ping Huang ◽  
Qiulian Wu ◽  
Jeremy Rich ◽  
James Liu
2014 ◽  
Vol 5 (4) ◽  
pp. 100 ◽  
Author(s):  
Joerg Hucklenbroich ◽  
Rebecca Klein ◽  
Bernd Neumaier ◽  
Rudolf Graf ◽  
Gereon Fink ◽  
...  

2019 ◽  
Vol 1 (Supplement_1) ◽  
pp. i4-i4
Author(s):  
JongMyung Kim ◽  
James Liu

Abstract To effectively target metastatic brain tumors (MBTs), the paradigm of treating MBTs after visualization on clinical imaging needs to be shifted to an understanding of the mechanisms that drive the formation and maintenance of brain metastasis-initiating cells (BMICs). Targeting this tumor sub-population, which may form as a result from activation of epithelial-mesenchymal transition, may allow for more effective means of isolating and targeting brain metastasis. In order to isolate BMICs, we have harvested cells from patient derived MBTs originating from lung cancer and cultured the cells using serum-free media conditions. In vivo phage display biopanning was used to isolate 12-amino acid length peptides that specifically target BMICs. Of the peptides recovered, one peptide, LBM4, was tested for specificity of binding to MBTs through in vitro and in vivo binding assays. When comparing patient derived metastatic brain tumors cells against brain metastasis cell lines, we found that both types of cells demonstrated similar morphology when grown in serum media conditions, but when grown in serum-free media, both demonstrated a tumor sphere morphology, similar to a stem cell-like state. LBM4 demonstrated specific binding to MBT cells over primary lung cancer cells in vitro through flow cytometry analysis and immunocytochemistry. Fluorescent tagged LBM4 intravenously injected into mice harboring intracranial BM demonstrated peptide localization to the tumor within the intracranial cavity visualized with live animal imaging. In vivo phage display biopanning is an effective tool that is able to isolate cell specific targeting peptides. MBT targeting peptides can potentially result in a shifting of the clinical treatment paradigm of brain metastases, through the development of more effective targeted therapeutics aimed at BMICs, as well as improving detection of MBT cells which may result in earlier tumor visualization as well as delineation of tumor recurrence versus radiation effects.


2015 ◽  
Vol 17 (suppl 5) ◽  
pp. v59.1-v59
Author(s):  
Ping Huang ◽  
Qiulian Wu ◽  
Jeremy Rich ◽  
James Liu

2019 ◽  
Vol 21 (Supplement_6) ◽  
pp. vi238-vi238
Author(s):  
JongMyung Kim ◽  
James Liu

Abstract To effectively target metastatic brain tumors (MBTs), the paradigm of initiating treatment against MBTs following detection on clinical imaging needs to be shifted to an understanding of the mechanisms that drive the formation and maintenance of brain metastasis-initiating cells (BMICs). Targeting this tumor sub-population, which may form as a result of activation of the epithelial-mesenchymal transition, may allow for more effective means of understanding and targeting brain metastases. In order to isolate BMICs, we have harvested cells from patient derived MBTs originating from lung cancer and cultured the cells using stem cell media conditions. We then performed in vitro and in vivo phage display biopanning to isolate 12-amino acid length peptides that specifically target BMICs. Several peptides were isolated from both in vitro and in vivo biopanning strategies. Of the peptides recovered, one peptide, LBM4, demonstrated specific binding to MBT cells over primary lung cancer cells in vitro through flow cytometry analysis and immunocytochemistry. Fluorescent tagged LBM4 intravenously injected into mice harboring intracranial brain metastases demonstrated peptide localization to the tumor within the intracranial cavity visualized with live animal imaging. Peptide imaging of tumor corresponded to MRI imaging confirming that the peptides could serve as an alternative to tumor imaging, with the potential for greater sensitivity resulting from the cellular targeting of MBTs. Our results demonstrate that we can use a combination of in vitro and in vivo phage display biopanning to isolate cell specific targeting peptides. MBT targeting peptides can potentially result in a shifting of the clinical treatment paradigm of brain metastases, through the development of more effective targeted therapeutics aimed at BMICs, as well as improving detection of MBT cells which may result in earlier tumor visualization, as well as delineation of tumor recurrence versus radiation effects.


1992 ◽  
Vol 47 (9) ◽  
pp. 1324-1332 ◽  
Author(s):  
Jens Freund ◽  
Afroditi Kapurniotu ◽  
Tadeusz A. Holak ◽  
Maryse Lenfant ◽  
Wolfgang Voelter

The solid phase synthesis of the inhibitor of hematopoietic stem cell proliferation, Ac–Ser–Asp–Lys–Pro–OH, and its derivative Ac–Ala–Asp–Lys–Pro–OH is described. 1H and 13C NMR investigations demonstrate that both peptides show no prefered conformation in water solution. Both peptides exist in a Pro-cis-trans equilibrium ratio of 9 (trans) : 1 (cis). Thymosin β4 is believed to be the precursor molecule of the tetrapeptide Ac–SDKP. The attachement of the random coil tetrapeptide to a rigid helical fragment could facilitate its in vivo enzymatic cleavage.


Sign in / Sign up

Export Citation Format

Share Document