Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
2.
PLoS One ; 4(12): e8314, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-20016813

ABSTRACT

BACKGROUND: Recombinant monoclonal antibodies have emerged as important tools for cancer therapy. Despite the promise shown by antibody-based therapies, the large molecular size of antibodies limits their ability to efficiently penetrate solid tumors and precludes efficient crossing of the blood-brain-barrier into the central nervous system (CNS). Consequently, poorly vascularized solid tumors and CNS metastases cannot be effectively treated by intravenously-injected antibodies. The inherent tumor-tropic properties of human neural stem cells (NSCs) can potentially be harnessed to overcome these obstacles and significantly improve cancer immunotherapy. Intravenously-delivered NSCs preferentially migrate to primary and metastatic tumor sites within and outside the CNS. Therefore, we hypothesized that NSCs could serve as an ideal cellular delivery platform for targeting antibodies to malignant tumors. METHODS AND FINDINGS: As proof-of-concept, we selected Herceptin (trastuzumab), a monoclonal antibody widely used to treat HER2-overexpressing breast cancer. HER2 overexpression in breast cancer is highly correlated with CNS metastases, which are inaccessible to trastuzumab therapy. Therefore, NSC-mediated delivery of trastuzumab may improve its therapeutic efficacy. Here we report, for the first time, that human NSCs can be genetically modified to secrete anti-HER2 immunoglobulin molecules. These NSC-secreted antibodies assemble properly, possess tumor cell-binding affinity and specificity, and can effectively inhibit the proliferation of HER2-overexpressing breast cancer cells in vitro. We also demonstrate that immunoglobulin-secreting NSCs exhibit preferential tropism to tumor cells in vivo, and can deliver antibodies to human breast cancer xenografts in mice. CONCLUSIONS: Taken together, these results suggest that NSCs modified to secrete HER2-targeting antibodies constitute a promising novel platform for targeted cancer immunotherapy. Specifically, this NSC-mediated antibody delivery system has the potential to significantly improve clinical outcome for patients with HER2-overexpressing breast cancer.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Breast Neoplasms/drug therapy , Drug Delivery Systems/methods , Neurons/cytology , Stem Cells/metabolism , Animals , Antibodies, Monoclonal, Humanized , Antibody Specificity/drug effects , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Culture Media, Conditioned/pharmacology , Female , Immunoglobulin G/immunology , Mice , Mice, Nude , Neurons/drug effects , Organ Specificity/drug effects , Receptor, ErbB-2/immunology , Stem Cells/drug effects , Trastuzumab , Xenograft Model Antitumor Assays
3.
Stem Cells ; 26(6): 1406-13, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18403751

ABSTRACT

Human neural and mesenchymal stem cells have been identified for cell-based therapies in regenerative medicine and as vehicles for delivering therapeutic agents to areas of injury and tumors. However, the signals required for homing and recruitment of stem cells to these sites are not well understood. Urokinase plasminogen activator (uPA) and urokinase plasminogen activator receptor (uPAR) are involved in chemotaxis and cell guidance during normal development and are upregulated in invasive tumors. Here we provided evidence that activation of uPA and uPAR in malignant solid tumors (brain, lung, prostate, and breast) augments neural and mesenchymal stem cell tropism. Expression levels of uPAR on human solid tumor cell lines correlated with levels of uPA and soluble uPAR in tumor cell-conditioned media. Cytokine expression profiles of these tumor-conditioned media were determined by protein arrays. Among 79 cytokines investigated, interleukin (IL)-6, IL-8, and monocyte chemoattractant protein-1 were the most highly expressed cytokines in uPAR-positive tumors. We provided evidence that human recombinant uPA induced stem cell migration, whereas depletion of uPA from PC-3 prostate cancer cell-conditioned medium blocked stem cell migration. Furthermore, retrovirus-mediated overexpression of uPA and uPAR in neuroblastoma (NB1691) cells induced robust migration of stem cells toward NB1691 cell-conditioned media, compared with media derived from wild-type NB1691 cells. We conclude that expression of uPA and uPAR in cancer cells underlies a novel mechanism of stem cell tropism to malignant solid tumors, which may be important for development of optimal stem cell-based therapies. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Mesenchymal Stem Cells/physiology , Neoplasms/physiopathology , Receptors, Cell Surface/physiology , Stem Cells/physiology , Urokinase-Type Plasminogen Activator/physiology , Brain Neoplasms/physiopathology , Breast Neoplasms/physiopathology , Cell Line, Tumor , Female , Humans , Lung Neoplasms/physiopathology , Male , Mesencephalon/embryology , Mesencephalon/physiopathology , Mesenchymal Stem Cells/cytology , Neuroblastoma , Polymerase Chain Reaction , Prostatic Neoplasms/physiopathology , Receptors, Cell Surface/genetics , Receptors, Urokinase Plasminogen Activator , Stem Cells/cytology , Urokinase-Type Plasminogen Activator/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...