Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Clin Cancer Res ; 23(12): 2951-2960, 2017 Jun 15.
Article in English | MEDLINE | ID: mdl-27979915

ABSTRACT

Purpose: Human neural stem cells (NSC) are inherently tumor tropic, making them attractive drug delivery vehicles. Toward this goal, we retrovirally transduced an immortalized, clonal NSC line to stably express cytosine deaminase (HB1.F3.CD.C21; CD-NSCs), which converts the prodrug 5-fluorocytosine (5-FC) to 5-fluorouracil (5-FU).Experimental Design: Recurrent high-grade glioma patients underwent intracranial administration of CD-NSCs during tumor resection or biopsy. Four days later, patients began taking oral 5-FC every 6 hours for 7 days. Study treatment was given only once. A standard 3 + 3 dose escalation schema was used to increase doses of CD-NSCs from 1 × 107 to 5 × 107 and 5-FC from 75 to 150 mg/kg/day. Intracerebral microdialysis was performed to measure brain levels of 5-FC and 5-FU. Serial blood samples were obtained to assess systemic drug concentrations as well as to perform immunologic correlative studies.Results: Fifteen patients underwent study treatment. We saw no dose-limiting toxicity (DLT) due to the CD-NSCs. There was 1 DLT (grade 3 transaminitis) possibly related to 5-FC. We did not see development of anti-CD-NSC antibodies and did not detect CD-NSCs or replication-competent retrovirus in the systemic circulation. Intracerebral microdialysis revealed that CD-NSCs produced 5-FU locally in the brain in a 5-FC dose-dependent manner. Autopsy data indicate that CD-NSCs migrated to distant tumor sites and were nontumorigenic.Conclusions: Collectively, our results from this first-in-human study demonstrate initial safety and proof of concept regarding the ability of NSCs to target brain tumors and locally produce chemotherapy. Clin Cancer Res; 23(12); 2951-60. ©2016 AACR.


Subject(s)
Cytosine Deaminase/genetics , Genetic Therapy , Glioma/drug therapy , Neural Stem Cells/transplantation , Adolescent , Adult , Cytosine Deaminase/administration & dosage , Cytosine Deaminase/adverse effects , Female , Flucytosine/administration & dosage , Fluorouracil/administration & dosage , Gene Transfer Techniques , Genetic Vectors , Glioma/genetics , Glioma/pathology , Humans , Male , Middle Aged , Neoplasm Grading
2.
Stem Cell Reports ; 7(3): 483-495, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27546534

ABSTRACT

Pre-clinical studies indicate that neural stem cells (NSCs) can limit or reverse CNS damage through direct cell replacement, promotion of regeneration, or delivery of therapeutic agents. Immortalized NSC lines are in growing demand due to the inherent limitations of adult patient-derived NSCs, including availability, expandability, potential for genetic modifications, and costs. Here, we describe the generation and characterization of a new human fetal NSC line, immortalized by transduction with L-MYC (LM-NSC008) that in vitro displays both self-renewal and multipotent differentiation into neurons, oligodendrocytes, and astrocytes. These LM-NSC008 cells were non-tumorigenic in vivo, and migrated to orthotopic glioma xenografts in immunodeficient mice. When administered intranasally, LM-NSC008 distributed specifically to sites of traumatic brain injury (TBI). These data support the therapeutic development of immortalized LM-NSC008 cells for allogeneic use in TBI and other CNS diseases.


Subject(s)
Cell Differentiation/genetics , Cell Self Renewal/genetics , Gene Expression , Genes, myc , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Animals , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/therapy , Cell Movement/genetics , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Disease Models, Animal , Gene Expression Profiling , Genome-Wide Association Study , Heterografts , Humans , Mice , Neural Stem Cells/pathology , Stem Cell Transplantation , Transcriptome , Transduction, Genetic , Transgenes
3.
Biochim Biophys Acta ; 1865(2): 237-44, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26957416

ABSTRACT

Cellular senescence is defined by an irreversible growth arrest and is an important biological mechanism for suppression of tumor formation. Although deletion/mutation to DNA sequences is one mechanism by which cancer cells can escape senescence, little is known about the epigenetic factors contributing to this process. Histone modifications and chromatin remodeling related to the function of a histone demethylase, jumonji domain-containing protein 3 (JMJD3; also known as KDM6B), play an important role in development, tissue regeneration, stem cells, inflammation, and cellular senescence and aging. The role of JMJD3 in cancer is poorly understood and its function may be at the intersection of many pathways promoted in a dysfunctional manner such as activation of the senescence-associated secretory phenotype (SASP) observed in aging.


Subject(s)
Cellular Senescence , Jumonji Domain-Containing Histone Demethylases/physiology , Neoplasms/pathology , Cellular Reprogramming , Epigenesis, Genetic , Humans , Neoplasms/genetics
4.
Article in English | MEDLINE | ID: mdl-26654402

ABSTRACT

The depletion of stem cell pools and the accumulation of senescent cells in animal tissues are linked to aging. Planarians are invertebrate flatworms and are unusual in that their stem cells, called neoblasts, are constantly replacing old and dying cells. By eliminating neoblasts in worms via irradiation, the biological principles of aging are exposed in the absence of wound healing and regeneration, making planaria a powerful tool for aging research.

5.
Mol Cancer Res ; 13(4): 636-50, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25652587

ABSTRACT

UNLABELLED: Jumonji domain-containing protein 3 (JMJD3/KDM6B) demethylates lysine 27 on histone H3 (H3K27me3), a repressive epigenetic mark controlling chromatin organization and cellular senescence. To better understand the functional consequences of JMJD3 its expression was investigated in brain tumor cells. Querying patient expression profile databases confirmed JMJD3 overexpression in high-grade glioma. Immunochemical staining of two glioma cell lines, U251 and U87, indicated intrinsic differences in JMJD3 expression levels that were reflected in changes in cell phenotype and variations associated with cellular senescence, including senescence-associated ß-galactosidase (SA-ß-gal) activity and the senescence-associated secretory phenotype (SASP). Overexpressing wild-type JMJD3 (JMJD3wt) activated SASP-associated genes, enhanced SA-ß-gal activity, and induced nuclear blebbing. Conversely, overexpression of a catalytically inactive dominant negative mutant JMJD3 (JMJD3mut) increased proliferation. In addition, a large number of transcripts were identified by RNA-seq as altered in JMJD3 overexpressing cells, including cancer- and inflammation-related transcripts as defined by Ingenuity Pathway Analysis. These results suggest that expression of the SASP in the context of cancer undermines normal tissue homeostasis and contributes to tumorigenesis and tumor progression. These studies are therapeutically relevant because inflammatory cytokines have been linked to homing of neural stem cells and other stem cells to tumor loci. IMPLICATIONS: This glioma study brings together actions of a normal epigenetic mechanism (JMJD3 activity) with dysfunctional activation of senescence-related processes, including secretion of SASP proinflammatory cytokines and stem cell tropism toward tumors.


Subject(s)
Brain Neoplasms/pathology , Cellular Senescence , Glioma/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Brain Neoplasms/metabolism , Cell Line, Tumor , Cytokines/genetics , Cytokines/metabolism , Epigenesis, Genetic , Glioma/pathology , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Neoplasm Grading , Neural Stem Cells/immunology , Tropism
6.
Pathol Oncol Res ; 20(4): 789-97, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25168767

ABSTRACT

Malignant gliomas are among the deadliest primary brain tumors. Despite multimodal therapy and advances in chemotherapy, imaging, surgical and radiation techniques, these tumors remain virtually incurable. Glioma stem cells may be responsible for resistance to traditional therapies and tumor recurrence. Therefore, elimination of glioma stem cells may be crucial for achieving therapeutic efficacy. Metformin, a small molecule drug widely used in the therapy of type 2 diabetes, has shown significant anti-tumor effects in patients with breast cancer and prostate cancer. Recent preclinical data suggest that metformin also has therapeutic effects against glioma. Here we review the markers and hallmarks of glioma stem cells, and the molecular mechanisms involved in therapeutic targeting of glioma stem cells by metformin.


Subject(s)
Biomarkers, Tumor/analysis , Glioma/drug therapy , Glioma/pathology , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Neoplastic Stem Cells/drug effects , Humans , Neoplastic Stem Cells/pathology , Prognosis
7.
Stem Cells Transl Med ; 2(12): 983-92, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24167321

ABSTRACT

CPT-11 (irinotecan) has been investigated as a treatment for malignant brain tumors. However, limitations of CPT-11 therapy include low levels of the drug entering brain tumor sites and systemic toxicities associated with higher doses. Neural stem cells (NSCs) offer a novel way to overcome these obstacles because of their inherent tumor tropism and ability to cross the blood-brain barrier, which enables them to selectively target brain tumor sites. Carboxylesterases (CEs) are enzymes that can convert the prodrug CPT-11 (irinotecan) to its active metabolite SN-38, a potent topoisomerase I inhibitor. We have adenovirally transduced an established clonal human NSC line (HB1.F3.CD) to express a rabbit carboxylesterase (rCE) or a modified human CE (hCE1m6), which are more effective at converting CPT-11 to SN-38 than endogenous human CE. We hypothesized that NSC-mediated CE/CPT-11 therapy would allow tumor-localized production of SN-38 and significantly increase the therapeutic efficacy of irinotecan. Here, we report that transduced NSCs transiently expressed high levels of active CE enzymes, retained their tumor-tropic properties, and mediated an increase in the cytotoxicity of CPT-11 toward glioma cells. CE-expressing NSCs (NSC.CEs), whether administered intracranially or intravenously, delivered CE to orthotopic human glioma xenografts in mice. NSC-delivered CE catalyzed conversion of CPT-11 to SN-38 locally at tumor sites. These studies demonstrate the feasibility of NSC-mediated delivery of CE to glioma and lay the foundation for translational studies of this therapeutic paradigm to improve clinical outcome and quality of life in patients with malignant brain tumors.


Subject(s)
Brain Neoplasms/therapy , Camptothecin/analogs & derivatives , Carboxylic Ester Hydrolases/metabolism , Glioma/therapy , Neural Stem Cells/enzymology , Neural Stem Cells/transplantation , Topoisomerase I Inhibitors/pharmacology , Adenoviridae/genetics , Animals , Biotransformation , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Camptothecin/pharmacokinetics , Camptothecin/pharmacology , Carboxylesterase/deficiency , Carboxylesterase/genetics , Carboxylic Ester Hydrolases/genetics , Cell Line, Tumor , Dose-Response Relationship, Drug , Feasibility Studies , Genetic Vectors , Glioma/enzymology , Glioma/genetics , Glioma/pathology , Humans , Irinotecan , Mice , Mice, Knockout , Mice, SCID , Neural Stem Cells/drug effects , Rabbits , Time Factors , Tissue Distribution , Topoisomerase I Inhibitors/pharmacokinetics , Transduction, Genetic , Transfection , Xenograft Model Antitumor Assays
8.
Stem Cells Transl Med ; 2(10): 766-75, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24014682

ABSTRACT

Numerous stem cell-based therapies are currently under clinical investigation, including the use of neural stem cells (NSCs) as delivery vehicles to target therapeutic agents to invasive brain tumors. The ability to monitor the time course, migration, and distribution of stem cells following transplantation into patients would provide critical information for optimizing treatment regimens. No effective cell-tracking methodology has yet garnered clinical acceptance. A highly promising noninvasive method for monitoring NSCs and potentially other cell types in vivo involves preloading them with ultrasmall superparamagnetic iron oxide nanoparticles (USPIOs) to enable cell tracking using magnetic resonance imaging (MRI). We report here the preclinical studies that led to U.S. Food and Drug Administration approval for first-in-human investigational use of ferumoxytol to label NSCs prior to transplantation into brain tumor patients, followed by surveillance serial MRI. A combination of heparin, protamine sulfate, and ferumoxytol (HPF) was used to label the NSCs. HPF labeling did not affect cell viability, growth kinetics, or tumor tropism in vitro, and it enabled MRI visualization of NSC distribution within orthotopic glioma xenografts. MRI revealed dynamic in vivo NSC distribution at multiple time points following intracerebral or intravenous injection into glioma-bearing mice that correlated with histological analysis. Preclinical safety/toxicity studies of intracerebrally administered HPF-labeled NSCs in mice were also performed, and they showed no significant clinical or behavioral changes, no neuronal or systemic toxicities, and no abnormal accumulation of iron in the liver or spleen. These studies support the clinical use of ferumoxytol labeling of cells for post-transplant MRI visualization and tracking.


Subject(s)
Cell Tracking/methods , Ferrosoferric Oxide , Magnetic Resonance Imaging/methods , Neural Stem Cells/transplantation , Staining and Labeling/methods , Stem Cell Transplantation/methods , Animals , Humans , Immunohistochemistry , Mice
9.
Sci Transl Med ; 5(184): 184ra59, 2013 May 08.
Article in English | MEDLINE | ID: mdl-23658244

ABSTRACT

High-grade gliomas are extremely difficult to treat because they are invasive and therefore not curable by surgical resection; the toxicity of current chemo- and radiation therapies limits the doses that can be used. Neural stem cells (NSCs) have inherent tumor-tropic properties that enable their use as delivery vehicles to target enzyme/prodrug therapy selectively to tumors. We used a cytosine deaminase (CD)-expressing clonal human NSC line, HB1.F3.CD, to home to gliomas in mice and locally convert the prodrug 5-fluorocytosine to the active chemotherapeutic 5-fluorouracil. In vitro studies confirmed that the NSCs have normal karyotype, tumor tropism, and CD expression, and are genetically and functionally stable. In vivo biodistribution studies demonstrated NSC retention of tumor tropism, even in mice pretreated with radiation or dexamethasone to mimic clinically relevant adjuvant therapies. We evaluated safety and toxicity after intracerebral administration of the NSCs in non-tumor-bearing and orthotopic glioma-bearing immunocompetent and immunodeficient mice. We detected no difference in toxicity associated with conversion of 5-fluorocytosine to 5-fluorouracil, no NSCs outside the brain, and no histological evidence of pathology or tumorigenesis attributable to the NSCs. The average tumor volume in mice that received HB1.F3.CD NSCs and 5-fluorocytosine was about one-third that of the average volume in control mice. On the basis of these results, we conclude that combination therapy with HB1.F3.CD NSCs and 5-fluorocytosine is safe, nontoxic, and effective in mice. These data have led to approval of a first-in-human study of an allogeneic NSC-mediated enzyme/prodrug-targeted cancer therapy in patients with recurrent high-grade glioma.


Subject(s)
Glioma/drug therapy , Glioma/therapy , Neural Stem Cells/cytology , Prodrugs/therapeutic use , Animals , Cell Line , Cytosine Deaminase/metabolism , Female , Flow Cytometry , Flucytosine/metabolism , Flucytosine/therapeutic use , Fluorouracil/metabolism , Humans , Male , Mice , Mice, Nude , Neural Stem Cells/metabolism , Prodrugs/metabolism
10.
PLoS One ; 7(4): e35150, 2012.
Article in English | MEDLINE | ID: mdl-22539956

ABSTRACT

BACKGROUND: Glioblastoma multiforme (GBM) is the most aggressive type of malignant primary brain tumors in adults. Molecular and genetic analysis has advanced our understanding of glioma biology, however mapping the cellular composition of the tumor microenvironment is crucial for understanding the pathology of this dreaded brain cancer. In this study we identified major cell populations attracted by glioma using orthotopic rodent models of human glioma xenografts. Marker-specific, anatomical and morphological analyses revealed a robust influx of host cells into the main tumor bed and tumor satellites. METHODOLOGY/PRINCIPAL FINDINGS: Human glioma cell lines and glioma spheroid orthotopic implants were used in rodents. In both models, the xenografts recruited large numbers of host nestin-expressing cells, which formed a 'network' with glioma. The host nestin-expressing cells appeared to originate in the subventricular zone ipsilateral to the tumor, and were clearly distinguishable from pericytes that expressed smooth muscle actin. These distinct cell populations established close physical contact in a 'pair-wise' manner and migrated together to the deeper layers of tumor satellites and gave rise to tumor vasculature. The GBM biopsy xenografts displayed two different phenotypes: (a) low-generation tumors (first in vivo passage in rats) were highly invasive and non-angiogenic, and host nestin-positive cells that infiltrated into these tumors displayed astrocytic or elongated bipolar morphology; (b) high-generation xenografts (fifth passage) had pronounced cellularity, were angiogenic with 'glomerulus-like' microvascular proliferations that contained host nestin-positive cells. Stromal cell-derived factor-1 and its receptor CXCR4 were highly expressed in and around glioma xenografts, suggesting their role in glioma progression and invasion. CONCLUSIONS/SIGNIFICANCE: Our data demonstrate a robust migration of nestin-expressing host cells to glioma, which together with pericytes give rise to tumor vasculature. Mapping the cellular composition of glioma microenvironment and deciphering the complex 'crosstalk' between tumor and host may ultimately aid the development of novel anti-glioma therapies.


Subject(s)
Brain Neoplasms/metabolism , Glioma/metabolism , Animals , Brain Neoplasms/pathology , Cell Communication , Cell Line, Tumor , Chemokine CXCL12/metabolism , Disease Models, Animal , Female , Glioma/pathology , Humans , Immunohistochemistry , Intermediate Filament Proteins/metabolism , Mice , Neoplasm Invasiveness , Neovascularization, Pathologic , Nerve Tissue Proteins/metabolism , Nestin , Phenotype , Rats , Receptors, CXCR4/metabolism , Transplantation, Heterologous , Tumor Microenvironment
11.
Stem Cells ; 30(2): 314-25, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22084033

ABSTRACT

Metastasis to multiple organs is the primary cause of mortality in breast cancer patients. The poor prognosis for patients with metastatic breast cancer and toxic side effects of currently available treatments necessitate the development of effective tumor-selective therapies. Neural stem cells (NSCs) possess inherent tumor tropic properties that enable them to overcome many obstacles of drug delivery that limit effective chemotherapy strategies for breast cancer. We report that increased NSC tropism to breast tumor cell lines is strongly correlated with the invasiveness of cancer cells. Interleukin 6 (IL-6) was identified as a major cytokine mediating NSC tropism to invasive breast cancer cells. We show for the first time in a preclinical mouse model of metastatic human breast cancer that NSCs preferentially target tumor metastases in multiple organs, including liver, lung, lymph nodes, and femur, versus the primary intramammary fat pad tumor. For proof-of-concept of stem cell-mediated breast cancer therapy, NSCs were genetically modified to secrete rabbit carboxylesterase (rCE), an enzyme that activates the CPT-11 prodrug to SN-38, a potent topoisomerase I inhibitor, to effect tumor-localized chemotherapy. In vitro data demonstrate that exposure of breast cancer cells to conditioned media from rCE-secreting NSCs (NSC.rCE) increased their sensitivity to CPT-11 by 200-fold. In vivo, treatment of tumor-bearing mice with NSC.rCE cells in combination with CPT-11 resulted in reduction of metastatic tumor burden in lung and lymph nodes. These data suggest that NSC-mediated enzyme/prodrug therapy may be more effective and less toxic than currently available chemotherapy strategies for breast cancer metastases.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Camptothecin/analogs & derivatives , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Neural Stem Cells/transplantation , Prodrugs/therapeutic use , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacokinetics , Biotransformation , Camptothecin/administration & dosage , Camptothecin/pharmacokinetics , Camptothecin/therapeutic use , Carboxylesterase/biosynthesis , Carboxylesterase/metabolism , Cell Line, Tumor , Cell Movement , Cell Survival/drug effects , Drug Delivery Systems , Female , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Irinotecan , Lung Neoplasms/drug therapy , Lymphatic Metastasis , Mammary Neoplasms, Experimental/drug therapy , Mice , Mice, Nude , Neoplasm Invasiveness , Neural Stem Cells/enzymology , Neural Stem Cells/metabolism , Prodrugs/administration & dosage , Prodrugs/pharmacokinetics , Rabbits , Xenograft Model Antitumor Assays
12.
Biochim Biophys Acta ; 1816(2): 191-8, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21767610

ABSTRACT

Antibodies and antibody conjugates have emerged as important tools for cancer therapy. However, a major therapeutic challenge for the use of antibodies is their inability to cross the blood-brain barrier (BBB) to reach tumors localized in the central nervous system (CNS). Multiple methods have been developed to enhance antibody delivery to the CNS, including direct injection, mechanical or biochemical disruption of the BBB, conjugation to a 'molecular Trojan horse', cationization, encapsulation in nanoparticles and liposomes, and more recently, stem cell-mediated antibody delivery. In this review, we discuss each of these approaches, highlighting their successes and the obstacles that remain to be overcome.


Subject(s)
Antibodies/administration & dosage , Brain/metabolism , Animals , Antibodies/metabolism , Blood-Brain Barrier , Diffusion , Humans , Liposomes , Nanoparticles , Stem Cells/physiology , Transcytosis
13.
Stem Cells ; 28(11): 2084-7, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21089119

ABSTRACT

Monoclonal antibodies are important tools for cancer therapy, however, three factors limit their effectiveness: toxicity, poor tumor penetration, and inability to cross the blood-brain barrier. This review discusses the emerging field of stem cell-mediated antibody delivery and how this approach may improve antibody therapy of cancer by overcoming these obstacles.


Subject(s)
Antibodies/immunology , Neoplasms/therapy , Stem Cells/metabolism , Animals , Humans , Models, Biological , Neoplasms/immunology , Stem Cells/cytology
14.
Curr Stem Cell Res Ther ; 5(3): 273-6, 2010 Sep.
Article in English | MEDLINE | ID: mdl-19951251

ABSTRACT

Neural stem cells (NSCs) have been investigated in preclinical models as delivery vehicles for therapeutic genes for treatment of tumors in the central nervous system and other organs. Melanoma at early stages is effectively treated with surgery and radiotherapy, however metastatic disease is almost universally fatal, thus novel therapeutic approaches are needed. We studied the use of HB1.F3.CD therapeutic NSCs, a well-characterized clonal cell line derived from human fetal telencephalon, for their potential of secreting prodrug-activating enzyme. HB1.F3.CD cells were transduced by adenovirus encoding rabbit carboxylesterase (rCE), which converts CPT-11 into SN-38, a potent topoisomerase 1 inhibitor. In vitro cell migration assays revealed robust migration of NSCs to conditioned media from melanoma cells. Cytokine profiles showed that IL-6, IL-8, MCP-1 and TIMP-2, known chemoattractants for stem cells, were highly expressed by melanoma cells. Exposure of melanoma cells to conditioned media from the HB1.F3.CD.rCE cells in the presence of CPT-11 increased the tumor cell-killing effect by approximately 100-fold when compared to CPT-11 alone. Our data demonstrate the rational for NSC-based enzyme/prodrug therapeutic approach to target metastatic melanoma. Future experiments will evaluate the therapeutic efficacy of NSC-mediated melanoma therapy in animal models, which will provide the basis for targeted therapy in patients with advanced melanoma.


Subject(s)
Camptothecin/analogs & derivatives , Carboxylesterase/metabolism , Melanoma/therapy , Neurons/enzymology , Neurons/physiology , Stem Cells/enzymology , Stem Cells/physiology , Animals , Antineoplastic Agents, Phytogenic/metabolism , Antineoplastic Agents, Phytogenic/therapeutic use , Camptothecin/metabolism , Camptothecin/therapeutic use , Carboxylesterase/genetics , Cell Line, Tumor , Cell Movement/physiology , Cytokines/metabolism , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/therapeutic use , Humans , Irinotecan , Melanoma/metabolism , Melanoma/pathology , Neurons/cytology , Rabbits , Stem Cells/cytology
15.
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
16.
PLoS One ; 4(9): e7218, 2009 Sep 29.
Article in English | MEDLINE | ID: mdl-19787043

ABSTRACT

BACKGROUND: Treatment strategies for the highly invasive brain tumor, glioblastoma multiforme, require that cells which have invaded into the surrounding brain be specifically targeted. The inherent tumor-tropism of neural stem cells (NSCs) to primary and invasive tumor foci can be exploited to deliver therapeutics to invasive brain tumor cells in humans. Use of the strategy of converting prodrug to drug via therapeutic transgenes delivered by immortalized therapeutic NSC lines have shown efficacy in animal models. Thus therapeutic NSCs are being proposed for use in human brain tumor clinical trials. In the context of NSC-based therapies, MRI can be used both to non-invasively follow dynamic spatio-temporal patterns of the NSC tumor targeting allowing for the optimization of treatment strategies and to assess efficacy of the therapy. Iron-labeling of cells allows their presence to be visualized and tracked by MRI. Thus we aimed to iron-label therapeutic NSCs without affecting their cellular physiology using a method likely to gain United States Federal Drug Administration (FDA) approval. METHODOLOGY: For human use, the characteristics of therapeutic Neural Stem Cells must be clearly defined with any pertubation to the cell including iron labeling requiring reanalysis of cellular physiology. Here, we studied the effect of iron-loading of the therapeutic NSCs, with ferumoxide-protamine sulfate complex (FE-Pro) on viability, proliferation, migratory properties and transgene expression, when compared to non-labeled cells. FE-Pro labeled NSCs were imaged by MRI at tumor sites, after intracranial administration into the hemisphere contralateral to the tumor, in an orthotopic human glioma xenograft mouse model. CONCLUSION: FE-Pro labeled NSCs retain their proliferative status, tumor tropism, and maintain stem cell character, while allowing in vivo cellular MRI tracking at 7 Tesla, to monitor their real-time migration and distribution at brain tumor sites. Of significance, this work directly supports the use of FE-Pro-labeled NSCs for real-time tracking in the clinical trial under development: "A Pilot Feasibility Study of Oral 5-Fluorocytosine and Genetically modified Neural Stem Cells Expressing Escherichia coli Cytosine Deaminase for Treatment of Recurrent High-Grade Gliomas".


Subject(s)
Brain Neoplasms/pathology , Glioma/pathology , Iron/metabolism , Magnetic Resonance Imaging/methods , Neurons/cytology , Stem Cells/cytology , Animals , Brain/metabolism , Brain Neoplasms/metabolism , Dextrans , Disease Models, Animal , Ferrosoferric Oxide/chemistry , Glioma/metabolism , Humans , Magnetite Nanoparticles , Mice , Neoplasm Transplantation , Protamines/chemistry
17.
Mol Cancer Res ; 6(12): 1819-29, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19074827

ABSTRACT

Hypoxia is a critical aspect of the microenvironment in glioma and generally signifies unfavorable clinical outcome. Effective targeting of hypoxic areas in gliomas remains a significant therapeutic challenge. New therapeutic platforms using neural stem cells (NSC) for tumor-targeted drug delivery show promise in treatment of cancers that are refractory to traditional therapies. However, the molecular mechanisms of NSC targeting to hypoxic tumor areas are not well understood. Therefore, we investigated the role of hypoxia in directed migration of NSCs to glioma and identified the specific signaling molecules involved. Our data showed that hypoxia caused increased migration of human HB1.F3 NSCs to U251 human glioma-conditioned medium in vitro. In HB1.F3 NSCs, hypoxia led to up-regulation of CXCR4, urokinase-type plasminogen activator receptor (uPAR), vascular endothelial growth factor receptor 2 (VEGFR2), and c-Met receptors. Function-inhibiting antibodies to these receptors inhibited the migration of HB1.F3 cells to glioma-conditioned medium. Small interfering RNA knockdown of hypoxia-inducible factor-1alpha in glioma cells blocked the hypoxia-induced migration of NSCs, which was due to decreased expression of stromal cell-derived factor-1 (SDF-1), uPA, and VEGF in glioma cells. Our in vivo data provided direct evidence that NSCs preferentially distributed to hypoxic areas inside intracranial glioma xenografts, as detected by pimonidazole hypoxia probe, as well as to the tumor edge, and that both areas displayed high SDF-1 expression. These observations indicate that hypoxia is a key factor in determining NSC tropism to glioma and that SDF-1/CXCR4, uPA/uPAR, VEGF/VEGFR2, and hepatocyte growth factor/c-Met signaling pathways mediate increased NSC-to-glioma tropism under hypoxia. These results have significant implications for development of stem cell-mediated tumor-selective gene therapies.


Subject(s)
Brain Neoplasms/therapy , Cell Movement/physiology , Glioma/therapy , Hypoxia/pathology , Stem Cells/cytology , Animals , Antibody Specificity , Brain Neoplasms/pathology , Cell Line, Transformed , Culture Media, Conditioned , Cytokines/genetics , Cytokines/immunology , Drug Delivery Systems/methods , Female , Gene Expression Regulation, Neoplastic , Genetic Therapy/methods , Glioma/pathology , Humans , Hypoxia/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , Mice, Nude , Neutralization Tests , RNA, Small Interfering , Stem Cell Transplantation , Stem Cells/immunology , Telencephalon/cytology , Xenograft Model Antitumor Assays
18.
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
19.
Stem Cells ; 26(6): 1575-86, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18339768

ABSTRACT

The utility of neural stem cells (NSCs) has extended beyond regenerative medicine to targeted gene delivery, as NSCs possess an inherent tropism to solid tumors, including invasive gliomas. However, for optimal clinical implementation, an understanding of the molecular events that regulate NSC tumor tropism is needed to ensure their safety and to maximize therapeutic efficacy. We show that human NSC lines responded to multiple tumor-derived growth factors and that hepatocyte growth factor (HGF) induced the strongest chemotactic response. Gliomatropism was critically dependent on c-Met signaling, as short hairpin RNA-mediated ablation of c-Met significantly attenuated the response. Furthermore, inhibition of Ras-phosphoinositide 3-kinase (PI3K) signaling impaired the migration of human neural stem cells (hNSCs) toward HGF and other growth factors. Migration toward tumor cells is a highly regulated process, in which multiple growth factor signals converge on Ras-PI3K, causing direct modification of the cytoskeleton. The signaling pathways that regulate hNSC migration are similar to those that promote unregulated glioma invasion, suggesting shared cellular mechanisms and responses. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Brain Neoplasms/pathology , Glioma/pathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/physiology , Brain Neoplasms/enzymology , Cell Line , Cell Line, Tumor , Cell Movement , Chemotaxis , Glioma/enzymology , Humans , Kidney , Neurons/enzymology , Neurons/physiology , Stem Cells/enzymology , Stem Cells/physiology , rac1 GTP-Binding Protein/metabolism
20.
Stem Cells ; 26(1): 292-8, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17962698

ABSTRACT

This report presents highlights of discussions that focused on the biology of cancer stem cells as conducted at the fifth Annual Meeting of the International Society for Stem Cell Research, held in Cairns, Australia, June 17-20, 2007. The function of adult stem cells is believed to depend on their niches, that is, the microenvironment in which these stem cells reside. A similar concept applies to understanding the development of cancer, as it is becoming increasingly clear that only a small subset of cancer cell populations is capable of initiating/sustaining tumor formation. These tumorigenic cells, commonly referred to as cancer stem cells, also appear to reside in particular niches, and they bear the known, albeit dysfunctional, stem cell characteristics of self-renewal and differentiation. Dysregulation of stem cell niches is thought to contribute to tumorigenesis by affecting the complex network of signaling interactions that occur between stem cells and their neighboring cells, thus imbalancing the physiological controls on self-renewal and differentiation processes. This hypothesis was widely explored at the conference to shed new light on the mechanisms of tumor origin and progression and to unveil novel antitumor therapeutic approaches.


Subject(s)
Adult Stem Cells , Neoplastic Stem Cells , Animals , Epigenesis, Genetic , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...