Elsevier

Methods

Volume 99, 15 April 2016, Pages 37-43
Methods

Neural stem cell therapy for cancer

https://doi.org/10.1016/j.ymeth.2015.08.013Get rights and content

Highlights

  • Engineered neural stem cells (NSCs) are a promising new approach to cancer therapy.

  • NSCs display unique tumor-homing properties that make them ideal drug delivery vehicles.

  • NSCs can be engineered to deliver a variety of therapeutic agents directly into local and invasive cancer foci.

  • Engineered NSC therapy has achieved dramatic reductions in tumor volumes in preclinical testing.

  • NSC therapy has recently entered human patient trials.

Abstract

Cancers of the brain remain one of the greatest medical challenges. Traditional surgery and chemo-radiation therapy are unable to eradicate diffuse cancer cells and tumor recurrence is nearly inevitable. In contrast to traditional regenerative medicine applications, engineered neural stem cells (NSCs) are emerging as a promising new therapeutic strategy for cancer therapy. The tumor-homing properties allow NSCs to access both primary and invasive tumor foci, creating a novel delivery platform. NSCs engineered with a wide array of cytotoxic agents have been found to significantly reduce tumor volumes and markedly extend survival in preclinical models. With the recent launch of new clinical trials, the potential to successfully manage cancer in human patients with cytotoxic NSC therapy is moving closer to becoming a reality.

Introduction

Over the past two decades, the dogma in the central nervous system has shifted from one of a static organ incapable of change to the current understanding of adult neurogenesis [1]. The key to much of this change was the discovery of neural stem cells (NSCs). NSCs are generated by the differentiation of embryonic tissue and can serve as a source for replenishing neurons and glial cells in the adult brain throughout life. NSCs are defined by the expression of classic markers, including Nestin and Sox2, as well as their expansion in growth factor rich media that contains fibroblast growth factor and epidermal growth factor. NSCs display the hallmarks of stem cells, both self-renewing as well as differentiating into neurons, astrocytes, and oligodendrocytes. This differentiation capacity has led to significant investigation into the use of NSCs for regenerative medicine applications to correct damage to the brain and central nervous system caused by physical trauma or disease states. These studies have shown that NSC transplants survive in the diseased or damaged brain, and have therapeutic benefits in certain disease models.

In contrast to these traditional NSC therapies, the use of NSCs as tumor-homing drug carriers is an emerging area of interest that holds promise for treating malignant brain cancer [2], [3], [4], [5]. Pioneering studies by Aboody et al. and Benedetti et al. first revealed the unique ability of NSCs to home to brain cancer [6], [7]. These studies showed that NSCs transplanted at different sites throughout the brain migrated through the non-diseased parenchyma to localize selectively with cancer foci. When the NSCs were engineered with anti-cancer gene products, cytotoxic NSC therapy significantly inhibited the progression of cancer xenografts. These studies opened the door to the possibility of harnessing drug-loaded NSCs as a tumor-homing therapy. Ensuing studies over the past 15 years have further developed this concept, exploring novel cytotoxic agents, different routes of administration, and numerous molecular assays to define the mechanisms of migration. This exciting work has rapidly moved cytotoxic NSC therapy from preclinical mouse studies to a recent first-in-human clinical trial.

Section snippets

Glioblastoma

Glioblastoma (GBM) is the most common primary brain tumor, yet effectively treating this aggressive form of cancer remains a daunting challenge. GBM is classified as a grade IV glioma by the World Health Organization [8], [9]. The current clinical standard of care for GBM is surgical resection followed by chemo- and radiation therapy. Yet, median survival for GBM remains only 12–15 months and only 5% of patients survive 5 years [10], [11], [12]. GBM survival has not significantly improved in

Endogenous NSCs

NSCs possess the capacity to both self-replicate as well as differentiate into the primary cell types found in the CNS: neurons, astrocytes, and oligodendrocytes. While NSCs are ubiquitous in the developing brain, small populations of dormant NSCs that respond to injury can also be harvested from the subventricular zone (SVZ) or the subgranular zone (SGZ) of the dentate gyrus (DG) in adults [1], [19]. Isolation procedures vary yet typically involve microdissection and enzymatic digestion of

Homing

The main function of NSCs in the brain is to replace lost or injured neurons and glia by differentiation after migration to the injured zone. This migration to injured tissue is triggered by hypoxia through the associated up-regulation of the transcription factor hypoxia-inducible factor-1α (HIF-1α), which in turn activates the expression of NSC chemoattractants. These include chemokines and pro-angiogenic growth factors such as stromal cell-derived factor 1 (SDF-1), monocyte chemotactic

Enzyme/prodrug

Enzyme/prodrug therapy was the first approach used for engineered NSC therapy and the first strategy to enter human patient testing [6], [32]. In this approach, the NSCs are engineered to express an enzyme that converts a non-toxic prodrug into a cytotoxic product. This allows more precise control of the timing, levels, and location of drug release. This approach also adds an additional layer of safety as the prodrug typically kills the NSC drug carrier [75]. Cytosine deaminase (CD) was used in

Routes of administration

Determining the most effective route to administer cytotoxic NSC therapies represents an important step for eventual human use. Direct injection into the established GBM has been the mainstay of cytotoxic NSC delivery, and as numerous studies have found, this method leads to efficient NSC transplant and robust tumor killing [2], [93], [100]. However, directly injecting NSCs into the immunosuppressive tumor niche improves the survival of human NSC transplants and neglects their defining ability

Conclusions

Tumoricidal NSC therapy is opening new doors for cancer therapy. The tumor-homing capacity of these cells creates a powerful drug delivery platform that provides access to invasive cancer foci which traditional surgery, chemotherapy, and radio-therapy cannot typically access. NSCs have been engineered with a wide range of therapeutic agents, and typically achieve tumor reductions of 70–90% in preclinical models. Despite the success of these studies, many challenges still remain. The treatment

Acknowledgements

This work was supported by the UNC Lineberger Comprehensive Cancer Center’s University Cancer Research Fund and the UNC Translational and Clinical Sciences Institute (KL2TR001109, UL1TR001111).

References (106)

  • K.M. Joo

    Human neural stem cells can target and deliver therapeutic genes to breast cancer brain metastases

    Mol. Ther.

    (2009)
  • J.X. Yang

    CXCR4 receptor overexpression in mesenchymal stem cells facilitates treatment of acute lung injury in rats

    J. Biol. Chem.

    (2015)
  • J. Balzarini et al.

    Differential mechanism of cytostatic effect of (E)-5-(2-bromovinyl)-2′-deoxyuridine, 9-(1,3-dihydroxy-2-propoxymethyl)guanine, and other antiherpetic drugs on tumor cells transfected by the thymidine kinase gene of herpes simplex virus type 1 or type 2

    J. Biol. Chem.

    (1993)
  • D.W. Stuckey et al.

    TRAIL on trial: preclinical advances in cancer therapy

    Trends Mol. Med.

    (2013)
  • A. Almasan et al.

    Apo2L/TRAIL: apoptosis signaling, biology, and potential for cancer therapy

    Cytokine Growth Factor Rev.

    (2003)
  • P.V. Dickson

    Intravascular administration of tumor tropic neural progenitor cells permits targeted delivery of interferon-beta and restricts tumor growth in a murine model of disseminated neuroblastoma

    J. Pediatr. Surg.

    (2007)
  • J. Hwang et al.

    Functional domains of Pseudomonas exotoxin identified by deletion analysis of the gene expressed in E. coli

    Cell

    (1987)
  • K.S. Aboody et al.

    Stem and progenitor cell-mediated tumor selective gene therapy

    Gene Ther.

    (2008)
  • A.U. Ahmed et al.

    The use of neural stem cells in cancer gene therapy: predicting the path to the clinic

    Curr. Opin. Mol. Ther.

    (2010)
  • J.S. Young

    Advances in stem cells, induced pluripotent stem cells, and engineered cells: delivery vehicles for anti-glioma therapy

    Expert Opin. Drug Deliv.

    (2014)
  • D.W. Stuckey et al.

    Stem cell-based therapies for cancer treatment: separating hope from hype

    Nat. Rev. Cancer

    (2014)
  • K.S. Aboody

    Neural stem cells display extensive tropism for pathology in adult brain: evidence from intracranial gliomas

    Proc. Natl. Acad. Sci. U.S.A.

    (2000)
  • S. Benedetti

    Gene therapy of experimental brain tumors using neural progenitor cells

    Nat. Med.

    (2000)
  • P.Y. Wen et al.

    Malignant gliomas in adults

    N. Engl. J. Med.

    (2008)
  • C. Adamson

    Glioblastoma multiforme: a review of where we have been and where we are going

    Expert Opin. Invest. Drugs

    (2009)
  • O.P. Erpolat

    Outcome of newly diagnosed glioblastoma patients treated by radiotherapy plus concomitant and adjuvant temozolomide: a long-term analysis

    Tumori

    (2009)
  • R. Stupp

    Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma

    N. Engl. J. Med.

    (2005)
  • H. Brem

    Biodegradable polymers for controlled delivery of chemotherapy with and without radiation therapy in the monkey brain

    J. Neurosurg.

    (1994)
  • A.J. Sawyer et al.

    New methods for direct delivery of chemotherapy for treating brain tumors

    Yale J. Biol. Med.

    (2006)
  • M.H. Cohen et al.

    FDA drug approval summary: bevacizumab (Avastin) as treatment of recurrent glioblastoma multiforme

    Oncologist

    (2009)
  • C.A. Oomen

    Opposite effects of early maternal deprivation on neurogenesis in male versus female rats

    PLoS One

    (2009)
  • W. Guo et al.

    Isolation of multipotent neural stem or progenitor cells from both the dentate gyrus and subventricular zone of a single adult mouse

    Nat. Protoc.

    (2012)
  • P. Ellis

    SOX2, a persistent marker for multipotential neural stem cells derived from embryonic stem cells, the embryo or the adult

    Dev. Neurosci.

    (2004)
  • P. Koch et al.

    A rosette-type, self-renewing human ES cell-derived neural stem cell with potential for in vitro instruction and synaptic integration

    Proc. Natl. Acad. Sci. U.S.A.

    (2009)
  • D. Corbeil et al.

    Prominin-1 (CD133): molecular and cellular features across species

    Adv. Exp. Med. Biol.

    (2013)
  • F.H. Gage

    Mammalian neural stem cells

    Science

    (2000)
  • S.A. Goldman

    Progenitor cell-based treatment of the pediatric myelin disorders

    Arch. Neurol.

    (2011)
  • R.W. Sandrock

    Isolation, characterization and preclinical development of human glial-restricted progenitor cells for treatment of neurological disorders

    Regen Med.

    (2010)
  • L.S. Shihabuddin et al.

    Adult spinal cord stem cells generate neurons after transplantation in the adult dentate gyrus

    J. Neurosci.

    (2000)
  • P.H. Schwartz

    Isolation and characterization of neural progenitor cells from post-mortem human cortex

    J. Neurosci. Res.

    (2003)
  • S.K. Kim

    Human neural stem cells target experimental intracranial medulloblastoma and deliver a therapeutic gene leading to tumor regression

    Clin. Cancer Res.

    (2006)
  • T. Cho

    Human neural stem cells: electrophysiological properties of voltage-gated ion channels

    Neuroreport

    (2002)
  • K.S. Aboody

    Neural stem cell-mediated enzyme/prodrug therapy for glioma: preclinical studies

    Sci. Trans. Med.

    (2013)
  • J. Yu

    Induced pluripotent stem cell lines derived from human somatic cells

    Science

    (2007)
  • H.W. Choi

    Neural stem cells differentiated from iPS cells spontaneously regain pluripotency

    Stem cells

    (2014)
  • E.X. Lee

    Glioma gene therapy using induced pluripotent stem cell derived neural stem cells

    Mol. Pharm.

    (2011)
  • T. Yamazoe

    Potent tumor tropism of induced pluripotent stem cells and induced pluripotent stem cell-derived neural stem cells in the mouse intracerebral glioma model

    Int. J. Oncol.

    (2015)
  • Q. Zhou et al.

    In vivo reprogramming of adult pancreatic exocrine cells to beta-cells

    Nature

    (2008)
  • T. Vierbuchen

    Direct conversion of fibroblasts to functional neurons by defined factors

    Nature

    (2010)
  • E. Lujan et al.

    Direct conversion of mouse fibroblasts to self-renewing, tripotent neural precursor cells

    Proc. Natl. Acad. Sci. U.S.A.

    (2012)
  • Cited by (47)

    • Cytopharmaceuticals: An emerging paradigm for drug delivery

      2020, Journal of Controlled Release
      Citation Excerpt :

      It should be stated that stem cells can home to tumors and other pathological sites, e.g. areas of neurodegeneration, which offers the opportunity for stem cell-based cytopharmaceuticals to treat various diseases [58,59]. However, concerns regarding the malignant potential of stem cells after in vivo transfusion might limit their use in cytopharmaceuticals [60]. In a pursuit of sheer convenience for clinical application, approved drugs are considered the first choice of cytopharmaceuticals preparation.

    View all citing articles on Scopus
    View full text