Biomaterials to suppress cancer stem cells and disrupt their tumoral niche

https://doi.org/10.1016/j.ijpharm.2016.12.013Get rights and content

Abstract

Lack of improvement in the treatment options of several types of cancer can largely be attributed to the presence of a subpopulation of cancer cells with stem cell signatures and to the tumoral niche that supports and protects these cells. This review analyses the main strategies that specifically modulate or suppress cancer stem cells (CSCs) and the tumoral niche (TN), focusing on the role of biomaterials (i.e. implants, nanomedicines, etc.) in these therapies. In the case of CSCs, we discuss differentiation therapies and the disruption of critical cellular signaling networks. For the TN, we analyze diverse strategies to modulate tumor hypervascularization and hypoxia, tumor extracellular matrix, and the inflammatory and tumor immunosuppressive environment. Due to their capacity to control drug disposition and integrate diverse functionalities, biomaterial-based therapies can provide important benefits in these strategies. We illustrate this by providing case studies where biomaterial-based therapies either show CSC suppression and TN disruption or improved delivery of major modulators of these features. Finally, we discuss the future of these technologies in the framework of these emerging therapeutic concepts.

Introduction

Conventional cancer treatment is based on two premises: first, that cancer cells are a homogeneous population that displays a distinct phenotype as compared to healthy cells, and that medicines can take advantage of these differences to eliminate the disease (Clevers, 2011). The second premise is that the tumoral niche is a clinically advantageous feature, at least for nanomedicine-based therapies, since its enhances permeability to macromolecules and nanocarriers and promotes their accumulation in the tumor (Schätzlein, 2006). Nowadays, there is growing evidence demonstrating that these two premises are incorrect or at least incomplete.

Tumor cell heterogeneity is now a widely accepted feature of cancer and can be discussed at the genetic and developmental levels, being both of these tightly connected. At the genetic level, tumor cells present intrinsic genetic variability, which results in several cancer subclones that evolve following Darwinian processes in an attempt to adapt towards the environment. This process leads to an enrichment of cells presenting advantageous mutations and more aggressive phenotype (Greaves and Maley, 2012). At the developmental level, it has been confirmed that tumor initiation and relapse is driven by a selected tumor cell subpopulation that has high resistance towards conventional therapies and that takes advantage of stem cell-specific features (Farrar, 2009, Marotta and Polyak, 2009). Antitumorals are designed to target rapidly cycling cells such as those from the tumor bulk, but will spare the quiescent (but deadly) cancer stem cells (CSCs) that will generate tumor relapse and metastasis (Clevers, 2011).

Tumor niche (TN) refers to the microenvironment that interacts with tumor cells and regulates their fate. TN has been revealed as a critical barrier for cancer treatment and it is analyzed in this manuscript through four different features: the vascular niche, the inflammatory and immunosuppresive niche, the hypoxic niche and the extracellular matrix, all of which are closely related among themselves, with the CSC phenotype (Fig. 1). The tumor microenvironment was mostly seen as an advantage in the past, since it enhances the permeability and retention of the nano-sized drugs in the tumor (i.e. the EPR effect). However, tumor vasculature is highly irregular and could be tight in some regions, while being leaky in others. This irregular growth of tumor vasculature also generates non-functional branches, leading to poorly irrigated regions that cannot be easily accessed with chemotherapy (Jain, 2005). Besides, the accumulation of stroma in the tumor and the high intratumoral pressure also prevent drug transport to the inner regions. A recent survey of the literature has indicated that only a 0.7% of the nanocarrier dose is delivered to solid tumors (Wilhelm et al., 2016), a results that suggests the failure of the overall concept of passive targeting as it is understood nowadays.

Besides its barrier effect to drug delivery, the tumor niche also provides important signaling, often related to the cancer stem cell phenotype, that promotes tumor spreading and protection. CSCs and their niche have been recognized as critical features of cancer progression in the last years, and are currently in the focus of intense programs for drug development. Indeed, some prototypes have been developed to the stage of clinical implementation or are in advanced clinical trials (Fig. 2). Most of the programs, however, are still focusing on separate aspects of CSCs and the TN, and as it will be illustrated in this review, those features are tightly interconnected (Fig. 1) and might not be effectively addressed separately.

The field of biomaterials and drug delivery, mostly in tissue engineering, has focused on pulsing important cell signaling routes, particularly those related to stem cell development, and understanding and mimicking the biological substrate (the “niche”). Concretely, scaffolds and other tissue engineering devices are frequently used to: (i) induce stem differentiation (Prabhakaran et al., 2009), (ii) deliver cell-cycle modulators (Nayab et al., 2007), (iii) modulate the inflammatory niche (Lisignoli et al., 2006), (iv) modulate tissue vasculature (Stegemann and Nerem, 2003) and (v) induce extracellular matrix remodeling (Schneider et al., 2010). This spectrum of biological activity fits perfectly the requirements of a new generation of antitumorals capable of modulating CSCs and their niche.

The objective of this review is to analyze the properties and implications of the CSC phenotype and the TN, and to cover the main therapies designed to address these characteristics, focusing on the potential role of biomaterial-based technologies (i.e. implants, nanomedicines, etc.) in such therapies.

Section snippets

Cancer stem cells

Cancer stem cells (CSCs) have been defined as a cell subpopulation in the tumor bulk that possesses stem cell capacities. CSCs may be derived from adult stem cells or progenitor cells, but also from terminally differentiated cells that undergo epigenetic changes (Marotta and Polyak, 2009, Hermann et al., 2010). In any case, malignant cells take advantage of stem cell-specific signaling to drive tumor development.

CSCs were isolated for the first time in the 1990′s in acute myeloid leukemia, and

Aberrant vasculature

The formation of abnormal blood and lymphatic vessels is one of the critical hallmarks of solid tumors, which is driven by the secretion of high concentrations of angiogenic factors by the tumor cells. On one hand, the role of this abnormal vasculature is the nutrition and oxygenation of the tumor, but it also has a regulatory role in the secretion of growth factors and cytokines that spur tumor growth. Tumor-induced angiogenesis results in different vessels from those in normal vasculature as

Outlook and conclusions

While there have been considerable advances in oncology over the last decades, several types of cancer still present very low survival rates. Many of these are cancers where a tumor initiating cell subpopulation with stem cell-like properties has been reported. Despite the existence of this population, therapies that where under clinical development in the last decade failed to recognize the importance of cancer stem cells (CSCs), while they also neglected the importance of the main traits of

Conflict of interest

The authors declare no conflicts of interest.

Authors contribution

CGM and NC performed the bibliographic search. MGF designed the paper outline. All authors contributed to data analysis, writing and revision of the manuscript.

Acknowledgments

The authors acknowledge funding from Fundación BBVA, Proyectos de Investigación en Biomedicina (2014-PO0110) and Ministerio de Economía y Competitividad (SAF2014-58189-R, FEDER Funds).

References (178)

  • J.O. Eloy et al.

    Co-loaded paclitaxel/rapamycin liposomes: Development, characterization and in vitro and in vivo evaluation for breast cancer therapy

    Colloids Surf. B Biointerfaces

    (2016)
  • M.S. Goldberg

    Immunoengineering: how nanotechnology can enhance cancer immunotherapy

    Cell

    (2015)
  • S.I. Grivennikov et al.

    Immunity, inflammation, and cancer

    Cell

    (2010)
  • S. Gurunathan et al.

    Antiangiogenic properties of silver nanoparticles

    Biomaterials

    (2009)
  • S. Hamdy et al.

    Co-delivery of cancer-associated antigen and Toll-like receptor 4 ligand in PLGA nanoparticles induces potent CD8+T cell-mediated anti-tumor immunity

    Vaccine

    (2008)
  • G. Helbig et al.

    NF- kB promotes breast cancer cell migration and metastasis by inducing the expression of the chemokine receptor CXCR4

    J. Biol. Chem.

    (2003)
  • P.C. Hermann et al.

    Cancer stem cells in solid tumors

    Semin. Cancer Biol.

    (2010)
  • Y. Hori et al.

    Engulfing tumors with synthetic extracellular matrices for cancer immunotherapy

    Biomaterials

    (2009)
  • Z. Huang et al.

    Anti-tumor immune responses of tumor-associated macrophages via toll-like receptor 4 triggered by cationic polymers

    Biomaterials

    (2013)
  • S.R. Hwang et al.

    Phospholipid-based microemulsion formulation of all-trans-retinoic acid for parenteral administration

    Int. J. Pharm.

    (2004)
  • Y.-I. Jeong et al.

    All-trans-retinoic acid release from core-shell type nanoparticles of poly(epsilon-caprolactone)/poly(ethylene glycol) diblock copolymer

    Int. J. Pharm.

    (2004)
  • A. Kallioniemi

    Bone morphogenetic protein 4-a fascinating regulator of cancer cell behavior

    Cancer Genet.

    (2012)
  • M. Kanapathipillai et al.

    Nanoparticle targeting of anti-cancer drugs that alter intracellular signaling or influence the tumor microenvironment

    Adv. Drug Deliv. Rev.

    (2014)
  • H. Kang et al.

    The elevated level of CXCR4 is correlated with nodal metastasis of human breast cancer

    Breast

    (2005)
  • S. Karthikeyan et al.

    Anticancer activity of resveratrol-loaded gelatin nanoparticles on NCI-H460 non-small cell lung cancer cells

    Biomed. Prev. Nutr.

    (2013)
  • B. Keith et al.

    Hypoxia-inducible factors, stem cells, and cancer

    Cell

    (2007)
  • M. Leszczyniecka et al.

    Differentiation therapy of human cancer: basic science and clinical applications

    Pharmacol. Ther.

    (2001)
  • R.J. Li et al.

    All-trans retinoic acid stealth liposomes prevent the relapse of breast cancer arising from the cancer stem cells

    J. Control. Release

    (2011)
  • L. Li et al.

    Pigment epithelium-derived factor gene loaded in cRGD-PEG-PEI suppresses colorectal cancer growth by targeting endothelial cells

    Int. J. Pharm.

    (2012)
  • S. Lim et al.

    Altered chemical and biological activities of all- trans retinoic acid incorporated in solid lipid nanoparticle powders

    J. Control. Release

    (2004)
  • F. Abedini et al.

    Effects of CXCR4 siRNA/dextran-spermine nanoparticles on CXCR4 expression and serum LDH levels in a mouse model of colorectal cancer metastasis to the liver

    Cancer Manag. Res.

    (2011)
  • F. Abedini et al.

    Cationized dextran nanoparticle-encapsulated CXCR4 -siRNA enhanced correlation between CXCR4 expression and serum alkaline phosphatase in a mouse model of colorectal cancer

    Int. J. Nanomed.

    (2012)
  • R. Abellán-Pose et al.

    Lymphatic targeting of nanosystems for anticancer drug therapy

    Curr. Pharm. Des.

    (2015)
  • R. Abellan-Pose et al.

    Polyaminoacid nanocapsules for drug delivery to the lymphatic system: effect of the particle size

    Int. J. Pharm.

    (2016)
  • B. Al-husein et al.

    Antiangiogenic therapy for cancer: an update

    Pharmacotherapy

    (2012)
  • O.A. Ali et al.

    In situ regulation of DC subsets and T cells mediates tumor regression in mice

    Sci. Transl. Med.

    (2009)
  • O.A. Ali et al.

    Biomaterial-based vaccine induces regression of established intracranial glioma in rats

    Pharm. Res.

    (2011)
  • O.A. Ali et al.

    Identification of immune factors regulating antitumor immunity using polymeric vaccines with multiple adjuvants

    Cancer Res.

    (2014)
  • C. Allen et al.

    Polycaprolactone- b -poly (ethylene oxide) block copolymer micelles as a novel drug delivery vehicle for neurotrophic agents FK506 and L-685, 818

    Bioconjug. Chem.

    (1998)
  • N.G. Amado et al.

    Flavonoids and Wnt/β-catenin signaling: potential role in colorectal cancer therapies

    Int. J. Mol. Sci.

    (2014)
  • S.A. Anderson et al.

    Magnetic resonance contrast enhancement of neovasculature with alpha(v)beta(3)-targeted nanoparticles

    Magn. Reson. Med.

    (2000)
  • F. Balkwill

    Cancer and the chemokine network

    Nat. Rev. Cancer

    (2004)
  • M.H. Barcellos-Hoff et al.

    The evolution of the cancer niche during multistage carcinogenesis

    Nat. Rev. Cancer

    (2013)
  • A.C.F. Barreto et al.

    Magnetic nanoparticles for a new drug delivery system to control quercetin releasing for cancer chemotherapy

    J. Nanopart. Res.

    (2011)
  • G. Bartholomeusz et al.

    In vivo therapeutic silencing of hypoxia-inducible factor 1 alpha (HIF-1) using single-walled carbon nanotubes noncovalently coated with siRNA

    Nano Res.

    (2009)
  • D. Benjamin et al.

    Rapamycin passes the torch: a new generation of mTOR inhibitors

    Nat. Rev. Drug Discov.

    (2011)
  • O. Benny et al.

    An orally delivered small-molecule formulation with antiangiogenic and anticancer activity

    Nat. Biotechnol.

    (2008)
  • E. Blanco et al.

    Colocalized delivery of rapamycin and paclitaxel to tumors enhances synergistic targeting of the PI3 K/Akt/mTOR pathway

    Mol. Ther.

    (2014)
  • E. Borrajo et al.

    Docetaxel-loaded polyglutamic acid-PEG nanocapsules for the treatment of metastatic cancer

    J. Control. Release

    (2016)
  • P. Carmeliet et al.

    Angiogenesis in cancer and other diseases

    Nature

    (2000)
  • Cited by (14)

    • Recent advances in drug delivery systems for targeting cancer stem cells

      2021, Acta Pharmaceutica Sinica B
      Citation Excerpt :

      There have been quite a lot of biomarkers described, such as CD44, CD133, and epithelial cell adhesion molecule (EpCAM)66. The strong binding between the existing biomarkers and their specific antibodies provides enhanced targeting based on the EPR effect, which is accompanied by a high cellular uptake and increased drug concentration in CSCs13,38,67. In this respect, targeting CSCs based on their specific biomarkers is unquestionably a rational choice, and correspondingly, a great amount of biomarkers-mediated delivery carriers and systems have been developed and explored.

    • Nanotheranostics: A tactic for cancer stem cells prognosis and management

      2020, Journal of Drug Delivery Science and Technology
      Citation Excerpt :

      Under such circumstances, nanotechnology serves as a powerful tool where a variety of nanomedicines are being formulated to target specific cells with reduced side effects. The therapeutic effects of a variety of nanoparticles against cancer cells are being investigated through clinical trials [126]. Drugs are extremely potent when they are successfully delivered at the target site at an effective concentration while overcoming the physical and biological barriers like mucosal and blood-brain barriers.

    • Suppression of cancer stem cells

      2020, Biomaterials for Cancer Therapeutics: Evolution and Innovation
    • Study of nanostructured fibroin/dextran matrixes for controlled protein release

      2019, European Polymer Journal
      Citation Excerpt :

      For these studies, we encapsulated horseradish peroxidase II conjugated to fluorescein (FITC-HRP) in the different SF/DSS films. This model protein was selected because it has a molecular weight and isoelectric point similar to many proteins of therapeutic interest (such as the bone morphogenetic protein or fibroblast growth factor) [61,62], thus providing a relevant model for drug delivery characterization studies. In order to evaluate the cargo effect in the release profile three different loadings of FITC-HRP were employed in the blend films: 0.33, 0.99 and 2.00% w/w in relation to the SF content.

    • Apoferritin nanocages loading mertansine enable effective eradiation of cancer stem-like cells in vitro

      2018, International Journal of Pharmaceutics
      Citation Excerpt :

      Gupta et al., 2009; Li et al., 2015; Liu et al., 2015; Sun et al., 2015) However, due to the rarity of CSCs and high heterogeneity of tumor mass, their applications are largely hampered by the poor delivery efficiency to CSCs. A handful ligands-modified nanosystems have been explored to improve drug delivery to the CSCs (Garcia-Mazas et al., 2017; Kamal and Nazzal, 2018; Kim et al., 2014b; Liu et al., 2015; Sharma et al., 2017; Shen et al., 2016; Zhao et al., 2013), but their specific recognition by CSCs still remains a great limit (Kim et al., 2014a; Rao et al., 2015; Shen et al., 2016). Therefore, it is urgently needed to find novel effective strategies to improve the recognition and eradication of CSCs for anticancer therapy.

    View all citing articles on Scopus
    View full text