Polymeric and lipid-based drug delivery systems for treatment of glioblastoma multiforme

https://doi.org/10.1016/j.jiec.2019.06.050Get rights and content

Abstract

Glioblastoma multiforme (GBM) is the most aggressive, malignant brain tumor found in adults, and has a short median survival time (MST). GBM is a heterogeneous group of brain tumors, is highly prone to develop resistance and likely to recur. In the context of GBM, the delivery of anti-cancer drugs is challenging because the blood brain barrier (BBB) restricts the passage of small molecules. Currently, nanomedicines based on liposomes, micelles, polymeric nanoparticles, and microparticles have attracted much attention, because they can cross the BBB and deliver anti-cancer drugs specifically to brain tumors. In this context, hydrogel-based systems incorporating nanoparticles, implantable carmustine wafers, microspheres, and lipid-based nanoparticles now appear to offer more effective, safer treatment strategies than conventional chemotherapeutic regimens. This review describes different polymeric hydrogel, chitosan, dendrimers, wafers, microspheres, and lipid-based nanoparticles like liposomes and solid-lipid nanoparticles that offers prominent strategies for the treatment and diagnosis of GBM.

Introduction

Glioblastoma multiforme (GBM) is a heterogeneous primary malignant brain tumor [1], [2]. GBM emerges from astrocytes and its cells rapidly reproduce due to the presence of a large network of blood vessels [3]. Uncontrolled cellular proliferation, resistance to radio and chemotherapy, growth of glial stem cells, invasion and infiltration of tumor cells, and apoptosis are characteristic of GBM [4]. GBM is also called the “octopus tumor” because it can extend tendrils to normal neighboring parenchymal cells [4], [5].

The World Health Organization (WHO) has classified astrocytoma into four grades (I, II, III and IV), and in grade IV, GBM has an incidence of 45–50%, although it may develop from low grade astrocytoma [6], [7]. The global incidence of GBM is 10 per 100,000 people [8]. According a report issued by the Central Brain Tumor Registry of the United States (CBTRUS, 2013), 12,760 new cases of GBM were predicted in 2018 [9]. In this report, relations between GBM incidence and age, gender were studied. Accordingly, due to expected increases in the size and mean age of the US population, the number of cases is expected to increase. Others have reported the incidence of GBM is highest in 75 to 84 year olds (at 15%) [8], [9], [10], and that it is greater for men than women [8], [11].

In the majority of cases, GBM is idiopathic, but some factors such as age, gender, family history, exposure to infections or strong electromagnetic fields, race, ethnicity, and a history of head injury or exposure to N-nitroso compounds are considered causes of GBM [12], [13], [14]. The clinical manifestations of GBM include edema, hemorrhage, and an altered mental status [15].

GBM is a highly aggressive neoplasm with an MST of 3 months if left untreated [16], though this can be improved by surgery plus radiotherapy or surgery plus chemotherapy or surgery plus radiotherapy and chemotherapy [17], [18], [19]. In 2004, the European Organization for Research and Treatment of Cancer stated GBM patients treated with radiotherapy plus temozolomide (TMZ) had an MST of 13–14 months, and radiotherapy alone had an MST of 11–13 months [8], [19].

GBM is aggressive, tends to recur, and is difficult to treat completely [20]. For newly diagnosed or recurrent GBM, the gold standard treatment is surgical resection followed by radiotherapy or chemotherapy with concomitant adjuvant TMZ chemotherapy [5] [Fig. 1]. Systemic treatments based on cytotoxic chemotherapy (e.g., TMZ [21], everolimus [22], or lomustine [23]) or hormonal therapy (e.g., using progesterone inhibitors, aromatase inhibitors, or hormone release growth hormone inhibitors [24]) are also commonly used to treat GBM [25].

In addition, molecular targeted therapies such as bevacizumab (targets vascular endothelial growth factor), cetuximab or nimotuzumab (target epidermal growth factor receptor), and CSF-1R inhibitor PLX3397 [26] or BLZ945 [27] (target colony stimulating factor-1 receptor) are emerging treatments for GBM [28]. Moreover, immunotherapies such as adoptive T-cell [29], tumor vaccine [30], and immune checkpoint [31] therapies have become a focus of current research. Programmed death-1 (PD-1), T-cell immunoglobulin mucin-3 (TIM-3), cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), and lymphocyte-activation gene 3 (LAG-3), which inhibit T-cell activation, are negative regulators of the immune system in GBM [32], and these ligands are also viewed as possible targets for GBM therapy [33]. In particular, programmed death ligand 1 (PD-L1) is overexpressed in GBM and monoclonal antibodies that inhibit PD-L1 or PD-1 receptor or its interactions with ligands offer other means of addressing its treatment [34]. Although, no Food and Drug Administration (FDA)-approved immunotherapy is available for GBM, the phase III trial of Ipilimumab and Nivolumab in GBM patients at different stages of treatment was initiated in 2014 (NCT02017717) [33].

As regards anti-cancer drugs, FDA approved TMZ as a first line drug for the treatment of GBM in 1999. TMZ is an alkylating anti-cancer drug that has been shown to increase patient survival [35], [36]. TMZ is converted intracellularly into MTIC (5-(3-methyltriazen-1-yl) imidazole-4-carboxamide) and methylates DNA at the N7 and O6 positions of guanine residues, which disrupts the cell repair mechanism and eventually causes cell death by breaking down double-stranded DNA [19], [37] [Fig. 2]. Standard TMZ based treatments include the combined use of TMZ, surgery, and radiation, but treatment efficacies are limited.

Development of different injectable hydrogel system have been developed for the treatment of diseases that are difficult to control like GBM. Such injectable hydrogel systems have potent capacity to encapsulate anti-cancer drugs and deliver successfully and efficiently at the site of action. The implantation of injectable hybrid hydrogel composed of protein-polymer conjugate to generate an effective platform for encapsulation and delivery of a DNA vaccine [38]. Similarly, pH- and temperature-responsive biodegradable copolymers were concomitant with human serum albumin to developed hybrid injectable hydrogels, that upgrade the stability and half-life of the biological drug [39]. Furthermore, the in situ forming injectable hydrogel [40] and stimuli‐sensitive injectable polymeric hydrogels [41] can also be the promising formulation for the delivery of therapeutic agents. These types of injectable formulation may control the anticancer drug release and subsequently eradicate the GBM.

Anatomical locations, high tumor heterogeneity leading to uncontrolled cellular proliferation, resistance to radiotherapy and chemotherapy, glial stem cell growth, invasion and infiltration of tumor cells, and apoptosis are the main reasons why GBM treatments are limited. On the other hand, resection has the shortcomings of causing collateral damage to neurological tissue, adversely affecting cognitive function of patients, and the different physical barrier in central nervous system (CNS) can delay in delivering of the anti-cancer agent to the tumor site [5]. Accordingly, different delivery strategies using drug carriers such as gold nanoparticles [42], [43], microspheres, or dendrimers that deliver anti-cancer drugs to tumor site without affecting neighboring healthy cells are being actively investigated [44], [45].

Cancer regrowth at original or different sites is problematic [46]. The main problem for this is that tumors can recur at original sites or migrate/metastasize to other parts of the body [47]. Primary treatment destroys most GBM cells, but some remain viable and continue to grow. GBM tumor cells have “finger-like tentacles”, which enable the disease to spread throughout the brain [48], [49], [50]. Furthermore, under the favorable microenvironment in the presence of neighboring cells, the vascular lymphatic network, hypoxic condition, and growth factor infiltered glial stem cells (GSCs) may grow into new tumor cells [49], [51] [Fig. 3].

Hypoxia is a stimulus found in the brain tumors. Hypoxia-responsive (HR) nanoparticles may show better anti-tumor effects in tumor treatment [52], [53], [54]. However, the treatment of GBM using HR-nanoparticles has not well-developed.

GBM recurrence is due to the ability of cancer cells to resist chemotherapy and radiotherapy [55], [56]. In one study, conventional therapies were found to cause GSCs to become temporarily latent [57]. The MST of patients that experience GBM recurrence is 8–9 months [55], which is extended by non-invasive stereotactic radiosurgery to 6.5–30 months. On the other hand, the MST of patients that undergo second surgery or are treated with TMZ for recurrence are 3.5–9 and 4.5 months, respectively [8], [58]. These reports shows TMZ is a moderately effective treatment for recurrent GBM [58], [59], [60], as re-surgery and re-irradiation may adversely affect quality of life.

Section snippets

Drug delivery systems (DDSs) and GBM treatment

TMZ, lomustine, carmustine, and bevacizumab are FDA approved for the treatment of GBM [61], but MST have not meaningfully improved due to recurrence [62]. These poor outcomes could be improved by the delivery of effective anti-cancer drugs through the BBB [63], [64]. Several developmental approaches have been devised based on chemical modifications of existing drugs, metallic or non-metallic nanoparticles [44], polymeric carriers [2], [65], [66], and lipid-based nanoparticles [28], [67], [68],

Polymeric DDSs used to treat GBM

Polymers play essential roles in modern drug delivery technology by providing a means to design the sustained release of hydrophilic and hydrophobic drugs [72]. Polymeric DDSs utilize different polymeric matrices such as hydrogels, nanoparticle, wafers, chitosan, and microspheres. These delivery systems provide the controlled release of different pharmaceutical active agents at high localized concentrations with limited systemic toxicities [71]. Polymeric DDSs have gained in popularity because

Wafers

Wafers are disc-shaped, synthetic or biodegradable implants containing an active pharmaceutical agent [159]. Wafers were developed to overcome the barrier effect of the BBB, and thus, enable the long-term delivery of drugs like carmustine [160], enhance bioavailabilities, reduce systemic toxic effects, and protect drugs.

Gliadel® wafers (also known as carmustine wafers) are composed of a biodegradable BCNU polymer impregnated with the alkylating agent carmustine [159], [161]. Gliadel® wafers are

Lipid-based nanoparticles

Lipid-based nanoparticles are potent carriers that improve drug bioavailability in or around disease targets in brain as they can cross the BBB. Such lipid-based nanoparticles are physiochemically stable, biocompatible, solubilize drugs well, and reduce drug-associated side effects [171], [172]. Liposomes [173], [174] and solid-lipid nanoparticles (SLNs) [67] are lipid-based nanoparticles with the potential to deliver anti-cancer drugs to brain and treat GBM. Liposomes are not all equal from

Conclusion and future perspectives

Researchers have made great progress at developing novel nanoparticle-based formulations with the aim of treating GBM, and there appears to be no end to the creativity displayed by those developing new therapeutics to prolong survival in GBM. In addition, combinations of current standard care and cell-targeting nano-carriers or polymeric or lipid-based nanoparticles have also been devised to address GBM cell resistance. Polymeric and lipid-based nanoparticles are viewed as promising carriers of

Acknowledgements

This study was supported by a grant from the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (Grant Nos: NRF-2018R1D1A1B07040858 and NRF-2016R1A6A1A03011325).

References (183)

  • C. Bastiancich et al.

    J. Control. Release

    (2017)
  • C. Bastiancich et al.

    J. Control. Release

    (2016)
  • S.A. Grossman et al.

    Semin. Oncol.

    (2004)
  • A. Malmström et al.

    Lancet Oncol.

    (2012)
  • X. Zhang et al.

    Exp. Ther. Med.

    (2012)
  • M. Wang et al.

    Glioma

    (2018)
  • K.K. Chow et al.

    Mol. Ther.

    (2013)
  • J. Zhang et al.

    Curr. Mol. Pharmacol.

    (2012)
  • M.S. Gil et al.

    J. Control. Release

    (2017)
  • T. Thambi et al.

    J. Control. Release

    (2017)
  • R. Stupp et al.

    Crit. Rev. Oncol. Hematol

    (2007)
  • T. Thambi et al.

    Biomaterials

    (2014)
  • T. Thambi et al.

    Acta Biomater.

    (2016)
  • S. Mallick et al.

    J. Egypt Natl. Canc. Inst.

    (2016)
  • O. Van Tellingen et al.

    Drug Resist. Updat.

    (2015)
  • L. Juillerat-Jeanneret

    Drug Discov Today

    (2008)
  • J. Yang et al.

    Bioact. Mater.

    (2016)
  • L. Battaglia et al.

    J. Pharm. Sci.

    (2014)
  • M.M. Nordling-David et al.

    J. Control. Release

    (2017)
  • Y. Malinovskaya et al.

    Int. J. Pharm.

    (2017)
  • X. Xu et al.

    Eur. J. Pharm. Biopharm.

    (2009)
  • C. Dufes et al.

    Adv. Drug Deliv. Rev.

    (2005)
  • M. Prabaharan

    Int. J. Biol. Macromol.

    (2015)
  • T. Fourniols et al.

    J. Control. Release

    (2015)
  • J.I. Kim et al.

    Biomaterials

    (2012)
  • M. Biondi et al.

    Adv. Drug Deliv. Rev.

    (2008)
  • L. Klouda

    Eur. J. Pharm. Biopharm.

    (2015)
  • M.E. Davis

    Clin. J. Oncol. Nurs.

    (2016)
  • S. Khawaldeh et al.

    Appl. Sci.

    (2017)
  • F.B. Furnari et al.

    Genes Dev.

    (2007)
  • D.N. Louis et al.

    Acta Neuropathol.

    (2016)
  • S.-W. Yoon et al.

    J. Pharm. Investig.

    (2019)
  • J.P. Thakkar et al.

    Cancer Epidemiol. Biomark. Prev.

    (2014)
  • Q.T. Ostrom et al.

    Neuro-oncology

    (2013)
  • K.R. Yabroff et al.

    Neuro Oncol.

    (2012)
  • B.P. McKinley et al.

    J. Neurosurg.

    (2000)
  • Q.T. Ostrom et al.

    Neuro Oncol.

    (2014)
  • M.C. Turner et al.

    Cancer Epidemiol. Biomark. Prev.

    (2014)
  • M. Wrensch et al.

    Neuro Oncol.

    (2002)
  • D.R. Johnson et al.

    Cancer

    (2012)
  • Neurosurgery

    (2008)
  • R. Stupp et al.

    N. Engl. J. Med.

    (2005)
  • M.H. Cohen et al.

    Oncologist

    (2009)
  • J.D. Hainsworth et al.

    Clin. Adv. Hematol. Oncol.

    (2012)
  • W. Wick et al.

    N. Engl. J. Med.

    (2017)
  • H.A. Burris

    Cancer Chemother. Pharmacol.

    (2013)
  • R. Baskar et al.

    Int. J. Med. Sci.

    (2012)
  • D. Yan et al.

    Oncogene

    (2017)
  • S.M. Pyonteck et al.

    Nat. Med.

    (2013)
  • L. Ampie et al.

    Front. Oncol.

    (2015)
  • Cited by (31)

    • Hybrid lipid-biopolymer nanocarrier as a strategy for GBM photodynamic therapy (PDT)

      2023, International Journal of Biological Macromolecules
    • Triple-negative breast cancer treatment meets nanoparticles: Current status and future direction

      2022, Journal of Drug Delivery Science and Technology
      Citation Excerpt :

      Dendrimers are hyper-branched, three-dimensional, symmetric, spherical, and nanosized synthetic structures composed of a core and inner and outer shells [65]. Dendrimers have a potential to conjugate high molecular weight hydrophilic agents by host-guest interactions or hydrophobic pharmaceutical agents through covalent conjugation [54]. Different types of dendrimers such as poly(etherhydroxylamine), poly(esteramine), polyamidoamine (PAMAM), poly(propyleneimine) (PPI), poly-l-lysine, melamine, and polyglycerol (PG) have been developed as a potential cargo [54,66].

    View all citing articles on Scopus
    View full text