ChapterPDF Available

Beyond the Blood–Brain Barrier: Facing New Challenges and Prospects of Nanotechnology-Mediated Targeted Delivery to the Brain

Authors:
  • SVKM’s NMIMS School of Pharmacy and Management Hyderabad Campus

Abstract and Figures

The blood–brain barrier (BBB) is a stealth barrier that protects the brain, and allows the entry of only a select pool of compounds into the brain and functions towards the maintenance of brain homeostasis. Brain tumors are one of the most prevalent tumors of the central nervous system that affect all age groups, and they have recently surpassed leukemia as the leading cause of cancer death. Due to the low permeability and high selectivity of the BBB to conventional anticancer drugs, effective treatments for brain tumors remain a challenge. In the last 20 years, tremendous efforts have been made to fabricate novel delivery strategies against brain cancer, and, amongst them, nanotechnology-mediated targeted delivery approaches have shown promising outcomes. In this chapter, we have summarized the role of the BBB and various nanotechnologies used to target drugs for the treatment of brain tumor. Herein, we also briefly discussed the hurdles, challenges, and safety considerations in using nanotechnology-mediated targeted delivery for brain tumors.
Content may be subject to copyright.
CHAPTER
15
Beyond the BloodBrain Barrier:
Facing New Challenges and Prospects
of Nanotechnology-Mediated Targeted
Delivery to the Brain
Mukesh Kumar
1
, Piyoosh Sharma
2
, Rahul Maheshwari
3
,
Muktika Tekade
4
, Sushant K. Shrivastava
2
and
Rakesh K. Tekade
3
1
Li Ka Shing Faculty of Medicine, School of Chinese Medicine, The University of Hong Kong,
Hong Kong
2
Department of Pharmaceutical Engineering & Technology, Indian Institute of
Technology (Banaras Hindu University), Varanasi, India
3
National Institute of Pharmaceutical
Education and Research (NIPER)- Ahmedabad, Gandhinagar, Gujarat, India
4
TIT College of
Pharmacy, Bhopal, Madhya Pradesh, India
OUTLINE
15.1 Introduction: Brain Tumor 398
15.2 Role of the BloodBrain Barrier
and Mechanism of Transport 401
15.3 Hurdles in Drug Delivery to the
Brain 404
15.4 Approaches in Delivering Drugs
to the Brain 405
15.4.1 Invasive Approach 406
15.4.2 Pharmacological Approach 407
15.4.3 Biological or Physiological
Approach 408
15.5 Nanotechnology and the
BloodBrain Barrier 409
15.6 Nanodevices for Brain Tumor
Targeting and Delivery 410
15.6.1 Liposomal Formulations 410
15.6.2 Polymeric Nanocarriers 412
15.6.3 Magnetic Nanoparticles 413
15.6.4 Dendrimeric Nanocarriers 414
397
Nanotechnology-based Targeted Drug Delivery Systems for Brain Tumors
DOI: https://doi.org/10.1016/B978-0-12-812218-1.00015-4 ©2018 Elsevier Inc. All rights reserved.
15.6.5 Carbon Nanotubes 416
15.6.6 Gold Nanoparticles 417
15.6.7 Viral Nanoparticles 418
15.6.8 Nucleic Acids-Based
Nanotechnology 418
15.7 Nanotechnology for Brain Tumor
Imaging 419
15.8 Challenges and Safety Considerations
of Nanotechnologies Used for
Brain-Targeted Delivery 421
15.9 Future Perspectives and
Conclusion 422
Acknowledgments 422
References 423
Further Reading 437
15.1 INTRODUCTION: BRAIN TUMOR
Cancer is one of the leading causes of mortality worldwide, with approximately 8.8 million
deaths in 2015 (Ferlay et al., 2013). Primary brain tumors are one of the most prevalent CNS
tumors, accounting for 8590% of cases and affecting all age groups (Mehta, Vogelbaum,
Chang, & Patel, 2011).Braincancerisaverycommonformofsolidmalignancyinchildrenand
adolescents (birth to 19 years), and it has surpassed leukemia as the leading cause of cancer
death (Adams et al., 2012; Siegel, Miller, & Jemal, 2016). According to a report of the cancer sta-
tistics for 2016, brain tumor is the second leading cause of cancer death in young males below
39 years of age and young females below 20 years of age (Adams et al., 2012; Siegel et al., 2016).
Brain tumors are classified by the WHO based on tumor morphology, cytogenetics, molecular
genetics, and immunologic markers into various types (Mehta et al., 2011). Based on tumor his-
tology and slow-growing and fast-growing properties, brain tumors are graded by their rate of
malignancy (Kleihues, Burger, & Scheithauer, 1993)(Table 15.1). The 2016 WHO classification
of brain tumors and their grading is summarized in Table 15.2 (Louis et al., 2016).
Recently, the American Cancer Society estimated 23,800 new cases and 16,700 deaths
from brain tumors and other nervous system tumors in the United States in the year 2017.
Brain tumors may be either noncancerous (benign) or cancerous (malignant). Of all the
brain tumors, glioblastoma is the most fatal and frequent malignant brain tumor (Adams
et al., 2012; Ellor, Pagano-Young, & Avgeropoulos, 2014). Glioblastomas and anaplastic
astrocytomas account for 38% of brain tumors and are the most common (Mehta et al.,
2011; Nagane, 2011; Tzeng & Green, 2013). Meningiomas and other mesenchymal tumors
account for 27% of brain tumors and are the second most common brain tumors (Mehta
et al., 2011). Glioblastoma or glioblastoma multiforme (GBM), one of the astrocytic grade
IV tumors, is a mitotically active, highly vascularized, and necrosis-prone tumor (Ellor
et al., 2014). Exposure to ionizing radiation, vinyl chloride, EpsteinBarr virus infection,
and HIV infection are risk factors for brain tumors (Perkins & Liu, 2016; McNeill, 2016;
Mehta et al., 2011; Schabet, 1999). Also, cellular phone use, radio frequency exposure, and
electromagnetic radiation have also been found to be risk factors for brain cancer (Group,
2010; Kheifets, Sussman, & Preston-Martin, 1999; Schu
¨z et al., 2006).
398 15. BEYOND THE BLOODBRAIN BARRIER
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
Interestingly, individuals with asthma and other allergic disorders such as hayfever,
eczema, and food allergies have a lower risk of brain cancer (Amirian et al., 2016; Wigertz
et al., 2007). However, the underlying mechanism of association between environmental,
occupational exposure and brain cancer risk is not entirely understood (da Silva, 2016). The
symptoms of brain tumors depend on the tumor type and anatomical regions of the lesions.
For example, meningioma, which is a very common intracranial brain tumor, originates in
the dura mater and is often asymptomatic (Aizer & Alexander, 2017; Vernooij et al., 2007),
while GBM, which is the most common intraparenchymal tumor of the CNS, is symptom-
atic. The general symptoms of brain tumor are headache, nausea, seizures, anorexia, nausea,
vomiting, lack of concentration, and mood disorders (Aizer & Alexander, 2017; Mehta et al.,
2011). Although the symptoms may correlate with the affected brain region, lesions in the
frontal lobe cause mood changes, lesions in the temporal lesions cause aphasia, while
cerebellar lesions cause ataxia and gait disturbances (Aizer & Alexander, 2017).
The ability of drugs to cross the bloodbrain barrier (BBB) and potentially target tumor
cells are a prerequisite for brain drug delivery that is unlikely to occur with most drugs
(Crawford, Rosch, & Putnam, 2016). Nanotechnology-based targeted drug delivery
systems are based on nanometer-size materials and devices which are used for biologic
applications and medicine (Lalu et al., 2017; Tekade, Maheshwari, Soni, Tekade, &
Chougule, 2017a; Tekade, Maheshwari, Soni, & Tekade, 2017b). Nanotechnology-based
drugs are designed in such a way that they cross the BBB and interact with tissue and cells
at the molecular level. Liposome, dendrimers, solid lipid nanoparticles and polymeric
nanoparticles are the majorly explored nanotechnology based platforms in recent years
TABLE 15.1 WHO Tumor Grading System Based on Histology and Proliferation Potential
WHO
Grade Criteria
IThe tumor cell has a low proliferative potential, and looks similar to a normal cell
The proliferation is slower than grades II, III, and IV tumor cells
There is a possibility of cure by surgical resection alone
II The tumor cell has a higher proliferative potential than grade I tumor cells with less mitotic
activity
Proliferation and metastasis are slower than grade III and IV tumor cells
The possibility of recurrence after surgery and becoming a higher-grade tumor is greater than
grade I tumor cells
III The tumor cell has a higher proliferation potential than grade I and II tumor cells and looks very
different from normal cells
Marked histopathological changes with an increased mitotic activity and highly metastatic in
nature
Treated with aggressive adjuvant chemotherapy
IV The tumor cell has a high proliferation potential with postoperative progression and fatal
outcomes
The tumor cells are mitotically active and necrosis-prone
Grade IV tumors cannot be cured, however, they are treated with aggressive adjuvant
chemotherapy
39915.1 INTRODUCTION: BRAIN TUMOR
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
(Maheshwari et al., 2012, 2015a; Soni et al., 2016b, 2017; Tekade et al., 2017a, 2017b;
Tekade, Maheshwari, Tekade, & Chougule, 2017c). Also, these drugs reach the brain tumor
and can be administered systemically, i.e., intravenous with minimal systemic side effects
(Crawford et al., 2016). Successful drug delivery in the case of brain tumor is very chal-
lenging, and requires multidisciplinary approaches comprising of engineering, chemistry,
cell biology, physiology, pharmacology, and medicine (Silva, 2007). In this chapter, we
TABLE 15.2 Drug Transporter Expressed in the Brain and Involved in Multidrug Resistance in Brain Cancer
SN Transporter Short Description Anticancer Drug
1. P-glycoprotein
(MDR1;
ABCB1)
This is a prototypic energy- and
Na
1
-dependent transporter involved in the
multidrug resistance of cancer cells. ABCB1
or multidrug resistance 1 (MDR1) gene is
responsible for the expression of
P-glycoprotein
Doxorubicin, Daunorubicin, Docetaxel,
Epirubicin, Etoposide, Idarubicin,
Methotrexate, Mitoxantrone, Paclitaxel,
Teniposide, Vinblastine, Vincristine
2 MRP1
(ABCC1)
The ABCC1 gene encodes multidrug
resistance-associated protein 1 (MRP1) in
humans. The ABCC1 transporter protein is
especially prevalent in neuroblastoma
(Munoz, Henderson, Haber, & Norris, 2007)
Daunorubicin, Doxorubicin, Epirubicin,
Etoposide, Melphalan, Methotrexate,
Teniposide, Vinblastine, Vincristine
3. MRP2
(ABCC2)
Multidrug resistance-associated protein 2
(MRP2), also known as canalicular
multispecific organic anion transporter
1 (cMOAT), is encoded by the ABCC2 gene
in humans
Cisplatin, Doxorubicin, Epirubicin,
Etoposide, Flavopiridol, Methotrexate,
Vincristine
4. MRP3
(ABCC3)
Multidrug resistance-associated protein
3 (MRP3) is a member of the superfamily of
ATP-binding cassette (ABC) transporters
and is encoded by the ABCC3 gene in
humans
Etoposide, Methotrexate, Teniposide
5. MRP4
(ABCC4)
The ABCC4 gene encodes multidrug
resistance-associated protein 4 (MRP4) in
humans
Methotrexate, 6-Mercaptopurine,
Thioguanine, Topotecan
6. MRP5
(ABCC5)
Multidrug resistance-associated protein
5 (MRP5) is encoded by the ABCC5 gene
in humans
6-Mercaptopurine, Thioguanine
7. MRP6
(ABCC6)
Multidrug resistance-associated protein
6 (MRP6), also known as multispecific
organic anion transporter E, is encoded
by the ABCC6 gene in human
Actinomycin D, Cisplatin, Daunorubicin,
Doxorubicin, Etoposide, Teniposide, BCRP
8. BCRP
(ABCG2)
Breast cancer resistance protein (BCRP) is
the second member of the G subfamily of
the large ATP-binding cassette transporter
superfamily (ABCG2). It is encoded by the
ABCG2 gene in humans
Doxorubicin, Bisantrene, Irinotecan,
Methotrexate, Mitoxantrone, Topotecan
400 15. BEYOND THE BLOODBRAIN BARRIER
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
have summarized the role of the bloodbrain barrier and various nanotechnologies used
for targeting drugs for the treatment of brain tumor. Herein, we also briefly discussed the
hurdles, challenges, and safety considerations in using nanotechnologies for brain tumors.
15.2 ROLE OF THE BLOODBRAIN BARRIER AND MECHANISM
OF TRANSPORT
The bloodbrain barrier (BBB) is a most critical barrier that separates brain tissue from
the peripheral circulation (Ballabh, Braun, & Nedergaard, 2004; Saunders, Ek, Habgood, &
Dziegielewska, 2008). The BBB is a complex structure comprising blood vessel endothelial
cells, junction complex (tight junctions and adherence junction), and astrocytes. The endo-
thelial cells of the BBB are different from endothelial cells of the peripheral blood circula-
tion as these cells are devoid of fenestrations, and have a more extensive junction complex
and thin pinocytic vesicular transport system. The endothelial cell tight junction complex
restricts the paracellular flux of polar molecules across the BBB, while small nonpolar
molecules diffuse freely across plasma membranes (Ballabh et al., 2004; Ransohoff, 2016).
This barrier acts as a selective gate for the entry of molecules into the brain, and it limits
the diffusion of potentially harmful substances into the brain. It also has a critical role in
the transportation of essential nutrients such as glucose, amino acids and larger molecules
such as insulin, leptin, and iron transferrin into the brain and removal of metabolites and
harmful substances from the brain via specific receptors and transporters (Ransohoff,
2016; Saunders, Habgood, Møllga
˚rd, & Dziegielewska, 2016). The tight junctions (TJ) and
adherence junctions (AJ) are the building blocks of the BBB. The TJ comprises of integral
membrane proteins such as claudin, occludin, junction adhesion molecules, and various
cytoplasmic accessory proteins, while AJ comprises of a cadherin, catenin, alpha-actinin,
and vinculin. Occludin and claudins form a complex of proteins spanning the intercellular
cleft with junctional adhesion molecules and several cytoplasmic scaffolding and regula-
tory proteins (Wolburg & Lippoldt, 2002; Wolburg, Noell, Mack, Wolburg-Buchholz, &
Fallier-Becker, 2009). The adherens junctions hold the cells together, giving the tissue
structural support via a complex formation of cadherin and scaffolding proteins alpha-,
beta-, and gamma-catenin. These are crucial for the integrity of tight junctions, and disrup-
tion of AJs leads to disruption of the BBB (Wolburg & Lippoldt, 2002). Astrocytes play a
significant role in the induction and maintenance of TJ and for polarized expression of
transporters in the luminal and abluminal endothelial membranes (Cheslow & Alvarez,
2016; Ronaldson & Davis, 2016; Wolburg et al., 2009).
Chemotherapeutic agents have limited efficacy in the treatment of brain tumors due to
the impediment to delivery across the intact BBB and brain-to-blood efflux of drugs.
Moreover, WHO grade III and IV brain tumor patients have a disrupted BBB. Early studies
have demonstrated a correlation between the reduction of TJ and tumor differentiation and
experimental evidence has emerged to place TJ in the frontline as the structure that cancer
cells must overcome in order to metastasize (Bauer, Erly, Moser, Maya, & Nael, 2015;
Liebner et al., 2000; Long, 1970, 1979;Papadopoulos et al., 2001). Glioma-derived factors
such as transforming growth factor beta-2, caveolin-1, reactive oxygen species, matrix metal-
loproteinases, aquaporins, and proinflammatory cytokines are responsible for the
40115.2 ROLE OF THE BLOODBRAIN BARRIER AND MECHANISM OF TRANSPORT
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
degradations of tight TJs (Ishihara et al., 2008; Schneider et al., 2004; Wolburg, Noell, Fallier-
Becker, Mack, & Wolburg-Buchholz, 2012). However, the degraded TJ and leaky BBB do not
result in optimal pharmacokinetics and dynamics of chemotherapeutic agents. Because in
tumor cells increased expression and activity of efflux transporter, drug metabolizing
enzymes and cerebral blood flow dynamics leads to inefficient drug concentrations in the
brain (Neuwelt et al., 1986; Wei et al., 2013). The BBB strictly regulates the brain ion concen-
tration and maintains it optimal for synaptic signaling activity. The concentration is main-
tained in spite of changes that can occur in plasma following exercise or disease conditions
(Hansen, 1985; Hladky & Barrand, 2016; Jeong et al., 2006; Nischwitz, Berthele, & Michalke,
2008). The BBB also regulates the excitatory amino acid glutamate levels in the brain by
keeping the peripheral plasma glutamate pools away from the central glutamate pool
(Abbott, Ro
¨nnba
¨ck, & Hansson, 2006; Bernacki, Dobrowolska, Nierwin
˜ska, & Malecki, 2008).
Moreover, the BBB prevents the entry of plasma proteins such as albumin, prothrombin,
and plasminogen into the brain. The endogenous metabolites or proteins or xenobiotics
which sometimes act as neurotoxins are also expelled from the brain via some energy-
dependent efflux transporters (Abbott, Patabendige, Dolman, Yusof, & Begley, 2010).
Molecules, including nutrients and drugs, can cross the BBB via several mechanisms of
transportation. Lipid-soluble/nonpolar and low-molecular-weight molecules can pass
freely across the BBB and enter the brain along with the concentration gradient (Liu, Tu,
Kelly, Chen, & Smith, 2004). Meanwhile highly polar molecules and high-molecular-
weight molecules which have a tendency to form more than six hydrogen bonds cannot
pass freely cross the BBB (Clark, 2003; Gleeson, 2008). However, lipid solubility and the
molecular weight of molecules are not absolute factors for passive transportation; there are
several drug molecules which do not follow the above rule of passive transportation across
the BBB (Bodor & Buchwald, 2003). Oxygen and carbon dioxide freely move across the
BBB, along with their concentration gradients, while bicarbonate ions which have a nega-
tive charge are poorly permeable to the BBB. This is because molecules that carry a posi-
tive charge have an advantage of interaction with negatively charged cell membrane
components which aids in the passage of these molecules across the BBB.
Solute carriers are another important mechanism for transportation across the BBB.
Highly polar molecules are not able to diffuse across the BBB, and thus there are large
numbers of solute carriers in the BBB (Suhy, Webb, Papp, Geier, & Sadee, 2017; Zhang,
Knipp, Ekins, & Swaan, 2002a; Zhou, Zhu, Wang, & Murray, 2016). These solute carriers
in the BBB facilitate many essential polar nutrients, such as glucose and amino acids, into
the brain. Some of these solute carriers depend on Na
1
gradients such as sodium-
dependent glucose transporter, sodium myoinositol cotransporter, an amino acid trans-
porter of glutamate and aspartate (EAAT1), and nucleosides, nucleotides, and nucleobases
(CNT1) transporter (Abbott et al., 2010; Rose & Verkhrastky, 2016).
ATP-binding cassette transporters (ABC transporters) in the BBB are increasingly recog-
nized as a key factor for the distribution and elimination of drugs from the brain (Chen
et al., 2016; Lo
¨scher & Potschka, 2005a). These transporters are responsible for the much
lower BBB passage of highly lipophilic drugs (Chen et al., 2016). These efflux transporters
are involved in the removal of potentially neurotoxic endogenous or xenobiotic molecules
from the brain and play a crucial role in detoxification of the brain (Dallas, Miller, &
Bendayan, 2006). The most common and extensively studied ABC efflux transporter is
402 15. BEYOND THE BLOODBRAIN BARRIER
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
P-glycoprotein (Pgp). Previous studies have demonstrated a key role of Pgp in the efflux
of a variety of lipid-soluble drugs out of the brain because several drugs are substrates
for these ABC efflux transporters (Begley, 2004; Chen et al., 2016). Other ABC efflux
transporters present in the BBB are the multidrug resistance protein (MRP) family and
breast cancer resistance protein (BCRP) (Fletcher, Williams, Henderson, Norris, & Haber,
2016; Szaka
´cs, Paterson, Ludwig, Booth-Genthe, & Gottesman, 2006).
The polar, larger-molecular-weight molecules, such as peptides, can cross the BBB by
transcytosis. Transcytosis is a type of transcellular transport by which larger-molecular-
weight molecules are transported across the BBB via an endocytic mechanism into the brain
(Goulatis & Shusta, 2017; Jones & Shusta, 2007). Transcytosis is of two types: receptor-
mediated transcytosis and adsorptive-mediated transcytosis (Abbott et al., 2010; Herve
´,
Ghinea, & Scherrmann, 2008). Receptor-mediated transcytosis involves the binding of
macromolecules (ligand) to specific receptors followed by endocytosis of ligandreceptor
complex across the BBB. Examples of receptor-mediated transcytosis are transport of trans-
ferrin, melanotransferrin, insulin, leptin, TNF-alpha, and epidermal growth factor (Fig. 15.1)
FIGURE 15.1 Schematic representation of the various transport process of molecules across the BBB. Adapted
with permission from Fernandes, Soni, & Patravale (2010).
40315.2 ROLE OF THE BLOODBRAIN BARRIER AND MECHANISM OF TRANSPORT
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
(Banks, Niehoff, Martin, & Farrell, 2002; Demeule et al., 2002a; Pan & Kastin, 1999; Pan,
Kastin, & Brennan, 2000; Visser, Voorwinden, Crommelin, Danhof, & de Boer, 2004).
In adsorptive-mediated transcytosis, an interaction between the positive charge on the
macromolecule and cell surface binding sites results in endocytosis of this complex across
the BBB (Sauer, Dunay, Weisgraber, Bienert, & Dathe, 2005). Transport of cationized
albumin and cell-penetrating peptides (Synb5/pAnt-(43-45)) across the BBB are examples
of adsorptive-mediated transcytosis (Drin, Cottin, Blanc, Rees, & Temsamani, 2003;
Pardridge, Triguero, Buciak, & Yang, 1990).
15.3 HURDLES IN DRUG DELIVERY TO THE BRAIN
Globally, approximately 1.5 billion people suffer from some central nervous system dis-
orders at any given time (Brasnjevic, Steinbusch, Schmitz, Martinez-Martinez, & Initiative,
2009). Bloodbrain barrier permeability is a major challenge for the scientist to meet the
high demand from patients for effective treatments. The numbers of people affected by
CNS disorders is a trend that will increase due to the increasing number of older adults
in the general population. If effective treatments fail to meet the demand, the number
of people with CNS disorders will increase to approximately 1.9 billion by 2020
(Pardridge, 2001).
The major hurdle in drug delivery to the brain is the bloodbrain barrier and enzymes
which restrict the entry of drugs into the brain. The bloodbrain barrier, together with
enzymes such as drug-metabolizing enzymes, play an important role in the homeostasis of
the brain by providing a gateway for the selective entry of any substances including drugs
(Pablo Rigalli, Ciriaci, Domingo Mottino, Alicia Catania, & Laura Ruiz, 2016). The BBB
acts as an anatomical and biochemical barrier which protects the brain from the entry of
pathogens and potentially harmful substances.
Brain endothelial cells are different from the microvasculature in the periphery, the endo-
thelial cells of brain capillaries are closely attached and form tight junctions, with limited
fenestrations and pinocytic vesicles. TJ also called zonulae occludes, restrict the passage of
hydrophilic molecules and macromolecules into the brain. Tight junctions with other pro-
teins such as claudins, occludins, and junctional adhesion molecules form a junctional com-
plex which maintains the integrity of endothelial cells (Agarwal, Sane, Oberoi, Ohlfest, &
Elmquist, 2011; Ballabh et al., 2004; Vartanian et al., 2014). Moreover, the endothelial cells
are surrounded by astrocyte foot processes, which further strengthens the BBB. This prop-
erty of the brain makes most of the treatments for CNS disorders such as brain tumor,
Alzheimer disease, Parkinson disease, and mood disorders ineffective and is pushing
researchers to develop novel and effective brain drug delivery systems. Interestingly, in a
report, one scientist reported that only 1% of pharmaceutical companies globally are
involved in brain drug delivery programs (Pardridge, 2005), which shows a considerable
demand for effective treatment for brain-related disorders.
The understanding of BBB physiology in normal/disease conditions and the nature of
various transport receptors at the BBB helps researchers to overcome the failure of effec-
tive brain drug delivery. There are several factors responsible for crossing the BBB to gain
entry into the brain (Upadhyay, 2014). Binding of the drug to a transporter, opening and
404 15. BEYOND THE BLOODBRAIN BARRIER
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
closing of ion channels due to ionic concentration lipophilicity, enzymatic degradation of
drugs, larger-molecular-weight drugs, the presence of functional groups, and the presence
of charged residues of the molecules are factors which decide the BBB permeability of any
drugs (Huttunen & Rautio, 2011; Schaddelee et al., 2003; Seelig, Gottschlich, & Devant,
1994). Though the drug enters the brain and is therapeutically available, it becomes
inactive by the presence of catabolic enzymes in the brain tissue (Foti et al., 2015; Tamai &
Tsuji, 1996). Another major hurdle in drug delivery to the brain is drug resistance
(Lo
¨scher & Potschka, 2005b). In general, lipophilicity is one of the prerequirements for
BBB penetration and to reach targets in the brain. However, several lipophilic drugs have
shown much less BBB permeability than would be predicted by their lipophilicity (Begley,
2004). This is due to the high binding affinity of these drugs to drug efflux transporters
and multidrug resistance protein family which are present at the BBB (de Lange, 2004;
Lingineni, Belekar, Tangadpalliwar, & Garg, 2017; Sun, Dai, Shaik, & Elmquist, 2003).
Particularly in the case of brain tumors, anticancer drugs with a high lipophilicity have
shown remarkably poor efficacy in the treatment of primary or metastatic brain tumors.
Because these drugs have a high affinity towards multidrug ABC transporters such as
Pgp or MRPs (Table 15.2), this makes the drugs less bioavailable to the tumor site
(Bart et al., 2000; Kemper, Boogerd, Thuis, Beijnen, & van Tellingen, 2004; Oberoi et al.,
2016; Wardill et al., 2016).
15.4 APPROACHES IN DELIVERING DRUGS TO THE BRAIN
Effective treatment of neurological disorders, such as brain tumors, psychological disor-
ders, and neurodegenerative disorders depends on successful drug delivery to the brain
(Chen, Dalwadi, & Benson, 2004; Choonara, Kumar, Modi, & Pillay, 2016; Oberoi et al.,
2016). The major challenge for successful drug delivery to the brain is to cross the protec-
tive barriers of the BBB and bloodcerebrospinal fluid (CSF) barrier (BCSFB). The compo-
sitions of the BBB and BCSFB are different, the BBB comprises of the tight junctions of
endothelial cells and the foot processes of astrocytes, while the BCSFB comprises of tight
junctions of choroid plexus cells with intracellular gaps and fenestration (Marques et al.,
2016). Although, the BBB and BCSFB have different physical compositions they both
participate in controlling the selective passage of molecules between the blood and brain
parenchyma or CSF (Dwibhashyam & Nagappa, 2008; Marques et al., 2016).
The circulating molecules in the blood enter the brain or CSF through the endothelial
cells or choroid plexus cells by passive diffusion or active transport or a combination of
both. Passive transport is a process in which molecules get to enter the brain or CSF
freely and is the primary pathway for many therapeutic agents. As a rule of thumb, only
lipid-soluble molecules with a molecular mass under 400600 Da can enter through the
tight junction of the BBB. However, water-soluble molecules and macromolecules with a
mass of ,600 Da may enter into the brain through carrier-mediated transport
(Vykhodtseva, McDannold, & Hynynen, 2008), receptor-mediated transport, or
absorptive-mediated transport (Goulatis & Shusta, 2017; Sai et al., 1998). Essential nutri-
ents such as amino acids, glucose, carboxylic acids, and nucleosides enter into the brain
through these transporters (Daneman & Prat, 2015). The receptor-mediated transporters
40515.4 APPROACHES IN DELIVERING DRUGS TO THE BRAIN
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
involved in the transport of surface receptor ligand-conjugated macromolecules include
transferrin and insulin (Burkhart et al., 2016; Goulatis & Shusta, 2017; May, Bedel, Shen,
Woods, & Liu, 2016).
In active transport, the molecules cross a cell membrane against the concentration gradi-
ent that is guided by cellular energy, i.e., adenosine triphosphate (ATP). Transporters such
as P-glycoprotein, multidrug-resistant protein, multidrug resistance-associated protein,
breast cancer resistance protein, organic anion transporter, and organic cation transporter
help in the active transportation of small hydrophilic molecules (Ro
¨mermann et al., 2013;
Tamai & Tsuji, 2000). As discussed earlier, several factors are responsible for successful
drug delivery across the BBB and the BCSFB. Due to the complexity of the BBB and
BCSFB, the systemic drug concentration is very high in conventional drug administration,
which significantly increases the risk of toxicity. To bypass the BBB and to deliver drugs
into the brain, without any systemic toxicity, three different approaches are currently
used: invasive, pharmacological, and biological or physiological.
15.4.1 Invasive Approach
The invasive approach is a physical-based technique in which peptides and nutrients
which have poor bioavailability in the brain after intravenous or oral administration are
made bioavailable (Gabathuler, 2010; Stepensky, 2016; Wu, Yoon, Wu, & Bodor, 2002).
These techniques include (1) intracerebroventricular infusion, (2) convection-enhanced
delivery, and (3) intracerebral injection or brain implants. In certain cases the BBB is
mechanically breached by applying osmotic shock, MRI-guided focused ultrasound BBB
disruption, and the use of a bradykinin analog to provide open access to the brain
(Dasgupta et al., 2016; Fortin, Gendron, Boudrias, & Garant, 2007; Kinoshita, McDannold,
Jolesz, & Hynynen, 2006; Wang, Frazier, & Brem, 2002).
15.4.1.1 Intracerebroventricular (ICV) Infusion
In ICV infusion, drug is directly injected into the cerebral lateral ventricles resulting in
diffusion of drugs into the brain through the cerebrospinal fluid (Passini & Wolfe, 2001).
The diffusion of the drug in the brain parenchyma from the CSF through the BCSFB is
very low; it is an effective drug delivery method only in the case that the target site is close
to the ventricles. This might be due to an anatomically different membrane barrier and dif-
ferent permeability profiles of the choroid plexus and brain parenchyma (Spector, Keep,
Snodgrass, Smith, & Johanson, 2015). ICV infusion of opioid peptides shows therapeutic
effects only by binding with the receptors, which are located near the ependymal surface
of the brain (Pardridge, 2007).
15.4.1.2 Convection-Enhanced Delivery
Convection-enhanced delivery (CED) is a stereotactically guided drug delivery method
in which the drug is delivered directly into targeted brain parenchymal cells. With the
help of stereotactic apparatus, the catheter(s) are inserted through cranial burr holes into
the brain and drug is actively pumped into the brain parenchyma through the catheter
(Cunningham et al., 2008; DiMeco et al., 2002). This method shares several advantages
406 15. BEYOND THE BLOODBRAIN BARRIER
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
over other invasive methods such as delivery of high-molecular-weight molecule drugs,
wide distribution of the drug, and it does not cause cerebral edema or measurable
increases in intracranial pressure (Bidros & Vogelbaum, 2009; Bobo et al., 1994; Chen,
Lonser, Morrison, Governale, & Oldfield, 1999). However, the precise placement of the
catheter is a critical step for successful drug delivery by CED (Jahangiri et al., 2017;
Vandergrift & Patel, 2006; Zhou, Singh, & Souweidane, 2017).
15.4.1.3 Intracerebral Injection or Brain Implants
Intracerebral injection is the most direct method for drug delivery to the target site. In
this method, intermittent bolus injections are administered locally in the brain, where the
drugs are diffused with minimal systemic exposure and toxicity (Bergonzi et al., 2016;
Greig, 1987). The use of brain implants is another strategy for drug delivery in which
biodegradable material impregnated with drugs is implanted into the brain. These
drug delivery approaches are commonly employed in the treatment of primary brain
tumors, and neurological disorders such as Parkinson disease (Brem & Gabikian, 2001;
Herzog et al., 2007; Lu et al., 2014; Marks et al., 2008).
15.4.2 Pharmacological Approach
The pharmacological approach comprises of techniques by which lipid-insoluble high-
molecular-weight drug molecules are modified and made bioavailable to the active site in
the brain. These techniques include (1) chemical techniques and (2) colloidal drug carrier
techniques.
15.4.2.1 Chemical Techniques
Chemical approaches are based on the modification of the chemical structure of the
drug molecule that improves their physicochemical property, i.e., lipophilicity thus
improves therapeutic activity (Lu et al., 2014). By reducing the relative number of polar
ends in a drug, this molecule increases the lipophilicity of the other molecule. In the same
way, by adding lipid groups to the polar ends of drug molecules better BBB-permeable
drug molecules are made than the original drug molecule. The prodrug method is another
example of chemical approaches in which drug molecules are modified in such a way
that, on systemic administration, it must convert into the active form by metabolic pro-
cesses (Kreuter, 2001; Lu et al., 2014).
15.4.2.2 Colloidal Drug Carrier Techniques
Colloidal drug approaches are based on colloidal drug carriers in which microscopically
dispersed drug particles are suspended in a suspension form (Kreuter, 2001). The most
common colloidal drug carriers are nanoparticles, micelles, liposomes, emulsions, and
dendrimers (Lu et al., 2014). Many of these colloidal drug carriers are effectively trans-
ported across the BBB by various transcellular mechanisms, such as endocytosis and trans-
cytosis, and have demonstrated significant pharmacokinetics and biodistribution in
different brain-related disorders including brain tumor (Batrakova & Kabanov, 2008;
Wong, Wu, & Bendayan, 2012). Targeted drug delivery to brain tumor by the oral route
40715.4 APPROACHES IN DELIVERING DRUGS TO THE BRAIN
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
has been successfully done using polymeric micelles (Bhujbal, de Vos, & Niclou, 2014;
Kabanov, Batrakova, & Miller, 2003).
15.4.3 Biological or Physiological Approach
Biological or physiological approaches are based on the mechanism of transport of an
essential molecule such as glucose, insulin, growth hormone, low-density lipoprotein, etc.,
in the brain. The specific transporter transports these molecules, such as insulin receptors,
transferrin receptors, etc. into the brain. The physiological approaches are (1) cell-
penetrating peptide-mediated drug delivery, (2) receptor-mediated drug delivery, and (3)
adsorptive-mediated drug delivery.
15.4.3.1 Cell-Penetrating Peptide-Mediated Drug Delivery
In this technique, highly basic amino acids with a positive charge are used to enhance
the brain drug delivery (Dissanayake, Denny, Gamage, & Sarojini, 2017; Temsamani &
Vidal, 2004). The interaction of the cell-penetrating peptide with the cell surface is
receptor-independent, and these can transport the drug molecules into the cytoplasm and
to the nucleus (Guidotti, Brambilla, & Rossi, 2017). They can enter into the cell by two
broad mechanisms: energy-independent direct penetration of the plasma membrane and
energy-dependent endocytosis (Fretz et al., 2007; Guidotti et al., 2017; Palm-Apergi, Lo
¨nn,
& Dowdy, 2012). This approach has been extensively studied preclinically and also tested
clinically in cancer patients (Jia et al., 2011; Warso et al., 2013).
15.4.3.2 Receptor-Mediated Drug Delivery
Receptors such as transferrin receptor, LDL receptor, and insulin receptor are highly
expressed on the endothelial cells of the BBB. In this approach, these receptors are targeted
by specific ligands, modified ligands, and antibodies which will transfer the drugs into the
brain (Pardridge, 2003). The antibody against transferrin receptor and insulin receptor
has demonstrated effective drug delivery of drugs via receptor-mediated transcytosis
(Jones & Shusta, 2007; Pinto, Arce, Yameen, & Vilos, 2017; Zhang & Pardridge, 2005).
Furthermore, the diphtheria toxin receptor, as a targeting vector and low-density lipopro-
tein receptor-related protein, is also used for drug delivery to the brain (Demeule et al.,
2008a; Demeule et al., 2008b; Gaillard, Brink, & de Boer, 2005).
15.4.3.3 Adsorptive-Mediated Drug Delivery
This is another approach to drug delivery in which endocytosis and transcytosis of a
drug molecule are different as compared to receptor-mediated endocytosis and transcyto-
sis. A nonspecific mechanism of endocytosis, adsorptive endocytosis delivers the peptides
and proteins from the luminal plasma membrane of the cell. Various peptide vectors have
been developed and studied in adsorptive-mediated drug delivery for brain tumors
(Byeon et al., 2016; Honary & Zahir, 2013; Jain et al., 2015).
408 15. BEYOND THE BLOODBRAIN BARRIER
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
15.5 NANOTECHNOLOGY AND THE BLOODBRAIN BARRIER
The BBB is the primary systemic barrier that restricts the access of drug molecules
to the brain (Deeken & Lo
¨scher, 2007; Pardridge, 1998). The drug molecules cannot pene-
trate the BBB efficiently due to the presence of a microvascular endothelial cellular net-
work in the brain. This network of cells is responsible for effluxing out a large number of
contents from the brain (Abbott et al., 2010; Demeule et al., 2002b). The tumor cells also
have an acidic environment that neutralizes the basic moiety to inactivate it. Apart from
this, the tumor cells could also impede the action of anticancer drugs by altering enzyme
activity and apoptotic regulation (Brigger, Dubernet, & Couvreur, 2002). Many available
anticancer drugs are largely hydrophilic in nature, and the BBB restricts their entry. The
only prospect for entry of these hydrophilic drugs into the brain is by conjugating or load-
ing them with nanodevices, which could provide efficient distribution of drug in the brain
(Be
´duneau, Saulnier, & Benoit, 2007; Ferrari, 2005; Lockman, Mumper, Khan, & Allen,
2002; Pardridge, 2002; Shi, Kantoff, Wooster, & Farokhzad, 2017).
Nanodevices are of various sizes, ranging from small molecules to large macromole-
cules. The potential applications of nanotechnology-mediated drug delivery include site-
specific drug targeting (Amiji, 2007; Koo, Rubinstein, & Onyuksel, 2005; Park et al., 2010)
and varying the pharmacokinetics of the anticancer drug to improve the therapeutic out-
come with reduced off-target effects (Davis & Shin, 2008; Gabizon, Shmeeda, & Barenholz,
2003). A nanotechnology-mediated drug delivery system is an efficient strategy to over-
come the difficulties of tumor cell-mediated resistance and triggering the release of anti-
cancer drugs into a specific tumor cell. Nanodevices provide protection to the anticancer
drug from the acidic environment of the tumor tissue and thereby increase the penetration
of anticancer drugs into tumor cells (Brannon-Peppas & Blanchette, 2012; Brigger et al.,
2002). Nanodevices can be utilized for delivery of prodrugs to the particular site
along with its activator enzyme (Gupta et al., 2009; Linderoth, Peters, Madsen, &
Andresen, 2009).
Nanotechnology provides greater promise for targeting delivery of anticancer drugs to
brain tumors. The primary reason behind the success is that they can be targeted through
both specific and nonspecific mechanisms. The specific targeting strategy is antigen-
dependent and based on the complexation of the nanomaterial with the specific antigen
and its interaction with a particular tumor cell. The nonspecific mechanism depends upon
the efficient delivery of drug molecules into the brain through vascular permeation
provided by BBB cessation due to a brain tumor. Nanoparticles provide better brain
tissue permeability and retention effect that makes nanodevices the ideal system for tar-
geted delivery of anticancer drugs towards brain tumors (Evans & Alexander, 1972;
Orringer et al., 2009; Peer et al., 2007).
The transportation of nanocarrier-complexed drug through the BBB can occur either via
passive diffusion or endocytosis. Passive diffusion is facilitated by larger drug concentra-
tions in the plasma that lead to a higher osmotic gradient in the BBB and thereby increases
the CNS entry of drug molecules. Endocytosis mainly relies on receptor mediation. The
specific receptor in the BBB interacts with the surface of nanoparticles and thus forms an
efficient mechanism for brain targeting (Gabathuler, 2010; Grabrucker et al., 2013; Kreuter,
2002; Wohlfart, Gelperina, & Kreuter, 2012).
40915.5 NANOTECHNOLOGY AND THE BLOODBRAIN BARRIER
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
15.6 NANODEVICES FOR BRAIN TUMOR TARGETING AND
DELIVERY
15.6.1 Liposomal Formulations
Liposomes are phospholipid bilayer membrane constructs consisting of diverse charac-
teristic groups such as phosphoric residues, hydrophobic chains, and a polar head.
Liposomes have varied physicochemical properties due to different phospholipid compo-
sitions, surficial charges, the size of the liposomal carrier, and the use of preparation meth-
odology (Maheshwari, Tekade, Sharma, & Kumar Tekade, R., 2015b; Sharma, Maheshwari,
Tekade, & Kumar Tekade, 2015). Liposomes are ideal nanodevices for drug delivery appli-
cations. Over the last several decades, liposomal formulations have been studied exten-
sively for the targeted delivery of molecules (Allen & Cullis, 2013; Daraee, Etemadi,
Kouhi, Alimirzalu, & Akbarzadeh, 2016; Lian & Ho, 2001; Torchilin, 2005). The liposomal
formulation provides regulated retentive entrapment and improved uptake of drugs by
targeted cells. The “stealth liposomes,” coated with biocompatible polymers, have been
used widely for targeted drug delivery applications (Allen & Cullis, 2013; Bae & Park,
2011; Gabizon, 2001). Macrophages are unable to recognize the stealth liposomes, and
thereby these types of nanodevices have a longer biological half-life (Cattel, Ceruti, &
Dosio, 2003; Deshpande, Biswas, & Torchilin, 2013; Moghimi & Szebeni, 2003; Silvander,
2002). In the last few years, rapid advancements have been made to utilize liposomal
nanoconstructs in targeted drug delivery for brain tumors.
The therapeutic index of convection-enhanced delivery of liposomes for targeting brain
tumor was investigated. The study showed that convection-enhanced delivery of
gadolinium-colabeled liposomes could be used successfully as visualizing agents of a rat
brain tumor model for in vivo MRI (Saito et al., 2004). Alternatively, immunoliposomes
were also utilized for targeting therapeutic genes for brain delivery in a brain tumor
model (Agarwal et al., 2011). Further, the PEGylated immunoliposomes carrying plasmid
DNA were targeted to mouse transferrin (Tfr) and human insulin receptors. Tfr receptor
targeting improves transport across the tumor vasculature, and insulin receptor targeting
causes transport across plasma and the nuclear membrane of human brain cancer cells
(Zhang, Zhu, & Pardridge, 2002b).
Siegal et al. investigated and compared the biodistribution and clinical efficiency of
doxorubicin (DOX) in free and stealth liposomal encapsulation form. The study was per-
formed for a secondary brain tumor model in Fischer rats. The comparative study con-
firmed the improved drug exposure and therapeutic efficiency with a stealth liposomal
carrier against early small- and large-sized brain tumors (Siegal, Horowitz, & Gabizon,
1995). Gao et al. have developed the dual-targeted DOX-liposomal carrier and anchored it
with folate (Fol) and Tfr. The folate-anchored liposomal system improves BBB penetration,
and Tfr conjugation efficiently targets brain glioma cells (Gao et al., 2013). Gaillard and
coworkers developed glutathione pegylated liposomal doxorubicin for a brain tumor.
Additional glutathione coating enhances the drug delivery of DOX in brain tumor as com-
pared to PEGylated liposomes (Gaillard et al., 2014). The focused ultrasound (FUS) pulse
can also be induced in and around the tumor with the simultaneous intravenous adminis-
tration of a microbubble contrasting agent. The FUS-introduced DOX-cationic liposomes
410 15. BEYOND THE BLOODBRAIN BARRIER
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
were investigated in a C6 glioma model. FUS when combined with microbubble contrast
agent, induced a noninvasive, local, and transient disruption of BBB without affecting
normal tissues. Combining the benefits of BBB opening using FUS and targeting using
cationic liposomes, this strategy can be effectively utilized further for targeting brain
tumor therapy (Lin et al., 2016). The microbubble DOX-liposomes also combined success-
fully with interleukin-4-receptor peptide for targeting in brain tumor therapy (Park et al.,
2016). In another development, DOX liposomes can be adequately anchored with pH-
responsive peptide H
7
K(R
2
)
2
as targeting ligand. This peptide specifically responds to the
acidic environment present in tumor tissue and can be successfully utilized as a targeting
strategy in brain tumor therapy (Zhao et al., 2016).
Multifunctional paclitaxel (PTX) liposomes were developed in several studies to
improve the therapeutic and functional targeting. The PTX liposomal system is anchored
with cyclic RGD peptide and cell penetration R8 peptide (RGD-R8-PTX-lipo). The study
showed that conjugation with RGD and R8 peptide possess multifunctions such as effec-
tive brain accumulation, targeting, and better tumor penetration in glioma-bearing mice
(Liu et al., 2014b). In another strategy for developing a multifunctional PTX liposomal sys-
tem, artemether was used additionally as a regulator of apoptosis and inhibitor of vasculo-
genic mimicry (VM) channels. The VM channels can cause a recurrence of brain glioma.
The liposomal system was developed by conjugating mannose-vitamin E derivative
(MAN) and dequalinium-lipid derivative (DQA) and proven to be an effective strategy for
treatment of brain tumor in in vitro and in vivo models (Li et al., 2014). Further, celecoxib
was also used along with epirubicin liposomes for the treatment of brain tumor and
destruction of VM channels. The results of the study indicate effective targeting using an
epirubicin and celecoxib combination to inhibit tumor VM channels and treat brain tumor
in glioma-bearing nude mice (Ju et al., 2016). A recent experiment also showed that vin-
cristine- and tetrandrine-combined liposomes along with Tfr tethering could increase cel-
lular uptake, inhibit multidrug resistance, and block cancer cell invasion and VM channels
to treat brain tumor (Song et al., 2017).
In an investigation by Yang et al., Angiopep-2 and neuropilin-1 receptor-specific pep-
tides have been anchored on vascular endothelial growth factor (VEGF) siRNA and doce-
taxel (DTX) liposomal systems. The dual peptides improve the binding ability and
internalization into glioma cells by a specific receptor-mediated endocytosis mechanism of
cellular uptake. The dual peptide anchored liposomes were shown to possess a better
targeting strategy for the treatment of brain tumor compared to nonmodified and single
peptide-modified liposomes (Yang, Li, Wang, Dong, & Qi, 2014). The comparative study
of six different peptide-based ligands (Angiopep-2, T7, Peptide-22, Cyclic RGDfK peptide,
D-SP5 cys peptide, and Pep-1) was performed on targeting efficiency of DOX liposomes.
An in vitro cellular uptake study proves that combination of Cyclic RGDfK and Peptide-22
synergistically improves the internalization more than any of the single ligand modifica-
tions. In vivo imaging also confirms higher brain tumor distribution by a Cyclic RGDfK
and Peptide-22 combination than single ligand-modified liposomes (Chen et al., 2017). In a
recent study, DTX-loaded nanoliposomes were developed, and cellular uptake was investi-
gated in C6 glioma cells (Shaw et al., 2017).
41115.6 NANODEVICES FOR BRAIN TUMOR TARGETING AND DELIVERY
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
15.6.2 Polymeric Nanocarriers
The polymeric nanocarrier-mediated drug delivery platform is a very versatile and effi-
cient strategy for modifying the properties of nanoparticles to meet specific requirements
(van Vlerken, Vyas, & Amiji, 2007). The versatile characteristics of the polymer matrices
offer significant advantages over other systems such as easy preparation, surface functio-
nalization, improved payload capacity, and protection (Chacko, Ventura, Zhuang, &
Thayumanavan, 2012; Ryu et al., 2010; Simone, Dziubla, & Muzykantov, 2008). There are
several classes of polymeric materials that could be effectively utilized for drug delivery in
brain tumor therapy, such as synthetic polymers poly(D,L-lactide-co-glycolide) (PLGA)
(Acharya & Sahoo, 2011; Chang et al., 2012), poly(L-lactic acid) (PLL) (del Burgo,
Herna
´ndez, Orive, & Pedraz, 2014; Liu, Xue, Ke, Xie, & Ma, 2014a), poly(phenylene ethy-
nylene) (PPE) (Kulkarni, Surnar, & Jayakannan, 2016), poly(epsilon-caprolactone) (PCL)
(Forrest et al., 2008; Nicolas, Mura, Brambilla, Mackiewicz, & Couvreur, 2013; Zhang et al.,
2012), polyalkylcyanoacrylates (PACA) (Gelperina et al., 2010; Yordanov, 2012), and natu-
ral polymers such as gelatin (Matsumoto et al., 2006), chitosan (Agrawal et al., 2017;
Garcia-Fuentes & Alonso, 2012; Kim, Choi, Kim, & Tae, 2013), hyaluronic acid (Choi,
Saravanakumar, Park, & Park, 2012; Choi et al., 2011), albumin (Comes Franchini et al.,
2010), and heparin (Yu & Zhang, 2009).
The most significant importance of using polymeric nanocarriers is that they sharply
increase the circulation time and decrease the RES accumulation (Klibanov, Maruyama,
Torchilin, & Huang, 1990; Torchilin, 1998). PEGylated liposomal systems have already been
discussed for targeted and effective delivery of DOX (Gabizon & Martin, 1997; Krauze et al.,
2007). Different polymeric nanocarriers could be prepared to apply varied type strategies.
The intravenous administration of DOX coated with Polysorbate-80 reduces tumor mass in
rats (Kreuter, 2001). The study reveals that PEGylated poly(methoxyPEG cyanoacrylate-
co-n-hexadecylcyanoacrylate) (PHDCA) with amphiphilic copolymer can penetrate the
brain to a larger extent than any other nanodevices (Calvo et al., 2001). The hybrid type
of nanoparticles combined with lipid and polymeric material also demonstrated effective
targeted delivery of chemotherapeutics (Dehaini et al., 2016; Zhang et al., 2017a).
The “polymeric wafers” are a new advanced strategy that has now been introduced to
deliver chemotherapeutics locally and bypass the BBB. These polymeric wafers are
implantable devices inserted at the tumor site to discharge and deliver the chemotherapeutic
agents. The polymer utilized in clinical application is polyanhydride poly[1,3-bis(carboxy-
phenoxy) propane- co-sebacic-acid] (PCPP:SA) along with the anticancer drug carmustine
(Brem et al., 1991; Brem et al., 2007; Mangraviti, Gullotti, Tyler, & Brem, 2016).
The major drawback with polymeric nanodevices is that they can be taken up by RES
and distributed in the liver, spleen, and bone marrow, which leads to very low t
1/2
of the
particles in the bloodstream (Le Ray, Vert, Gautier, & Benoit, 1994; Verrecchia et al., 1995).
To overcome this issue, several modifications can be incorporated, such as conjugation of
polymeric nanocarriers with antibodies (Duncan, 2006; Masood, 2016; Reddy et al., 2006),
peptides (Arnold, Czupiel, & Shoichet, 2017; Jiang et al., 2017; Shen et al., 2011; Wang
et al., 2015; Zhang et al., 2016), proteins (Wang et al., 2016), nucleotides (Guo et al., 2011;
Mead et al., 2016), or receptor-specific ligands such as Tfr (Jain et al., 2015), and folic acid
(Wang et al., 2016).
412 15. BEYOND THE BLOODBRAIN BARRIER
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
15.6.3 Magnetic Nanoparticles
Magnetic nanoparticles can be successfully employed as drug delivery vehicles for
effective targeting of therapeutic agents. In this strategy, therapeutic agents can be
attached to the surface or encapsulated inside a complex of polymer and magnetic nano-
particles (Mody et al., 2014b). This therapeutic drug delivery strategy is based on the
application of a magnetic field to localize the specific target in the body. The underlying
mechanism behind this strategy depends upon generated heat from the moving magnetic
nanomaterials using a strong magnetic field. This therapeutic strategy is noninvasive and
safer to target the magnetic nanoparticles to the brain tumor tissues (Silva et al., 2011).
Chen and coworkers have developed doxorubicin (DOX) magnetic nanoparticles, in
which DOX are chemically bonded to ferric oxide nanoparticles and embedded in PEG
functionalized with porous silica (Chen, Zhang, Chen, Zhang, & Zhang, 2010). Magnetic
nanoparticles can also be employed along with focused ultrasound (FUS) to improve drug
targeting synergistically. This synergistic combination increases the local magnetic nano-
particle concentration in the targeted tissue. Liu and coworkers successfully employed this
strategy in the delivery of epirubicin with iron oxide magnetic nanoparticles (Liu et al.,
2010). In another experiment, PEI-modified magnetic nanoparticles were delivered suc-
cessfully into a brain tumor. The study showed a 30-fold increase in targeting by intracaro-
tid administration along with magnetic targeting in comparison to intravenous
administration (Chertok, David, & Yang, 2010). Recently, FUS has also been utilized with
microbubbles to enhance drug targeting in brain tumor. The combination of FUS with
microbubbles increases brain permeability and disrupts the BBB in the condition of a brain
tumor. To utilize the same effect, Fan et al. developed a theranostic complex of superpara-
magnetic iron oxide nanoparticles loaded with microbubbles. The magnetically labeled
system enhanced targeted drug (DOX) deposition in brain glioma cells (Fan et al., 2016).
In a similar experiment, dynamic contrast enhancement (DCE)-based magnetic resonance
imaging was characterized by FUS-induced brain permeability and utilized for targeted
delivery of DOX in 9L gliosarcoma cells of rats (Park, Aryal, Vykhodtseva, Zhang, &
McDannold, 2017).
Some studies have also been proven to be successful in the low-frequency magnetic
field (Golovin, Golovin, Klyachko, Majouga, & Kabanov, 2017a; Golovin, Klyachko,
Majouga, Sokolsky, & Kabanov, 2017b). In such a study, Kim et al. developed magnetic
vortex microdisks for targeting brain tumor. The results showed that mechanical stimulus
generated by microdisk oscillations could selectively target brain tumor cells and kill them
(Kim et al., 2010). In a recent experiment, biogenic nanosized cell-derived microvesicles
have been targeted effectively by magnetic and folate functionalization. In this study,
microvesicles were conjugated with streptavidin-modified iron oxide nanoparticles and
effectively transformed as targeted drug delivery nanovectors for delivery of anticancer
agents in the treatment of brain tumor (Zhang et al., 2017b). The Tfr receptor-binding
peptide T7 mediated drug targeting was also utilized along with the application of an
applied magnetic field. This dual-targeted magnetic PLGA nanoparticle was employed for
delivery of paclitaxel and curcumin. Dual targeting results in a 10-fold increase in cellular
uptake and over fivefold increase in brain targeting in an orthotopic glioma model
(Cui et al., 2016).
41315.6 NANODEVICES FOR BRAIN TUMOR TARGETING AND DELIVERY
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
15.6.4 Dendrimeric Nanocarriers
Dendrimeric structures have unique characteristic properties such as monodispersity
(Mody, Tekade, Mehra, Chopdey, & Jain, 2014a), modifiable surface functionality (Patri,
Kukowska-Latallo, & Baker, 2005), membrane transport efficiency, high drug payload
(Satija, Gupta, & Jain, 2007), biocompatibility, and well-defined molecular structure and
composition (Jain & Asthana, 2007). These unique properties of dendrimers make them
highly suitable nanocarriers for drug targeting. Usually dendrimer architecture consists of
three topological domains: (1) a central core comprising of an atom or a molecule with a
minimum of two identical functional groups; (2) branches, with several interior repeating
units structured geometrically in a sequence of radically aligned layers known as “genera-
tions” ; and (3) terminal groups which determine the surface properties of the dendritic
structure (Kesharwani, Jain, & Jain, 2014a; Kesharwani, Tekade, & Jain, 2015a; Martinho
et al., 2014)(Fig. 15.2). Dendrimeric architectures are prepared using divergent and con-
vergent approaches. In both approaches, dendrimers are synthesized by step-by-step
growth and branching generations. Dendrimers are depicted and classified by their gener-
ation numbers. The central core group represented G0 (generation 0), although following
additions of branching units were termed as higher generations of dendrimers (G1, G2,
G3, etc.). This exponential increase with the addition of each group results in sterically
crowded dendrimers, which leads to geometrical changes in the dendrimeric structures.
With the growth in generations, dendrimers take a globular structure due to increasing
steric hindrance (Kesharwani, Tekade, & Jain, 2014b; Kesharwani, Tekade, & Jain, 2015b).
Several strategies were developed to transport the therapeutic agents across the brain in
a site-specific manner. Amongst all the approaches, dendrimers have emerged as promis-
ing vectors with great potential for delivering drugs into the CNS. Various types of den-
drimers, such as polyamidoamine (PAMAM), poly-L-lysine (PLL), and poly(propylimine)
(PPI) have been explored successfully in the treatment and diagnosis of brain tumors.
Sarin and coworkers have developed a G5 PAMAM dendrimer conjugated with DOX and
in chelation with Gd-DTPA. The results demonstrated significant improvement in target-
ing as compared to free DOX in a rodent malignant glioma model (Sarin et al., 2008, 2009).
A PEGylation strategy is also employed to successfully deliver anticancer agents to the tar-
geted site. He et al. have developed PEGylated PAMAM dendrimers as dual targeting
nanocarriers in brain tumor. They prepared PEGylated G4 PAMAM dendrimers with Tfr
and WGA loaded with DOX. The results demonstrated reduced cytotoxicity and increased
accumulation of DOX in the tumor site due to targeting effects of Tfr and WGA (He et al.,
2011). Li et al. explored the targeting potential of G4 PAMAM dendrimers anchored with
Tfr and encapsulated with tamoxifen in brain gliomas. The results indicated faster drug
release in weakly acidic conditions (Li et al., 2012). Recently, the PEGylated PAMAM den-
drimers were also anchored with various peptides such as angiopep-2 (Xu et al., 2016) and
homing peptides (Pep-1) (Jiang et al., 2016) as an effective targeting strategy in the treat-
ment of brain tumors.
Similarly, PEGylated PLL-based dendrimers loaded with DOX have been effectively uti-
lized for brain tumor targeting. In an experiment by Kaminskas et al., nanocarriers were
linked to an acid-labile 4-(hydrazinosulfonyl) benzoic acid (HSBA) linker that causes the
release of DOX in acidic conditions. The in vivo results in rats bearing Walker 256 tumors
414 15. BEYOND THE BLOODBRAIN BARRIER
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
demonstrated higher uptake in tumor tissue as compared to control tissue of muscle and
heart (pH 5) (Kaminskas et al., 2011). Ligand-anchored poly(propyleneimine) dendrimers
were also evaluated for their brain-targeting potential by Jain and coworkers. They
anchored various ligand molecules, such as sialic acid (S), glucosamine (G), and concanav-
alin A (C) to the surface of poly(propyleneimine) (PPI) dendrimers encapsulated with pac-
litaxel. The results of in vivo pharmacokinetics and biodistribution studies in rats showed
a significantly higher accumulation of paclitaxel in the brain as compared to free pacli-
taxel. The order of targeting potential of various ligands under investigation was found as
sialic acid .glucosamine .concanavalin A (Patel, Gajbhiye, Kesharwani, & Jain, 2016).
Researchers have recently developed a spatially controlled multistage nanocarrier by
FIGURE 15.2 Structured diagram of dendrimers presenting its major domains: (1) core, (2) generations, (3) ter-
minal groups, and (4) internal voids.
41515.6 NANODEVICES FOR BRAIN TUMOR TARGETING AND DELIVERY
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
encapsulating small polyamidoamine (PAMAM) dendrimers (B5 nm) within large gelatin
NPs (B200 nm). This strategy showed the higher stability of nanodevices in the systemic
circulation and enhanced permeation through the tumor vasculature. The PAMAM den-
drimer was released in response to MMP-2 enzymes in the tumor microenvironment and
resulted in higher intracellular uptake and deeper tissue penetration in a tumor model.
The results have also demonstrated effective and targeting delivery of methotrexate with
this system (Fan et al., 2017).
15.6.5 Carbon Nanotubes
In recent years, CNTs have emerged as a potential delivery system for biologically
active molecules to enhance the therapeutic benefits. Currently, functionalized CNT-
mediated selective and specific delivery of biomolecules has gained huge attention as a
potential, promising nano-architecture due to its unique physicochemical properties in the
treatment of various deadly diseases including cancer (Bianco, Kostarelos, & Prato, 2005;
Bottini et al., 2006; Sajid et al., 2016; Zhang, Bai, & Yan, 2010).
Ren et al. employed angiopep-2 surface-modified oxidized-MWCNTs for a dual-
targeting drug delivery system for the treatment of brain glioma. These authors reported
that the ultrahigh surface area of MWCNTs resulted in a loading potential of higher order
to load doxorubicin (DOX). Angiopep-2 is an activatable cell-penetrating peptide with the
ability to selectively bind to the low-density lipoprotein receptor-related protein receptor,
generally overexpressed on the BBB and glioma cells. The outcomes suggested that
MWCNTs had lower cardiotoxicity than DOX alone (Ren et al., 2012). Another example of
using angiopep-2, presented by Kafa et al., has also displayed significant outcomes as a
targeting ligand for brain delivery using MWCNTs. In particular, these authors demon-
strated better results with functionalized MWCNTs in comparison to the nontargeted for-
mulation in each aspect including in vitro and in vivo performance employing primary
porcine brain endothelial cells and primary rat astrocytes, following intravenous adminis-
tration. Enhanced whole-brain uptake, increased uptake in glioma brain cells, and
enhanced brain accumulation were found with angiopep-2-functionalized MWCNTs (Kafa
et al., 2016). It was also reported that functionalization does not significantly affect the
dimension of MWCNTs, indicating the importance of functionalized MWCNT diameter
towards their accumulation at desired sites. Continuing with cancer, Mehra et al., demon-
strated the pharmacokinetic and pharmacodynamic investigations of the cancer-targeting
potential of the DOX-loaded D-α-tocopheryl polyethylene glycol 1000 succinate-decorated
surface-functionalized MWCNT nano-formulation and compared it with pristine
MWCNTs and free doxorubicin solution. The authors concluded that the investigated
MWCNTs’ nano-architect had greater cancer-targeting potential on tumor-bearing Balb/c
mice (Mehra, Verma, Mishra, & Jain, 2014). In a different investigation, Lu et al. employed
MWCNTs and magnetic NPs as a dual approach for treating brain cancer. In this case, the
functionalization was done with poly(acrylic acid) via free radical polymerization linked
with folic acid and DOX was used as the chemotherapeutic agent. Outcomes using
advanced microscopic techniques suggest that DOX-loaded folic acid-conjugated
MWCNTs efficiently accumulated within U-87 cells following the desired release of DOX
416 15. BEYOND THE BLOODBRAIN BARRIER
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
intracellularly and then effectively transported into the nucleus with the MWCNTs left in
the cytoplasm (Lu, Wei, Ma, Yang, & Chen, 2012; McDevitt et al., 2007). Apart from
MWCNTs, McDevitt et al. used SWCNTs modified with rituximab and lintuzumab to
demonstrate the capability to selectively target cancerous cells. Based on pharmacody-
namic studies, the authors claimed higher biodistribution in a murine xenograft model of
lymphoma, suggesting further SWCNTs as a potential delivery platform for selective
delivery (McDevitt et al., 2007).
15.6.6 Gold Nanoparticles
Biologically active enteritis can also be decorated onto the exterior of AuNPs (Wolburg
et al., 2012). The huge exterior area/volume ratio of AuNPs allows their surface to be
encrusted with a variety of molecules like therapeutic agents, site-specific ligand, and anti-
fouling polymers (Catherine, & Olivier, 2017). An interesting pilot study was conducted
using a cell culture technique employing an animal model of glioblastoma multiforme in
which PEG-based gold NPs supported radiation therapy. The dual action gold NPs
enhanced survival of mice with orthotopic GBM tumors. The authors also demonstrated
enhanced extravasation and greater accumulation of gold NPs, indicating that this dual
approach significantly disrupts the BBB and could be leveraged to enhance cancer cell tar-
geting. There findings indicate that pegylated gold NPs can be significantly combined
with radiation therapy for the treatment of cancerous cells (Joh et al., 2013). In another
study, Hainfeld et al. demonstrated the use of gold NPs for high-resolution computed
tomography tumor imaging with radiation therapy using 30 Gy 100 kVp X-rays. The gold
NPs were taken up by tumor cells 19 3time more than the normal brain cells. Also, mice
injected with gold NPs were capable of inducing tumor-free survival for more than a year
in comparison to mice receiving radiation only (Hainfeld, Smilowitz, O’Connor,
Dilmanian, & Slatkin, 2013). Ruan et al., considering the problem of limited accumulation
of drugs and NPs within brain cells, proposed a legumain base approach to enhance the
retention of chemotherapeutics in brain tumors. They devised a nano-platform, gold NPs-
A&C, which was comprised of Ala-Ala-Asn-Cys-Lys-modified gold NPs, and 2-cyano-6-
aminobenzothiazole-modified gold NPs. This approach increased the accumulation of the
gold NPs in glioma cells both in vitro and in vivo due to the blocking of NP exocytosis
and minimizing NP backflow to the bloodstream. The authors also employed DOX as a
chemotherapeutic agent to further improve the efficiency against glioma. The median sur-
vival time for the DOX-linked AuNPs-A&C increased to more than 285% in comparison to
the saline group. They concluded that this approach has potential to elevate NP tumor
accumulation and therefore may have a better clinical outcome (Ruan et al., 2016).
Gromnicova et al. studied the ability of nanosized (4 nm) glucose-modified gold NPs to
penetrate human brain endothelium selectively and subsequently to enter astrocytes. The
transfer rate of these NPs across primary human brain endothelium was not less than
three times faster than across nonbrain endothelia. The authors further reported that
movement of these NPs occurred across the apical and basal plasma membranes through
the cytosol with relatively little vesicular or paracellular migration; antibiotics that inter-
fere with vesicular transport did not block the migration. The transfer rate also relied on
41715.6 NANODEVICES FOR BRAIN TUMOR TARGETING AND DELIVERY
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
surface modification of the NPs and incubation temperature. They claimed that the
glucose-modified NPs traverse the endothelium, move through the extracellular matrix,
and localize in astrocytes, and it was finally concluded that these NPs have the ability to
cross the bloodbrain barrier and deliver the therapeutic goods (Gromnicova et al., 2013).
15.6.7 Viral Nanoparticles
Viral vectors are the most recognizable system for delivering exogenous genetic materi-
als to specific cells, following traversing them to the cell nucleus and thereby activating
the genomic (During, 1997; Handbook; Vile, Tuszynski, & Castleden, 1996). Liu et al. dis-
played RVG29 (rabies virus glycoprotein) as a promising ligand for proficient brain-
targeting gene delivery. For the purpose, RVG29 was treated on PAMAM dendrimer using
bifunctional PEG, then complexed with DNA, yielding multiconstructed NPs. The results
exhibited that a high potential of BBB disruption ability in an in vitro BBB model was
achieved using RVG29 ligand containing formulation, whereas in vivo results demon-
strated higher accumulation and cellular internalization in the brain. This dual-functional
strategy provides a safer and noninvasive mode for gene delivery through the BBB (Liu
et al., 2009).
In another example of gene therapy, Zhang et al. employed an antisense technique with
an artificial virus that uses a receptor-specific monoclonal antibody gene delivery system
to treat a brain tumor. Mice implanted with intracranial U-87 human glial brain tumors
were treated with a nonviral expression plasmid encoding antisense mRNA against the
human epidermal growth factor receptor gene. The plasmid DNA is packaged within the
interior of PEG-functionalized immunoliposomes and administered to the cancerous cells
with MAbs that target the mouse transferrin receptor and the human insulin receptor.
The mouse TRFR MAb enables transport across the tumor vasculature, which is of mouse
brain origin, and the INSR MAb causes transport across the plasma membrane and the
nuclear membrane of the human brain cancer cell. As an outcome, the lifespan of the mice
treated weekly with an intravenous administration of the EGFR antisense gene therapy
packaged within the artificial virus was increased 100% compared with mice treated either
with a luciferase gene or with saline (Zhang et al., 2002b).
Lee et al. tried to achieve better cellular internalization and enhance the bioavailability
employing rabies virus-based silica-modified gold nanorods based on a photothermal
principle to treat brain cancer. The prepared functionalized nanorod was injected and
could induce a hyperthermal effect in response to NIR laser (808 nm) irradiation, based on
localized surface plasmon resonance, to effectively suppress brain tumors of mice.
Together, these results support the assertion that the rabies virus mimetic gold nanorods
are a potential prototype delivery platform for treating brain tumors (Lee et al., 2017a).
15.6.8 Nucleic Acids-Based Nanotechnology
Oligonucleotides annealed with targeted mRNA sequences can be utilized successfully
as a delivery vector to the targeted site. Galectin-1 is a naturally occurring galactose-
binding protein, which is upregulated in the case of GBM. Galectin-1 is involved in cancer
418 15. BEYOND THE BLOODBRAIN BARRIER
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
progression and is a potent immune suppressor in the tumor microvasculature. Recently,
Woensel et al. developed siRNA-tailored chitosan NPs to target Galectin-1 for the treat-
ment of GBM via intranasal delivery. This study was the first of its type and involved con-
centrated chitosan NP suspensions for the delivery of siRNA into brain tumor a few hours
after intranasal administration. These NPs could complex siRNA targeting Galectin-1 to a
high percentage and avoid RNAse degradation. The results indicated that siRNA nano-
formulation significantly depressed the expression of Galectin-1 in both murine and
human GBM cell lines. Sequence-specific RNA-interference displayed a more than 50%
Galectin-1 reduction in tumor-bearing mice. This investigation indicates that the intranasal
pathway is an underexplored transport route for delivering siRNA-based therapies target-
ing Galectin-1 in the treatment of GBM (Van Woensel et al., 2016).
In another application of the siRNA delivery system, Wei et al. investigated the biomed-
ical significance of intravenously administered T7 peptide-based coreshell NPs employ-
ing an siRNA approach against brain tumors. As a methodology, the authors employed
layer-by-layer deposition of protamine, chondroitin sulfate, siRNA, and cationic lipid vesi-
cles followed by T7 peptide modification to construct a site-specific siRNA-based delivery
system. The pharmacokinetic study revealed a higher cellular internalization of fluores-
cence intensity of siRNA in brain microvascular endothelial cells and U87 glioma cells
when tested with T7-siRNA-based liposomal construct and compared with PEG-siRNA
based system. In vivo, the experiment exhibited significant silencing of EGFR protein
expression in the U87 glioma cells after treatment with the T7-siRNA-based liposomal con-
struct. It was also noticed that this construct was advantageous in deep penetration of the
tumor spheroid compared with the non-T7-modified formulation. The authors concluded
that transferrin receptor-mediated coreshell NPs are promising siRNA delivery systems
for selective brain-targeting applications (Wei et al., 2016).
Different to siRNA, Lee et al., demonstrated downregulation of oncogenic miRNA-21
for the treatment of glioblastoma by rescuing tumor suppressors, PTEN and PDCD4. The
authors constructed three-way junction (3WJ)-based RNA NPs artificially collected from
pRNA of bacteriophage phi29 DNA packaging motor and conjugated with folic acid. The
formed nano-construct was shown to be promising in silencing miR-21 expression in glio-
blastoma cells both in vitro and in vivo, with favorable biodistribution. Systemically
injected FA-3WJ-LNA-miR21 RNP efficiently rescued PTEN and PDCD4, resulting in glio-
blastoma cell apoptosis and tumor growth regression. Overall, the survival rate was also
significantly improved by FA-3WJ-LNA-miR21 RNP. These results are indicative of the
clinical benefit of FA-3WJ RNP-based gene therapy for the successful targeted therapy of
developing and even recurring glioblastoma (Lee et al., 2017b).
15.7 NANOTECHNOLOGY FOR BRAIN TUMOR IMAGING
Magnetic resonance imaging is an excellent example of the utilization of nanotechnol-
ogy for the imaging of brain cancer cells since numerous NPs have been reported as MRI
contrast agents (Babes, Denizot, Tanguy, Le Jeune, & Jallet, 1999; Hyeon, Piao, & Park,
2016; Kim, Lee, Kwak, & Kim, 2005; Na, Song, & Hyeon, 2009; Wei et al., 2017a). Mostly,
NP-based imaging agents have been fabricated with a core of iron oxide crystals with/
41915.7 NANOTECHNOLOGY FOR BRAIN TUMOR IMAGING
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
without a shell of organic material, like PEG. The main advantage of using NPs as the
contrast agent is the extent of information they provide about the stage/level of the tumor
(Wei, Liao, Mahmood, Xu, & Zhou, 2017b). However, gadolinium-based contrast agents
and NP-based contrast agents cause an elevation of tumors by passing through areas of
disrupted BBB, where they change the Mr signal intensity (Boxerman, Schmainda, &
Weisskoff, 2006; Sorensen, Tievsky, Ostergaard, Weisskoff, & Rosen, 1997). As an advan-
tage over gadolinium-based imaging, NP-based contrast agents, for example, ultra-small
superparamagnetic iron oxide NPs are taken up by the phagocytes found at tumor mar-
gins, and thus areas of tumor not seen with gadolinium-enhanced MRI can be detected
using iron oxide-based NPs. Also, iron oxide-based NPs spend a longer time inside the
tumor and more accurately delineate tumor margins. NPs are also capable of being used
for selective molecular tumor targeting (Enochs, Harsh, Hochberg, & Weissleder, 1999; Lee
et al., 2008; Thorek, Chen, Czupryna, & Tsourkas, 2006).
Recently, Cho et al. developed a novel imaging agent using peptide-decorated AuNPs
toward brain glioma stem cell marker CD133 with stimulus responsive ability. The peptide
CBP4 is relatively small and was screened by a phage display method, and showed attach-
ment with the target CD133 in comparison to an antibody. The authors employed AuNPs
as a quencher, and the targeting peptide was linked to AuNPs with greater efficacy. Using
a quenching effect, the peptide-decorated AuNPs displayed signal onoff characters
depending upon the presence of the target. Also, the formed system showed excellent
biocompatibility when localized in the cytosol. The authors concluded that the peptide-
decorated AgNPs could be used as an imaging agent for correct diagnosis of GBM and
have potential to be explored as a drug carrier (Cho et al., 2017).
Timbie et al. designed an Mr image-guided delivery system of cisplatin-incorporated
brain-penetrating NPs to invasive glioma with selective ultrasound to analyze whether
brain-penetrating NPs modified with PEG would be able to be delivered through both the
bloodtumor and BBB to producing therapeutic effectiveness. This is an example of using
an Mr-guided delivery approach for the treatment of brain tumors (Timbie et al., 2017).
Wang et al. used a folic acid-based targeting strategy to deliver fluorescent magnetic
bovine serum albumin-modified NPs to enhance intracellular dual modal imaging in
human glioma U251 cells. The outcomes revealed that the prepared system was not toxic
to cells with better biocompatibility and higher cellular uptake. The formulation was shown
to be effectively internalized for MRI and intracellular visualization after fluorescein iso-
thiocyanate (dye) labeling in the targeted U251 cells. These investigators finally concluded
that the developed formulation effectively enables dual modal imaging without significant
toxicity and a site-specific carrier for the treatment of brain cancer in the future. In an inter-
esting and novel approach, Han et al. employed autocatalytic brain tumor-targeting, in
which half of the portion of nanoparticles was introduced through BBB leakage, receptor-
mediated transcytosis, carrier-mediated transcytosis, and adsorptive-mediated transcytosis
and the remaining portion was introduced when these NPs reached the tumor site and
released BBB modulators which enhanced the permeability of the BBB (Wang et al., 2016).
Thus, the efficiency of NP accumulation in tumors autocatalytically increases with time
and subsequent administrations. The overall conclusion can be drawn from different
investigations that NPs have a promising and meaningful role as imaging or contrast
agents for the detection of progression of various diseases.
420 15. BEYOND THE BLOODBRAIN BARRIER
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
15.8 CHALLENGES AND SAFETY CONSIDERATIONS OF
NANOTECHNOLOGIES USED FOR BRAIN-TARGETED DELIVERY
Nanotechnology approaches comprise of engineered nanomaterials and nanodevices
which interact more specifically with biological systems at molecular levels and are used
in diagnostics as well as therapeutics (Bawa, Audette, & Rubinstein, 2016). The applica-
tions of nanotechnology in basic and clinical neuroscience are developing day by day.
Besides the various advantages of nanotechnology in the understanding of neurobiology,
neuropathophysiology, and intervention at the molecular level, there are numerous chal-
lenges associated with nanotechnology (Howard, 2016; Soni, Garg, Patel, & Yadav, 2016a).
In the field of cancer, early detection of precancerous and neoplastic lesions remains
a major challenge for nanotechnology (Fruscella, Ponzetto, Crema, & Carloni, 2016).
Conventional cancer imaging technologies are not able to detect the precancerous stage
based on lesion anatomy. This is because of low spatial resolution—to increase the spatial
resolution, imaging techniques require a signal-amplifying material conjugate, known as a
contrast agent (Namen & Luke, 2017). Nanoparticles have been used as multifunctional
and molecularly targeted contrast agents for imaging smaller and early-stage cancers
(Fruscella et al., 2016; Harisinghani et al., 2003; Sullivan & Ferrari, 2004).
In the case of multidimensional CNS disorders, another important challenge for nano-
technologies is the requirement for multitargeting molecules which can interact with mul-
tiple receptors and play specific cellular and physiological functions (Kemp, Shim, Heo, &
Kwon, 2016; Wang, 2016). Another major challenge for nanotechnology is the anatomically
restrictive BBB of the CNS. Nanotechnologies designed for brain disorders including brain
cancer should efficiently deliver at the target site with minimal disruption of the BBB and
minimal systemic and local side effects (Giles, 2003; Oberdo
¨rster, Oberdo
¨rster, &
Oberdo
¨rster, 2005; Wang, 2016). On the other hand, the inherent complexity of the CNS,
cellular heterogeneity, and multidimensional cellular connections in the brain are also a
challenge for nanotechnology (Shah, 2016). Nanotechnology products are designed to
interact physically with target cells at a molecular level. However, due to the complexity
of the CNS at present, there are only a few applications of this type (Shah, 2016). As nano-
technology advances and broadens its applications in medicine and diagnostics, the safety
concerns about nanotechnologies is also an emerging area of research (Braydich-Stolle,
Hussain, Schlager, & Hofmann, 2005; Dobrovolskaia & McNeil, 2007; Maynard & Aitken,
2016; Voura, Jaiswal, Mattoussi, & Simon, 2004). The morphological characteristics of
nanoparticles can modulate pharmacokinetics and biodistribution of drug molecules and
can cause systemic toxicity (Moghimi, Hunter, & Murray, 2001; Moghimi, Hunter, &
Murray, 2005). However, there is a lack of systemic studies on the safety issue, particularly
for brain-related disorders. Previous studies reported that the nanoparticles could
modulate the activity of P-glycoprotein efflux pumps (Batrakova, Li, Alakhov, Miller, &
Kabanov, 2003; Huang et al., 2011; Miller, Bauer, & Hartz, 2008).
Earlier studies also reported the carcinogenic potential of nanoparticles in various
in vitro and in vivo studies. Nanoparticles are noncarcinogenic, however photo-dependent
carcinogenesis has been observed in some fullerenes and fullerene derivatives (Magaye,
Zhao, Bowman, & Ding, 2012; Rancan et al., 2002; Remya, Syama, Sabareeswaran, &
42115.8 CHALLENGES AND SAFETY CONSIDERATIONS OF NANOTECHNOLOGIES USED
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
Mohanan, 2016; Roller, 2009). The environmental hazard of nanotechnologies is another
important safety aspect because of their size and possible nonbiodegradable nature; they
can rapidly distribute and accumulate in the environment (Brandeburova
´et al., 2017;
Oukarroum, Zaidi, Samadani, & Dewez, 2017).
15.9 FUTURE PERSPECTIVES AND CONCLUSION
In the last 20 years, tremendous efforts have been observed to fabricate novel delivery
strategies against brain cancer, ranging from invasive to noninvasive strategies. However,
as far as the prognosis is concerned, little or few improvements have been noted, which
does not enable the enhancement of the median survival of patients. However, some inno-
vative reports are encouraging and may be path breaking. For instance, temozolomide,
alone or in combination with other alkylating agents, seems to be a better platform in the
treatment of brain tumors, owing to its good BBB permeability properties and low toxicity.
BBB permeability is significant, which enables greater accumulation in the CNS for
successful antitumor therapy. In recognition of the important roles of the BBB in brain
tumor chemotherapy and a better understanding of transport mechanisms and their mod-
ulators, it is possible to overcome the limitations presented by traditional chemotherapy.
Research in this field is now focused on the development of noninvasive, more specific,
and targeted strategies that exploit the knowledge of the pathogenesis of brain cancers.
Tumor growth critically depends on the formation of new blood vessels. Thus, the inhibi-
tion of angiogenesis pathways constitutes an attractive strategy for targeted therapy,
which has been investigated by single or combined agents. Further improvements will
result from a better understanding of tumor biology and the pathways involved therein.
Another promising and noninvasive tool for the delivery of therapeutic drugs to brain
tumors employs drug-loaded nanocarrier systems that take advantage of the disrupted
BBB at tumor sites with disorganized vasculature and leakier capillaries to achieve selec-
tive tumor delivery. Modification of the nanocarrier surface properties improves uptake
by endothelial cells. Also, to selectively deliver chemo drugs in brain tumors, magnetic
NPs are of great interest, with the possibility of monitoring and quantifying the process by
Mr imaging. Furthermore, SLNs have been found to be advantageous in delivering chemo
drugs due to their good BBB permeability, low intrinsic cytotoxicity, and the biodegrad-
ability of lipids used in their preparation.
In another dimension to the delivery strategies, the invasive strategies, local delivery of
chemotherapeutic agents to brain tumors by CED improves drug distribution compared to
other strategies only driven by diffusion. However, this technique is also confounded by
notable side effects. Overall, despite several issues that need to be addressed, targeted
therapy and particulate systems are promising strategies that are worth investigating
further for efficient CNS delivery of chemo drugs.
Acknowledgments
The authors would like to acknowledge the Science and Engineering Research Board, Department of Science
and Technology, Government of India, for a grant allocated to Dr Tekade for research work on gene delivery
422 15. BEYOND THE BLOODBRAIN BARRIER
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
(ECR/2016/001964) and N-PDF funding (Dr. Maheshwari; PDF/2016/003329). RT would also like to thank
NIPER-Ahmedabad for providing research support for research into cancer and arthritis. The authors acknow-
ledge support by the Fundamental Research Grant (FRGS) scheme of the Ministry of Higher Education, Malaysia,
to support research into gene delivery. The authors are also thankful to Department of Health Research (DHR),
Ministry of Health and Family Welfare, Govt. of India, New Delhi for sanctioning the Young Scientist Grant to
Mr. Piyoosh Sharma (25011/215-HRD/2016-HR).
References
Abbott, N. J., Patabendige, A. A., Dolman, D. E., Yusof, S. R., & Begley, D. J. (2010). Structure and function of the
bloodbrain barrier. Neurobiology of Disease,37(1), 1325.
Abbott, N. J., Ro
¨nnba
¨ck, L., & Hansson, E. (2006). Astrocyteendothelial interactions at the bloodbrain barrier.
Nature Reviews Neuroscience,7(1), 4153.
Acharya, S., & Sahoo, S. K. (2011). PLGA nanoparticles containing various anticancer agents and tumour delivery
by EPR effect. Advanced Drug Delivery Reviews,63(3), 170183.
Adams, S., Braidy, N., Bessesde, A., Brew, B. J., Grant, R., Teo, C., & Guillemin, G. J. (2012). The kynurenine path-
way in brain tumor pathogenesis. Cancer Research,72(22), 56495657.
Agarwal, S., Sane, R., Oberoi, R., Ohlfest, J. R., & Elmquist, W. F. (2011). Delivery of molecularly targeted therapy
to malignant glioma, a disease of the whole brain. Expert Reviews in Molecular Medicine,13, e17.
Agrawal, P., Singh, R. P., Kumari, L., Sharma, G., Koch, B., Rajesh, C. V., ... Muthu, M. S. (2017). TPGS-chitosan
cross-linked targeted nanoparticles for effective brain cancer therapy. Materials Science and Engineering: C,74,
167176.
Aizer, A., & Alexander, B. (2017). Brain tumors—epidemiology.Pathology and Epidemiology of Cancer (pp. 279290).
Springer.
Allen, T. M., & Cullis, P. R. (2013). Liposomal drug delivery systems: From concept to clinical applications.
Advanced Drug Delivery Reviews,65(1), 3648.
Amiji, M. M. (2007). Nanotechnology—improving targeted delivery. Drug Delivery,17,5356.
Amirian, E. S., Zhou, R., Wrensch, M. R., Olson, S. H., Scheurer, M. E., Il’Yasova, D., ... Lau, C. C. (2016).
Approaching a scientific consensus on the association between allergies and glioma risk: A report from the gli-
oma international case-control study. Cancer Epidemiology and Prevention Biomarkers,25(2), 282290.
Arnold, A. E., Czupiel, P., & Shoichet, M. (2017). Engineered polymeric nanoparticles to guide the cellular inter-
nalization and trafficking of small interfering ribonucleic acids. Journal of Controlled Release,259,315.
Babes, L., Denizot, B. T., Tanguy, G., Le Jeune, J. J., & Jallet, P. (1999). Synthesis of iron oxide nanoparticles used
as MRI contrast agents: A parametric study. Journal of Colloid and Interface Science,212(2), 474482.
Bae, Y. H., & Park, K. (2011). Targeted drug delivery to tumors: Myths, reality and possibility. Journal of Controlled
Release,153(3), 198.
Ballabh, P., Braun, A., & Nedergaard, M. (2004). The bloodbrain barrier: An overview: Structure, regulation, and
clinical implications. Neurobiology of Disease,16(1), 113.
Banks, W. A., Niehoff, M. L., Martin, D., & Farrell, C. L. (2002). Leptin transport across the bloodbrain barrier of
the Koletsky rat is not mediated by a product of the leptin receptor gene. Brain Research,950(1), 130136.
Bart, J., Groen, H., Hendrikse, N., van der Graaf, W., Vaalburg, W., & de Vries, E. (2000). The blood-brain barrier
and oncology: New insights into function and modulation. Cancer Treatment Reviews,26(6), 449462.
Batrakova, E. V., & Kabanov, A. V. (2008). Pluronic block copolymers: Evolution of drug delivery concept from
inert nanocarriers to biological response modifiers. Journal of Controlled Release,130(2), 98106.
Batrakova, E. V., Li, S., Alakhov, V. Y., Miller, D. W., & Kabanov, A. V. (2003). Optimal structure requirements
for pluronic block copolymers in modifying P-glycoprotein drug efflux transporter activity in bovine brain
microvessel endothelial cells. Journal of Pharmacology and Experimental Therapeutics,304(2), 845854.
Bauer, A. H., Erly, W., Moser, F. G., Maya, M., & Nael, K. (2015). Differentiation of solitary brain metastasis from
glioblastoma multiforme: A predictive multiparametric approach using combined MR diffusion and perfusion.
Neuroradiology,57(7), 697703.
Bawa, R., Audette, G. F., & Rubinstein, I. (2016). Handbook of clinical nanomedicine: Nanoparticles, imaging, therapy,
and clinical applications. Pan Stanford.
423REFERENCES
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
Be
´duneau, A., Saulnier, P., & Benoit, J.-P. (2007). Active targeting of brain tumors using nanocarriers. Biomaterials,
28(33), 49474967.
Begley, D. J. (2004). ABC transporters and the blood-brain barrier. Current Pharmaceutical Design,10(12),
12951312.
Bergonzi, M. C., Guccione, C., Grossi, C., Piazzini, V., Torracchi, A., Luccarini, I., ... Bilia, A. R. (2016). Albumin
nanoparticles for brain delivery: A comparison of chemical versus thermal methods and in vivo behavior.
ChemMedChem,11(16), 18401849.
Bernacki, J., Dobrowolska, A., Nierwin
˜ska, K., & Malecki, A. (2008). Physiology and pharmacological role of the
blood-brain barrier. Pharmacological Reports,60(5), 600622.
Bhujbal, S. V., de Vos, P., & Niclou, S. P. (2014). Drug and cell encapsulation: Alternative delivery options for the
treatment of malignant brain tumors. Advanced Drug Delivery Reviews,67, 142153.
Bianco, A., Kostarelos, K., & Prato, M. (2005). Applications of carbon nanotubes in drug delivery. Current Opinion
in Chemical Biology,9(6), 674679.
Bidros, D. S., & Vogelbaum, M. A. (2009). Novel drug delivery strategies in neuro-oncology. Neurotherapeutics,6
(3), 539546.
Bobo, R. H., Laske, D. W., Akbasak, A., Morrison, P. F., Dedrick, R. L., & Oldfield, E. H. (1994). Convection-
enhanced delivery of macromolecules in the brain. Proceedings of the National Academy of Sciences,91(6),
20762080.
Bodor, N., & Buchwald, P. (2003). Brain-targeted drug delivery. American Journal of Drug Delivery,1(1), 1326.
Bottini, M., Bruckner, S., Nika, K., Bottini, N., Bellucci, S., Magrini, A., ...Mustelin, T. (2006). Multi-walled carbon
nanotubes induce T lymphocyte apoptosis. Toxicology Letters,160(2), 121126.
Boxerman, J., Schmainda, K., & Weisskoff, R. (2006). Relative cerebral blood volume maps corrected for contrast
agent extravasation significantly correlate with glioma tumor grade, whereas uncorrected maps do not.
American Journal of Neuroradiology,27(4), 859867.
Brandeburova
´, P., Bı
´roˇ
sova
´, L., Vojs, M., Kromka, A., Ga
´l, M., Tichy
´, J., ... Mackuˇ
lak, T. (2017). The influence of
selected nanomaterials on microorganisms. Monatshefte fu
¨r Chemie-Chemical Monthly,148(3), 525530.
Brannon-Peppas, L., & Blanchette, J. O. (2012). Nanoparticle and targeted systems for cancer therapy. Advanced
Drug Delivery Reviews,64, 206212.
Brasnjevic, I., Steinbusch, H. W., Schmitz, C., Martinez-Martinez, P., & Initiative, E. N. R. (2009). Delivery of pep-
tide and protein drugs over the bloodbrain barrier. Progress in Neurobiology,87(4), 212251.
Braydich-Stolle, L., Hussain, S., Schlager, J. J., & Hofmann, M.-C. (2005). In vitro cytotoxicity of nanoparticles in
mammalian germline stem cells. Toxicological Sciences,88(2), 412419.
Brem, H., & Gabikian, P. (2001). Biodegradable polymer implants to treat brain tumors. Journal of Controlled
Release,74(1), 6367.
Brem, H., Mahaley, M. S., Jr, Vick, N. A., Black, K. L., Schold, S. C., Jr, Burger, P. C., ... Cozzens, J. W. (1991).
Interstitial chemotherapy with drug polymer implants for the treatment of recurrent gliomas. Journal of
Neurosurgery,74(3), 441446.
Brem, S., Tyler, B., Li, K., Pradilla, G., Legnani, F., Caplan, J., & Brem, H. (2007). Local delivery of temozolomide
by biodegradable polymers is superior to oral administration in a rodent glioma model. Cancer Chemotherapy
and Pharmacology,60(5), 643650.
Brigger, I., Dubernet, C., & Couvreur, P. (2002). Nanoparticles in cancer therapy and diagnosis. Advanced Drug
Delivery Reviews,54(5), 631651.
Burkhart, A., Skjørringe, T., Johnsen, K. B., Siupka, P., Thomsen, L. B., Nielsen, M. S., ... Moos, T. (2016).
Expression of iron-related proteins at the neurovascular unit supports reduction and reoxidation of iron for
transport through the blood-brain barrier. Molecular Neurobiology,53(10), 72377253.
Byeon, H. J., Lee, S., Min, S. Y., Lee, E. S., Shin, B. S., Choi, H.-G., & Youn, Y. S. (2016). Doxorubicin-loaded nano-
particles consisted of cationic-and mannose-modified-albumins for dual-targeting in brain tumors. Journal of
Controlled Release,225, 301313.
Calvo, P., Gouritin, B., Chacun, H., Desmae
¨le, D., D’Angelo, J., Noel, J.-P., ... Couvreur, P. (2001). Long-
circulating PEGylated polycyanoacrylate nanoparticles as new drug carrier for brain delivery. Pharmaceutical
Research,18(8), 11571166.
Catherine, L., & Olivier, P. (2017). Gold nanoparticles for physics, chemistry and biology. World Scientific.
Cattel, L., Ceruti, M., & Dosio, F. (2003). From conventional to stealth liposomes: A new frontier in cancer chemo-
therapy. Tumori,89(3), 237249.
424 15. BEYOND THE BLOODBRAIN BARRIER
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
Chacko, R. T., Ventura, J., Zhuang, J., & Thayumanavan, S. (2012). Polymer nanogels: A versatile nanoscopic drug
delivery platform. Advanced drug Delivery Reviews,64(9), 836851.
Chang, J., Paillard, A., Passirani, C., Morille, M., Benoit, J.-P., Betbeder, D., & Garcion, E. (2012). Transferrin
adsorption onto PLGA nanoparticles governs their interaction with biological systems from blood circulation
to brain cancer cells. Pharmaceutical Research,29(6), 14951505.
Chen, C., Duan, Z., Yuan, Y., Li, R., Pang, L., Liang, J., ... Wang, J. (2017). Peptide-22 and cyclic RGD functionalized
liposomes for glioma targeting drug delivery overcoming BBB and BBTB.ACS Applied Materials & Interfaces.
Chen, F.-H., Zhang, L.-M., Chen, Q.-T., Zhang, Y., & Zhang, Z.-J. (2010). Synthesis of a novel magnetic drug deliv-
ery system composed of doxorubicin-conjugated Fe 3 O 4 nanoparticle cores and a PEG-functionalized porous
silica shell. Chemical Communications,46(45), 86338635.
Chen, M. Y., Lonser, R. R., Morrison, P. F., Governale, L. S., & Oldfield, E. H. (1999). Variables affecting
convection-enhanced delivery to the striatum: A systematic examination of rate of infusion, cannula size, infu-
sate concentration, and tissue—cannula sealing time. Journal of Neurosurgery,90(2), 315320.
Chen, Y., Dalwadi, G., & Benson, H. (2004). Drug delivery across the blood-brain barrier. Current Drug Delivery,
1(4), 361376.
Chen, Z., Shi, T., Zhang, L., Zhu, P., Deng, M., Huang, C., ... Li, J. (2016). Mammalian drug efflux transporters of
the ATP binding cassette (ABC) family in multidrug resistance: A review of the past decade. Cancer Letters,
370(1), 153164.
Chertok, B., David, A. E., & Yang, V. C. (2010). Polyethyleneimine-modified iron oxide nanoparticles for brain
tumor drug delivery using magnetic targeting and intra-carotid administration. Biomaterials,31(24), 63176324.
Cheslow, L., & Alvarez, J. I. (2016). Glial-endothelial crosstalk regulates bloodbrain barrier function. Current
Opinion in Pharmacology,26,3946.
Cho, J.-H., Kim, A.-R., Kim, S.-H., Lee, S.-J., Chung, H., & Yoon, M.-Y. (2017). Development of a novel imaging
agent using peptide-coated gold nanoparticles toward brain glioma stem cell marker CD133. Acta Biomaterialia,
47, 182192.
Choi, K. Y., Saravanakumar, G., Park, J. H., & Park, K. (2012). Hyaluronic acid-based nanocarriers for intracellular
targeting: Interfacial interactions with proteins in cancer. Colloids and Surfaces B: Biointerfaces,99,8294.
Choi, K. Y., Yoon, H. Y., Kim, J.-H., Bae, S. M., Park, R.-W., Kang, Y. M., ... Jeong, S. Y. (2011). Smart nanocarrier
based on PEGylated hyaluronic acid for cancer therapy. ACS Nano,5(11), 85918599.
Choonara, Y. E., Kumar, P., Modi, G., & Pillay, V. (2016). Improving drug delivery technology for treating neuro-
degenerative diseases. Expert Opinion on Drug Delivery,13(7), 10291043.
Clark, D. E. (2003). In silico prediction of bloodbrain barrier permeation. Drug Discovery Today,8(20), 927933.
Comes Franchini, M., Baldi, G., Bonacchi, D., Gentili, D., Giudetti, G., Lascialfari, A., ... Micotti, E. (2010). Bovine
serum albumin-based magnetic nanocarrier for MRI diagnosis and hyperthermic therapy: A potential thera-
nostic approach against cancer. Small,6(3), 366370.
Crawford, L., Rosch, J., & Putnam, D. (2016). Concepts, technologies, and practices for drug delivery past the
bloodbrain barrier to the central nervous system. Journal of Controlled Release,240, 251266.
Cui, Y., Zhang, M., Zeng, F., Jin, H., Xu, Q., & Huang, Y. (2016). Dual-targeting magnetic PLGA nanoparticles for
codelivery of paclitaxel and curcumin for brain tumor therapy. ACS Applied Materials & Interfaces,8(47),
3215932169.
Cunningham, J., Pivirotto, P., Bringas, J., Suzuki, B., Vijay, S., Sanftner, L., ... Bankiewicz, K. S. (2008).
Biodistribution of adeno-associated virus type-2 in nonhuman primates after convection-enhanced delivery to
brain. Molecular Therapy,16(7), 12671275.
da Silva, J. (2016). DNA damage induced by occupational and environmental exposure to miscellaneous chemi-
cals. Mutation Research/Reviews in Mutation Research,770, 170182.
Dallas, S., Miller, D. S., & Bendayan, R. (2006). Multidrug resistance-associated proteins: Expression and function
in the central nervous system. Pharmacological Reviews,58(2), 140161.
Daneman, R., & Prat, A. (2015). The bloodbrain barrier. Cold Spring Harbor Perspectives in Biology,7(1), a020412.
Daraee, H., Etemadi, A., Kouhi, M., Alimirzalu, S., & Akbarzadeh, A. (2016). Application of liposomes in medi-
cine and drug delivery. Artificial Cells, Nanomedicine, and Biotechnology,44(1), 381391.
Dasgupta, A., Liu, M., Ojha, T., Storm, G., Kiessling, F., & Lammers, T. (2016). Ultrasound-mediated drug delivery
to the brain: Principles, progress and prospects. Drug Discovery Today: Technologies,20,4148.
Davis, M. E., & Shin, D. M. (2008). Nanoparticle therapeutics: An emerging treatment modality for cancer. Nature
Reviews Drug Discovery,7(9), 771782.
425REFERENCES
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
de Lange, E. C. (2004). Potential role of ABC transporters as a detoxification system at the bloodCSF barrier.
Advanced Drug Delivery Reviews,56(12), 17931809.
Deeken, J. F., & Lo
¨scher, W. (2007). The blood-brain barrier and cancer: Transporters, treatment, and Trojan
horses. Clinical Cancer Research,13(6), 16631674.
Dehaini, D., Fang, R. H., Luk, B. T., Pang, Z., Hu, C.-M. J., Kroll, A. V., ... Zhang, L. (2016). Ultra-small
lipidpolymer hybrid nanoparticles for tumor-penetrating drug delivery. Nanoscale,8(30), 1441114419.
del Burgo, L. S., Herna
´ndez, R. M., Orive, G., & Pedraz, J. L. (2014). Nanotherapeutic approaches for brain cancer
management. Nanomedicine: Nanotechnology, Biology and Medicine,10(5), e905e919.
Demeule, M., Currie, J. C., Bertrand, Y., Che, C., Nguyen, T., Regina, A., ... Beliveau, R. (2008a). Involvement of
the low-density lipoprotein receptor-related protein in the transcytosis of the brain delivery vector Angiopep-
2. Journal of Neurochemistry,106(4), 15341544.
Demeule, M., Poirier, J., Jodoin, J., Bertrand, Y., Desrosiers, R. R., Dagenais, C., ... Kennard, M. (2002a). High
transcytosis of melanotransferrin (P97) across the bloodbrain barrier. Journal of Neurochemistry,83(4),
924933.
Demeule, M., Regina, A., Che, C., Poirier, J., Nguyen, T., Gabathuler, R., ... Beliveau, R. (2008b). Identification
and design of peptides as a new drug delivery system for the brain. Journal of Pharmacology and Experimental
Therapeutics,324(3), 10641072.
Demeule, M., Re
´gina, A., Jodoin, J., Laplante, A., Dagenais, C., Berthelet, F., ... Be
´liveau, R. (2002b). Drug trans-
port to the brain: Key roles for the efflux pump P-glycoprotein in the bloodbrain barrier. Vascular
Pharmacology,38(6), 339348.
Deshpande, P. P., Biswas, S., & Torchilin, V. P. (2013). Current trends in the use of liposomes for tumor targeting.
Nanomedicine,8(9), 15091528.
DiMeco, F., Li, K. W., Tyler, B. M., Wolf, A. S., Brem, H., & Olivi, A. (2002). Local delivery of mitoxantrone for the
treatment of malignant brain tumors in rats. Journal of Neurosurgery,97(5), 11731178.
Dissanayake, S., Denny, W. A., Gamage, S., & Sarojini, V. (2017). Recent developments in anticancer drug delivery
using cell penetrating and tumor targeting peptides. Journal of Controlled Release,250,6276.
Dobrovolskaia, M. A., & McNeil, S. E. (2007). Immunological properties of engineered nanomaterials. Nature
Nanotechnology,2(8), 469478.
Drin, G., Cottin, S., Blanc, E., Rees, A. R., & Temsamani, J. (2003). Studies on the internalization mechanism of
cationic cell-penetrating peptides. Journal of Biological Chemistry,278(33), 3119231201.
Duncan, R. (2006). Polymer conjugates as anticancer nanomedicines. Nature Reviews Cancer,6(9), 688701.
During, M. J. (1997). Adeno-associated virus as a gene delivery system. Advanced Drug Delivery Reviews,27(1),
8394.
Dwibhashyam, V., & Nagappa, A. (2008). Strategies for enhanced drug delivery to the central nervous system.
Indian journal of Pharmaceutical Sciences,70(2), 145.
Ellor, S. V., Pagano-Young, T. A., & Avgeropoulos, N. G. (2014). Glioblastoma: Background, standard treatment para-
digms, and supportive care considerations.SAGE Publications Sage. CA: Los Angeles, CA.
Enochs, W. S., Harsh, G., Hochberg, F., & Weissleder, R. (1999). Improved delineation of human brain tumors on
MR images using a long-circulating, superparamagnetic iron oxide agent. Journal of Magnetic Resonance
Imaging,9(2), 228232.
Evans, R., & Alexander, P. (1972). Mechanism of immunologically specific killing of tumour cells by macrophages.
Nature,236(5343), 168170.
Fan, C.-H., Cheng, Y.-H., Ting, C.-Y., Ho, Y.-J., Hsu, P.-H., Liu, H.-L., & Yeh, C.-K. (2016). Ultrasound/magnetic
targeting with SPIO-DOX-microbubble complex for image-guided drug delivery in brain tumors. Theranostics,
6(10), 1542.
Fan, Y., Yuan, S., Huo, M., Chaudhuri, A. S., Zhao, M., Wu, Z., & Qi, X. (2017). Spatial controlled multistage nano-
carriers through hybridization of dendrimers and gelatin nanoparticles for deep penetration and therapy into
tumor tissue. Nanomedicine: Nanotechnology, Biology and Medicine.
Ferlay, J., Soerjomataram, I., Ervik, M., Dikshit, R., Eser, S., Mathers, C., ... Bray, F. (2013). Cancer incidence and
mortality worldwide: IARC Cancer Base No. 11 GLOBOCAN 2012v1. 0. Lyon: International Agency for Research
on Cancer.
Fernandes, C., Soni, U., & Patravale, V. (2010). Nano-interventions for neurodegenerative disorders.
Pharmacological Research,62(2), 166178.
426 15. BEYOND THE BLOODBRAIN BARRIER
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
Ferrari, M. (2005). Cancer nanotechnology: Opportunities and challenges. Nature Reviews Cancer,5(3), 161171.
Fletcher, J. I., Williams, R. T., Henderson, M. J., Norris, M. D., & Haber, M. (2016). ABC transporters as mediators
of drug resistance and contributors to cancer cell biology. Drug Resistance Updates,26,19.
Forrest, M. L., Ya
´n
˜ez, J. A., Remsberg, C. M., Ohgami, Y., Kwon, G. S., & Davies, N. M. (2008). Paclitaxel prodrugs
with sustained release and high solubility in poly (ethylene glycol)-b-poly (ε-caprolactone) micelle nanocar-
riers: Pharmacokinetic disposition, tolerability, and cytotoxicity. Pharmaceutical Research,25(1), 194206.
Fortin, D., Gendron, C., Boudrias, M., & Garant, M. P. (2007). Enhanced chemotherapy delivery by intraarterial
infusion and blood-brain barrier disruption in the treatment of cerebral metastasis. Cancer,109(4), 751760.
Foti, R. S., Tyndale, R. F., Garcia, K. L., Sweet, D. H., Nagar, S., Sharan, S., & Rock, D. A. (2015). “Target-site”
drug metabolism and transport. Drug Metabolism and Disposition,43(8), 11561168.
Fretz, M. M., Penning, N. A., Al-Taei, S., Futaki, S., Takeuchi, T., Nakase, I., ... Jones, A. T. (2007). Temperature-,
concentration-and cholesterol-dependent translocation of L-and D-octa-arginine across the plasma and nuclear
membrane of CD34 1leukaemia cells. Biochemical Journal,403(2), 335342.
Fruscella, M., Ponzetto, A., Crema, A., & Carloni, G. (2016). The extraordinary progress in very early cancer diag-
nosis and personalized therapy: The role of oncomarkers and nanotechnology. Journal of Nanotechnology,2016.
Gabathuler, R. (2010). Approaches to transport therapeutic drugs across the bloodbrain barrier to treat brain
diseases. Neurobiology of Disease,37(1), 4857.
Gabizon, A., & Martin, F. (1997). Polyethylene glycol-coated (pegylated) liposomal doxorubicin. Drugs,54(4), 1521.
Gabizon, A., Shmeeda, H., & Barenholz, Y. (2003). Pharmacokinetics of pegylated liposomal doxorubicin. Clinical
Pharmacokinetics,42(5), 419436.
Gabizon, A. A. (2001). Stealth liposomes and tumor targeting: One step further in the quest for the magic bullet. AACR.
Gaillard, P. J., Appeldoorn, C. C., Dorland, R., van Kregten, J., Manca, F., Vugts, D. J., ... Maussang, D. (2014).
Pharmacokinetics, brain delivery, and efficacy in brain tumor-bearing mice of glutathione pegylated liposomal
doxorubicin (2B3-101). PloS one,9(1), e82331.
Gaillard, P. J., Brink, A., & de Boer, A. G. (2005). Diphtheria toxin receptor-targeted brain drug delivery.International
Congress Series (pp. 185198). Elsevier.
Gao, J.-Q., Lv, Q., Li, L.-M., Tang, X.-J., Li, F.-Z., Hu, Y.-L., & Han, M. (2013). Glioma targeting and bloodbrain
barrier penetration by dual-targeting doxorubincin liposomes. Biomaterials,34(22), 56285639.
Garcia-Fuentes, M., & Alonso, M. J. (2012). Chitosan-based drug nanocarriers: Where do we stand? Journal of
Controlled Release,161(2), 496504.
Gelperina, S., Maksimenko, O., Khalansky, A., Vanchugova, L., Shipulo, E., Abbasova, K., ... Kreuter, J. (2010).
Drug delivery to the brain using surfactant-coated poly (lactide-co-glycolide) nanoparticles: Influence of the
formulation parameters. European Journal of Pharmaceutics and Biopharmaceutics,74(2), 157163.
Giles, J. (2003). Nanotechnology: What is there to fear from something so small? Nature,426(6968), 750, 750.
Gleeson, M. P. (2008). Generation of a set of simple, interpretable ADMET rules of thumb. Journal of Medicinal
Chemistry,51(4), 817834.
Golovin, Y., Golovin, D., Klyachko, N., Majouga, A., & Kabanov, A. (2017a). Modeling drug release from functio-
nalized magnetic nanoparticles actuated by non-heating low frequency magnetic field. Journal of Nanoparticle
Research,19(2), 64.
Golovin, Y. I., Klyachko, N. L., Majouga, A. G., Sokolsky, M., & Kabanov, A. V. (2017b). Theranostic multimodal
potential of magnetic nanoparticles actuated by non-heating low frequency magnetic field in the new-
generation nanomedicine. Journal of Nanoparticle Research,19(2), 63.
Goulatis, L. I., & Shusta, E. V. (2017). Protein engineering approaches for regulating bloodbrain barrier transcy-
tosis. Current Opinion in Structural Biology,45, 109115.
Grabrucker, A. M., Chhabra, R., Belletti, D., Forni, F., Vandelli, M. A., Ruozi, B., & Tosi, G. (2013). Nanoparticles as
BloodBrain Barrier permeable CNS targeted drug delivery systems.The Blood Brain Barrier (BBB) (pp. 7189).
Springer.
Greig, N. H. (1987). Optimizing drug delivery to brain tumors. Cancer Treatment Reviews,14(1), 128.
Gromnicova, R., Davies, H. A., Sreekanthreddy, P., Romero, I. A., Lund, T., Roitt, I. M., ... Male, D. K. (2013).
Glucose-coated gold nanoparticles transfer across human brain endothelium and enter astrocytes in vitro.
PLoS One,8(12), e81043.
Group, I. S. (2010). Brain tumour risk in relation to mobile telephone use: Results of the INTERPHONE interna-
tional casecontrol study. International Journal of Epidemiology, dyq079.
427REFERENCES
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
Guidotti, G., Brambilla, L., & Rossi, D. (2017). Cell-penetrating peptides: From basic research to clinics. Trends in
Pharmacological Sciences,38(4), 406424.
Guo, J., Gao, X., Su, L., Xia, H., Gu, G., Pang, Z., ... Chen, H. (2011). Aptamer-functionalized PEGPLGA nano-
particles for enhanced anti-glioma drug delivery. Biomaterials,32(31), 80108020.
Gupta, P. B., Onder, T. T., Jiang, G., Tao, K., Kuperwasser, C., Weinberg, R. A., & Lander, E. S. (2009).
Identification of selective inhibitors of cancer stem cells by high-throughput screening. Cell,138(4), 645659.
Hainfeld, J. F., Smilowitz, H. M., O’Connor, M. J., Dilmanian, F. A., & Slatkin, D. N. (2013). Gold nanoparticle
imaging and radiotherapy of brain tumors in mice. Nanomedicine,8(10), 16011609.
Handbook, M.B. Retroviral Vectors From Laboratory Tools to Molecular Medicines.
Hansen, A. J. (1985). Effect of anoxia on ion distribution in the brain. Physiological Reviews,65(1), 101148.
Harisinghani, M. G., Barentsz, J., Hahn, P. F., Deserno, W. M., Tabatabaei, S., van de Kaa, C. H., ... Weissleder, R.
(2003). Noninvasive detection of clinically occult lymph-node metastases in prostate cancer. New England
Journal of Medicine,2003(348), 24912499.
He, H., Li, Y., Jia, X.-R., Du, J., Ying, X., Lu, W.-L., ... Wei, Y. (2011). PEGylated Poly (amidoamine) dendrimer-
based dual-targeting carrier for treating brain tumors. Biomaterials,32(2), 478487.
Herve
´, F., Ghinea, N., & Scherrmann, J.-M. (2008). CNS delivery via adsorptive transcytosis. The AAPS Journal,
10(3), 455472.
Herzog, C. D., Dass, B., Holden, J. E., Stansell, J., Gasmi, M., Tuszynski, M. H., ... Kordower, J. H. (2007). Striatal
delivery of CERE-120, an AAV2 vector encoding human neurturin, enhances activity of the dopaminergic
nigrostriatal system in aged monkeys. Movement Disorders,22(8), 11241132.
Hladky, S. B., & Barrand, M. A. (2016). Fluid and ion transfer across the bloodbrain and bloodcerebrospinal
fluid barriers; a comparative account of mechanisms and roles. Fluids and Barriers of the CNS,13(1), 19.
Honary, S., & Zahir, F. (2013). Effect of zeta potential on the properties of nano-drug delivery systems-a review
(Part 1). Tropical Journal of Pharmaceutical Research,12(2), 255264.
Howard, K. A. (2016). Nanomedicine: Working towards defining the field.Nanomedicine (pp. 112). Springer.
Huang, I.-P., Sun, S.-P., Cheng, S.-H., Lee, C.-H., Wu, C.-Y., Yang, C.-S., ... Lai, Y.-K. (2011). Enhanced chemother-
apy of cancer using pH-sensitive mesoporous silica nanoparticles to antagonize P-glycoproteinmediated
drug resistance. Molecular Cancer Therapeutics,10(5), 761769.
Huttunen, K. M., & Rautio, J. (2011). Prodrugs-an efficient way to breach delivery and targeting barriers. Current
Topics in Medicinal Chemistry,11(18), 22652287.
Hyeon, T., Piao, Y., Park, Y.I. 2016. Method of preparing iron oxide nanoparticles coated with hydrophilic mate-
rial, and magnetic resonance imaging contrast agent using the same, Google Patents.
Ishihara, H., Kubota, H., Lindberg, R. L., Leppert, D., Gloor, S. M., Errede, M., ... Frei, K. (2008). Endothelial cell
barrier impairment induced by glioblastomas and transforming growth factor β2 involves matrix metallopro-
teinases and tight junction proteins. Journal of Neuropathology & Experimental Neurology,67(5), 435448.
Jahangiri, A., Chin, A. T., Flanigan, P. M., Chen, R., Bankiewicz, K., & Aghi, M. K. (2017). Convection-enhanced
delivery in glioblastoma: A review of preclinical and clinical studies. Journal of Neurosurgery,126(1), 191200.
Jain, A., Jain, A., Garg, N. K., Tyagi, R. K., Singh, B., Katare, O. P., ... Soni, V. (2015). Surface engineered
polymeric nanocarriers mediate the delivery of transferrinmethotrexate conjugates for an improved under-
standing of brain cancer. Acta Biomaterialia,24, 140151.
Jain, N. K., & Asthana, A. (2007). Dendritic systems in drug delivery applications. Expert Opinion on Drug
Delivery,4(5), 495512.
Jeong, S., Hahm, K., Shin, J., Leem, J., Lee, C., & Han, S. (2006). Changes in magnesium concentration in the serum
and cerebrospinal fluid of neuropathic rats. Acta Anaesthesiologica Scandinavica,50(2), 211216.
Jia, L., Gorman, G. S., Coward, L. U., Noker, P. E., McCormick, D., Horn, T. L., ... Ganesh, B. (2011). Preclinical
pharmacokinetics, metabolism, and toxicity of azurin-p28 (NSC745104) a peptide inhibitor of p53 ubiquitina-
tion. Cancer Chemotherapy and Pharmacology,68(2), 513524.
Jiang, Y., Lv, L., Shi, H., Hua, Y., Lv, W., Wang, X., ... Xu, Q. (2016). PEGylated polyamidoamine dendrimer
conjugated with tumor homing peptide as a potential targeted delivery system for glioma. Colloids and Surfaces
B: Biointerfaces,147, 242249.
Jiang, Y., Wang, X., Lv, W., Liu, X., Zhang, H., Zhang, M., ... Xu, Q. (2017). Enhanced anti-glioma efficacy of
ultrahigh loading capacity paclitaxel prodrug conjugate self-assembled targeted nanoparticles. ACS Applied
Materials & Interfaces.
428 15. BEYOND THE BLOODBRAIN BARRIER
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
Joh, D. Y., Sun, L., Stangl, M., Al Zaki, A., Murty, S., Santoiemma, P. P., ... Bhang, D. (2013). Selective targeting of
brain tumors with gold nanoparticle-induced radiosensitization. PloS one,8(4), e62425.
Jones, A. R., & Shusta, E. V. (2007). Bloodbrain barrier transport of therapeutics via receptor-mediation.
Pharmaceutical Research,24(9), 17591771.
Ju, R.-J., Zeng, F., Liu, L., Mu, L.-M., Xie, H.-J., Zhao, Y., ... Lu, W.-L. (2016). Destruction of vasculogenic mimicry
channels by targeting epirubicin plus celecoxib liposomes in treatment of brain glioma. International Journal of
Nanomedicine,11, 1131.
Kabanov, A. V., Batrakova, E. V., & Miller, D. W. (2003). Pluronicsblock copolymers as modulators of drug
efflux transporter activity in the bloodbrain barrier. Advanced Drug Delivery Reviews,55(1), 151164.
Kafa, H., Wang, J. T.-W., Rubio, N., Klippstein, R., Costa, P. M., Hassan, H. A., ... Abbott, N. J. (2016).
Translocation of LRP1 targeted carbon nanotubes of different diameters across the bloodbrain barrier
in vitro and in vivo. Journal of Controlled Release,225, 217229.
Kaminskas, L. M., Kelly, B. D., McLeod, V. M., Sberna, G., Owen, D. J., Boyd, B. J., & Porter, C. J. (2011).
Characterisation and tumour targeting of PEGylated polylysine dendrimers bearing doxorubicin via a pH
labile linker. Journal of Controlled Release,152(2), 241248.
Kemp, J. A., Shim, M. S., Heo, C. Y., & Kwon, Y. J. (2016). “Combo” nanomedicine: Co-delivery of multi-modal
therapeutics for efficient, targeted, and safe cancer therapy. Advanced Drug Delivery Reviews,98,318.
Kemper, E. M., Boogerd, W., Thuis, I., Beijnen, J. H., & van Tellingen, O. (2004). Modulation of the bloodbrain
barrier in oncology: Therapeutic opportunities for the treatment of brain tumours? Cancer Treatment Reviews,
30(5), 415423.
Kesharwani, P., Jain, K., & Jain, N. K. (2014a). Dendrimer as nanocarrier for drug delivery. Progress in Polymer
Science,39(2), 268307.
Kesharwani, P., Tekade, R. K., & Jain, N. K. (2014b). Generation dependent cancer targeting potential of poly (pro-
pyleneimine) dendrimer. Biomaterials,35(21), 55395548.
Kesharwani, P., Tekade, R. K., & Jain, N. K. (2015a). Dendrimer generational nomenclature: The need to harmo-
nize. Drug Discovery Today,20(5), 497499.
Kesharwani, P., Tekade, R. K., & Jain, N. K. (2015b). Generation dependent safety and efficacy of folic acid conju-
gated dendrimer based anticancer drug formulations. Pharmaceutical Research,32(4), 14381450.
Kheifets, L. I., Sussman, S. S., & Preston-Martin, S. (1999). Childhood brain tumors and residential electromagnetic fields
(EMF).Reviews of environmental contamination and toxicology (pp. 111129). Springer.
Kim, D.-H., Rozhkova, E. A., Ulasov, I. V., Bader, S. D., Rajh, T., Lesniak, M. S., & Novosad, V. (2010).
Biofunctionalized magnetic-vortex microdiscs for targeted cancer-cell destruction. Nature Materials,9(2),
165171.
Kim, E. H., Lee, H. S., Kwak, B. K., & Kim, B.-K. (2005). Synthesis of ferrofluid with magnetic nanoparticles by
sonochemical method for MRI contrast agent. Journal of Magnetism and Magnetic Materials,289, 328330.
Kim, J.-Y., Choi, W. I., Kim, Y. H., & Tae, G. (2013). Brain-targeted delivery of protein using chitosan-and RVG
peptide-conjugated, pluronic-based nano-carrier. Biomaterials,34(4), 11701178.
Kinoshita, M., McDannold, N., Jolesz, F. A., & Hynynen, K. (2006). Targeted delivery of antibodies through the
bloodbrain barrier by MRI-guided focused ultrasound. Biochemical and Biophysical Research Communications,
340(4), 10851090.
Kleihues, P., Burger, P. C., & Scheithauer, B. W. (1993). The new WHO classification of brain tumours. Brain
pathology,3(3), 255268.
Klibanov, A. L., Maruyama, K., Torchilin, V. P., & Huang, L. (1990). Amphipathic polyethyleneglycols effectively
prolong the circulation time of liposomes. FEBS Letters,268(1), 235237.
Koo, O. M., Rubinstein, I., & Onyuksel, H. (2005). Role of nanotechnology in targeted drug delivery and imaging:
A concise review. Nanomedicine: Nanotechnology, Biology and Medicine,1(3), 193212.
Krauze, M. T., Noble, C. O., Kawaguchi, T., Drummond, D., Kirpotin, D. B., Yamashita, Y., ... Bankiewicz, K. S.
(2007). Convection-enhanced delivery of nanoliposomal CPT-11 (irinotecan) and PEGylated liposomal doxoru-
bicin (Doxil) in rodent intracranial brain tumor xenografts. Neuro-Oncology,9(4), 393403.
Kreuter, J. (2001). Nanoparticulate systems for brain delivery of drugs. Advanced Drug Delivery Reviews,47(1),
6581.
Kreuter, J. (2002). Transport of drugs across the blood-brain barrier by nanoparticles. Current Medicinal Chemistry-
Central Nervous System Agents,2(3), 241249.
429REFERENCES
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
Kulkarni, B., Surnar, B., & Jayakannan, M. (2016). Dual functional nanocarrier for cellular imaging and drug deliv-
ery in cancer cells based on π-conjugated core and biodegradable polymer arms. Biomacromolecules,17(3),
10041016.
Lalu, L., Tambe, V., Pradhan, D., Nayak, K., Bagchi, S., Maheshwari, R., ... Tekade, R. K. (2017). Novel nanosys-
tems for the treatment of ocular inflammation: Current paradigms and future research directions. Journal
Controlled Release,268,1939.
Le Ray, A., Vert, M., Gautier, J., & Benoit, J. (1994). Fate of [14C] poly (DL-lactide-co-glycolide) nanoparticles after
intravenous and oral administration to mice. International Journal of Pharmaceutics,106(3), 201211.
Lee, C., Hwang, H. S., Lee, S., Kim, B., Kim, J. O., Oh, K. T., ... Youn, Y. S. (2017a). Rabies virus-inspired silica-
coated gold nanorods as a photothermal therapeutic platform for treating brain tumors. Advanced Materials,29
(13). Available from https://doi.org/10.1002/adma.201605563.
Lee, H.-Y., Li, Z., Chen, K., Hsu, A. R., Xu, C., Xie, J., ... Chen, X. (2008). PET/MRI dual-modality tumor imaging
using arginine-glycine-aspartic (RGD)conjugated radiolabeled iron oxide nanoparticles. Journal of Nuclear
Medicine,49(8), 13711379.
Lee, T. J., Yoo, J. Y., Shu, D., Li, H., Zhang, J., Yu, J.-G., ... Cui, R. (2017b). RNA nanoparticle-based targeted
therapy for glioblastoma through inhibition of oncogenic miR-21. Molecular Therapy,25(7), 15441555.
Li, X.-Y., Zhao, Y., Sun, M.-G., Shi, J.-F., Ju, R.-J., Zhang, C.-X., ... Zeng, F. (2014). Multifunctional liposomes
loaded with paclitaxel and artemether for treatment of invasive brain glioma. Biomaterials,35(21), 55915604.
Li, Y., He, H., Jia, X., Lu, W.-L., Lou, J., & Wei, Y. (2012). A dual-targeting nanocarrier based on poly (amidoa-
mine) dendrimers conjugated with transferrin and tamoxifen for treating brain gliomas. Biomaterials,33(15),
38993908.
Lian, T., & Ho, R. J. (2001). Trends and developments in liposome drug delivery systems. Journal of Pharmaceutical
Sciences,90(6), 667680.
Liebner, S., Fischmann, A., Rascher, G., Duffner, F., Grote, E.-H., Kalbacher, H., & Wolburg, H. (2000). Claudin-1
and claudin-5 expression and tight junction morphology are altered in blood vessels of human glioblastoma
multiforme. Acta Neuropathologica,100(3), 323331.
Lin, Q., Mao, K.-L., Tian, F.-R., Yang, J.-J., Chen, P.-P., Xu, J., ... Zheng, L. (2016). Brain tumor-targeted delivery
and therapy by focused ultrasound introduced doxorubicin-loaded cationic liposomes. Cancer Chemotherapy
and Pharmacology,77(2), 269280.
Linderoth, L., Peters, G. H., Madsen, R., & Andresen, T. L. (2009). Drug delivery by an enzyme-mediated cycliza-
tion of a lipid prodrug with unique bilayer-formation properties. Angewandte Chemie International Edition,48
(10), 18231826.
Lingineni, K., Belekar, V., Tangadpalliwar, S. R., & Garg, P. (2017). The role of multidrug resistance protein
(MRP-1) as an active efflux transporter on bloodbrain barrier (BBB) permeability. Molecular Diversity,111.
Liu, H.-L., Hua, M.-Y., Yang, H.-W., Huang, C.-Y., Chu, P.-C., Wu, J.-S., ... Chen, P.-Y. (2010). Magnetic resonance
monitoring of focused ultrasound/magnetic nanoparticle targeting delivery of therapeutic agents to the brain.
Proceedings of the National Academy of Sciences,107(34), 1520515210.
Liu, T., Xue, W., Ke, B., Xie, M.-Q., & Ma, D. (2014a). Star-shaped cyclodextrin-poly (l-lysine) derivative co-deli-
vering docetaxel and MMP-9 siRNA plasmid in cancer therapy. Biomaterials,35(12), 38653872.
Liu, X., Tu, M., Kelly, R. S., Chen, C., & Smith, B. J. (2004). Development of a computational approach to predict
blood-brain barrier permeability. Drug Metabolism and Disposition,32(1), 132139.
Liu, Y., Huang, R., Han, L., Ke, W., Shao, K., Ye, L., ... Jiang, C. (2009). Brain-targeting gene delivery and cellular
internalization mechanisms for modified rabies virus glycoprotein RVG29 nanoparticles. Biomaterials,30(25),
41954202.
Liu, Y., Ran, R., Chen, J., Kuang, Q., Tang, J., Mei, L., ... He, Q. (2014b). Paclitaxel loaded liposomes decorated
with a multifunctional tandem peptide for glioma targeting. Biomaterials,35(17), 48354847.
Lockman, P., Mumper, R., Khan, M., & Allen, D. (2002). Nanoparticle technology for drug delivery across the
blood-brain barrier. Drug Development and Industrial Pharmacy,28(1), 113.
Long, D. M. (1970). Capillary ultrastructure and the blood-brain barrier in human malignant brain tumors. Journal
of Neurosurgery,32(2), 127144.
Long, D. M. (1979). Capillary ultrastructure in human metastatic brain tumors. Journal of Neurosurgery,51(1), 5358.
Lo
¨scher, W., & Potschka, H. (2005a). Blood-brain barrier active efflux transporters: ATP-binding cassette gene
family. NeuroRx,2(1), 8698.
430 15. BEYOND THE BLOODBRAIN BARRIER
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
Lo
¨scher, W., & Potschka, H. (2005b). Drug resistance in brain diseases and the role of drug efflux transporters.
Nature Reviews Neuroscience,6(8), 591602.
Louis, D. N., Perry, A., Reifenberger, G., von Deimling, A., Figarella-Branger, D., Cavenee, W. K., ... Ellison,
D. W. (2016). The 2016 World Health Organization classification of tumors of the central nervous system: A
summary. Acta Neuropathologica,131(6), 803820.
Lu, C.-T., Zhao, Y.-Z., Wong, H. L., Cai, J., Peng, L., & Tian, X.-Q. (2014). Current approaches to enhance CNS
delivery of drugs across the brain barriers. International Journal of Nanomedicine,9, 22412257.
Lu, Y.-J., Wei, K.-C., Ma, C.-C. M., Yang, S.-Y., & Chen, J.-P. (2012). Dual targeted delivery of doxorubicin to can-
cer cells using folate-conjugated magnetic multi-walled carbon nanotubes. Colloids and Surfaces B: Biointerfaces,
89,19.
Magaye, R., Zhao, J., Bowman, L., & Ding, M. (2012). Genotoxicity and carcinogenicity of cobalt-, nickel-and
copper-based nanoparticles (Review). Experimental and Therapeutic Medicine,4(4), 551561.
Maheshwari, R. G., Tekade, R. K., Sharma, P. A., Darwhekar, G., Tyagi, A., Patel, R. P., & Jain, D. K. (2012).
Ethosomes and ultradeformable liposomes for transdermal delivery of clotrimazole: A comparative assess-
ment. Saudi Pharm J,20(2), 161170.
Maheshwari, R. G., Thakur, S., Singhal, S., Patel, R. P., Tekade, M., & Tekade, R. K. (2015a). Chitosan encrusted
nonionic surfactant based vesicular formulation for topical administration of ofloxacin. Science of Advanced
Materials,7(6), 11631176.
Maheshwari, R., Tekade, M., Sharma, P. A., & Kumar Tekade, R. (2015b). Nanocarriers assisted siRNA gene
therapy for the management of cardiovascular disorders. Current Pharmaceutical Design,21(30), 44274440.
Mangraviti, A., Gullotti, D., Tyler, B., & Brem, H. (2016). Nanobiotechnology-based delivery strategies: New fron-
tiers in brain tumor targeted therapies. Journal of Controlled Release,240, 443453.
Marks, W. J., Ostrem, J. L., Verhagen, L., Starr, P. A., Larson, P. S., Bakay, R. A., ... Olanow, C. W. (2008). Safety
and tolerability of intraputaminal delivery of CERE-120 (adeno-associated virus serotype 2neurturin) to
patients with idiopathic Parkinson’s disease: An open-label, phase I trial. The Lancet Neurology,7(5), 400408.
Marques, F., Sousa, J. C., Brito, M. A., Pahnke, J., Santos, C., Correia-Neves, M., & Palha, J. A. (2016). The choroid
plexus in health and in disease: Dialogues into and out of the brain. Neurobiology of Disease.
Martinho, N., Florindo, H., Silva, L., Brocchini, S., Zloh, M., & Barata, T. (2014). Molecular modeling to study den-
drimers for biomedical applications. Molecules,19(12), 2042420467.
Masood, F. (2016). Polymeric nanoparticles for targeted drug delivery system for cancer therapy. Materials Science
and Engineering: C,60, 569578.
Matsumoto, G., Kushibiki, T., Kinoshita, Y., Lee, U., Omi, Y., Kubota, E., & Tabata, Y. (2006). Cationized gelatin
delivery of a plasmid DNA expressing small interference RNA for VEGF inhibits murine squamous cell carci-
noma. Cancer Science,97(4), 313321.
May, A. A., Bedel, N. D., Shen, L., Woods, S. C., & Liu, M. (2016). Estrogen and insulin transport through the
blood-brain barrier. Physiology & Behavior,163, 312321.
Maynard, A. D., & Aitken, R. J. (2016). ‘Safe handling of nanotechnology’ ten years on. Nature Research.
McDevitt, M. R., Chattopadhyay, D., Kappel, B. J., Jaggi, J. S., Schiffman, S. R., Antczak, C., ... Scheinberg, D. A.
(2007). Tumor targeting with antibody-functionalized, radiolabeled carbon nanotubes. Journal of Nuclear
Medicine,48(7), 11801189.
McNeill, K. A. (2016). Epidemiology of brain tumors. Neurologic Clinics,34(4), 981998.
Mead, B. P., Mastorakos, P., Suk, J. S., Klibanov, A. L., Hanes, J., & Price, R. J. (2016). Targeted gene transfer to
the brain via the delivery of brain-penetrating DNA nanoparticles with focused ultrasound. Journal of
Controlled Release,223, 109117.
Mehra, N. K., Verma, A. K., Mishra, P., & Jain, N. (2014). The cancer targeting potential of D-α-tocopheryl poly-
ethylene glycol 1000 succinate tethered multi walled carbon nanotubes. Biomaterials,35(15), 45734588.
Mehta, M., Vogelbaum, M. A., Chang, S., & Patel, N. (2011). Neoplasms of the central nervous system. In V. T.
DeVita, Jr, T. S. Lawrence, & S. A. Rosenberg (Eds.), Cancer: Principles and Practice of Oncology. Philadelphia
(pp. 17001749). PA:Lippincott Williams & Wilkins.
Miller, D. S., Bauer, B., & Hartz, A. M. (2008). Modulation of P-glycoprotein at the blood-brain barrier:
Opportunities to improve central nervous system pharmacotherapy. Pharmacological Reviews,60(2), 196209.
Mody, N., Tekade, R. K., Mehra, N. K., Chopdey, P., & Jain, N. K. (2014a). Dendrimer, liposomes, carbon nanotubes
and PLGA nanoparticles: One platform assessment of drug delivery potential. Aaps Pharmscitech,15(2), 388399.
431REFERENCES
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
Mody, V. V., Cox, A., Shah, S., Singh, A., Bevins, W., & Parihar, H. (2014b). Magnetic nanoparticle drug delivery
systems for targeting tumor. Applied Nanoscience,4(4), 385392.
Moghimi, S. M., Hunter, A. C., & Murray, J. C. (2001). Long-circulating and target-specific nanoparticles: Theory
to practice. Pharmacological Reviews,53(2), 283318.
Moghimi, S. M., Hunter, A. C., & Murray, J. C. (2005). Nanomedicine: Current status and future prospects. The
FASEB Journal,19(3), 311330.
Moghimi, S. M., & Szebeni, J. (2003). Stealth liposomes and long circulating nanoparticles: Critical issues in phar-
macokinetics, opsonization and protein-binding properties. Progress in Lipid Research,42(6), 463478.
Munoz, M., Henderson, M., Haber, M., & Norris, M. (2007). Role of the MRP1/ABCC1 multidrug transporter pro-
tein in cancer. IUBMB Life,59(12), 752757.
Na, H. B., Song, I. C., & Hyeon, T. (2009). Inorganic nanoparticles for MRI contrast agents. Advanced Materials,21
(21), 21332148.
Nagane, M. (2011). Neuro-oncology: Continuing multidisciplinary progress. The Lancet Neurology,10(1), 18.
Namen, A. V., & Luke, G. P. (2017). Spectroscopic photoacoustic imaging of gold nanorods. Biomedical
Nanotechnology: Methods and Protocols, 179194.
Neuwelt, E., Howieson, J., Frenkel, E., Specht, D. H., Weigel, R., Buchan, C. G., & Hill, S. (1986). Therapeutic effi-
cacy of multiagent chemotherapy with drug delivery enhancement by blood-brain barrier modification in glio-
blastoma. Neurosurgery,19(4), 573582.
Nicolas, J., Mura, S., Brambilla, D., Mackiewicz, N., & Couvreur, P. (2013). Design, functionalization strategies
and biomedical applications of targeted biodegradable/biocompatible polymer-based nanocarriers for drug
delivery. Chemical Society Reviews,42(3), 11471235.
Nischwitz, V., Berthele, A., & Michalke, B. (2008). Speciation analysis of selected metals and determination of
their total contents in paired serum and cerebrospinal fluid samples: An approach to investigate the perme-
ability of the human blood-cerebrospinal fluid-barrier. Analytica Chimica Acta,627(2), 258269.
Oberdo
¨rster, G., Oberdo
¨rster, E., & Oberdo
¨rster, J. (2005). Nanotoxicology: An emerging discipline evolving from
studies of ultrafine particles. Environmental Health Perspectives, 823839.
Oberoi, R. K., Parrish, K. E., Sio, T. T., Mittapalli, R. K., Elmquist, W. F., & Sarkaria, J. N. (2016). Strategies to
improve delivery of anticancer drugs across the bloodbrain barrier to treat glioblastoma. Neuro-Oncology,
18(1), 2736.
Orringer, D. A., Koo, Y., Chen, T., Kopelman, R., Sagher, O., & Philbert, M. (2009). Small solutions for big pro-
blems: The application of nanoparticles to brain tumor diagnosis and therapy. Clinical Pharmacology &
Therapeutics,85(5), 531534.
Oukarroum, A., Zaidi, W., Samadani, M., & Dewez, D. (2017). Toxicity of nickel oxide nanoparticles on a freshwa-
ter green algal strain of chlorella vulgaris. BioMed Research International,2017.
Pablo Rigalli, J., Ciriaci, N., Domingo Mottino, A., Alicia Catania, V., & Laura Ruiz, M. (2016). modulation of
expression and activity of ABC transporters by the phytoestrogen genistein. Impact on Drug Disposition.
Current Medicinal Chemistry,23(13), 13701389.
Palm-Apergi, C., Lo
¨nn, P., & Dowdy, S. F. (2012). Do cell-penetrating peptides actually “penetrate” cellular mem-
branes?. Molecular Therapy,20(4), 695697.
Pan, W., & Kastin, A. J. (1999). Entry of EGF into brain is rapid and saturable. Peptides,20(9), 10911098.
Pan, W., Kastin, A. J., & Brennan, J. M. (2000). Saturable entry of leukemia inhibitory factor from blood to the cen-
tral nervous system. Journal of Neuroimmunology,106(1), 172180.
Papadopoulos, M., Saadoun, S., Woodrow, C., Davies, D., Costa-Martins, P., Moss, R., ...Bell, B. (2001). Occludin
expression in microvessels of neoplastic and non-neoplastic human brain. Neuropathology and Applied
Neurobiology,27(5), 384395.
Pardridge, W. M. (1998). Blood-brain barrier carrier-mediated transport and brain metabolism of amino acids.
Neurochemical Research,23(5), 635644.
Pardridge, W. M. (2001). Brain drug targeting: The future of brain drug development. Cambridge University Press.
Pardridge, W. M. (2002). Drug and gene delivery to the brain: The vascular route. Neuron,36(4), 555558.
Pardridge, W. M. (2003). Blood-brain barrier drug targeting: The future of brain drug development. Molecular
Interventions,3(2), 90.
Pardridge, W. M. (2005). The blood-brain barrier: Bottleneck in brain drug development. NeuroRx,2(1), 314.
Pardridge, W. M. (2007). Drug targeting to the brain. Pharmaceutical Research,24(9), 17331744.
432 15. BEYOND THE BLOODBRAIN BARRIER
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
Pardridge, W. M., Triguero, D., Buciak, J., & Yang, J. (1990). Evaluation of cationized rat albumin as a potential
blood-brain barrier drug transport vector. Journal of Pharmacology and Experimental Therapeutics,255(2),
893899.
Park, J., Aryal, M., Vykhodtseva, N., Zhang, Y.-Z., & McDannold, N. (2017). Evaluation of permeability, doxorubi-
cin delivery, and drug retention in a rat brain tumor model after ultrasound-induced blood-tumor barrier
disruption. Journal of Controlled Release,250,7785.
Park, J. H., von Maltzahn, G., Ong, L. L., Centrone, A., Hatton, T. A., Ruoslahti, E., ... Sailor, M. J. (2010).
Cooperative nanoparticles for tumor detection and photothermally triggered drug delivery. Advanced
Materials,22(8), 880885.
Park, S.-H., Yoon, Y. I., Moon, H., Lee, G.-H., Lee, B.-H., Yoon, T.-J., & Lee, H. J. (2016). Development of a novel
microbubble-liposome complex conjugated with peptide ligands targeting IL4R on brain tumor cells. Oncology
Reports,36(1), 131136.
Passini, M. A., & Wolfe, J. H. (2001). Widespread gene delivery and structure-specific patterns of expression in
the brain after intraventricular injections of neonatal mice with an adeno-associated virus vector. Journal of
Virology,75(24), 1238212392.
Patel, H. K., Gajbhiye, V., Kesharwani, P., & Jain, N. K. (2016). Ligand anchored poly (propyleneimine) dendri-
mers for brain targeting: Comparative in vitro and in vivo assessment. Journal of Colloid and Interface Science,
482, 142150.
Patri, A. K., Kukowska-Latallo, J. F., & Baker, J. R. (2005). Targeted drug delivery with dendrimers: Comparison
of the release kinetics of covalently conjugated drug and non-covalent drug inclusion complex. Advanced Drug
Delivery Reviews,57(15), 22032214.
Peer, D., Karp, J. M., Hong, S., Farokhzad, O. C., Margalit, R., & Langer, R. (2007). Nanocarriers as an emerging
platform for cancer therapy. Nature Nanotechnology,2(12), 751760.
Perkins, M., & Liu, G. (2016). Primary brain tumors in adults: Diagnosis and treatment. American Family Physician,
93(3), 211217.
Pinto, M. P., Arce, M., Yameen, B., & Vilos, C. (2017). Targeted brain delivery nanoparticles for malignant glio-
mas. Nanomedicine,12(1), 5972.
Rancan, F., Rosan, S., Boehm, F., Cantrell, A., Brellreich, M., Schoenberger, H., ... Moussa, F. (2002). Cytotoxicity
and photocytotoxicity of a dendritic C 60 mono-adduct and a malonic acid C 60 tris-adduct on Jurkat cells.
Journal of photochemistry and Photobiology B: Biology,67(3), 157162.
Ransohoff, R. M. (2016). How neuroinflammation contributes to neurodegeneration. Science,353(6301), 777783.
Reddy, G. R., Bhojani, M. S., McConville, P., Moody, J., Moffat, B. A., Hall, D. E., ... Sugai, J. V. (2006).
Vascular targeted nanoparticles for imaging and treatment of brain tumors. Clinical Cancer Research,12(22),
66776686.
Remya, N., Syama, S., Sabareeswaran, A., & Mohanan, P. (2016). Toxicity, toxicokinetics and biodistribution of
dextran stabilized Iron oxide Nanoparticles for biomedical applications. International Journal of Pharmaceutics,
511(1), 586598.
Ren, J., Shen, S., Wang, D., Xi, Z., Guo, L., Pang, Z., ... Jiang, X. (2012). The targeted delivery of anticancer drugs
to brain glioma by PEGylated oxidized multi-walled carbon nanotubes modified with angiopep-2. Biomaterials,
33(11), 33243333.
Roller, M. (2009). Carcinogenicity of inhaled nanoparticles. Inhalation Toxicology,21(sup1), 144157.
Ro
¨mermann, K., Wanek, T., Bankstahl, M., Bankstahl, J. P., Fedrowitz, M., Mu
¨ller, M., ... Langer, O. (2013). R)-[11
C] verapamil is selectively transported by murine and human P-glycoprotein at the bloodbrain barrier, and
not by MRP1 and BCRP. Nuclear Medicine and Biology,40(7), 873878.
Ronaldson, P. T., & Davis, T. P. (2016). Glial support of bloodbrain barrier integrity: Molecular targets for novel thera-
peutic strategies in stroke.Non-neuronal mechanisms of brain damage and repair afterstroke (pp. 4580). Springer.
Rose, C. R., & Verkhrastky, A. (2016). Principles of sodium homeostasis and sodium signalling in astroglia. Glia,
64(10), 16111627.
Ruan, S., Hu, C., Tang, X., Cun, X., Xiao, W., Shi, K., ... Gao, H. (2016). Increased gold nanoparticle retention in
brain tumors by in situ enzyme-induced aggregation. ACS Nano,10(11), 1008610098.
Ryu, J.-H., Chacko, R. T., Jiwpanich, S., Bickerton, S., Babu, R. P., & Thayumanavan, S. (2010). Self-cross-linked
polymer nanogels: A versatile nanoscopic drug delivery platform. Journal of the American Chemical Society,132
(48), 1722717235.
433REFERENCES
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
Sai, Y., Kajita, M., Tamai, I., Wakama, J., Wakamiya, T., & Tsuji, A. (1998). Adsorptive-mediated endocytosis of a
basic peptide in enterocyte-like Caco-2 cells. American Journal of Physiology-Gastrointestinal and Liver Physiology,
275(3), G514G520.
Saito, R., Bringas, J. R., McKnight, T. R., Wendland, M. F., Mamot, C., Drummond, D. C., ... Bankiewicz, K. S.
(2004). Distribution of liposomes into brain and rat brain tumor models by convection-enhanced delivery mon-
itored with magnetic resonance imaging. Cancer Research,64(7), 25722579.
Sajid, M. I., Jamshaid, U., Jamshaid, T., Zafar, N., Fessi, H., & Elaissari, A. (2016). Carbon nanotubes from synthe-
sis to in vivo biomedical applications. International Journal of Pharmaceutics,501(1), 278299.
Sarin, H., Kanevsky, A. S., Wu, H., Brimacombe, K. R., Fung, S. H., Sousa, A. A., ... Aronova, M. A. (2008).
Effective transvascular delivery of nanoparticles across the blood-brain tumor barrier into malignant glioma
cells. Journal of Translational Medicine,6(1), 80.
Sarin, H., Kanevsky, A. S., Wu, H., Sousa, A. A., Wilson, C. M., Aronova, M. A., ... Vo, H. Q. (2009). Physiologic
upper limit of pore size in the blood-tumor barrier of malignant solid tumors. Journal of Translational Medicine,
7(1), 51.
Satija, J., Gupta, U., & Jain, N. K. (2007). Pharmaceutical and biomedical potential of surface engineered dendri-
mers. Critical Reviewstin Therapeutic Drug Carrier Systems,24(3), 257306.
Sauer, I., Dunay, I. R., Weisgraber, K., Bienert, M., & Dathe, M. (2005). An apolipoprotein E-derived peptide med-
iates uptake of sterically stabilized liposomes into brain capillary endothelial cells. Biochemistry,44(6),
20212029.
Saunders, N. R., Ek, C. J., Habgood, M. D., & Dziegielewska, K. M. (2008). Barriers in the brain: A renaissance?
Trends in Neurosciences,31(6), 279286.
Saunders, N. R., Habgood, M. D., Møllga
˚rd, K., & Dziegielewska, K. M. (2016). The biological significance of brain
barrier mechanisms: Help or hindrance in drug delivery to the central nervous system? F1000Research,5.
Schabet, M. (1999). Epidemiology of primary CNS lymphoma. Journal of Neuro-Oncology,43(3), 199201.
Schaddelee, M. P., Voorwinden, H. L., Groenendaal, D., Hersey, A., Ijzerman, A. P., Danhof, M., & De Boer, A. G.
(2003). Bloodbrain barrier transport of synthetic adenosine A 1 receptor agonists in vitro: Structure transport
relationships. European journal of Pharmaceutical Sciences,20(3), 347356.
Schneider, S. W., Ludwig, T., Tatenhorst, L., Braune, S., Oberleithner, H., Senner, V., & Paulus, W. (2004).
Glioblastoma cells release factors that disrupt blood-brain barrier features. Acta Neuropathologica,107(3), 272276.
Schu
¨z, J., Jacobsen, R., Olsen, J. H., Boice, J. D., McLaughlin, J. K., & Johansen, C. (2006). Cellular telephone use
and cancer risk: Update of a nationwide Danish cohort. Journal of the National Cancer Institute,98(23),
17071713.
Seelig, A., Gottschlich, R., & Devant, R. M. (1994). A method to determine the ability of drugs to diffuse through
the blood-brain barrier. Proceedings of the National Academy of Sciences,91(1), 6872.
Shah, S. (2016). The nanomaterial toolkit for neuroengineering. Nano Convergence,3(1), 25.
Sharma, P., Maheshwari, R., Tekade, M., & Kumar Tekade, R. (2015). Nanomaterial based approaches for the
diagnosis and therapy of cardiovascular diseases. Current Pharmaceutical Design,21(30), 44654478.
Shaw, T. K., Mandal, D., Dey, G., Pal, M. M., Paul, P., Chakraborty, S., ... Mandal, M. (2017). Successful delivery
of docetaxel to rat brain using experimentally developed nanoliposome: A treatment strategy for brain tumor.
Drug Delivery,24(1), 346357.
Shen, J., Zhan, C., Xie, C., Meng, Q., Gu, B., Li, C., ... Lu, W. (2011). Poly (ethylene glycol)-block-poly (D, L-
lactide acid) micelles anchored with angiopep-2 for brain-targeting delivery. Journal of Drug Targeting,19(3),
197203.
Shi, J., Kantoff, P. W., Wooster, R., & Farokhzad, O. C. (2017). Cancer nanomedicine: Progress, challenges and
opportunities. Nature Reviews Cancer,17(1), 2037.
Siegal, T., Horowitz, A., & Gabizon, A. (1995). Doxorubicin encapsulated in sterically stabilized liposomes for the
treatment of a brain tumor model: Biodistribution and therapeutic efficacy. Journal of Neurosurgery,83(6),
10291037.
Siegel, R. L., Miller, K. D., & Jemal, A. (2016). Cancer statistics, 2016. CA: A Cancer Journal for Clinicians,66(1), 730.
Silva, A. C., Oliveira, T. R., Mamani, J. B., Malheiros, S. M., Malavolta, L., Pavon, L. F., ... Vidoto, E. L. G. (2011).
Application of hyperthermia induced by superparamagnetic iron oxide nanoparticles in glioma treatment.
International Journal of Nanomedicine,6, 591603.
Silva, G. A. (2007). Nanotechnology approaches for drug and small molecule delivery across the blood brain bar-
rier. Surgical Neurology,67(2), 113116.
434 15. BEYOND THE BLOODBRAIN BARRIER
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
Silvander, M. (2002). Steric stabilization of liposomes—A review.Lipid and Polymer-Lipid Systems (pp. 3540).
Springer.
Simone, E. A., Dziubla, T. D., & Muzykantov, V. R. (2008). Polymeric carriers: Role of geometry in drug delivery.
Expert Opinion on Drug Delivery,5(12), 12831300.
Song, X.-L., Liu, S., Jiang, Y., Gu, L.-Y., Xiao, Y., Wang, X., ... Li, X.-T. (2017). Targeting vincristine plus tetran-
drine liposomes modified with DSPE-PEG 2000-transferrin in treatment of brain glioma. European Journal of
Pharmaceutical Sciences,96, 129140.
Soni, R., Garg, A., Patel, V., & Yadav, A. K. (2016a). Nanoparticles: A regnant approach for drug delivery and can-
cer management. Asian Journal of Biomaterial Research,2(1), 921.
Soni, N., Soni, N., Pandey, H., Maheshwari, R., Kesharwani, P., & Tekade, R. K. (2016b). Augmented delivery of
gemcitabine in lung cancer cells exploring mannose anchored solid lipid nanoparticles. J Colloid Interface Sci,
481, 107116.
Soni, N., Tekade, M., Kesharwani, P., Bhattacharya, P., Maheshwari, R., Dua, K., Hansbro, P. M., & Tekade, R. K.
(2017). Recent Advances in Oncological Submissions of Dendrimer. Curr Pharm Des,23(21), 30843098.
Sorensen, A. G., Tievsky, A. L., Ostergaard, L., Weisskoff, R. M., & Rosen, B. R. (1997). Contrast agents in func-
tional MR imaging. Journal of Magnetic Resonance Imaging,7(1), 4755.
Spector, R., Keep, R. F., Snodgrass, S. R., Smith, Q. R., & Johanson, C. E. (2015). A balanced view of choroid
plexus structure and function: Focus on adult humans. Experimental Neurology,267,7886.
Stepensky, D. (2016). Delivery of peptides and proteins to the brain using nano-drug delivery systems and other formula-
tions.Nanomedicine (pp. 201220). Springer.
Suhy, A. M., Webb, A., Papp, A. C., Geier, E. G., & Sadee, W. (2017). Expression and splicing of ABC and SLC
transporters in the human blood-brain barrier measured with RNAseq. European Journal of Pharmaceutical
Sciences,103,4751.
Sullivan, D. C., & Ferrari, M. (2004). Nanotechnology and tumor imaging: Seizing an opportunity. Molecular
Imaging,3(4), 15353500200404139.
Sun, H., Dai, H., Shaik, N., & Elmquist, W. F. (2003). Drug efflux transporters in the CNS. Advanced Drug Delivery
Reviews,55(1), 83105.
Szaka
´cs, G., Paterson, J. K., Ludwig, J. A., Booth-Genthe, C., & Gottesman, M. M. (2006). Targeting multidrug
resistance in cancer. Nature reviews Drug Discovery,5(3), 219234.
Tamai, I., & Tsuji, A. (1996). Drug delivery through the blood-brain barrier. Advanced Drug Delivery Reviews,19(3),
401424.
Tamai, I., & Tsuji, A. (2000). Transporter-mediated permeation of drugs across the bloodbrain barrier. Journal of
Pharmaceutical Sciences,89(11), 13711388.
Tekade, R. K., Maheshwari, R., Soni, N., Tekade, M., & Chougule, M. B. (2017a). Chapter 1 - Nanotechnology for
the development of nanomedicine A2 - Mishra, Vijay. In P. Kesharwani, M. C. I. M. Amin, & A. Iyer (Eds.),
Nanotechnology-Based Approaches for Targeting and Delivery of Drugs and Genes. Academic Press.
Tekade, R. K., Maheshwari, R., Soni, N., & Tekade, M. (2017b). Carbon nanotubes in targeting and delivery of
drugs A2 - Mishra, Vijay. In P. Kesharwani, M. C. I. M. Amin, & A. Iyer (Eds.), Nanotechnology-Based
Approaches for Targeting and Delivery of Drugs and Genes. Academic Press.
Tekade, R. K., Maheshwari, R., Tekade, M., & Chougule, M. B. (2017c). Solid lipid nanoparticles for targeting and
delivery of drugs and genes A2 - Mishra, Vijay. In P. Kesharwani, M. C. I. M. Amin, & A. Iyer (Eds.),
Nanotechnology-Based Approaches for Targeting and Delivery of Drugs and Genes. Academic Press.
Temsamani, J., & Vidal, P. (2004). The use of cell-penetrating peptides for drug delivery. Drug Discovery Today,9
(23), 10121019.
Thorek, D. L., Chen, A. K., Czupryna, J., & Tsourkas, A. (2006). Superparamagnetic iron oxide nanoparticle probes
for molecular imaging. Annals of biomedical engineering,34(1), 2338.
Timbie, K. F., Afzal, U., Date, A., Zhang, C., Song, J., Miller, G. W., ... Price, R. J. (2017). MR image-guided deliv-
ery of cisplatin-loaded brain-penetrating nanoparticles to invasive glioma with focused ultrasound. Journal of
Controlled Release,263, 120131.
Torchilin, V. (1998). Polymer-coated long-circulating microparticulate pharmaceuticals. Journal of
Microencapsulation,15(1), 119.
Torchilin, V. P. (2005). Recent advances with liposomes as pharmaceutical carriers. Nature Reviews Drug Discovery,
4(2), 145160.
Tzeng, S. Y., & Green, J. J. (2013). Therapeutic nanomedicine for brain cancer. Therapeutic Delivery,4(6), 687704.
435REFERENCES
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
Upadhyay, R. K. (2014). Drug delivery systems, CNS protection, and the blood brain barrier. BioMed Research
International,2014.
Vandergrift, W. A., & Patel, S. J. (2006). Convection-enhanced delivery of immunotoxins and radioisotopes for
treatment of malignant gliomas. Neurosurgical Focus,20(4), E13.
Vartanian, A., Singh, S. K., Agnihotri, S., Jalali, S., Burrell, K., Aldape, K. D., & Zadeh, G. (2014). GBM’s multifaceted
landscape: Highlighting regional and microenvironmental heterogeneity. Neuro-Oncology,16(9), 11671175.
Vernooij, M. W., Ikram, M. A., Tanghe, H. L., Vincent, A. J., Hofman, A., Krestin, G. P., ... van der Lugt, A.
(2007). Incidental findings on brain MRI in the general population. New England Journal of Medicine,357(18),
18211828.
Verrecchia, T., Spenlehauer, G., Bazile, D., Murry-Brelier, A., Archimbaud, Y., & Veillard, M. (1995). Non-stealth
(poly (lactic acid/albumin)) and stealth (poly (lactic acid-polyethylene glycol)) nanoparticles as injectable drug
carriers. Journal of Controlled Release,36(12), 4961.
Vile, R. G., Tuszynski, A., & Castleden, S. (1996). Retroviral vectors. Molecular Biotechnology,5(2), 139158.
Visser, C. C., Voorwinden, L. H., Crommelin, D. J., Danhof, M., & de Boer, A. G. (2004). Characterization and
modulation of the transferrin receptor on brain capillary endothelial cells. Pharmaceutical Research,21(5),
761769.
Voura, E. B., Jaiswal, J. K., Mattoussi, H., & Simon, S. M. (2004). Tracking metastatic tumor cell extravasation with
quantum dot nanocrystals and fluorescence emission-scanning microscopy. Nature Medicine,10(9), 993998.
Vykhodtseva, N., McDannold, N., & Hynynen, K. (2008). Progress and problems in the application of focused
ultrasound for bloodbrain barrier disruption. Ultrasonics,48(4), 279296.
Wang, B., Lv, L., Wang, Z., Jiang, Y., Lv, W., Liu, X., ... Xu, Q. (2015). Improved anti-glioblastoma efficacy by IL-
13Rα2 mediated copolymer nanoparticles loaded with paclitaxel. Scientific Reports,5.
Wang, H. (2016). Development of multi-modality polymer-coated quantum dots for cancer imaging. Atlanta, GA: Georgia
Institute of Technology.
Wang, P. P., Frazier, J., & Brem, H. (2002). Local drug delivery to the brain. Advanced Drug Delivery Reviews,54(7),
9871013.
Wang, X., Tu, M., Tian, B., Yi, Y., Wei, Z., & Wei, F. (2016). Synthesis of tumor-targeted folate conjugated fluores-
cent magnetic albumin nanoparticles for enhanced intracellular dual-modal imaging into human brain tumor
cells. Analytical Biochemistry,512,817.
Wardill, H. R., Mander, K. A., Van Sebille, Y. Z., Gibson, R. J., Logan, R. M., Bowen, J. M., & Sonis, S. T. (2016).
Cytokine-mediated blood brain barrier disruption as a conduit for cancer/chemotherapy-associated neurotox-
icity and cognitive dysfunction. International Journal of Cancer,139(12), 26352645.
Warso, M., Richards, J., Mehta, D., Christov, K., Schaeffer, C., Bressler, L. R., ... Beattie, C. (2013). A first-in-class,
first-in-human, phase I trial of p28, a non-HDM2-mediated peptide inhibitor of p53 ubiquitination in patients
with advanced solid tumours. British Journal of Cancer,108(5), 10611070.
Wei, H., Bruns, O. T., Kaul, M. G., Hansen, E. C., Barch, M., Wi´
sniowska, A., ... Okada, S. (2017a). Exceedingly
small iron oxide nanoparticles as positive MRI contrast agents. Proceedings of the National Academy of Sciences,
201620145.
Wei, K.-C., Chu, P.-C., Wang, H.-Y. J., Huang, C.-Y., Chen, P.-Y., Tsai, H.-C., ... Feng, L.-Y. (2013). Focused
ultrasound-induced bloodbrain barrier opening to enhance temozolomide delivery for glioblastoma treat-
ment: A preclinical study. PloS one,8(3), e58995.
Wei, L., Guo, X.-Y., Yang, T., Yu, M.-Z., Chen, D.-W., & Wang, J.-C. (2016). Brain tumor-targeted therapy by sys-
temic delivery of siRNA with Transferrin receptor-mediated core-shell nanoparticles. International Journal of
Pharmaceutics,510(1), 394405.
Wei, Y., Liao, R., Mahmood, A. A., Xu, H., & Zhou, Q. (2017b). pH-responsive pHLIP (pH low insertion peptide)
nanoclusters of superparamagnetic iron oxide nanoparticles as a tumor-selective MRI contrast agent. Acta
Biomaterialia,55, 194203.
Wigertz, A., Lo
¨nn, S., Schwartzbaum, J., Hall, P., Auvinen, A., Christensen, H. C., ... Schoemaker, M. J. (2007).
Allergic conditions and brain tumor risk. American Journal of Epidemiology,166(8), 941950.
van Vlerken, L. E., Vyas, T. K., & Amiji, M. M. (2007). Poly (ethylene glycol)-modified nanocarriers for tumor-
targeted and intracellular delivery. Pharmaceutical Research,24(8), 14051414.
Van Woensel, M., Wauthoz, N., Rosie
`re, R., Mathieu, V., Kiss, R., Lefranc, F., ... Mathivet, T. (2016). Development
of siRNA-loaded chitosan nanoparticles targeting Galectin-1 for the treatment of glioblastoma multiforme via
intranasal administration. Journal of Controlled Release,227,7181.
436 15. BEYOND THE BLOODBRAIN BARRIER
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
Wohlfart, S., Gelperina, S., & Kreuter, J. (2012). Transport of drugs across the bloodbrain barrier by nanoparti-
cles. Journal of Controlled Release,161(2), 264273.
Wolburg, H., & Lippoldt, A. (2002). Tight junctions of the bloodbrain barrier: Development, composition and
regulation. Vascular Pharmacology,38(6), 323337.
Wolburg, H., Noell, S., Fallier-Becker, P., Mack, A. F., & Wolburg-Buchholz, K. (2012). The disturbed bloodbrain
barrier in human glioblastoma. Molecular Aspects of Medicine,33(5), 579589.
Wolburg, H., Noell, S., Mack, A., Wolburg-Buchholz, K., & Fallier-Becker, P. (2009). Brain endothelial cells and
the glio-vascular complex. Cell and Tissue Research,335(1), 7596.
Wong, H. L., Wu, X. Y., & Bendayan, R. (2012). Nanotechnological advances for the delivery of CNS therapeutics.
Advanced Drug Delivery Reviews,64(7), 686700.
Wu, J., Yoon, S. H., Wu, W. M., & Bodor, N. (2002). Synthesis and biological evaluations of brain-targeted chemi-
cal delivery systems of [Nva2]-TRH. Journal of Pharmacy and Pharmacology,54(7), 945950.
Xu, Z., Wang, Y., Ma, Z., Wang, Z., Wei, Y., & Jia, X. (2016). A poly (amidoamine) dendrimer-based nanocarrier
conjugated with Angiopep-2 for dual-targeting function in treating glioma cells. Polymer Chemistry,7(3),
715721.
Yang, Z.-Z., Li, J.-Q., Wang, Z.-Z., Dong, D.-W., & Qi, X.-R. (2014). Tumor-targeting dual peptides-modified cat-
ionic liposomes for delivery of siRNA and docetaxel to gliomas. Biomaterials,35(19), 52265239.
Yordanov, G. (2012). Poly (alkyl cyanoacrylate) nanoparticles as drug carriers: 33 years later. Bulgarian Journal of
Chemistry,1(2), 6172.
Yu, W.-Y., & Zhang, N. (2009). Surface modification of nanocarriers for cancer therapy. Current Nanoscience,5(2),
123134.
Zhang, E. Y., Knipp, G. T., Ekins, S., & Swaan, P. W. (2002a). Structural biology and function of solute transpor-
ters: Implications for identifying and designing substrates. Drug Metabolism Reviews,34(4), 709750.
Zhang, P., Hu, L., Yin, Q., Zhang, Z., Feng, L., & Li, Y. (2012). Transferrin-conjugated polyphosphoester hybrid
micelle loading paclitaxel for brain-targeting delivery: Synthesis, preparation and in vivo evaluation. Journal of
Controlled Release,159(3), 429434.
Zhang, R. X., Ahmed, T., Li, L. Y., Li, J., Abbasi, A. Z., & Wu, X. Y. (2017a). Design of nanocarriers for nanoscale
drug delivery to enhance cancer treatment using hybrid polymer and lipid building blocks. Nanoscale,9(4),
13341355.
Zhang, W., Yu, Z.-L., Wu, M., Ren, J.-G., Xia, H.-F., Sa, G.-L., ... Chen, G. (2017b). Magnetic and folate functionali-
zation enables rapid isolation and enhanced tumor-targeting of cell-derived microvesicles. ACS Nano,11(1),
277290.
Zhang, X., Wang, X., Zhong, W., Ren, X., Sha, X., & Fang, X. (2016). Matrix metalloproteinases-2/9-sensitive
peptide-conjugated polymer micelles for site-specific release of drugs and enhancing tumor accumulation:
Preparation and in vitro and in vivo evaluation. International Journal of Nanomedicine,11, 1643.
Zhang, Y., Bai, Y., & Yan, B. (2010). Functionalized carbon nanotubes for potential medicinal applications. Drug
Discovery Today,15(11), 428435.
Zhang, Y., & Pardridge, W. M. (2005). Delivery of β-galactosidase to mouse brain via the blood-brain barrier
transferrin receptor. Journal of Pharmacology and Experimental Therapeutics,313(3), 10751081.
Zhang, Y., Zhu, C., & Pardridge, W. M. (2002b). Antisense gene therapy of brain cancer with an artificial virus
gene delivery system. Molecular Therapy,6(1), 6772.
Zhao, Y., Ren, W., Zhong, T., Zhang, S., Huang, D., Guo, Y., ... Zhang, X. (2016). Tumor-specific pH-responsive
peptide-modified pH-sensitive liposomes containing doxorubicin for enhancing glioma targeting and anti-
tumor activity. Journal of Controlled Release,222,5666.
Zhou, F., Zhu, L., Wang, K., & Murray, M. (2016). Recent advance in the pharmacogenomics of human Solute
Carrier Transporters (SLCs) in drug disposition. Advanced Drug Delivery Reviews,116,2136.
Zhou, Z., Singh, R., & Souweidane, M. M. (2017). Convection-Enhanced Delivery for Diffuse Intrinsic Pontine
Glioma Treatment. Current Neuropharmacology,15(1), 116128.
Further Reading
American Cancer Society. (2017). American cancer society: Cancer facts and figures 2017.Atlanta:American Cancer
Society.
437FURTHER READING
NANOTECHNOLOGY-BASED TARGETED DRUG DELIVERY SYSTEMS FOR BRAIN TUMORS
... Although the majority of current studies demonstrating successful delivery of nanoparticles across the BBB encapsulate or conjugate antibodies or nanobodies for enhanced site specificity, this success encourages further study into the delivery of therapeutic antibodies into the brain onboard nanoparticle delivery complexes. It is important to note, however, that only a few nanoparticles targeting CNS diseases have ever reached the clinical trial stage, and to our knowledge all have failed due to technical and cost limitations, as well as exhibiting long-term toxicity as described in other reviews [69,70]. ...
... Dendrimers are typically much smaller than liposomes, with some AuNPs only reaching up to 10 nm in width, and they therefore rarely encapsulate drug molecules as liposomes do. Instead, therapeutics can be conjugated to their branched surfaces for delivery [69]. Cross-BBB delivery of dendrimers has been achieved through carotid injection [75], olfactory nerve transport [76], and less invasively via exploitation of carrier-mediated transport and endocytosis mechanisms [77]. ...
... Cross-BBB delivery of dendrimers has been achieved through carotid injection [75], olfactory nerve transport [76], and less invasively via exploitation of carrier-mediated transport and endocytosis mechanisms [77]. Polyamidoamine (PAMAM) is one of the more established dendrimers currently being studied for use in cross-BBB drug delivery, recently demonstrating very promising in vitro and in vivo results [45,69]. A PA-MAM dendrimer loaded with siLSNCT5, a type of non-coding RNA that contributes to the suppression of tumour growth and glioma metastasis, was conjugated to cell-penetrating peptide, tLyp-1, and administered intravenously in glioma-bearing mice. ...
Article
Full-text available
For the treatment of neurological diseases, achieving sufficient exposure to the brain parenchyma is a critical determinant of drug efficacy. The blood–brain barrier (BBB) functions to tightly control the passage of substances between the bloodstream and the central nervous system, and as such poses a major obstacle that must be overcome for therapeutics to enter the brain. Monoclonal antibodies have emerged as one of the best-selling treatment modalities available in the pharmaceutical market owing to their high target specificity. However, it has been estimated that only 0.1% of peripherally administered antibodies can cross the BBB, contributing to the low success rate of immunotherapy seen in clinical trials for the treatment of neurological diseases. The development of new strategies for antibody delivery across the BBB is thereby crucial to improve immunotherapeutic efficacy. Here, we discuss the current strategies that have been employed to enhance antibody delivery across the BBB. These include (i) focused ultrasound in combination with microbubbles, (ii) engineered bi-specific antibodies, and (iii) nanoparticles. Furthermore, we discuss emerging strategies such as extracellular vesicles with BBB-crossing properties and vectored antibody genes capable of being encapsulated within a BBB delivery vehicle.
... TJs are stabilized by adherens junctions (AJs), which are close to the basolateral membrane and comprise cadherin, catenin, alpha-actinin, and vinculin, forming homophilic endothelial-to-endothelial contacts roughly 20 nm wide and participating in the development and preservation of TJs [15]. AJs are connected to the EC cytoskeleton, modulate receptor signaling, and regulate the transendothelial migration of immune cells [14]. ...
Article
Full-text available
The state of well-being and health of our body is regulated by the fine osmotic and biochemical balance established between the cells of the different tissues, organs, and systems. Specific districts of the human body are defined, kept in the correct state of functioning, and, therefore, protected from exogenous or endogenous insults of both mechanical, physical, and biological nature by the presence of different barrier systems. In addition to the placental barrier, which even acts as a linker between two different organisms, the mother and the fetus, all human body barriers, including the blood–brain barrier (BBB), blood–retinal barrier, blood–nerve barrier, blood–lymph barrier, and blood–cerebrospinal fluid barrier, operate to maintain the physiological homeostasis within tissues and organs. From a pharmaceutical point of view, the most challenging is undoubtedly the BBB, since its presence notably complicates the treatment of brain disorders. BBB action can impair the delivery of chemical drugs and biopharmaceuticals into the brain, reducing their therapeutic efficacy and/or increasing their unwanted bioaccumulation in the surrounding healthy tissues. Recent nanotechnological innovation provides advanced biomaterials and ad hoc customized engineering and functionalization methods able to assist in brain-targeted drug delivery. In this context, lipid nanocarriers, including both synthetic (liposomes, solid lipid nanoparticles, nanoemulsions, nanostructured lipid carriers, niosomes, proniosomes, and cubosomes) and cell-derived ones (extracellular vesicles and cell membrane-derived nanocarriers), are considered one of the most successful brain delivery systems due to their reasonable biocompatibility and ability to cross the BBB. This review aims to provide a complete and up-to-date point of view on the efficacy of the most varied lipid carriers, whether FDA-approved, involved in clinical trials, or used in in vitro or in vivo studies, for the treatment of inflammatory, cancerous, or infectious brain diseases.
... Opening of BBB due to the release of interleukin-6 from astrocytes is observed in the brain due to traumatic conditions [51]. Therefore, BBB is a significant obstacle to the treatment of brain diseases with disrupted or leaky BBB, which pushes researchers to develop novel and effective brain drug delivery systems [12,55,56]. ...
Article
In recent years, unique physicochemical properties of amphiphilic block copolymers have been utilized to design the polymeric micelles for brain-specific delivery of drugs, proteins, peptides, and genes. Their unique properties such as nano-size, charge-switching ability, stimuli-responsive cargo release, flexible structure, and self-assembly enable them to overcome limitations of conventional dosage forms that include rapid drug release, drug efflux, and poor brain bioavailability, and poor stability. These limitations hinder their therapeutic efficacy in treating brain diseases. Their ease of functionalization and enhanced penetration and retention effect make them suitable nanocarriers for the diagnosis of various brain diseases. In this context, the present manuscript provides an insight into the progress made in the functionalization of micelles such as the incorporation of stimuli-sensitive moieties in copolymers, conjugation of cargo molecules with the core-forming block via responsive smart linkers, and conjugation of active ligands and imaging moieties with the corona forming block for brain targeting and imaging. Further, the review also expounds on the role of polymeric micelles in delivering neurotherapeutic to the brain. Some patents related to polymeric micelles formulated for brain delivery are also enlisted.
... Chitosan has been thought about as a possible material for nerve regeneration thanks to its distinctive properties like growth, medicinal drug activity, biodegradability, and biocompatibility. Haipeng et al. according to those neurons civilized on the chitosan membrane will grow well which chitosan tube will greatly promote the repair of the peripheral nervous system [29,114,115]. Apart from that, bio-nanotechnology with nano-bioelectronics has several benefits in brain analysis and neurobiology. ...
Article
Full-text available
The modern age is an arena of interdisciplinary research and knowledge domain which involves versatile field of sciences to work cooperatively for the improvement of the mankind. Biotechnology is providing for more personalized healthcare and continued analysis of the human body. As biotechnology advances day by day, we have to uphold the pace to discover future medical applications of it. Biotechnology is a huge and rapidly growing field. Biomedical technology involves the application of engineering and technology principles to the domain of living or biological systems. Generally biomedical denotes larger stress on issues related to human health and diseases. Different kinds of live expression systems like plant or insect cells, transgenic animals, mammals, yeast, Escherichia coli and more are particularly beneficial because biotechnology-derived medicines from them. This type of expressed gene or protein incorporates the identical nucleotide sequence as endogenous form of humans. Application of biotechnology in different domain of biomedical fields has already brought about a substantial difference which denotes the superiority over traditional ways of treatment. It is very easy to understand that how biotechnology can be played a crucial role in medical purposes. This paper will try to highlight the glimpse of multifaceted application of biotechnology in different field as well as from different angle of application.
... Further experiments to analyze the nature of this unidirectional flow of miRNA would allow for a better understanding of its mechanism. With the intracerebral method of drug delivery, the BBB is no longer an obstacle, as the drug is delivered directly to the ventricles, where the BBB has already been encountered [52]. ...
Article
Full-text available
Glioblastoma multiforme (GBM) is perhaps the most devastating tumor of the central nervous system (CNS), with an approximately 7% five-year survival rate. The micro-RNAs (miRNA) miRNA-200a and are downregulated in GBM. We hypothesize that miPEP-200a and miPEP-200b, discovered in our lab, has potential to restore levels of miRNA-200a and miRNA-200b and function as inhibitors of migration of glioma cells. For this work, a literature search was conducted, identifying various upregulated and downregulated members of the miRNA family in GBM. A search for the miPEP-200 family was also performed. An in-depth analysis of the blood-brain barrier (BBB) components was conducted to determine feasible candidates for therapeutic treatment of GBM. There has recently been a substantial increase in studies examining the role of miRNA in GBM. A thorough review of existing literature showed that in GBM, 73% (256) of miRNAs involved are upregulated and 27% (95) are downregulated. The latter group contains miRNA-200a and miRNA-200b. It's been shown that miRNA has excellent stability in the blood stream. Although miRNA can cross the BBB from the cerebrospinal fluid (CSF) to the blood, the reverse movement has not yet been shown. Considering the characteristics of the BBB combined with experimental data from various studies showing the ability of miRNA to cross the BBB, miRNA has potential for use as a therapeutic agent in GBM. Specifically, miRNA-200a and miRNA-200b are known to be downregulated in GBM. Interestingly, our laboratory recently discovered the novel proteins miPEP-200a and miPEP-200b, both products of pri-miRNA translation. These proteins may demonstrate great potential for upregulating miRNA-200a and miRNA-200b, given data suggesting this positive feedback mechanism found in one study. Since miPEP-200a and 200b were shown to inhibit migration of cancer cells and Epithelial to Mesenchymal Transition (EMT) of cancer cells, we propose that these miPEP-200a and miPEP-200b may function as therapeutic agents for targeting the migration of glioma cells.
Article
Our present work demonstrates the molecular hybridization-assisted design, synthesis, and biological evaluation of 22 benzylpiperazine-linked 1,2,4-triazole compounds (PD1-22) as AD modifying agents. All the compounds were tested for their in vitro hChEs, hBACE-1, and Aβ-aggregation inhibition properties. Among them, compound PD-08 and PD-22 demonstrated good hChE and hBACE-1 inhibition as compared to standards donepezil and rivastigmine. Both compounds displaced PI from PAS at 50 µM concentration which was comparable to donepezil and also demonstrated anti-Aβ aggregation properties in self- and AChE-induced thioflavin T assay. Both compounds have shown excellent BBB permeation via PAMPA-BBB assay and were found to be non-neurotoxic at 80 µM concentration against differentiated SH-SY5Y cell lines. Compound PD-22 demonstrated an increase in rescued eye phenotype in Aβ-phenotypic drosophila AD model and amelioration of behavioral deficits in the Aβ-induced rat model of AD. The in-silico docking studies of compound PD-22 revealed a good binding profile towards CAS and PAS residues of AChE and the catalytic dyad of the BACE-1. The 100 ns molecular dynamics simulation studies of compound PD-22 complexed with AChE and BACE-1 enzymes suggested stable ligand-protein complex throughout the simulation run. Based on our findings compound PD-22 could further be utilized as a lead to design a promising candidate for AD therapy.
Article
Central nervous system (CNS) disorders, including brain tumor, ischemic stroke, Alzheimer's disease, and Parkinson's disease, threaten human health. And the existence of the blood-brain barrier (BBB) hinders the delivery of drugs and the design of drug targeting delivery vehicles. Over the past decades, great interest has been given to cell membrane-based biomimetic vehicles since the rise of targeting drug delivery systems and biomimetic nanotechnology. Cell membranes are regarded as natural multifunction biomaterials, and provide potential for targeting delivery design and modification. Cell membrane-based biomimetic vehicles appear timely with the participation of cell membranes and nanoparticles, and raises new lights for BBB recognition and transport, and effective therapy with its biological multifunction and high biocompatibility. This review summarizes existing challenges in CNS target delivery and recent advances of different kinds of cell membrane-based biomimetic vehicles for effective CNS target delivery, and deliberates the BBB targeting mechanism. It also discusses the challenges and possibility of clinical translation, and presents new insights for development.
Article
This review covers nanotherapeutic strategies for solving the global problems associated with Alzheimer's disease (AD). The most dramatic factor contributing humanistic, social and economic urgency of the situation is the incurability of the disease, with the drug intervention addressing only AD symptoms and retarding their progress. Key sources behind these challenges are the inability of the early diagnosis of AD, the lack of comprehensive information on the molecular mechanism of the pathogenesis, the blood-brain barrier obstacles, and the insufficient effectiveness of currently available drugs and therapeutic strategies. The application of nanocarriers allows part of these problems to be solved, together with the improvement of drug bioavailability, prolonged circulation, and overcoming/bypassing the biological barriers. To this date, numerous types and sub-types of nanocarriers are developed and reviewed, the majority of which can be adapted for the treatment of various diseases. Therefore, herein, nanotherapy strategies are specifically categorized in term of the administration routes of AD medicines, with the noninvasive, i.e., transdermal, oral, and intranasal routes emphasized. Further, benefits/limitations of various nanocarriers are discussed, and perspectives of their application are highlighted.
Article
Full-text available
Neurological disorders are a leading cause of morbidity worldwide, giving rise to a growing need to develop treatments to revert their symptoms. This review highlights the great potential of recent advances in cell therapy for the treatment of neurological disorders. Through the administration of pluripotent or stem cells, this novel therapy may promote neuroprotection, neuroplasticity, and neuroregeneration in lesion areas. The review also addresses the administration of these therapeutic molecules by the intranasal route, a promising, non-conventional route that allows for direct access to the central nervous system without crossing the blood–brain barrier, avoiding potential adverse reactions and enabling the administration of large quantities of therapeutic molecules to the brain. Finally, we focus on the need to use biomaterials, which play an important role as nutrient carriers, scaffolds, and immune modulators in the administration of non-autologous cells. Little research has been conducted into the integration of biomaterials alongside intranasally administered cell therapy, a highly promising approach for the treatment of neurological disorders.
Article
Full-text available
A freshwater microalga strain of Chlorella vulgaris was used to investigate toxic effects induced by nickel oxide nanoparticles (NiO-NPs) in suspension. Algal cells were exposed during 96 h to 0–100 mg L ⁻¹ of NiO-NPs and analyzed by flow cytometry. Physicochemical characterization of nanoparticles in tested media showed a soluble fraction (free Ni ²⁺ ) of only 6.42% for 100 mg L ⁻¹ of NiO-NPs, indicating the low solubility capacity of these NPs. Toxicity analysis showed cellular alterations which were related to NiO-NPs concentration, such as inhibition in cell division (relative cell size and granularity), deterioration of the photosynthetic apparatus (chlorophyll synthesis and photochemical reactions of photosynthesis), and oxidative stress (ROS production). The change in cellular viability demonstrated to be a very sensitive biomarker of NiO-NPs toxicity with EC 50 of 13.7 mg L ⁻¹ . Analysis by TEM and X-ray confirmed that NiO-NPs were able to cross biological membranes and to accumulate inside algal cells. Therefore, this study provides a characterization of both physicochemical and toxicological properties of NiO-NPs suspensions in tested media. The use of the freshwater strain of C. vulgaris demonstrated to be a sensitive bioindicator of NiO-NPs toxicity on the viability of green algae.
Article
Full-text available
Statement of significance: Detection and diagnosis of tumors at early stage are critical for the improvement of the survival rate of cancer patients. However, the challenge remains when the tumor specific markers are unto be determined, especially in early tumor detection. pH low insertion peptide (pHLIP) has been used as a specific ligand to target the tumor acidic microenvironment for tumors at early and metastatic stages. Superparamagnetic iron nanoparticles (SPION) are contrast enhancing agents used in the noninvasive magnetic resonance imaging for tumors. This research has demonstrated that pH-responsive pHLIP nanoclusters of SPION were able to target different tumors and facilitate the noninvasive diagnosis of tumors by MRI.
Book
The fascination with gold is a story which spans millennia, however scientists have recently found a new interest for gold when it is divided into miniscule grains, such as gold nanoparticles. This scientific enthusiasm started in various fields of science in the middle of the 1980s and the present book offers a panorama of the major scientific achievements obtained with gold nanoparticles. Various topics are reviewed such as: gold nanoparticle preparation methods, their plasmon resonance and thermo-optical properties, their catalytic properties, their use in biology and medicine as well as their possible toxicity and, finally, their future technological applications. The book also contains an in-depth study of the use of gold nanoparticles throughout the ages, starting from times where the concept of nanoparticles was beyond the realm of human imagination. All these topics are presented by world-class scientists within a set of self-contained chapters. This book may be used as an advanced textbook by graduate students and scientists who need an introduction to gold nanoparticles. It is also suitable for experts in the related areas of chemistry, biology, material science, optics and physics, who are interested in broadening their knowledge and who wish to have an overview of the subject. Each chapter gradually leads the reader from the basics of a topic towards some of the current scientific challenges in the area. The necessary background material to achieve a solid understanding of each topic and the scientific literature to go further in the field is provided.
Article
Ocular discomforts involve anterior/posterior-segment diseases, symptomatic distress and associated inflammations and severe retinal disorders. Conventionally, the formulations such as eye drops, eye solutions, eye ointments and lotions, etc. were used as modalities to attain relief from such ocular discomforts. However, eye allows limited access to these traditional formulations due to its unique anatomical structure and dynamic ocular environment and therefore calls for improvement in disease intervention. To address these challenges, development of nanotechnology based nanomedicines and novel nanosystems (liposomes, cubosomes, polymeric and lipidic nanoparticles, nanoemulsions, spanlastics and nano micelles) are currently in progress (some of them are already marketed such as Eye-logic liposomal eye spray@Naturalife, Ireland). Today, it is one of the central concept in designing more accessible formulations for deeper segments of the eyes. These nanosystems has largely enabled the availability of medicaments at required site in a required concentration without inversely affecting the eye tissues; and therefore, attaining the excessive considerations from the formulation scientists and pharmacologists worldwide. The entrapment of drugs, genes, and proteins inside these novel systems is the basis that works at the bio-molecular level bestows greater potential to eradicate disease causatives. In this review, we highlighted the recent attempts of nanotechnology-based systems for treating and managing various ocular ailments. The progress described herein may pave the way to new, highly effective and vital ocular nanosystems.
Chapter
Carbon nanotubes (CNTs) are the novel nanocarrier system for the delivery of drugs and other biomolecules of clinical importance. Many researchers credited CNTs mechanical, chemical, biological and optical properties for the successful transformation of biomolecules inside the target sites or sites of desire. Moreover, the dimension of this novel delivery system that ranging from 1-100 nm should also be credited for providing excellent surface properties. The advent of CNTs will provide the opportunity to encapsulate and target the drug with higher stability (since the stability is the problem with many newly discovered delivery systems) than the available systems and can revolutionize the way we will detect and treat the human diseases in the future. Today, many of the variants of CNTs like single walled and multi walled are under clinical investigations or under research and developmental stage. In this chapter we tried to better explain the synthesis, properties, functionalization, regulatory aspects and biomedical applications of CNTs. The basic objective of this chapter is to discuss various aspects of CNTs to provide complete understanding about the many undiscovered potential applications of CNTs.
Chapter
A technology of 21st century, nanotechnology has the potential to transform almost every area of applications right from making a needle to robot. It is basically involves manipulating physical, chemical, biological and optical properties at the nanoscale, often involving dimensions that ranging from 1-100 nm. In the field of medicine, it has already moved from being used in passive structures to active structures, through target specific delivery systems like dendrimers, liposomes, quantum dots, polymeric nanoparticles and metallic nanoparticles. These novel delivery systems have already been shown to be more effective than conventional delivery tools and also reported to have fewer side effects. The use of nanotechnology in the field of medicine could revolutionize the way we detect and treat the human diseases in the future. Today, many of these novel nanoscale delivery devices are under clinical practice or under research and developmental stage. The number of products approaching the approval and review process is likely to grow as time moves forward and as new nanotechnology medical applications are developed. Notably, the National Cancer Institute (NCI) and European medical agencies (EMA) has signed the alliance to develop nanodevice technology for early detection, diagnosis, and treatment of various diseases like cancer, HIV, cardiac, respiratory and depression. This basic objective of this chapter is to discuss various nanotechnology based approaches that have attained significant attention in the biomedical field.
Chapter
Most of the biologically active compounds are struggle with low solubility and therefore low bioavailability, biological degradation and inadvertent intrinsic side effects. To overcome such difficulties, the emergence of potential and novel drug carrier system is of supreme importance in terms of their efficient applicability through different routes of administration like skin, oral, topical, parenteral and pulmonary. Considering this, Solid lipid nanoparticles have attracted huge attention for the successful delivery of drugs and genes and also employed as an effective strategy for targeting therapeutics. The potential advantages of solid lipid nanoparticles over other nanoparticles are because of their high biocompatibility, higher drug loading capacity and scalability. In this chapter, we summarize the complete understanding about morphology, methodology, characterization, hybridization and biomedical applications of solid lipid nanoparticles.
Article
Background: Disseminated metastatic cancer requires insistent management owing to its reduced responsiveness for chemotherapeutic agents, toxicity to normal cells consequently lower survival rate and hampered quality of life of patients. Methods: Dendrimer mediated cancer therapy is advantageous over conventional chemotherapy, radiotherapy and surgical resection due to reduced systemic toxicity, and molecular level cell injury to cancerous mass, for an appreciable survival of the subject. Recently used dendrimer mediated nanotechnology for oncology aims to conquer these challenges. Dendrimers based nano-constructs are having architectures comparable to that of biological vesicles present in the human body. Results: Operating with dendrimer technology, proffers the exclusive and novel strategies with numerous applications in cancer management involving diagnostics, therapeutics, imaging, and prognostics by sub-molecular interactions. Dendrimers are designed to acquire the benefits of the malignant tumor morphology and characteristics, i.e. leaky vasculature of tumor, expression of specific cell surface antigen, and rapid proliferation. Conclusion: Dendrimers mediated targeted therapy recommends innovatory function equally in diagnostics (imaging, immune-detection) as well as chemotherapy. Currently, dendrimers as nanomedicine has offered a strong assurance and advancement in drastically varying approaches towards cancer imaging and treatment. The present review discusses different approaches for cancer diagnosis and treatment such as, targeted and control therapy, photodynamic therapy, photo-thermal therapy, gene therapy, antiangiogenics therapy, radiotherapy etc.
Article
Systemically administered chemotherapeutic drugs are often ineffective in the treatment of invasive brain tumors due to poor therapeutic index. Within gliomas, despite the presence of heterogeneously leaky microvessels, dense extracellular matrix and high interstitial pressure generate a “blood-tumor barrier” (BTB), which inhibits drug delivery and distribution. Meanwhile, beyond the contrast MRI-enhancing edge of the tumor, invasive cancer cells are protected by the intact blood-brain barrier (BBB). Here, we tested whether brain-penetrating nanoparticles (BPN) that possess dense surface coatings of polyethylene glycol (PEG) and are loaded with cisplatin (CDDP) could be delivered across both the blood-tumor and blood-brain barriers with MR image-guided focused ultrasound (MRgFUS), and whether this treatment could control glioma growth and invasiveness. To this end, we first established that MRgFUS is capable of significantly enhancing the delivery of ~ 60 nm fluorescent tracer BPN across the blood-tumor barrier in both the 9 L (6-fold improvement) gliosarcoma and invasive F98 (28-fold improvement) glioma models. Importantly, BPN delivery across the intact BBB, just beyond the tumor edge, was also markedly increased in both tumor models. We then showed that a CDDP loaded BPN formulation (CDDP-BPN), composed of a blend of polyaspartic acid (PAA) and heavily PEGylated polyaspartic acid (PAA-PEG), was highly stable, provided extended drug release, and was effective against F98 cells in vitro. These CDDP-BPN were delivered from the systemic circulation into orthotopic F98 gliomas using MRgFUS, where they elicited a significant reduction in tumor invasiveness and growth, as well as improved animal survival. We conclude that this therapy may offer a powerful new approach for the treatment invasive gliomas, particularly for preventing and controlling recurrence.
Article
The presence of cell and tissue barriers together with the low biomembrane permeability of various therapeutics often hampers systemic drug distribution; thus, most of the available molecules are of limited therapeutic value. Opportunities to increase medicament concentrations in areas that are difficult to access now exist with the advent of cell-penetrating peptides (CPPs), which can transport into the cell a wide variety of biologically active conjugates (cargoes). Numerous preclinical evaluations with CPP-derived therapeutics have provided promising results in various disease models that, in some cases, prompted clinical trials. The outcome of these investigations has thus opened new perspectives for CPP application in the development of unprecedented human therapies that are well tolerated and directed to intracellular targets.