Glyconanoparticles Activity 5

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 19

Seminars in Cancer Biology 69 (2021) 24–42

Contents lists available at ScienceDirect

Seminars in Cancer Biology


journal homepage: www.elsevier.com/locate/semcancer

Review

Glyco-nanoparticles: New drug delivery systems in cancer therapy


Haroon Khan a, *, Hamid Reza Mirzaei b, Atefeh Amiri c, Esra Kupeli Akkol d,
Syed Muhammad Ashhad Halimi e, Hamed Mirzaei f, *
a
Department of Pharmacy, Abdul Wali Khan University, Mardan, 23200, Pakistan
b
Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran. Iran
c
Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
d
Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, 06330, Etiler, Ankara, Turkey
e
Department of Pharmacy, University of Peshawar, 21200, Pakistan
f
Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran

A R T I C L E I N F O A B S T R A C T

Keywords: Cancer is known as one of the most common diseases that are associated with high mobility and mortality in the
Glyco-nanoparticles world. Despite several efforts, current cancer treatment modalities often are highly toxic and lack efficacy and
Cancer specificity. However, the application of nanotechnology has led to the development of effective nanosized drug
Drug delivery system
delivery systems which are highly selective for tumors and allow a slow release of active anticancer agents.
Different Nanoparticles (NPs) such as the silicon-based nano-materials, polymers, liposomes and metal NPs have
been designed to deliver anti-cancer drugs to tumor sites. Among different drug delivery systems, carbohydrate-
functionalized nanomaterials, specially based on their multi-valent binding capacities and desirable bio-
compatibility, have attracted considerable attention as an excellent candidate for controlled release of thera­
peutic agents. In addition, these carbohydrate functionalized nano-carriers are more compatible with con­
struction of the intracellular delivery platforms like the carbohydrate-modified metal NPs, quantum dots, and
magnetic nano-materials. In this review, we discuss recent research in the field of multifunctional glycol-
nanoparticles (GNPs) intended for cancer drug delivery applications.

1. Introduction among the prominent ingredients of the living organisms. There is suf­
ficient knowledge of the structures, interactions, and functions of the
It is commonly accepted that cancer is an important public health nucleic acid and protein. Nevertheless, there is not enough knowledge
problem with a high mortality throughout the world [1]. In spite of the about the contribution of carbohydrate to the cells. In fact, a dense
broad used of common chemotherapeutic agents for treating cancer, coating of carbohydrate, known as glycocalyx, covers the the surface of
these agents have some shortcomings, including inadequate tumor tis­ mammalian cells [15]. Another study showed that carbohydrate
sues recognition leading to damage to the healthy tissues and low appeared basically conjugated to the lipids and proteins (i.e., glyco­
bio-compatibility caused by their hydrophobicity, which results in the protein, glycolipid, & proteoglycan) in the glycocalyx. Indeed, it acts as
aggregation [2,3]. Hence, researchers should necessarily provide effi­ a glycoconjugate, which develops its biological functions. We know that
cient therapeutic tools for recognizing the attributes of the healthy cells such complex oligosaccharides play a role in controlling several normal
and cancer cells in the clinical contexts, and concurrently target the and pathological procedures [16]. However, multivalent ligands
cancerous tissues. Notably, clinics need intelligent, sensible, and compensate for the excessive low affinities of the carbohydrates to the
particular nano-carrier systems for the targeted delivery of the chemo­ biological objects. Therefore, we designed a novel integrated strategy
therapeutic agents [3–8]. Some studies have addressed a number of called glyco-nanotechnology strategy 4 for examining and interfering in
nano-carrier which could employed as effective delivery systems such as the carbohydrate to protein-mediated and carbohydrate to carbohydrate
the polymeric NPs, dendrimers micelles, and liposomes, for optimizing interactions. It is notable that glycol-nanotechnology is one of the
the treatment regimens for cancer [9–14]. strategies to tailor the sugar bio-functional gold nano-clusters (glyco­
According to research, carbohydrate, nucleic acid, and protein are l-nanoparticles or GNPs) in a convenient and flexible manner [17–19].

* Corresponding authors.
E-mail addresses: haroonkhan@awkum.edu.pk (H. Khan), Mirzaei-h@kaums.ac.ir (H. Mirzaei).

https://doi.org/10.1016/j.semcancer.2019.12.004
Received 3 October 2019; Received in revised form 28 November 2019; Accepted 2 December 2019
Available online 20 December 2019
1044-579X/© 2019 Elsevier Ltd. All rights reserved.
H. Khan et al. Seminars in Cancer Biology 69 (2021) 24–42

Actually, GNPs present a glycocalix-like surface with multi-valent car­ Based on the aforesaid discussion, nanotechnology would have a lot of
bohydrate representation and globular form [18]. The GNPs have a se­ usages in the health care management (Fig. 1). Over the last twenty
ries of benefits in comparison to the formerly procured colloids that are years, FDA confirmed multiple nano-medicines to treat cancer, diabetes,
simplified procurement and treatment, exceptional little core dimension hepatitis, autoimmune diseases, cardio-vascular illnesses, neurological
and narrow distribution size, control over the ligand numbers and NPs diseases, the increased cholesterol, Parkinson’s, and specific infectious
sizes, water solubility, higher storage stability with no flocculation, illnesses (Tables 1, 2) [34]. Hundreds of nano-carrier-based products
singular physical features, and lack of cyto-toxicity [20]. Herein, we have been recently provided at different phases of pre-clinical and
summarize the GNPs used in treatment of cancer. clinical developments [35]. Hence, it seems that developing nano-based
therapies open new horizons in the medical field.
2. Nanotherapeutics: a new way for treatment
3. Advantage of nano-technology for cancer
Nano-therapeutics is one of the current applications of nanotech­
nology with a great effect on the medical fields [21–26]. It is notable that biological processes such as the procedures essential
Nano-technology is the source of nano-drug [27]. Nanomedicine is a for life and the ones, which result in the cancer, take place at the nano-
relatively developing area of science which dates back to 1959 accord­ scale. Actually, the human’s body consists of multiple biological nano-
ing to the predictions of Richard P. Feynman [28]. According to the machines [49]. In fact, nanotechnology offers opportunities for study­
studies, a nano-meter is 1 millionth part of a milli-meter, and the term ing and manipulating the macro-molecules in the real time and over the
nano refers to the dwarf [29]. Therefore, nanotechnology addresses initial phases of cancer development [50]. Nanotechnology is able to
examination, control, and modifications of the molecular and/or atomic detect rapidly and sensitively the cancer-associated molecules that en­
structures in a range from 1 to 100 nm in size. It should be noted that the ables the scientists for detecting molecular modifications even when
nano-drug branch is using diverse nanotechnological strategies such as they take place just in a small percentage of cells [49,51–53]. Moreover,
nano-biosensors and biological instruments [21]. Notably, the quantum it enjoys potentials for generating thoroughly new and strongly efficient
impacts the nano level affect the chemical, biological, optical physical, treatment factors. Eventually, using the nano-scale materials for cancer
and mechanical features that allow the experts in the field to utilize the results from their capability to be easily functionalized and readily
advantages of these phenomena [30]. Moreover, nano drug involves adjusted; their capability for delivering and functioning as diagnostic
novel notions and uses of the molecular nano technology in order to and therapeutic agents; and their capability for passively accumulating
design the nano-machines termed nano-robots. In addition, basic at the tumor area in order to have active targeting to the cancer cells and
working power of the nano-structures is provided by biological delivering through the conventional biological obstacles in the body like
macro-molecules and structures or xenobiotic chemical medicines. the blood-brain barrier (BBB) or pancreas dense stromal tissue, which
However, the most important reality of the nano-materials refers to their largely modulates delivering the bio-molecules to create our central
size that is the same as numerous biological macro-molecules that fa­ nervous system [49]. In the below, we highlighted different advantages
cilitates the application of the nano-materials in-vivo and in-vitro. which are associated with nano-based therapies in the treatment of
Therefore, multiple diagnostic kits, analytical devices, physio-therapy cancer.
uses, and drug delivery systems have been presented so far by amal­
gamating the nano-technology with the biological materials. Thus, as a 3.1. Passive tumor accumulation
branch of medicine, nano-therapeutic reserves numerous studies and
advancements. In spite of the traditional techniques of medicine, ac­ According to recent studies, an efficient cancer medicine delivery
cording to the new method, the medicine would bind on NPs that must gain strong accumulation in the environment around a tumor and
empower it for acting with higher efficiency and accuracy and fewer not any adverse effects on the surrounding normal cells [54]. It has been
consequences. As demonstrated by researchers, nano-therapeutics offers found that passive localization of numerous medicines and medicine
novel opportunities for enhancing safe and efficient traditional treat­ carriers because of the respective extra-vasation via enhanced perme­
ments [31]. For this reason, diverse international and domestic orga­ ability and retention (EPR) effect that is a leaky vasculature, has an
nizations and pharmaceutical firms have investment on this area for extremely good performance for tumors [55,56]. With the rapid growth
generating novel in-vivo imaging methods, gene treatment, and drug of the tumor mass, a network of the blood vessels should be expanded
delivery systems. It should be mentioned that the sale rate of the rapidly for accommodating the needs of the tumor cells for oxygen and
nano-drug has been $16 just in nanotechnology. Given that the nutrients [54]. It should be noted that such an abnormal and weakly
nano-medicine products are expected to obtain 350.8 billion dollars by modulated vessel formations, i.e. angiogenesis, would lead to the ves­
2025 which this suggests its considerable impacts on the universal sel’s walls with big pores. It has been indicated that the leaky vessels
economies [21]. Presently, those involved in the healthcare industries would permit the comparatively great NPs for extravasating into the
are attempting to achieve greater productivity, accessibility, and ther­ tumor masses. Since the rapid developing tumor mass lacks a functional
apeutic quality at less expenditure. Of course, one of the challenges for lymphatic system, clearing such NPs would be limited and additionally
the healthcare experts has been the permanent and severe neurological improves the accumulation. Using the EPR effect, the NPs greater than
dysfunctions like cancer, HIV or AIDS, diabetes, and heart diseases. One 8 nm may passively select the cancer cells via free passage across the
of the other challenges is the infectious illnesses, in which traditional great pores and attain greater intra-tumoral accumulation (Fig. 2) [57].
anti-microbial factors applied for their healing enhance reverse conse­ Most recent nano-medicines for treating the solid tumor rely on the EPR
quences and higher resistance to the drugs. However, targeted speci­ effect for ensuring higher medicine accumulations and improvement of
ficity is a key barrier to achieve the treatment efficacy [21]. It has been the therapeutic efficiency. In fact, in the case of the absence of the tar­
approved that NPs are reasonable and motivating means to deliver the geting cell kinds, which express the targeting interest ligand, this drug
medicines in a controlled and targeted manner. In addition, nano-drugs delivery system would be named the passive targeting [54]. Prior to
contribute significantly to resolving these as nanotechnology based reaching the adjacency of the tumor site for the passive targeting to
formulations would improve the pharmaco-kinetic traits, occur, the EPR effect needs the drug delivery systems to have a lengthy
bio-availability, and medicine targeting in multiple dysfunctions [21]. circulation for allowing adequate levels of drugs to the target areas. For
Some researchers believe that nano-drug can prevent and treat illnesses, designing nano-medicines, which are able to stay in the blood longer, we
and also enjoys potent utilizations in diagnosing, monitoring treating, can mask such nano-medicines via modification of the surface with
discovering drugs, performing surgical operation, and doing gene de­ using different polymeric materials such as poly-ethylene glycol (PEG).
livery through molecular information of the human systems [32,33]. Actually, PEG is frequently employed for making the water-insoluble

25
H. Khan et al. Seminars in Cancer Biology 69 (2021) 24–42

Fig. 1. Various Nano-therapeutics in Healthcare.

more rapid uptakes of the NPs with a negative charge, the


Table 1
nano-medicines containing a positive or neutral charge would be esti­
Selected nano-materials in cancer therapy.
mated to possess a more prolonged plasma half-life [58]. Using the EPR
Type of Nano- Cancer Outcome Reference effect for the passive tumor targeting drug delivery would have its’ own
material
problems and issues. Even though the EPR effect would be one of the
Nano-capsules Colon Enhancement of [36] specific phenomena in solid tumors, the greater tumor mass and central
therapeutic efficiency of area of the metastasis would not show the EPR effect due to an extreme
drug
Lipid nano-capsules Melanoma, Improve anticancer [37,38]
hypoxic condition. Therefore, some techniques are employed in clinical
Ovarian cancer potential of drug, Effective settings for artificially enhancing the EPR effect: gradual angiotensin II
in lung cancer therapy infusion for increasing the systolic blood pressure, topical utilization of
Polyamidoamine Hepatic cancer Enhance treatment of [39] the NO-releasing factors for expanding the blood and photo-dynamic
dendrimers hepatic cancer
treatment or hyperthermia-mediated vascular per-meabilization in the
Modified solid lipid Cervical cancer Synergistic anticancer [40]
nanoparticles effect on cervical cancer solid tumors [58]. It should be noted that the passive accumulation via
Hyaluronan solid Ovarian cancer Improve paclitaxel [38] the EPR impact is very important reasonable drug delivery system for
nanoemulsions targeting solid tumors therapy. Nonetheless, the sizes or the molecular weights of
Cyclodextrin- Prostate cancer Improve delivery of the [41] NPs is not just a determining factor of the EPR effect, but also another
nanosponges drug
Conjugated Pancreatic Improve efficacy of drug [42]
factor, including the surface charge, bio-compatibility, and in vivo sur­
Dendrimers cancer veillance system for macro-molecules must be considered when devel­
GNPs GBM Anti-cancer effects [43] oping a nano-medicine for effective passive tumor accumulation [57].
Chitosan based NPs Liver cancer Improve efficacy of drug [44]
Mesoporous nano- – Improve efficacy of drug [45]
silica MCM-41 3.2. Active tumor targeting
Nano-Fe3O4/CA Breast cancer Induce apoptosis [46]
Nano-magnetic pancreatic Improve efficacy of drug, [47]
formulation malignancy and anti-cancer effects
It is usually accepted that the EPR effect that is used as a particle
Nano-graphene breast cancer Improve efficacy of drug, [48] "passive tumor targeting" scheme has control over the particles in the
oxide and anti-cancer effects tumor area. Nevertheless, the EPR effect would not result in higher
uptakes of NPs into the cells, but it is necessary to internalize the NPs or
drugs cell for a number of the therapeutic modalities depending on the
NPs in order to be water-soluble in several preclinical study laboratories.
medicine activations into the cell nucleus or cytosol [60]. In a similar
The PEG-coated liposomal doxorubicin (Doxil) would be applied in the
way, delivering the nucleic acids (siRNA, DNA, & miRNA) in the genetic
clinics for the breast cancer holding passive tumor accumulations. Since
treatments would require the escape of such molecules from the endo­
the in vivo surveillance platforms for macro-molecules (e.g., scavenger
some; therefore, these can achieve favorable sub-cellular compartments.
receptors in the reticuloendothelial system) are apparently indicated
Additionally, the EPR is heterogeneous and its strength is variable

26
H. Khan et al. Seminars in Cancer Biology 69 (2021) 24–42

Table 2
FDA-approved nano-medicines.
Year of Disease Drug Component Nanoparticle benefits
approval

2000 Iron deficiency in chronic kidney Venofer® Iron sucrose Permits improved dose
disease
2003 Menopausal therapy Estrasorb™ Micellar Estradiol Controlled delivery of therapeutic
2005 Breast cancer Abraxane®/ABI-007 Albumin-bound paclitaxelnanoparticles Enhanced solubility; Enhanced delivery
to
tumor bioavailability; Increased release
2007 Anemia associated with Mircera® or Methoxy Chemically synthesized Elevated aptamers stability as a result of
chronic kidney disease PEGepoetin erythrocyteinducing PEGylation
agent
2008 Imaging agent Feridex®/Endorem® SPION-dex Superparamagnetic character
2008 multiple myeloma Doxil®/Caelyx™ (Janssen) Liposomal doxorubicin Elevated delivery to tumor site; lowered
systemic toxicity which arises from side-
effects
2008 Crohn’s disease Cimzia® Antibody conjugate Improve efficacy of drug
2009 Rheumatoid arthritis Cimzia® Antibody conjugate Improve efficacy of drug
2009 Imaging agent GastroMARK™; umirem® SPION-silicone Super paramagnetic character
2009 Deficiency anemia iron deficiency in Feraheme™ SPION withpolyglucose Magnetite suspension contributes to
chronic sorbitolcarboxymethylether prolonged
kidney disease steady release and decreases dose
numbers
2009 Schizophrenia Invega® Paliperidone Improve efficacy of drug
2009 Bone substitute EquivaBone® Hydroxyapatite Mimics bone structure
2010 Chronic gout Krystexxa PEGylated PEGylated
porcine-like uricase porcine-like uricase
2012 Non-small-cell lung carcinoma Abraxane®/ABI-007 Albumin-bound paclitaxel NPs Improve efficacy of drug
2012 Acute Lymphoblastic Marqibo® Liposomal Vincristine Improve efficacy of drug
Leukemia
2013 Pancreatic cancer Abraxane®/ABI-007 Albumin-bound paclitaxel nanoparticles Improve efficacy of drug
2013 Psoriatic Arthritis Cimzia® Antibody conjugate Improve efficacy of drug
2013 Ankylosing Spondylitis Cimzia® Antibody conjugate Improve efficacy of drug
2014 Malignant hypothermia Ryanodex® Dantrolene sodium Improve efficacy of drug
2014 Multple Sclerosis Plegridy® PEGylated IFN Improve efficacy of drug
beta-1a
2015 Hemophilia Adynovate Polymer-protein conjugate Improve efficacy of drug
2015 Pancreatic Cancer Onivyde® Liposomal Irinotecan Improve efficacy of drug

amongst various tumors and or patients. Thus, the active targeting 3.3. Transport across the tissue obstacles
would be regarded as a crucial trait for the next generation NPs drugs
(Fig. 2) [60]. It should be noted that this would empower specific mo­ NPs or nano-medicine delivery would be impeded by tissue obstacles
dalities of treatments which could not be achieved with the EPR and prior to the medicine and reach the tumor region [63]. It has been
enhance the therapeutic efficacy that may be done through the EPR, but revealed that there are a variety of tissue barriers for obtaining an
with lower acceptable impacts [61]. Based on the results, it is possible to effective transfer of the NPs to the tumor microenvironment such as
attain NPs’ active targeting to the tumor cell, micro-environment or biological obstacles, tumor endothelium obstacles, and the functional
vasculature and the directed delivery to intra-cellular compartments by obstacles. It should be mentioned that the biological obstacles are the
changing the NPs surface with fewer molecules, anti-bodies, affi-bodies, physical constructs or cell formations restricting the NPs motions.
peptides and aptamers [61]. Moreover, the EPR effects is a path for NPs’ Moreover, the functional obstacles may have an effect on the transfer of
extravasating from the circulation to tumor microenvironment via using the intact NPs or nano-medicine within the tumor masses, higher
leaky vasculature tumors. Then, the therapeutic agents which are car­ interstitial fluid pressure, and acidic environments. Thus, designing NPs
ried by NPs could be released in an extra-cellular matrix and diffuse and approaches for resolving such obstacles would be of high impor­
across the tumor tissues [62]. Afterwards, the NPs would transfer a tance for improving the cancer therapeutic efficiency [64].
variety of surface ligands which are associated with the active targeting According to the studies, the tumor micro-environment (TME) is one
of the NPs to the target cells or tissues. However, it is estimated that the of the dynamic systems consisting of irregular vasculature, fibro-blasts,
active targeting enhances the accumulations of NPs or medicines in the and immune cells so that each of them has been placed in an extra-
tumor and promotes their prospective cell uptake via the cellular matrix (ECM). In fact, TME creates the two biological and
receptor-induced endocytosis. In addition, particles engineered for functional obstacles to the nano-medicine delivery in treating cancers
vascular targeting would combine the ligands binding to the endothelial [65]. Notably, the abnormal vasculature and increased level of the cell
cell-surface receptors [62]. According to the predictions, vascular tar­ density would affect on the interstitial fluid pressure into tumor
geting provides synergistic approach that uses the targeting of the microenvironment. Consequently, this pressure gradient would be un­
vascular tissue and cells into the illed tissues. It should be mentioned desirable for freely diffusing the NPs and would be frequently a limita­
that a majority of the nanotechnology-based approaches that have been tion for greater EPR effects. It should be noted that as the numbers of the
confirmed to be used clinically or have been in the developed clinical tumor masses reaches 106 cells, the metabolic strains would ensue.
trials rely e on the EPR effects. Therefore, the next generation Many times, the cells in the core of such a growing cluster have a dis­
nano-therapies would apply the targeting for enabling and enhancing tance of 100–200 u m from the nutrient source; that is, 200um would be
the intra-cellular uptakes, intra-cellular trafficking, and penetrating the a limiting distance for the oxygen diffusion. Therefore, in the core of
physiological obstacles that obstruct the medicine accessibility to a tumors, tumor cells have a hypoxic condition and could be lived at the
number of tumors [62]. 2.5–10 mmHg concentrations of pO2 [65]. In fact, the anoxic metabolic
pathways may kick in and produce lactic acid. NPs would have

27
H. Khan et al. Seminars in Cancer Biology 69 (2021) 24–42

Fig. 2. Passive vs. active targeting approaches for anti-cancer drug delivering systems (Top). Through enhancing the retention effects as well as permeability, the NPs
would diffuse via the leaky vasculature and aggregate in the tumor tissue. Therefore, it is possible to release the medicine in extra-cellular matrix and consequently
diffuse across the tissues (a). (Down) Considering the active targeting, when the particles have been extravasated in the tumor tissues, targeting ligands existence
(such as carbohydrate and anti-body on the NP surface contributes to their interactions with the receptors present on the tumor cell resulting in the greater ac­
cumulations and preferential cellular uptakes across the receptor-mediated endocytosis (b).

instability in the acidic environment. Thus, the medicine delivery to the endothelial cells sealed in the continual tight junction surrounding the
target tumor cells could not be anticipated. In addition, the tumor ECM capillaries. Indeed, the astrocytes cover outside the layer of the epithe­
would provide a nutrient for the stromal and cancer cells. Actually, it lial cells, which additionally play a role in the selectiveness of the pas­
would be a set of the fibrous proteins and poly-saccharides that would sage of materials. However, since the BBB keeps detrimental materials
expand quickly in the aggressive cancer as a result of fast growth of the from the brain, it constrains the drugs delivery for the brain illnesses like
stromal cells. Cancer stroma is known as a cancer with abnormal and the brain tumors and other neurological illnesses. Notably, a lot of
weakly functioning vasculature, modified extra-cellular matrix, rapid studies have been done for resolving the BBB to deliver drugs. Moreover,
growth of the fibroblasts and infiltration of the macrophages. Therefore, multivalent property of the NPs would make the nano-carriers inter­
both tumor-stroma interactions would increase the pancreatic tumor cell esting for developing the BBB-crossing delivering approaches. In addi­
invasions and metastasis; however, the tumor stroma and TME would tion, a hopeful NP design has transferrin receptor-targeting moiety for
provide an undesirable environment for delivering the drugs and other facilitating such NPs transfer through BBB [68].
kinds of the cancer therapies [66].
Since the EPR effect is one of the clinically related phenomena for 4. Nanoparticles and Cancer therapy
permeation of the nano-carriers tumor, some approaches have been
designed for addressing the tumor endothelium barriers [67]. Notably, Based on the research, traditional cancer chemotherapy possesses
the approaches for declining the interstitial fluid pressures for the cancer treatment factors, which distribute non-specifically in the
improvement of the tumor permeation are the ECM-targeting pharma­ humans bodies. Therefore, such medicines would have an effect on the
cological interventions for normalizing the vasculature into the TME, cancerous and normal cells. Such nonspecific distribution of the medi­
hypertonic solutions for shrinking the hyperthermia, ECM cells, cines would limit the drug dosage into the cancer cells while creating
high-intensity focused ultrasound (HIFU), or radiofrequency (RF) for very high toxicity to the normal cells, tissues, and parts of the body, so
enhancing the nanomedicine transfers and accumulations. Moreover, that it causes different reverse consequences such as hair loss, weakness,
such approaches diminish the hypoxic conditions in the greater tumor and organ dysfunctions, which result in lower quality of life for patients
masses [67]. Even though the tumor masses and TME provide a severe with cancer [69]. Some researchers believe that NPs have been signifi­
and an acidic environment for nano-based delivery system stabilization, cantly considered during the past ten years, because they would have a
the designing of pH-responsive nano-carrier could be useful. Multiple lot of advantages for drug delivery for overcoming the restrictions in
procedures that have been presented earlier would be applied for traditional chemotherapy [60,70]. In fact, NPs may be formed in
addressing the tumor stroma barriers [67]. different sizes ranging between 1 and 1000 nm. Moreover, they may
One of the other formidable tissue barriers for the medicines and NPs consist of different substances such as the polymers (bio-degradable
delivery is the BBB [68]. It should be noted that BBB is one of the polymeric NPs and dendrimers), lipids, (the solid-lipid NPs and lipo­
physical barriers in the central nervous system for preventing detri­ somes), in-organic substances (metal NPs and quantum dots), and bio­
mental materials from entering the brain. In fact, it contains the logical substances (the viral NPs and albumin NPs). Additionally, they

28
H. Khan et al. Seminars in Cancer Biology 69 (2021) 24–42

can be adapted for concurrently carrying the medicines and imaging noted that development of the novel substances, selection of the proper
probes and developed for specifically targeting the diseased tissues substances for all specific treatments, and other parameters should be
molecules [60]. As it is known, the NPs for the delivery of anticancer optimally chosen for designing more acceptable targeted NPs [76]. As
agents reached the 1st clinical trial in the mid-1980s. In fact, the 1st NPs, reported by researchers, the above parameters are the NPs’ dimension,
such as liposomal with encapsulated doxorubicin (DOX) entered the from, sedimentation, the medicine encapsulation efficiency, favorable
pharmaceutical markets in 1995. Ever since, multiple novel NPs for medicine release profiles, distribution in the body, circulation, and
cancer medicine delivery have been confirmed and/or are recently being costs. As an example, researchers demonstrated about the particles size
developed as a result of their numerous benefits [60]. Benefits of NPs are that the clearance rate of very little NPs could be high, and the majority
the higher solubility of the hydrophobic medicines, more lengthy cir­ of the NPs could finish in the liver and spleen and result in infeasible and
culation time, lower nonspecific uptakes, prevention of unfavorable inefficient utilization of the targeted NPs [76]. Put differently, greater
off-target and consequences, greater intra-cellular permeation, and NPs are possibly too large for going across the little capillaries for
more specific cancer-targeting. Some studies showed that the utilization delivering the drug [76]. Therefore, selection of the proper substances
of the targeted NPs for delivering chemotherapeutic factors in treating and particles size is one of the other significant aspects in the targeted
cancers would be accompanied by several benefits for improving the NPs for cancer therapy. Although researchers performed a lot of studies
medicine or gene delivery and overcoming multiple issues related to for developing novel targeted NPs, just a few of them are used clinically,
traditional chemotherapy [71,72]. As an instance, NPs through the such as the Abraxane®, Doxil®, and MyocetTM that are confirmed by
passive targeting or active targeting, would promote intra-cellular the FDA. One of the main accounts for gradual progression of the effi­
concentrations of the genes and therapeutic agents in the tumor cells cient targeted NPs results from the absence of information of distrib­
while stopping toxicity in the normal cells. Moreover, the functionalized uting and localizing the targeted NPs following the oral administrations
NPs may be developed as the temperature-sensitive or pH-sensitive or injections [77]. As an instance, the majority of the studies conducted
carriers. Furthermore, the pH-sensitive delivery system of drugs have not explored the NPs’ targeting efficacy in real time in-vivo.
possibly delivers and releases the medicines into more acidic Therefore, there are insufficient data on the accurate bio-distribution
micro-environment of the tumor cells and/or elements in the cancer and consequent treatment impacts. Hence, eradication of cancer (ma­
cells. Researchers showed that temperature-sensitive systems are lignant) cells in the body and control over the therapeutic impacts on the
capable of carrying and releasing medicines with modifications in the cells in real time would be one of the other challenges that should be
temperature locally in a tumor area supplied by resources like the ul­ eliminated for developing effective targeted NPs [78].
trasound waves, magnetic fields, and so forth in order to use the com­
bined therapies, including the chemotherapy and hyperthermia. 5. Glyco-nanoparticles
Notably, the NPs targeting to the tumors through the cancer-specific
properties and moieties declined the impacts of the compositions, Researchers extensively dealt with integrating nanotechnology with
sizes, and molecular masses of the NPs on their efficiency [73]. More­ carbohydrates over the past ten years. In fact, advancements in the
over, the targeted NPs are able to make additional modifications or glycol-nanotechnology resulted in creating diverse bio-active glycol-
functionalization for reducing toxicity. As an instance, modification of nanostructures for different health associated utilizations like the drug
NPs surface chemistry could decline their toxicity and immuno-toxicity delivery, bio-materials, gene therapies, pathogen detection, suppressors
[74]. Even though the targeted NPs are considered as one of the stra­ of toxins, and lectin-based bio-sensors [79–82]. Researchers indicated
tegies for overcoming the absence of the traditional chemotherapy that the NPs functionalized with the carbohydrates would provide a
specificity, probable hazards and challenges related to the new approach largely multi-valent technique of interactions with lectins and pro­
would be followed. However, a number of the cancer cell types would curement of the increased local ligands level on small surfaces. In fact,
enhance the medicine resistance to the medicine therapy period and researchers extremely paid attention to inserting their biological traits
render the medicines released from the targeted NPs to be inefficient. It into the nano-structured substances to the carbohydrates because of
should be noted that the combined treatments such as using the targeted their utilization in the biomimetic purposes that contribute crucially to
NPs to deliver the chemo-therapeutics and gene therapeutics are prob­ the biorecognition procedures at molecular levels and functionally to
ably delivered efficiently and particularly targeted to cancer cells and the living systems [83]. Indeed, their significant performance is to be
tissues for overcoming the drug resistance and stopping the tumor used as the recognition markers [84]. Even though there is a weak
development. One of the other strategies for overcoming this medicine binding between the carbohydrates and lectins, it is possible to enhance
resistance is the development of the multi-functional targeted NPs. it largely through the multivalent impact of densely packaged carbo­
Moreover, the targeted NPs for treating the cancer, as the other novel hydrate molecules with certain functionalities that is called glycocluster
technologies, would be followed by numerous challenges and issues. effect [85,86]. It should be mentioned that glycol NPs as the
One of the challenges of the targeted NPs is the probable alteration in the carbohydrate-based systems would work similarly for mimicking the
solubility, stability, and pharmaco-kinetic features of the medicines behaviors of the naturally available glycocalyx. Hence, making and
carried by the NPs. Nonetheless, researchers did not highly examine engineering the highly innovative glycol NPs with certain
such issues. In addition, shelf life, accumulation, leakage, and toxicity of physio-chemical features would assist the additional increase of their
the substances employed for making NPs are the other restrictions for specific recognition features on the multi-valent scaffolds in glyco­
their utilization. A number of substances applied for making the NPs like science. However, glycol-polymers are the synthetic macro-molecules
aspoly(lactic-co-glycolic acid)(PLGA) are of lower toxicity; however, containing sugar moiety and high potentials to mimic oligosaccharides
they degrade rapidly and would not circulate in the tissues long enough [87–89]. Based on the results, oligosaccharides contribute crucially to
for the prolonged medicine and gene delivery. In other words, additional multiple biological procedures like signal transmission, inter-cellular
substances like the carbon nano-tubes and quantum dots would have recognition, and fertilization [90–92]. Other studies have developed
higher durability, and could preserve in the body for weeks, months, or the self-assembled nano-products functionalized with the different type
even years; this makes them potentially poisonous and limits their uti­ of oligosaccharides for studying and evaluating the
lization for the iterated therapies [75]. In fact, the newly developed protein-carbohydrate and carbohydrate–carbohydrate interactions
substances for making the targeted NPs like silicon and silica, such as the [93]. Actually, conjugating glycol-polymers with bio-molecules,
solid, porous, and hollow silicon NPs, have been designed but their including ligands, phospholipids, lipids, deoxyribonucleic acid plas­
application for delivering the drug to the cancer patients has been taken mids, oligopeptides, and proteins has been done for yielding a big family
off gradually as a result of the probable health hazards related to the of the neo-glycoconjugates with the intended features to make the
introduction of novel substances in the humans bodies. It should be self-assembled GNPs [94]. In addition, the glycol-polymer as

29
H. Khan et al. Seminars in Cancer Biology 69 (2021) 24–42

nano-carriers has been procured in various kinds such the vesicles, mi­ glycopolymer–lectin interactions [98]. For example, Chen et al. showed
celles, NPs, and nano-fibers in order to deliver the bio-active molecules an approach that directed the functionalization of ZnS:Mn2+ and ZnS
like the folic acid, biotin–avidin, antibody, and DNA sequences. Based QDs containing chitosan. Researchers attained QDs functionalized by
on the new advancements in the conventional synthetic techniques of chitosan with a mean size of 4.5 nm when a mix of Zn(Ac)2, Mn(Ac)2,
the complicated glycol-conjugates, at present, it is feasible to provide chitosan, and Na2S2O3 aqueous solution was irradiated with a 1.1 × 1015
novel polyvalent systems that offer multi-valent carbohydrate–receptor Bq 60Co γ-ray source at the atmospheric pressure and room temperature
interactions [94]. In fact, multiple kinds of glycol NPs such as metallic, [99]. Finally, Surolia et al. could reveal that melibiose-QDs selectively
semi-conductor glycol-quantum dots, magnetic, and self-assembled bind to soybean agglutinin (SBA) and have been particulaly
glycol NPs are found [94].In the below, we highlighted different kinds de-agglutinated through α-galactose [100]. Furthermore, binding abil­
of GNPs as well as their application in cancer treatment. ities of the maltotriose-QDs with Con A have been analyzed by con­
trolling the light diffused at 600 nm (Fig. 3).
6. Types of glyco-nanoparticles
6.1.1. Metal glyco-nanoparticles
6.1. Glyco-quantum dots Researchers reported that magnetic NPs (MNPs), which may be
manipulated through an exterior magnetic field, was followed by novel
It is commonly agreed that the fluorescent semi-conductor nano- opportunities such as improving quality of the MRI, site-specific drug
crystals (also called quantum dots (QDs)) have been greatly considered deliveries, hyper-thermic treatments for tumor cells, and current studies
over the past ten years as a result of their specific size-dependent optical attention to the manipulation of the cell membranes. In addition, they
features [95]. It should be noted that quantum electronic and mechan­ are so potent that they can be employed for clinical diagnoses and
ical features of the substances have considerable differences from the treatments as a result of their specific physical traits. However, the
bulk solid features. In spite of the possible use of the optical imaging, the magnetic cores like the cobalt ferrites, iron oxide, manganese, and nickel
GNPs containing semi-conductor nano-crystals, the early hydrophobic may be functionalized with proper glycol-conjugates. Overall, co-
QDs indicated higher cytotoxicity and weaker solubility in various types precipitation, thermal decomposition, and microemulsion have been
of biological systems. In fact, different protection procedures must be considered as the most widely applied techniques of the preparation of
conducted for conjugating with antibodies, proteins, and DNA for the magnetic NPs. For example, Kasteren et al. utilized the cross-linked
conferring the water solubility and stability. Moreover, the glyco-QDs iron oxide (CLIO) amine-functionalized dextran-coated NPs as a ground
have been applied as a label for in vivo imaging concurrently. Notably, to combine several copies of sialyl LewisX [102]. Moreover, metal oxide
Fang et al. initially used the glyco-QDs as in vitro bio-labels [96]. In surfaces may be readily altered by chloro-, alkoxy-, and
addition, optical features of the mannose-encapsulated CdSe/ZnS core aminoalkyl-silanes for additional coupling of the bio-molecules. In fact,
and shell QDs have been specified by the confocal microscope imaging the silica-coated magnetite NPs functionalized with the α-d-mannosides
for staining the living cells. Based on the outputs extracted, in spite of have been attained by a triazole (Huisgen-type reaction) or an amide
the prevalence of the mannose-QDs over the whole sperm body, the (peptidic coupling) link. In addition, terminal alkynyl or carboxy func­
sperm heads have a high concentration of the GlcNAc-QDs because of tional groups entered the silica oxide-coated magnetite and azidopropyl-
diverse distributions of mannose receptor and GlcNAc on the sperm or aminopropyl-armed α-d-mannosides have been applied for uniform
surfaces. A study applied the chitosan-QD (CS-QD) hybrid nano-spheres orientation of carbohydrates at the surface of the NPs. It has been found
to bio-image and bio-label [97]. It is notable that a non-solvent-assisted that the binding efficiency with mannose-MGNPs has been greater than
counterion complexation technique has been used to prepare the hybrid the other magnetic functionalized particles containing lectins [103].
nanospheres where CS aqueous solution used for adding the glutaral­ Pieters and colleagues have recently revealed that magnetic NPs con­
dehyde aqueous solution to QDs. The above two research indicated that jugated to the mono-valent galabiose conjugates and tetravalent
QD proper systems may display powerful fluorescence emissions and galabiose-linked dendrimers through the biotin–streptavidin coupling
longer stability as compared to other imaging systems. However, Sun are probably employed for detection of Streptococcus suis [104]. The
et al. showed coupling the commercially available QD-streptavidin with magnetic NPs coated with the mono-valent galabiose conjugates had
a biotin end-terminated lactose glycol-polymer. Actually, the Ricinus greater interactions in comparison to the tetra-valent particles. It should
communis agglutinin (RCA120)-immobilized agarose beads approved for be noted that Davis group utilized the iron oxide NPs (IONPs) for
the fluorescent staining by Confocal microscopy because of functionalization of the 3 distinct glycol-polymers (α-D-mannose,

Fig. 3. Accumulation of the sugar-QDs via Con A tetramer.

30
H. Khan et al. Seminars in Cancer Biology 69 (2021) 24–42

α-D-glucose and β-D-glucose bearing glycol-polymers) by grafting to the effects for the cancer cells (K562) and higher and more specific binding
IONPs [105]. One of the studies in the field showed the abilities with Con A. Additionally, in vitro examination demonstrated
photo-chemically-induced coupling of the unmodified that the glycol-micelles are able to kill cancer cells under light irradia­
mono-saccharides over the actuated spindle-type hematite and spherical tion and in the light treatment length-dependent approach. Hence, this
magnetite NPs [106]. Initially, 4-azido-2,3,5,6-tetra-fluorobenzamido study is of high importance to develop the utilizations for the cancer
derivatives functionalized the iron oxide NPs by sonication and termi­ imaging and treatments [109].
nal phosphate groups have been coupled to the metal oxide surface of Reineke et al. synthesized the cationic poly(methacrylamido­
the NPs that generate the Fe–O–P structures. In the next stage, coupling trehalose) (poly(trehalose)) through the RAFT polymerization [110].
the D-mannose has been done to the functionalized NPs via They made the nano-complexes of the cationic polymers with siRNA for
photochemically-induced CH with no chemical derivatization of car­ developing the delivery system as an effective targeting system to GBM
bohydrates (Fig. 4). It has been found that the synthesized mannose cells. Reineke et al. also published the other flexible strategies for syn­
conjugated magnetic NPs had higher recognition abilities toward the thesizing the glycol-polymers via the condensation polymerization of
Con A. However, one of the newly designed strategies produced the esterified carbohydrates and diamines. Such glycopolymers created
glycol-polymer modified magnetic NPs via incorporating the polymer a nano-complex containing nucleic acid and have been applied to deliver
grafting from strategy with glycosylation through the click chemistry the genes [111,112]. For example, Zhou et al. used glycopolymers
[107,108]. In other words, the poly(N-carboxyanhydrides) has been grafted onto poly(l-lysine) (PLL) as gene delivery system [113].
synthesized with ring-opening polymerization in the presence of 3-ami­ Although PLL possesses considerable condensations potential with
nopropyl-triethoxysilane (APTS)-functionalized magnetic NPs which plasmid DNA because of the hydrophilic amino groups with positive
could lead to providing clickable alkyne groups. Notably, charge in water, it would not be generally selected as one of the gene
azide-functionalized galactose has been bound to such NPs for making delivery vectors due to higher cytotoxicity and lower transfection effi­
the glycol-MNPs through the Huisgen click reaction. cacy. Nonetheless, the PLL has been modified by the saccharide-with
polymers for reducing cytotoxicity and increasing transfection efficacy
6.2. Polymeric glycol-nanoparticles of the PLL. Indeed, glycopolymer modified PLL had less cytotoxicity in
comparison to the PLL based on the MTT assays in HepG2 and NIH3T3
Research showed that synthetic paths which were very new and cell lines. Outputs indicated that a lower substitution degree on the PLL
significant prompted the polymer chemists to preparation of the poly­ is able to enhance the pDNA condensation capacities. Such a situation
meric glycol NPs with distinct chemical functional groups and diverse resulted in the higher usefulness of PLL for delivering the genes with
morphologies. Such polymeric glycol NPs are hopeful for creating greater biological features [113].
diverse bio-active glycol-polymer structures for different health associ­ PLGA is known as a bio-degradable polymer with the highest rate of
ated utilizations like the drug delivery, bio-materials, biotechnology, success utilization because its hydrolysis results in the metabolite
and nanotechnology, and gene treatments. For example, Chen et al. monomers, glycolic acid and lactic acid. Based on the research, since the
presented an approach for synthesizing the self-assembled porphyr­ above 2 monomers are as endogenous molecules and could be readily
in–glycopolymer conjugates by incorporating the Reversible Addi­ metabolized through Krebs cycle in the body. There is a relationship
tion− Fragmentation chain Transfer (RAFT) polymerization and one-pot between the minimum systemic toxicity and PLGA utilization for the
conjugation reaction as one of the one-pots of multireactions [109]. drug delivery or bio-material uses [114]. It confirmed the PLGA could be
Such porphyrin–PMAG conjugates had self-assembly behaviors for employed as proper drug delivery systems for human. Various polymers
forming the micelles in the water as a result of the hydrophobic are found in the markets with various molecular weights and co-polymer
porphyrin in the middle and hydrophilic glycopolymer at the two ends. compounds. However, degradation time is variable from numerous
It should be mentioned that Con A has been used to test the binding months to multiple years that depends on the copolymer ratios and
abilities of the synthesized glycol NPs. Results indicated the anticancer molecular weights [115,116]. Of course, the monomers ratio applied

Fig. 4. Functionalizing the hematite NPs with 1 accompanied by coupling the D-mannose and consequent binding with Con A.

31
H. Khan et al. Seminars in Cancer Biology 69 (2021) 24–42

would identify the PLGA types. As an instance, the PLGA 50:50 would the respective flexibility in the control of chemical, physiological, and
identify a copolymer whose composition equals 50 % glycolic acid and biological characteristics [123]. According to some studies, researchers
50 % lactic acid. Moreover, poly(lactic acid) (PLA) has been utilized to a applied the core-shell structured and self-assembled block copolymer
lower extent in comparison to the PLGA because of the decreased NPs for medicines, proteins, gene deliveries, and the imaging factors
degradation rates. [124–128]. The majority of the synthesized NPs are single-functional.
As shown in the Figure, PLGA-NPs are internalized in the cells Therefore, these NPs would be inadequate in a complicated physiolog­
partially via the fluid phase pinocytosis as well as the clathrin-mediated ical context. This suggests the requirement for developing innovative
endocytosis. Furthermore, the PLGA-NPs could escape the endo- multi-functional NPs [123]. Commonly, we have crucial documents for
lysosomes and enter the cytoplasm (Fig. 5). Such a condition would indicating that easy, effective, and stable ligands critically contribute to
facilitate the NPs interaction with the vesicular membranes that results the active selection of the cancer cells. However, the function of the
in the temporal and localized de-stabilization of the membranes leading effective medicine releases for the targeted cancer cells via utilization of
to escaping the NPs into the cytosol [117]. In fact, researchers approved the nano-carriers may be influenced by a number of the factors,
the PLGA NPs capacities as the drug delivery systems for a lot of treat­ including stimuli-responsive medicine releases, tumor-targeting capa­
ment factors such as chemotherapy, antibiotic, antiseptic, bility, cellular internalizations, medicine loading levels, and circulation
anti-inflammatory and antioxidant medicines, and proteins, and may be times. Actually, many examinations have tried to using of specific li­
desirable for tumor- and or DNA-targeting [118,119]. gands that have a direct relationship to the cancer cellular uptakes of the
nano-carriers for reducing injuries in the normal tissues [123]. Anyway,
7. Glyco-nanoparticles and cancer therapy the nano-carriers extracted from self- assembly of the amphiphilic block
co-polymers offer wide possibilities [129]. Although they have been
It is commonly accepted that one of the most severe medical issues synthesized via the controlled polymerization methods, the experts in
resulting in mortality throughout the world is cancer. In spite of the the field have discovered the glycopolymers for over 50 years so that
broad utilization of the traditional chemo-therapeutic agents for treating their significance has been increased for becoming a helpful means in
cancer, they suffer from a number of shortcomings like improper the drug delivery utilizations due to the respective recognition features
recognition of tumor tissues leading to damages to the healthy tissues [130,131]. Moreover, glycopolymers enjoy a key potent for increasing
and lower bio-compatibility as a result of their hydrophobicity, which their specificity via interaction with the cell surfaces and proteins in the
result in aggregation. Hence, it is highly necessary for the clinics to have physiologic events. In addition, the crosstalk between
an efficient treatment instrument for recognizing the features of the carbohydrate-binding proteins and carbohydrates would be fully weak
healthy and cancer cells, and concurrently select the cancerous tissues. in the biological procedures, though it could be improved via applica­
Intelligent, sensible, and particular nano-carrier platforms for the tar­ tion of the multi-meric carbohydrate molecules called cluster glycoside
geted delivery of the anti-cancer agents could be necessary for the effect [131,132]. Taken together, it seems that glycol-nanoparticles
clinical utilization. Some researchers examined various nano-carrier- could employed as potential delivery system for targeting different
based medicine delivery systems containing the polymeric NPs, lipo­ anti-cancer-agents. Table 3 illustrates various glycol-nanoparticles
somes, micelles, and dendrimers for optimizing the treatment regimens which are applied in cancer therapy.
for cancer [9–11,13,121,122]. Co-polymers with the hydrophilic and
hydrophobic chains would self-assemble into developed NPs with
7.1. Inorganic glycol-nanoparticles
diverse morphology; for example, dimension, shapes, compositions,
structures, and so on have been significantly considered as the result of
Notably, the gold glycol-nanoparticles (GNPs) have been considered

Fig. 5. A schema of nanoparticle internalization in the cells.

32
H. Khan et al. Seminars in Cancer Biology 69 (2021) 24–42

Table 3
Selected glycol-nanoparticles which are used in cancer therapy. A variety of GNPs such as magnetic, and gold NPs enable to carry different anti-cancer agents.
Cancer Nanoparticle Drug Model Type of cell line Ref

Human tumors β‑Cyclodextrin-Bearing Gold Glyco-nanoparticles Methotrexate In vitro – [133]


Melanoma cancer Gold glycol-nanoparticles In vivo B16OVA, A37, Mel JuSo, B16.F10, MeWo, [134]
In vitro SKMel24, CHO
Prostate cancer Gold nanoparticles + NaBH4+Either thio-glucose or In vitro DU-145 [135]
sodium citrate
adenocarcinoma Gold glyconanoparticles + glucose + biotin + siRNA In vivo in CMT/167 [136]
vitro
Leukemia cancer Gold nanoparticles+ in vitro THP-1, MCF-7 [137]
thio-PEG + thio-glucose+
Glycopolymer-Stabilized Gold Nanoparticles [138]
Breast cancer Metformin Loaded gold glycol-nanoparticles Metformin In vitro MCF-7 [139]
Neuroblastoma Glycol-polymer-coated gold nanoparticles DOX In vitro SH-SY5Y [140]
Prostate cancer Galacto-glycogen nanoparticles – In vitro PC3 [141]
Hepatocellular Magnetic iron oxide nanoparticles + polydopamine + Hypericin In vitro HepG2, MCF-7 [142]
carcinoma Hypericin +
Lac
Breast cancer Iron oxide nanoparticles + polydopamine + glucose – In vitro MDA-MB-231, MCF-10A, 4T1 [143]
oxidase In vivo
Multiple cancers Magnetic glyco-nanoparticle – In vitro TA3-ST TA3-HA, MCF-7, B16-F1, B16-F10, SKOV- [144]
3, HT29, A549, A498, 184B5
Lung cancer Fluorescein isothiocyanate- (FITC)-doped mesoporous DOX In vitro A549, PCC [145]
silica
Nanoparticles
Hepatocarcinoma Galactose-based glycopolymer-drug conjugates DOX In vitro HepG2, COS7 [146]
nanoparticle
Breast cancer Glucose-conjugated chitosan DOX In vitro 4T1 [147]
Nanoparticles

significantly as a result of the respective organized properties as the good stability and dispersibility in the aqueous environment, fair tar­
water-soluble carbohydrate-functionalized nano-clusters with the geting capability for the ASGP-R-overexpressing tumor cells, and
excellent capacity for chemical glycobiology, bio-medicine, diagnostics, insignificant cytotoxicity in the absence of light, and a good function in
and clinical utilizations. Over the past decade, de la Fuente et al. pub­ the PDT. Researchers also presented an approach to deliver Hy that is
lished a leading integrated glycol-nanotechnology method. The re­ highly flexible and can be readily expanded so that Hy can be replaced
searchers examined and assessed the carbohydrate and with other hydrophobic PSs; however, it can conjugate the targeting
carbohydrate–protein interactions according to the utilization of such moiety and the scaffold for performing various performances [142].
NPs. They also applied the NPs as the potent means in the anti-adhesive Researchers used the spectrophotometry analyses and showed that
treatment for cell to cell adhesion examinations and for preventing the the amount of Hy in the PHMs was 72 μmol g-1 PHMs. In fact, the
pathogen invasion [148–151]. However, small carbohydrates like developed Hy-entrapped glycol NPs (Lac-PHM) indicated very good
glucose may be bound to the gold NPs (AuNPs) and applied for sensitive stability, water dispersibility, and selectiveness for the asialoglycopro­
colorimetric assays [152]. tein receptors overexpressing HepG2 cells. Moreover, the atomic
For example, Conde et al. assessed the anti-cancer effects of the adsorption spectroscopy analyses demonstrated that the amounts of the
siRNAGlycoNPs (AuNP@PEG@Glucose@siRNA) as compared to the Lac-PHMs taken in the HepG2 cells has been 2.1-fold greater than that of
PEGylated GlycoNPs (AuNP@PEG@Glucose) both in vitro and in vivo the triethylene glycol-modified PHMs. In addition, the outputs of the
[153]. In fact, siRNA GlycoNPs are associated with the expression of the intra-cellular reactive oxygen specimens production, detection, cyto-
pro-apoptotic proteins (i.e., caspases 3 and 9 and Fas/CD95) in a toxicity examination, and apoptosis determination revealed that the
dosage-dependent way that is not dependent on the inflammatory re­ Lac-PHMs had a good photo-dynamic impact on the HepG2 cells [142].
sponses. In addition, the targeting of siRNA GlycoNPs could target Some researchers claimed that engineering the multi-valent glycol-
expressing the c-Myc gene in animal modelthat is one of the essential nanoparticles for effective selection of the carbohydrate binding pro­
regulators of the cell rapid growth and apoptosis through in-vivo RNAi teins in the mammalian cells was significantly favored for molecular
in the tumor tissues that results in about 80 % decline in the tumor size recognition examinations as well as treatment utilizations. In fact, the
with no related inflammation. multi-valent receptor ligand presentation in the biological systems is one
Research indicates that photo-dynamic treatment is one of the safe, of the strategies used for compensating the poor and lower affinity
non-invasive modalities to treat cancer, where photosensitizer (PS) is binding between the proteins and carbohydrates [157]. Due to such a
one of the essential components [154]. In general, hypericin (Hy) is one multi-valency, general binding of a multi-functional ligand may be
of the promising l PSs, but its clinical utilization is considerably considerably more powerful than a combined distinct binding events of
restricted by its weak hydrophilicity [155]. For example, Shao et al. the single ligands [158]. According to the studies, the biologically
provided an easy and new approach for designing a nano-carrier to inspired engineering of the synthetic systems provided with the carbo­
deliver Hy. In fact, Hy has been initially entrapped into the hydrate ligands in a multi-valent form has been extremely investigated
poly-dopamine (PDA) film via the dopamine polymerization procedure over the past ten years [159,160]. Indeed, the GNPs function as a
on the surface of the magnetic iron oxide nanoparticles (MNPs) [156]. multi-valent scaffold that could carry several copies of the carbohydrate
Therefore, there may be a specific interaction between lactose and ligands. Therefore, they increase probable binding with the recognition
asialoglycoprotein receptors (ASGP-R) that has been conjugated to the receptors such as lectins [161].
surface of the PDA of the as-constructed PDA–Hy–MNP composite NPs Due to the carbohydrate-lectin interactions, the glycol-
(PHMs) by adding Michael or Schiff base reaction under alkaline con­ nanomaterials could probably intervene in several cellular activities
ditions [156]. The resulting constructed Hy-entrapped glyco­ such as cell to cell communications, immune responses, and bacterial
l-nanoparticles (expressed as Lac-PHMs) possess such e features as very and viral infections [162,163]. Particularly, the multi-valent

33
H. Khan et al. Seminars in Cancer Biology 69 (2021) 24–42

carbohydrate binding proteins, which have a mediatory role in the cytosol are much greater than the ones in the normal cells or
malignant cellular activities, are attractive molecular targets. For extra-cellular fluids [181]. Finally, via incorporation of the two
example, Besford et al. used the glycogen NPs as a biosourced redox-responsive and pH sensitive features into the GPD NPs, DOX may
glyco-scaffold to engineer the multivalent glycol-nanoparticles [164]. be precisely and systematically released from the NPs in the hepatoma
Actually, the glycogen NPs that is one of the naturally occurring cells’ cytoplasm that are reductive and acidic.
greatly-branched glucose polymers, has been functionalized with lactose According to the studies, the cancer cells can be reproduced with
and attained by copper(I)-catalyzed alkyne-azide cycloaddition chem­ higher speed and prolonged living compared to the normal cells as they
istry for the targeted interactions with lectins ex-situ and on the prostate apply certain proteins and glucose for supporting their continual growth
cancer cells. Therefore, lactosylated glycogen consisting of the terminal [182]. However, their capability of covalently conjugating with diverse
β-galactoside moieties has been called galacto-glycogen (GG). It has a bio-molecules through the thiol groups is an interesting features of GNPs
strong interaction with the peanut agglutinin (PNA) that is a β-gal­ [183]. In fact, the GNPs capped with proper functional bio-molecules
actoside-specific lectin which has been seen by dynamic light scattering, may increase their uptakes through the cancer cells but not enclosing
optical wave guide lightmode spectroscopy, and quartz crystal the normal cells; for example, macrophage and endothelial cell, and thus
micro-balance measurement. With regard to the results, GG NPs had has been considered for detecting and treating tumors [184–187].
multivalent bindings to the PNA with an affinity constant of 3.4 × 105 Actually, glucose is one of the major sources of the metabolic energies
M− 1, and the GG-PNA complex could not be displaced by lactose. This for the cells. Therefore, the cancer cells consume considerably higher
condition demonstrates a competitive binding of GG to the lectin. Such glucose than the normal cells. In particular, greater numbers of the
GG NPs have been experimented in terms of the relationships with the glucose molecules are internalized by GLUT receptors found on the
cell membranes of prostate cancer in-vitro, in which the particles had cancer cell surfaces [188,189]. Hence, some researchers believe that
higher affinity for the membrane, which has been confirmed by obser­ glucose tagging is one of the efficient ways for facilitating the GNPs
vations from the flow cytometry and confocal microscopy. The above introduction into the cells. in vitro and in vivo examinations showed that
finding could be related to the results from particular extra-cellular the pegylated glucose coated GNPs (or Glu-GNPs) with a diameter of
galectin-1 targeting. Moreover, the GG NPs cause accumulation be­ 20 nm would have higher efficiency in selecting the solid cancer such as
tween the prostate cancer cells. Such findings revealed an approach to the prostate, breast, ovarian, and liver cancers [186,190]. Moreover,
engineer a biosourced polysaccharide with the surface moieties, which images indicated that Glu-GNPs enteres the cancer cell more than the
indicate influential multi-valent interactions with lectins and the tar­ surrounding normal cell in the animal examinations [176,191]. Even
geted interactions with the prostate cancer cells [164]. though treatment efficiency of the Glu-GNPs against the naked GNPs has
As we know, the hepatic galactose/N-acetylglucosamine receptor been significantly considered by researchers, multiple studies have
called asialoglycoprotein receptor (ASGPR) is particularly subjected considered the NPs uptake in the bound-growing cells (e.g., MCF-7 and
significantly to the hepatoma cells surfaces [165,166]. For this reason, HeLa cells) and solid tumors. Researchers applied the suspension cells,
the ASGPR has been employed as one of the autoantigens for achieving specifically THP-1 cells, for modelling the metastatic cells and cancer
the targeted hepatopathy therapies by specifically recognizing the stem cells [192]. They assessed the Glu-GNPs cellular uptake and made a
galactose [167]. In addition, the tumor micro-environment like hypoxia, comparison between it and the uptake of the bound cells, specifically the
acidity, higher glutathione (GSH), and over-expressed enzymes, inspired MCF-7 cells. Based on the outputs, Glu-GNPs had a greater effect on the
the reasonable development of the intelligent NPs for responding to THP-1 cells as compared to the MCF-7 cells. Moreover, average gold
diverse physic-chemical and biochemical stimuli [168–172]. To achieve concentrations in all THP-1 cells could be up to ~35 % higher than the
the above-mentioned objective, the authors presented the cleavable MCF-7 cells. Additionally, one to two hour starvation would be opti­
linkages like the pH-sensitive bonds (i.e., boronate ester & “Schiff” base) mum, and differential targeting impacts would be lost in a case of the
and redox-responsive linkages (i.e., disulfide & Se–Se bonds] for making cells starvation more than three hours. Of course, X-ray outputs showed
the NPs. This would distinguish the carcinoma cells from the normal that 9 Gy was an optimum irradiation dose. In fact, the Glu-GNPs plus
ones and regulate the medicine release processes. It also exactly satisfies X-rays could obtain more reasonable cancer killing impacts (at least 20
the mechanisms of diverse factors, including the extra-cellular and % more) in comparison with the GNPs plus X-rays or X-rays itself. The
intra-cellular releases [173–176]. Hence, for realizing a valid and above findings can contribute to more acceptable cancer therapies of the
effective NP-induced HCC therapy, the stable loading of the hydropho­ cancer metastasis as well as cancer stem cells [192].
bic factors in the blood circulation with no leakages, selective transports,
and drugs release into the hepatoma cells is required. For example, Wu 7.2. Polymeric glycol-nanoparticles
et al. designed a crucial cross-linking glycol-polymerdrug conjugates
(GPDs) NPs with certain dual-responsive traits for attaining the selective It is widely accepted that the mammary glands healthy cells is
transfer and program release of anti-cancer drugs to treat HCC [177]. It described by presenting the surface of the branched, O-linked core 2
should be noted that Wu et al. addressed certain recognition between glycans with higher amounts of N-acetyl-D-glucosamine (GlcNAc).
galactose and ASGPR, and the cluster glycoside impact that may effi­ Nonetheless, proteins presented on breast tumors surface provide basi­
ciently cause the carbohydrate ligand to improve the affinity for their cally linear, truncated core 1 mucin-type glycans like α-N-acetyl-D-
protein receptors [178,179]. They utilized galactose for building the galactosamine (αGalNAc, the Tnantigenglycan) with complete or near
glycopolymer for achieving the higher ASGPR-induced hepatoma the complete lack of the core 2 residues [193]. Notably, such differences
cellular binding and internalizations [177]. Indeed, a disulfide bond has would be selected as one of the strategies for the cancer immuno-therapy
been proposed for the side-chain of glycopolymer through a dynamically [194]. Correspondingly, multi-valent glycol-conjugates have been pro­
covalent boronate ester between the galactose moiety and cured where the mucin glycans have been provided on different scaf­
phenyl-boronic acid that shows the pH-modulated traits. Then, the hy­ folds such as the peptides, dendrimers, lipo-peptides, and proteins
drophobic model anti-cancer medicine DOX has been conjugated with [195–198]. A number of such strategies have been designed as far as
glycol-polymer for forming diverse amphiphilic conjugates by a clinical trials [199]. Nano-materials show a different ground to present
self-eliminating disulfide bond [180]. The above procedure can ensure glycans8, which result in the higher synthetic controls and greater
the traceless releases of DOX that keeps the original chemical structures densities compared to the recent protein scaffolds. Carbohydrates which
and pharmacological actions of DOX. Additionally, hydrophobic core of present AuNPs decorated with little molecule thiolated glycans have
the self-assembly GPD NPs has been internally cross-linked by a disulfide been applied as the means for studying the
bond for stabilizing the architecture and avoiding the drug permeation carbohydrate-to-carbohydrate interactions in anti-adhesive treatments
in a physiological environment. Notably, GSH levels in the cancer cells’ and as cancer vaccine and anti-HIV candidates [151,200–203].

34
H. Khan et al. Seminars in Cancer Biology 69 (2021) 24–42

Parry et al.’s study described designing and constructing the peptide- the other approaches for improving efficiency and selectivity of the
free multi-valent glycosylated nano-scale constructs as the potent syn­ cancer therapy is to employ hyperthermia when combining with com­
thetic cancer vaccines, which produce the considerable titers of anti- mon cancer drugs like radiation therapies and chemo-therapy [212,
bodies that are selective for the abnormal mucin glycans [204]. A 213]. It should be noted that hyperthermia results in the higher sensi­
poly-merizable version of Tn-antigen glycan has been procured and tivity of a number of cancer cells to the radiation. Moreover, it may
transformed into the organized glycol-polymers through a RAFT poly­ increase the effects of specific anti-cancer medicines and allow applying
merization. Afterwards, polymers have been combined with gold NPs lower chemotherapy dosages [212]. Indocyanine green is one of the
that yield the multi-copy multivalent nano-scale glycol-conjugates. optical tracers, which is able to produce heat via absorption of
Immunological examinations showed that such nano-materials caused near-infrared light. In addition, Manchanda et al.’s research is important
influential and lengthy generation of antibody, which would be selective because of the multi-functional PLGA NPs synthesis and combination of
to the Tn-antigen glycan and cross reactive towards the mucin proteins the medicines with various physical features (indocyanine green that is
presenting Tn. As revealed by the findings, the proof-of concept of an amphiphilic and DOX that is hydrophobic). Such indocyanine
easy and modular strategy towards the synthetic anti-cancer vaccines on green-DOX NPs would possess potent utilizations as the drug delivery
the basis of multi-valent glycosylated nano-materials with no require­ systems for the incorporated the chemotherapy and localized hyper­
ment for a normal vaccine protein element [204]. thermia [214].
PLGA NPs connected to the targeting ligands would be employed for In a study, researchers procured the cisplatin NPs with the average
targeting the severe tumors with high affinities [205]. Moreover, the dimension between 150 and 160 nm and about 2 % w/w cisplatin
PLGA NPs possess great surface areas and functional groups to conjugate contents [215]. It has been shown that cisplatin-loaded PLGA-mono­
to several diagnostic factors; for example, radioisotopic, optical, or methoxy (m)PEG NPs are efficient for hindering the tumor development
magnetic [206]. NPs carriers enjoy higher stability in the biological in the animal model of colon cancer which suffer serious combined
fluids. They also have higher ability for avoiding the enzymatic me­ immune deficiencies. It should be noted that the mice given treatment
tabolisms compared to additional colloidal carriers like the liposomes or with the cisplatin-loaded NPs have the greater survival rates in com­
lipid vesicles [207]. Of course, a majority of the anti-cancer medicines parison to the free cisplatin group [215]. Furthermore, the
examined in the PLGA NPs procurements would be presented here. Cisplatin-loaded PLGA-mPEG NPs led to increasing of residence time of
Table 4 is a summary of them. The PLGA NPs with the docetaxel with the cisplatin in the systemic circulation in animal model of prostate cancer
favorable dimension and medicine-loading features appropriate for [216]. Taken together, these studies indicated that PLGA NPs have very
intravenous injections may be procured without the use of the important properties which could be introduced it as powerful tools for
Tween®80. For example, a study revealed that cellular cyto-toxicity of targeting different tumors with high affinity.
NPs has been greater than free drugs. Moreover, docetaxel -loaded NPs
had acceptable levels of plasmas in-vivo as compared to the traditional 8. Glycoside-nanoparticles and cancer therapy
formulation of docetaxel (Taxotere®) [208]. Some researcher showed
the use of nano-precipitation procedure to create the docetaxel-loaded Researchers introduced a type of the synthetic glycosides as the
NPs [209,210]. The study conducted by Cheng et al. demonstrated suppressors of adeno-carcinoma and glioma cell proliferations [236,
that diminishing the medicine-loading to 1 % (w/w) diminished the 237]. Notably, glycosides are as a derivative of the N-acyl-D-glucos­
particles accumulation and had docetaxel-loaded PLGA NPs with nar­ amine. Moreover, earlier findings showed that their activities would
rower size distribution [210]. enhance as the prolonged hydrocarbon chain has been present at the C-1
Betancourt et al. formulated NPs via nano-precipitation of the acid- position of the glycoside moiety. Given that conjugating a sugar with an
ended PLGA for controlling DOX release in the pH-dependent way and oleyl chain would improve conjugate anti-tumoral activities [238].
delivering great loads of active therapeutic agent to a breast cancer cell However, this mechanism of the growth suppression would suggest
line. It could be concluded that pH-dependent release behaviors may changes in the lipid metabolisms. Actually, results showed that the
result from the fast degradation of polymer and decrease the ionic in­ glycoside-therapies led to significant alterations in the levels of glyco­
teractions between medicine and polymer at an acidic pH [211]. One of lsphingolipids with modulatory contributions to the tumor development

Table 4
Selected poly lactic-co-glycolic acid (PLGA)-nanoparticles used in cancer therapy. PLGA are able to carry different anti-cancer agent.
Cancer Polymer for nanoparticle Drug Cell line Reference
Preparation

Glioma PLGA Paclitaxel glioma C6 cells [217]


Colon carcinoma PLGA ethylene Paclitaxel HCT116 [218]
oxide fumarate
Colon cancer PLGA Paclitaxel HT-29 [219]
Breast -colorectal adenocarcinoma DMAB-modified Docetaxel MCF-7, Caco-2 [220]
PLGA-TPGS
Cervical cancer PLGA-d-a-TPGS Docetaxel HeLa [221]
Lymphoma PLGA Zinc (II) P388-D1 [222]
Phthalocyanine
Glioma PLGA Paclitaxel C6 rat glioma [223]
Breast cancer PLGA Vincristine MCF-7/ADR [224]
Verapamil
Ovarian adenocarcinoma PLGA Hypericin NuTu-19 [225]
Breast cancer PLGA Docetaxel MCF-7 TAX30 [226]
Ovarian adenocarcinoma -Breast cancer PLGA Curcumin A2780CP, MDA-MB-231 [227]
Prostate cancer PLGA Curcumin LNCaP, PC3, DU-145 [228]
Breast cancer PLGA DOX MDA-MB-231 [229]
Colon cancer PLGA-mPEG Cisplatin HT29 cells [230]
Prostate cancer PLGA-mPEG Cisplatin LNCaP [231]
Lung cancer PLGA Paclitaxel NCI-H69 (SCLC) [232,233]
Glioma PLGA Paclitaxel C6 glioma [234]
Cervical cancer PEGylated PLGA Paclitaxel HeLa [235]

35
H. Khan et al. Seminars in Cancer Biology 69 (2021) 24–42

and play a role in the paths of the cell mortality or rapid growth 9. Clinical status
[239–241]. Nevertheless, initial in-vivo tests with the most acceptable
candidates have been accompanied by moderate outputs [242]. Due to There is evidence of the history of the colloidal gold utilization for
their lengthy alkyl chain, compositions have been weakly soluble in the enjoying its health advantages for many centuries [250]. One of the
aqueous physiological media. Moreover, a number of them have been earliest approved successes of the health advantages of the colloidal gold
exposed to the enzymatic degradation that declined their biological is its application as one of sources of radiation for brachy-therapy and
stabilities [243]. In fact, using the intra-tumoral injections of the med­ arthritis treatment [251]. Surely, such a success has enhanced the
icines distributed in a dimethyl sulfoxide (DMSO)/H2O mixture has been attention to the use of the gold and further noble metals for health care
feasible with the Bovine Serum Albumin (BSA) as a carrier. However, uses. For example, a number of AuNPs, including Aurimune that is a
just 1 enzyme-resistant thioglycoside derivative experienced consider­ tumor necrosis factor-bound AuNP, finished the major clinical trials
able suppressive activities of the tumor growth with highly recurrent (NCT00356980 & NCT00436410 from www.clinicaltrials.gov) so that
dosages [244]. It could be found that IONP-based drug delivery systems they provide the ground for several systems presented in this research.
possibly affect the stabilization and selection of the bio-active glycosides Although any AuNP products have been not clinically confirmed, each of
to the tumor site and result in the greater treatment activities. them holds light adsorption features of AuNPs and is examined in terms
As stated by researchers, treatment agents in a majority of the IONP of diverse treatment utilizations ranging from the healing solid tumors
medicne delivery systems would have a covalent binding or electro- to acne. For instance, AuroLase® that has been provided by Nano­
static adsorption over the IONP surface, distributed into a polymer spectra, is the silica-gold nano-shells coated with (poly)ethylene glycol
matrix, or encapsulated within the amphiphilic nano-structures [245]. (PEG) developed for thermal ablation of the solid tumors after triggering
Overall, each solution requires a pre-available hydrophilic coating with with a near-infrared energy source [250]. As stated by researchers, silica
functional moiety over IONP scaffold. In order to create a simplified core is utilized as a dielectric core, gold shell provides thermal ablation
procedure, the technique on the basis of co-precipitation yielding capability after intense adsorption of the near-infrared NIR light, and
aqueous IONP would be prioritized because of possible additional sur­ PEG layer causes general particle stability. Researchers published the
face tailoring, even though the NP achieved via the technique would above technology more than 10 years ago [252]. Their major results that
have lower qualities [246]. In addition, more stages would be needed to is a foundation for AuroLase® indicated that AuroLase® might be used
integrate the medicines [247]. Indeed, thermal degradation technique inducing photo-thermal cell mortality in-vitro after triggering with a
would provide the most reasonable solution with regard to the NIR energy source and increasing the solid tumor temperature for in­
physic-chemical features (i.e., the size or dimension, the size distribu­ duction of irreparable thermal damages to the tumors in the mice after
tion, crystallinity and reproducibility) of the iron oxide cores; however, intra-tumoral injections [252]. Some studies indicated the use of
it would yield IONP just stable in the organic solvent. As a result, a AuroLase® for a follow-up for treating the thermal ablation of the brain
further stage would be necessary for aqueously stabilizer the NP prior to and prostate cancers so that prostate cancer exhibited potentials for total
the medicine integration [248]. thermal ablation of the solid tumors after an organized injection of
A study conducted by Groult et al. dealt with possible direct stabi­ AuNPs [253,254]. Then, AuroLase® introduced into 2 individual clin­
lization of the oleic acid-coated iron oxide NPs (OA-IONP) procured by ical trials (Table 1). The first clinical trial has been updated in September
thermally degrading via micellation with bio-active glycosides 2014 (NCT00848042). Reports showed that it could be completed to
mentioned earlier [249]. It should be noted that hydrophobic alkyl treat the patients suffering from refractory and or recurrent tumors for
glycoside tails in such colloidal structures would enclose aliphatic chain the head and neck cancers (www.clinicaltrials.gov). The other clinical
of OA-coated IONP via hydrophobic interactions, whereas hydrophilic trial that is recently active but not recruiting is examining the Auro­
sugar would be dispersed on the micelle outer surface and improves the Lase® to treat the primary and or metastatic lung tumors, in which the
water-dispersibility via creating a coverage of the hydrophobic layer. airway has been blocked. AuroLase® is one of the examples of the
Such a configuration, in which the treatment cargo works as a micellar in-organic NPs, which has been regularly studies and approved effi­
coating would require further experiments for studying the drug de­ ciency in ablating the tumor at the pre-clinical phase and is currently
livery system viability. For the first stage, researchers should test if being explored in the clinics. AuroLase® has several benefits for treating
bio-active coatings would meet the requirements of the resultant hy­ tumors. It should be mentioned that AuroLase® is capable of treating
drophilic matrix for in vivo application of the probes or not; that is, tumors and avoiding several consequences related to more common
prevention of the opsonization, improvement of the colloidal stability, tumor treatments such as chemotherapy via a localized tumor treatment
bio-compatibility, and proper relaxometric features for imaging uses. that would selectively treat the tumor and restrict damages to the
Afterwards, it should be monitored if antitumor activities would be healthy tissues [255,256]. In addition, AuroLase® would provide the
maintained into the micellar formulations of such therapeutic com­ ground for exploring an obvious limitation in the recent tumor treat­
pounds. Next, glycoside-coated IONP micelles have been experimented ment. Therefore, it would be a means for providing a treatment, which
as MRI contrast agents and anti-mitotics on the glioma and lung cancer could not be currently feasible. Notably, AuroLase at pre-clinical phase
cell lines. Then, a comparison has been made between the micelle would be examined as a supplementary treatment for radiation and
anti-mitotic activities and activities of the relative free glycosides. imaging and some other commercial utilization [257,258]. However, it
Overall, glycoside-coated IONP micelles formulation of the above gly­ has specific biological and technical challenges prior to the clinical ap­
cosides had their own antitumor impacts; just in 1 case, it exhibited an provals. For example, it should be proved that the system is able to be
uncommon therapeutic improvement. Eventually, micelles showed op­ administered intra-venously, and the target tumors can utilize the
timum relaxometric features for their application as the T2-weighed enhanced permeation and retention (EPR) effects as one of the active
MRI contrast agents. Their outputs suggested that the bio-active hy­ targeting moieties. Researchers approved that EPR effect can assist in
drophilic nano-formulations are the theranostic agents with synergistic the tumor aggregation of NPs in the pre-clinical animal examinations;
features achieved from 2 entities that individually would not be pre­ however, it has lower validity in the clinical settings. Therefore,
pared for in vivo utilizations, and reinforce possible use of the dependence on the EPR’s targeting effect would be accompanied by
bio-molecules as both medicines for treatment and a coating for challenges for AuroLase® [259]. Moreover, as the AuroLase® is one of
OA-IONP micellar stabilization [249]. It seems that glycoside-NPs could the local therapies developed for treating the solid tumors at the tumor
be employed as new delivery system for targeting various anti-cancer site, it would be hard to translate the technology for treating the sys­
agents. In this regard, more studies for approving it as a proper drug temic malignancies. For this reason, many polymeric NPs undergo
delivery system in the treatment of cancer are needed. pre-clinical stage of advancement; however, they possess potentials for
the targeted drug delivery of the anti-cancer medicines as a result the

36
H. Khan et al. Seminars in Cancer Biology 69 (2021) 24–42

convenience, by which the ligands are able to bind. Furthermore, the combining multi-valent presentation of carbohydrates (glycol-clusters)
albumin-bound NPs of paclitaxel (Abraxane) had successful utilization with specific chemicophysical features of the nanosized metallic core.
for delivering paclitaxel to treat the breast cancer after a fail in of Notably, feasible binding of various kinds of carbohydrates and other
various combined chemotherapy utilization for this disease or relapse molecules; for example, luminescent probe, peptide, and magnetic
within six months after adjuvant chemotherapy. Thus Abraxane is chelate, onto a similar gold nanoparticle in a controlled manner (multi-
associated with different advantages. For example, albumin could be as functional GNPs) and modification of the core for obtaining GNPs with
non-toxic agent and immune system completely tolerates it, because magnetic or fluorescence features would make such a multi-valent
albumin is a plasma protein which has 66 kDa molecular weight. In glyco-scaffold appropriate to do research of the carbohydrate medi­
addition, its application would eliminate the needs for poison solvent ated interactions and utilizations in molecular imaging and therapies.
(Cremophor EL poly-oxyethylated castor oil); that is, it limits dosing
Taxol [260]. Moreover, it enjoys features such as the albumin Funding source
pharmaco-kinetics, particularly its prolonged half-life that would be
specifically interesting for designing the medicine carriers for passive None.
targeting. Researchers indicated that Abraxane targets cancer the tissues
due to higher metabolic demands and active transports of the plasma Declaration of Competing Interest
proteins for anabolic procedures [260]. Apparently, albumin would help
endothelial transcytosis of the protein bound and unbound plasma None.
constituents by attaching to a cell surface receptor (gp60). Conse­
quently, the Gp60 would bind to the caveolin-1 with consequent crea­ References
tion of the transcytolic vesicles. Furthermore, Abraxane might be
transferred into the tumor via the secreted protein acidic that is rich in [1] H.R. Mirzaei, A. Sahebkar, R. Salehi, J.S. Nahand, E. Karimi, M.R. Jaafari,
cysteine (SPARC) or osteonectin, which binds albumin due to a sequence H. Mirzaei, Boron neutron capture therapy: moving toward targeted cancer
therapy, J. Cancer Res. Ther. 12 (2) (2016) 520–525.
homology with gp60. It should be noted that the SPARC, as caveolin-1,
[2] F. Bray, J. Ferlay, I. Soerjomataram, R.L. Siegel, L.A. Torre, A. Jemal, Global
would be frequently expressed in a number cancer such as the breast, cancer statistics 2018: GLOBOCAN estimates of incidence and mortality
lung, and prostate, which explains why albumin accumulates in a worldwide for 36 cancers in 185 countries, CA Cancer J. Clin. 68 (6) (2018)
394–424.
number tumors and therefore makes easy the intra-tumoral accumula­
[3] A. Dag, Glyconanoparticles for targeted tumor therapy of platinum anticancer
tion of the albumin-bound medicines. Finally and at the Phase III of the drug, Biomacromolecules 20 (8) (2019) 2962–2972.
research, Abraxane led to greater tumor response rate, a more accept­ [4] R. Rampado, S. Crotti, Nanovectors design for theranostic applications in
able safety profile, and greater survival in comparison to the common colorectal Cancer, J. Oncol. 2019 (2019), 2740923.
[5] S. Boca, D. Gulei, A.A. Zimta, A. Onaciu, L. Magdo, A.B. Tigu, C. Ionescu,
paclitaxel in the patients taking the 2nd phase of chemotherapy [261]. A A. Irimie, R. Buiga, I. Berindan-Neagoe, Nanoscale delivery systems for
few nanoparticle-based therapies are approved by FDA for cancer microRNAs in cancer therapy, Cell. Mol. Life Sci. (2019).
therapy. There are not any glycol-nanoparticles-based therapies in the [6] I.I. Lungu, A.M. Grumezescu, Nanobiomaterials used in Cancer therapy: an up-to-
date overview, Molecules 24 (19) (2019).
clinical in the treatment of various diseases such as cancer. Hence, it [7] F.Y. Du, Q.F. Zhou, W.J. Sun, G.L. Chen, Targeting cancer stem cells in drug
seems that clinical studies could release these NPs as new and effective discovery: current state and future perspectives, World J. Stem Cells 11 (7)
cancer therapies. (2019) 398–420.
[8] X. Wu, H. Yang, W. Yang, X. Chen, J. Gao, X. Gong, H. Wang, Y. Duan, D. Wei,
J. Chang, Nanoparticle-based diagnostic and therapeutic systems for brain
10. Conclusion tumors, J. Mater. Chem. B 7 (31) (2019) 4734–4750.
[9] O.S. Muddineti, B. Ghosh, S. Biswas, Current trends in using polymer coated gold
nanoparticles for cancer therapy, Int. J. Pharm. 484 (1-2) (2015) 252–267.
It has been known that recent therapeutic options for diverse cancers [10] K. Babiuch, A. Dag, J. Zhao, H. Lu, M.H. Stenzel, Carbohydrate-specific uptake of
are surgical operation, hormone therapies, radiation, and chemo­ fucosylated polymeric micelles by different Cancer cell lines, Biomacromolecules
therapy. Even though the above traditional alternatives enhanced the 16 (7) (2015) 1948–1957.
[11] B.S. Bolu, R. Sanyal, A. Sanyal, Drug delivery systems from self-assembly of
patients’ survival rate, they still suffer from different constraints. In fact, dendron-polymer conjugates (dagger), Molecules (Basel, Switzerland) 23 (7)
targeting the pharmaceutical is one of the quickly emerging strategies (2018).
for overcoming problems in the drug delivery, particularly to the tumor [12] N.S. Templeton, Nonviral delivery for genomic therapy of cancer, World J. Surg.
33 (4) (2009) 685–697.
site. Despite of the conventional drug delivery systems, NPs-based
[13] M. Piffoux, A.K.A. Silva, C. Wilhelm, Modification of extracellular vesicles by
compounds gain higher accumulation in the tumor site through the fusion with liposomes for the design of personalized biogenic drug delivery
respective active or passive mechanisms. It should be noted that the systems, ACS Nano 12 (7) (2018) 6830–6842.
nano-sized carriers support lengthier circulation time, simple perme­ [14] P.N. Navya, A. Kaphle, S.P. Srinivas, S.K. Bhargava, V.M. Rotello, H.K. Daima,
Current trends and challenges in cancer management and therapy using designer
ation into the cellular membrane, and effective site-specific targeting as nanomaterials, Nano Converg. 6 (1) (2019) 23.
a result of their exclusive features such the little sizes, larger surface to [15] A. Frey, K.T. Giannasca, R. Weltzin, P.J. Giannasca, H. Reggio, W.I. Lencer, M.
volume ratios, adjustable surface chemistry, and capability of encapsu­ R. Neutra, Role of the glycocalyx in regulating access of microparticles to apical
plasma membranes of intestinal epithelial cells: implications for microbial
lation of different medicines. However, current progressions in the attachment and oral vaccine targeting, J. Exp. Med. 184 (3) (1996) 1045–1059.
polymeric nano-medicines include the targeting of the polymer-based [16] R.A. Dwek, Glycobiology: toward understanding the function of sugars, Chem.
NPs, polyplexes, micelles, polymersomes, dendrimers, medicines/pro­ Rev. 96 (2) (1996) 683–720.
[17] A.G. Barrientos, J.M. de la Fuente, T.C. Rojas, A. Fernandez, S. Penades, Gold
teins conjugates, and the lipid hybrid systems to the tumor pathological glyconanoparticles: synthetic polyvalent ligands mimicking glycocalyx-like
site. As NPs have various functional moieties, they enhance functions surfaces as tools for glycobiological studies, Chemistry (Weinheim an der
with regard to the target capability, circulation durability, greater intra- Bergstrasse, Germany) 9 (9) (2003) 1909–1921.
[18] F. Compostella, O. Pitirollo, A. Silvestri, L. Polito, Glyco-gold nanoparticles:
cellular permeation, stimuli sensitivity, and carrier mediated visualiza­ synthesis and applications, Beilstein J. Org. Chem. 13 (2017) 1008–1021.
tions. Of course, one of the goals of the synthetic chemists when the [19] T. Mosaiab, D.C. Farr, M.J. Kiefel, T.A. Houston, Carbohydrate-based
multi-valent nature of the carbohydrate mediated interaction has been nanocarriers and their application to target macrophages and deliver
antimicrobial agents, Adv. Drug Deliv. Rev. (2019) [152-152].
discovered is a long search for constructing multi-valent carbohydrate
[20] S. Penadés, M. Jesus, Á.G. Barrientos, C. Clavel, O. Martínez-Ávila, D. Alcántara,
systems with accurate geometries, which are largely effective in inter­ Multifunctional Glyconanoparticles: Applications in Biology and Biomedicine,
acting with the carbohydrate binding proteins. Nonetheless, one of the Nanomaterials for Application in Medicine and Biology, Springer, 2008,
challenges to do this would be the controlling of the spatial and topo­ pp. 93–101.
[21] M. Prasad, U.P. Lambe, B. Brar, I. Shah, M. J, K. Ranjan, R. Rao, S. Kumar,
logical requirements for such systems. GNPs are the sugar-coated gold, S. Mahant, S.K. Khurana, H.M.N. Iqbal, K. Dhama, J. Misri, G. Prasad,
iron oxide or semi-conductor NPs with certain thiol-ending glycosides Nanotherapeutics: An insight into healthcare and multi-dimensional applications

37
H. Khan et al. Seminars in Cancer Biology 69 (2021) 24–42

in medical sector of the modern world, Biomed. Pharmacother. 97 (2018) tumor vasculature-specific nano-graphene oxide, Biomaterials 104 (2016)
1521–1537. 361–371.
[22] F.F. Sahle, T.L. Lowe, Design strategies for programmable oligonucleotide [49] O.C. Farokhzad, R. Langer, Impact of nanotechnology on drug delivery, ACS Nano
nanotherapeutics, Drug Discov. Today (2019) in press. 3 (1) (2009) 16–20.
[23] C.F. Anderson, M.E. Grimmett, C.J. Domalewski, H. Cui, Inhalable [50] T.J. Anchordoquy, Y. Barenholz, D. Boraschi, M. Chorny, P. Decuzzi, M.
Nanotherapeutics to Improve Treatment Efficacy for Common Lung Diseases, A. Dobrovolskaia, Z.S. Farhangrazi, D. Farrell, A. Gabizon, H. Ghandehari,
2019, e1586. Mechanisms and barriers in cancer nanomedicine: addressing challenges, looking
[24] M. Shariati, W. Willaert, W. Ceelen, Aerosolization of nanotherapeutics as a for solutions, ACS Publications 11 (1) (2017) 12–18.
newly emerging treatment regimen for peritoneal carcinomatosis, Cancers (Basel) [51] B. Rajitha, R.R. Malla, R. Vadde, P. Kasa, G.L.V. Prasad, B. Farran, S. Kumari,
11 (7) (2019). E. Pavitra, M.A. Kamal, G.S.R. Raju, S. Peela, G.P. Nagaraju, Horizons of
[25] G.S.R. Raju, B. Dariya, S.K. Mungamuri, G. Chalikonda, S.M. Kang, I.N. Khan, P. nanotechnology applications in female specific cancers, Semin. Cancer Biol.
S. Sushma, G.P. Nagaraju, E. Pavitra, Y.K. Han, Nanomaterials multifunctional (2019) in press.
behavior for enlightened cancer therapeutics, Semin. Cancer Biol. (2019) in press. [52] E. Pavitra, B. Dariya, G. Srivani, S.M. Kang, A. Alam, P.R. Sudhir, M.A. Kamal, G.
[26] G. Seeta Rama Raju, E. Pavitra, G.P. Nagaraju, K. Ramesh, B.F. El-Rayes, J.S. Yu, S.R. Raju, Y.K. Han, B. Lakkakula, G.P. Nagaraju, Y.S. Huh, Engineered
Imaging and curcumin delivery in pancreatic cancer cell lines using PEGylated nanoparticles for imaging and drug delivery in colorectal cancer, Semin. Cancer
alpha-Gd2(MoO4)3 mesoporous particles, Dalton Transactions (Cambridge, Biol. (2019) in press.
England : 2003) 43 (8) (2014) 3330–3338. [53] G.S.R. Raju, E. Pavitra, N. Merchant, H. Lee, G.L.V. Prasad, G.P. Nagaraju, Y.
[27] S. Thakkar, D. Sharma, K. Kalia, R.K. Tekade, Tumor microenvironment targeted S. Huh, Y.K. Han, Targeting autophagy in gastrointestinal malignancy by using
nanotherapeutics for cancer therapy and diagnosis: a review, Acta Biomaterialia nanomaterials as drug delivery systems, Cancer Lett. 419 (2018) 222–232.
101 (2020) 43–68. [54] J.L. Perry, K.G. Reuter, J.C. Luft, C.V. Pecot, W. Zamboni, J.M. DeSimone,
[28] R.P. Feynman, There’s plenty of room at the bottom, California Inst. Technol. Mediating passive tumor accumulation through particle size, tumor type, and
Eng. Sci. Mag. XXIII (5) (1960). location, Nano Lett. 17 (5) (2017) 2879–2886.
[29] D.J. Bharali, S.A. Mousa, Emerging nanomedicines for early cancer detection and [55] V.P. Torchilin, Passive and active drug targeting: drug delivery to tumors as an
improved treatment: current perspective and future promise, Pharmacol. Ther. example, in: Handbook of Experimental Pharmacology, 197, 2010, pp. 3–53.
128 (2) (2010) 324–335. [56] B. Farran, E. Pavitra, P. Kasa, S. Peela, G.S. Rama Raju, G.P. Nagaraju, Folate-
[30] R. Seigneuric, L. Markey, D.S. Nuyten, C. Dubernet, C.T. Evelo, E. Finot, targeted immunotherapies: passive and active strategies for cancer, Cytokine
C. Garrido, From nanotechnology to nanomedicine: applications to cancer Growth Factor Rev. 45 (2019) 45–52.
research, Curr. Mol. Med. 10 (7) (2010) 640–652. [57] J. Fang, H. Nakamura, H. Maeda, The EPR effect: unique features of tumor blood
[31] B. Flühmann, I. Ntai, G. Borchard, S. Simoens, S. Mühlebach, Nanomedicines: the vessels for drug delivery, factors involved, and limitations and augmentation of
magic bullets reaching their target? Eur. J. Pharm. Sci. 128 (2019) 73–80. the effect, Adv. Drug Deliv. Rev. 63 (3) (2011) 136–151.
[32] B.Y. Kim, J.T. Rutka, W.C. Chan, Nanomedicine, N. Engl. J. Med. 363 (25) (2010) [58] M. Durymanov, T. Kamaletdinova, S.E. Lehmann, J. Reineke, Exploiting passive
2434–2443. nanomedicine accumulation at sites of enhanced vascular permeability for non-
[33] O.C. Farokhzad, R. Langer, Nanomedicine: developing smarter therapeutic and cancerous applications, J. Control. Release 261 (2017) 10–22.
diagnostic modalities, Adv. Drug Deliv. Rev. 58 (14) (2006) 1456–1459. [60] S. Swain, P.K. Sahu, S. Beg, S.M. Babu, Nanoparticles for Cancer targeting:
[34] S. Bhaskar, F. Tian, T. Stoeger, W. Kreyling, J.M. de la Fuente, V. Grazu, P. Borm, current and future directions, Curr. Drug Deliv. 13 (8) (2016) 1290–1302.
G. Estrada, V. Ntziachristos, D. Razansky, Multifunctional Nanocarriers for [61] G. Lin, S. Chen, P. Mi, Nanoparticles targeting and remodeling tumor
diagnostics, drug delivery and targeted treatment across blood-brain barrier: microenvironment for Cancer theranostics, J. Biomed. Nanotechnol. 14 (7)
perspectives on tracking and neuroimaging, Part. Fibre Toxicol. 7 (2010) 3. (2018) 1189–1207.
[35] T.J. Webster, Projections for nanomedicine into the next decade: but is it all about [62] R. Bazak, M. Houri, S. El Achy, S. Kamel, T. Refaat, Cancer active targeting by
pharmaceuticals? Int. J. Nanomedicine 3 (1) (2008) i. nanoparticles: a comprehensive review of literature, J. Cancer Res. Clin. Oncol.
[36] M. Feng, L. Zhong, Z. Zhan, Z. Huang, J. Xiong, Enhanced antitumor efficacy of 141 (5) (2015) 769–784.
resveratrol-loaded nanocapsules in colon cancer cells: physicochemical and [63] G. De Rosa, G. Salzano, M. Caraglia, A. Abbruzzese, Nanotechnologies: a strategy
biological characterization, Eur. Rev. Med. Pharmacol. Sci. 21 (2) (2017) to overcome blood-brain barrier, Curr. Drug Metab. 13 (1) (2012) 61–69.
375–382. [64] A.G. Cattaneo, R. Gornati, E. Sabbioni, M. Chiriva-Internati, E. Cobos, M.
[37] P. Resnier, N. Galopin, Y. Sibiril, A. Clavreul, J. Cayon, A. Briganti, P. Legras, R. Jenkins, G. Bernardini, Nanotechnology and human health: risks and benefits,
A. Vessières, T. Montier, G. Jaouen, Efficient ferrocifen anticancer drug and Bcl-2 J. Appl. Toxicol. 30 (8) (2010) 730–744.
gene therapy using lipid nanocapsules on human melanoma xenograft in mouse, [65] E.S. Kawasaki, A. Player, Nanotechnology, nanomedicine, and the development
Pharmacol. Res. 126 (2017) 54–65. of new, effective therapies for cancer, Nanomed. Nanotechnol. Biol. Med. 1 (2)
[38] J.-e. Kim, Y.-J. Park, Paclitaxel-loaded hyaluronan solid nanoemulsions for (2005) 101–109.
enhanced treatment efficacy in ovarian cancer, Int. J. Nanomedicine 12 (2017) [66] S.E. McNeil, Unique Benefits of Nanotechnology to Drug Delivery and
645. Diagnostics, Characterization of Nanoparticles Intended for Drug Delivery,
[39] S.P. Kuruvilla, G. Tiruchinapally, M. ElAzzouny, M.E. ElSayed, N- Springer, 2011, pp. 3–8.
acetylgalactosamine-targeted delivery of dendrimer-doxorubicin conjugates [67] S. Nizzero, A. Ziemys, M. Ferrari, Transport barriers and oncophysics in cancer
influences doxorubicin cytotoxicity and metabolic profile in hepatic Cancer cells, treatment, Trends Cancer 4 (4) (2018) 277–280.
Adv. Healthc. Mater. 6 (5) (2017), 1601046. [68] A.M. Grabrucker, B. Ruozi, D. Belletti, F. Pederzoli, F. Forni, M.A. Vandelli,
[40] B. Liu, L. Han, J. Liu, S. Han, Z. Chen, L. Jiang, Co-delivery of paclitaxel and TOS- G. Tosi, Nanoparticle transport across the blood brain barrier, Tissue Barriers 4
cisplatin via TAT-targeted solid lipid nanoparticles with synergistic antitumor (1) (2016), e1153568.
activity against cervical cancer, Int. J. Nanomedicine 12 (2017) 955. [69] E. Perez-Herrero, A. Fernandez-Medarde, Advanced targeted therapies in cancer:
[41] C.L. Gigliotti, R. Minelli, R. Cavalli, S. Occhipinti, G. Barrera, S. Pizzimenti, drug nanocarriers, the future of chemotherapy, Eur. J. Pharm. Biopharm. 93
G. Cappellano, E. Boggio, L. Conti, R. Fantozzi, In vitro and in vivo therapeutic (2015) 52–79.
evaluation of camptothecin-encapsulated β-cyclodextrin nanosponges in prostate [70] S.C. Baetke, T. Lammers, F. Kiessling, Applications of nanoparticles for diagnosis
cancer, J. Biomed. Nanotechnol. 12 (1) (2016) 114–127. and therapy of cancer, Br. J. Radiol. 88 (1054) (2015), 20150207.
[42] K. Öztürk, G. Esendağlı, M.U. Gürbüz, M. Tülü, S. Çalış, Effective targeting of [71] J.D. Heidel, M.E. Davis, Clinical developments in nanotechnology for cancer
gemcitabine to pancreatic cancer through PEG-cored Flt-1 antibody-conjugated therapy, Pharm. Res. 28 (2) (2011) 187–199.
dendrimers, Int. J. Pharm. 517 (1-2) (2017) 157–167. [72] I. Ali, U. Rahis, K. Salim, M.A. Rather, W.A. Wani, A. Haque, Advances in nano
[43] B. Mujokoro, M. Adabi, E. Sadroddiny, M. Adabi, M. Khosravani, Nano-structures drugs for cancer chemotherapy, Curr. Cancer Drug Targets 11 (2) (2011)
mediated co-delivery of therapeutic agents for glioblastoma treatment: a review, 135–146.
Mater. Sci. Eng. C 69 (2016) 1092–1102. [73] M. Saad, O.B. Garbuzenko, E. Ber, P. Chandna, J.J. Khandare, V.P. Pozharov,
[44] N.M. Elbaz, I.A. Khalil, A.A. Abd-Rabou, I.M. El-Sherbiny, Chitosan-based nano- T. Minko, Receptor targeted polymers, dendrimers, liposomes: which nanocarrier
in-microparticle carriers for enhanced oral delivery and anticancer activity of is the most efficient for tumor-specific treatment and imaging? J. Control. Release
propolis, Int. J. Biol. Macromol. 92 (2016) 254–269. 130 (2) (2008) 107–114.
[45] A.A. Rafi, M. Mahkam, S. Davaran, H. Hamishehkar, A Smart pH-responsive [74] R. Subbiah, M. Veerapandian, K.S. Yun, Nanoparticles: functionalization and
nano-carrier as a drug delivery system: a hybrid system comprised of mesoporous multifunctional applications in biomedical sciences, Curr. Med. Chem. 17 (36)
nanosilica MCM-41 (as a nano-container) & a pH-sensitive polymer (as smart (2010) 4559–4577.
reversible gatekeepers): preparation, characterization and in vitro release studies [75] A.K. Jain, M. Das, N.K. Swarnakar, S. Jain, Engineered PLGA nanoparticles: an
of an anti-cancer drug, Eur. J. Pharm. Sci. 93 (2016) 64–73. emerging delivery tool in cancer therapeutics, Crit. Rev. Ther. Drug Carrier Syst.
[46] K. He, Y. Ma, B. Yang, C. Liang, X. Chen, C. Cai, The efficacy assessments of 28 (1) (2011) 1–45.
alkylating drugs induced by nano-Fe3O4/CA for curing breast and hepatic cancer, [76] K.T. Nguyen, Targeted Nanoparticles for Cancer Therapy: Promises and
Spectrochim. Acta A. Mol. Biomol. Spectrosc. 173 (2017) 82–86. Challenge, 2011.
[47] A.K. Rochani, S. Balasubramanian, A.R. Girija, S. Raveendran, A. Borah, [77] B.A. Cisterna, N. Kamaly, W.I. Choi, A. Tavakkoli, O.C. Farokhzad, C. Vilos,
Y. Nagaoka, Y. Nakajima, T. Maekawa, D.S. Kumar, Dual mode of cancer cell Targeted nanoparticles for colorectal cancer, Nanomedicine (London, England)
destruction for pancreatic cancer therapy using Hsp90 inhibitor loaded polymeric 11 (18) (2016) 2443–2456.
nano magnetic formulation, Int. J. Pharm. 511 (1) (2016) 648–658. [78] B. Bahrami, M. Hojjat-Farsangi, H. Mohammadi, E. Anvari, G. Ghalamfarsa,
[48] D. Yang, L. Feng, C.A. Dougherty, K.E. Luker, D. Chen, M.A. Cauble, M.M.B. Holl, M. Yousefi, F. Jadidi-Niaragh, Nanoparticles and targeted drug delivery in cancer
G.D. Luker, B.D. Ross, Z. Liu, In vivo targeting of metastatic breast cancer via therapy, Immunol. Lett. 190 (2017) 64–83.

38
H. Khan et al. Seminars in Cancer Biology 69 (2021) 24–42

[79] J. Luczkowiak, A. Munoz, M. Sanchez-Navarro, R. Ribeiro-Viana, A. Ginieis, B. [107] T.D. Farr, C.H. Lai, D. Grunstein, G. Orts-Gil, C.C. Wang, P. Boehm-Sturm, P.
M. Illescas, N. Martin, R. Delgado, J. Rojo, Glycofullerenes inhibit viral infection, H. Seeberger, C. Harms, Imaging early endothelial inflammation following stroke
Biomacromolecules 14 (2) (2013) 431–437. by core shell silica superparamagnetic glyconanoparticles that target selectin,
[80] S.J. Richards, M.W. Jones, M. Hunaban, D.M. Haddleton, M.I. Gibson, Probing Nano Lett. 14 (4) (2014) 2130–2134.
bacterial-toxin inhibition with synthetic glycopolymers prepared by tandem post- [108] T. Borase, T. Ninjbadgar, A. Kapetanakis, S. Roche, R. O’Connor, C. Kerskens,
polymerization modification: role of linker length and carbohydrate density, A. Heise, D.F. Brougham, Stable aqueous dispersions of glycopeptide-grafted
Angew. Chem. Int. Ed. Engl. 51 (31) (2012) 7812–7816. selectably functionalized magnetic nanoparticles, Angew. Chemie Int. Ed. 52 (11)
[81] R. Ribeiro-Viana, M. Sanchez-Navarro, J. Luczkowiak, J.R. Koeppe, R. Delgado, (2013) 3164–3167.
J. Rojo, B.G. Davis, Virus-like glycodendrinanoparticles displaying quasi- [109] W. Lu, W. Ma, J. Lu, X. Li, Y. Zhao, G. Chen, Microwave-assisted synthesis of
equivalent nested polyvalency upon glycoprotein platforms potently block viral glycopolymer-functionalized silver nanoclusters: combining the bioactivity of
infection, Nat. Commun. 3 (2012) 1303. sugar with the fluorescence and cytotoxicity of silver, Macromol. Rapid Commun.
[82] M. Marradi, F. Chiodo, I. Garcia, S. Penades, Glyconanoparticles as 35 (8) (2014) 827–833.
multifunctional and multimodal carbohydrate systems, Chem. Soc. Rev. 42 (11) [110] A. Sizovs, L. Xue, Z.P. Tolstyka, N.P. Ingle, Y. Wu, M. Cortez, T.M. Reineke, Poly
(2013) 4728–4745. (trehalose): sugar-coated nanocomplexes promote stabilization and effective
[83] D.P. Gamblin, E.M. Scanlan, B.G. Davis, Glycoprotein synthesis: an update, Chem. polyplex-mediated siRNA delivery, J. Am. Chem. Soc. 135 (41) (2013)
Rev. 109 (1) (2009) 131–163. 15417–15424.
[84] S.K. Podder, A. Surolia, B.K. Bachhawat, On the specificity of carbohydrate-lectin [111] M. Tranter, Y. Liu, S. He, J. Gulick, X. Ren, J. Robbins, W.K. Jones, T.M. Reineke,
recognition. The interaction of a lectin from Ricinus communis beans with simple In vivo delivery of nucleic acids via glycopolymer vehicles affords therapeutic
saccharides and concanavalin A, Eur. J. Biochem. 44 (1) (1974) 151–160. infarct size reduction in vivo, Mol. Ther. 20 (3) (2012) 601–608.
[85] K. Matsuura, M. Hibino, T. Ikeda, Y. Yamada, K. Kobayashi, Self-organized [112] V.P. Taori, H. Lu, T.M. Reineke, Structure-activity examination of poly
glycoclusters along DNA: effect of the spatial arrangement of galactoside residues (glycoamidoguanidine)s: glycopolycations containing guanidine units for nucleic
on cooperative lectin recognition, Chemistry (Weinheim an der Bergstrasse, acid delivery, Biomacromolecules 12 (6) (2011) 2055–2063.
Germany) 10 (2) (2004) 352–359. [113] D. Zhou, C. Li, Y. Hu, H. Zhou, J. Chen, Z. Zhang, T. Guo, Glycopolymer
[86] C.R. Becer, The glycopolymer code: synthesis of glycopolymers and multivalent modification on physicochemical and biological properties of poly(L-lysine) for
carbohydrate-lectin interactions, Macromol. Rapid Commun. 33 (9) (2012) gene delivery, Int. J. Biol. Macromol. 50 (4) (2012) 965–973.
742–752. [114] A. Kumari, S.K. Yadav, S.C. Yadav, Biodegradable polymeric nanoparticles based
[87] K. Babiuch, R. Wyrwa, K. Wagner, T. Seemann, S. Hoeppener, C.R. Becer, drug delivery systems, Colloids Surf. B, Biointerfaces 75 (1) (2010) 1–18.
R. Linke, M. Gottschaldt, J. Weisser, M. Schnabelrauch, U.S. Schubert, [115] M. Vert, J. Mauduit, S. Li, Biodegradation of PLA/GA polymers: increasing
Functionalized, biocompatible coating for superparamagnetic nanoparticles by complexity, Biomaterials 15 (15) (1994) 1209–1213.
controlled polymerization of a thioglycosidic monomer, Biomacromolecules 12 [116] A. Prokop, J.M. Davidson, Nanovehicular intracellular delivery systems,
(3) (2011) 681–691. J. Pharm. Sci. 97 (9) (2008) 3518–3590.
[88] S. Slavin, J. Burns, D.M. Haddleton, C.R. Becer, Synthesis of glycopolymers via [117] J.K. Vasir, V. Labhasetwar, Biodegradable nanoparticles for cytosolic delivery of
click reactions, Eur. Polym. J. 47 (4) (2011) 435–446. therapeutics, Adv. Drug Deliv. Rev. 59 (8) (2007) 718–728.
[89] Q. Zhang, L. Su, J. Collins, G. Chen, R. Wallis, D.A. Mitchell, D.M. Haddleton, C. [118] M. Berthet, Y. Gauthier, C. Lacroix, B. Verrier, C. Monge, Nanoparticle-based
R. Becer, Dendritic cell lectin-targeting sentinel-like unimolecular dressing: the future of wound treatment? Trends Biotechnol. 35 (8) (2017)
glycoconjugates to release an anti-HIV drug, J. Am. Chem. Soc. 136 (11) (2014) 770–784.
4325–4332. [119] F. Danhier, E. Ansorena, J.M. Silva, R. Coco, A. Le Breton, V. Préat, PLGA-based
[90] Y. Gou, S. Slavin, J. Geng, L. Voorhaar, D.M. Haddleton, C.R. Becer, Controlled nanoparticles: an overview of biomedical applications, J. Control. Release 161 (2)
alternate layer-by-layer assembly of lectins and glycopolymers using QCM-D, ACS (2012) 505–522.
Macro Lett. 1 (1) (2011) 180–183. [121] D. Pooja, R. Sistla, H. Kulhari, Dendrimer-drug Conjugates: Synthesis Strategies,
[91] M. Semsarilar, V. Ladmiral, S. Perrier, Highly branched and hyperbranched Stability and Application in Anticancer Drug Delivery, Design of Nanostructures
glycopolymers via reversible addition− fragmentation chain transfer for Theranostics Applications, Elsevier, 2018, pp. 277–303.
polymerization and click chemistry, Macromolecules 43 (3) (2010) 1438–1443. [122] C.A. Figg, A. Simula, K.A. Gebre, B.S. Tucker, D.M. Haddleton, B.S. Sumerlin,
[92] I. Kurtulus, G. Yilmaz, M. Ucuncu, M. Emrullahoglu, C.R. Becer, V. Bulmus, A new Polymerization-induced thermal self-assembly (PITSA), Chem. Sci. 6 (2) (2015)
proton sponge polymer synthesized by RAFT polymerization for intracellular 1230–1236.
delivery of biotherapeutics, Polym. Chem. 5 (5) (2014) 1593–1604. [123] J. An, X. Dai, Z. Wu, Y. Zhao, Z. Lu, Q. Guo, X. Zhang, C. Li, An acid-triggered
[93] G. Yilmaz, C.R. Becer, Glycopolymer code based on well-defined glycopolymers degradable and fluorescent nanoscale drug delivery system with enhanced
or glyconanomaterials and their biomolecular recognition, Front. Bioeng. cytotoxicity to cancer cells, Biomacromolecules 16 (8) (2015) 2444–2454.
Biotechnol. 2 (2014) 39. [124] H.S. Oberoi, N.V. Nukolova, F.C. Laquer, L.Y. Poluektova, J. Huang, Y. Alnouti,
[94] G. Yilmaz, C.R. Becer, Glyconanoparticles and their interactions with lectins, M. Yokohira, L.L. Arnold, A.V. Kabanov, S.M. Cohen, T.K. Bronich, Cisplatin-
Polym. Chem. 6 (31) (2015) 5503–5514. loaded core cross-linked micelles: comparative pharmacokinetics, antitumor
[95] E. Pavitra, G. Seeta Rama Raju, G.P. Nagaraju, G. Nagaraju, Y.K. Han, Y.S. Huh, J. activity, and toxicity in mice, Int. J. Nanomedicine 7 (2012) 2557–2571.
S. Yu, TPAOH assisted size-tunable Gd2O3@mSi core-shell nanostructures for [125] V.T. Huynh, J.Y. Quek, P.L. de Souza, M.H. Stenzel, Block copolymer micelles
multifunctional biomedical applications, Chemical Communications (Cambridge, with pendant bifunctional chelator for platinum drugs: effect of spacer length on
England) 54 (7) (2018) 747–750. the viability of tumor cells, Biomacromolecules 13 (4) (2012) 1010–1023.
[96] A. Robinson, J.M. Fang, P.T. Chou, K.W. Liao, R.M. Chu, S.J. Lee, Probing lectin [126] M.S. Shim, Y.J. Kwon, Stimuli-responsive polymers and nanomaterials for gene
and sperm with carbohydrate-modified quantum dots, ChemBioChem 6 (10) delivery and imaging applications, Adv. Drug Deliv. Rev. 64 (11) (2012)
(2005) 1899–1905. 1046–1059.
[97] Y. Lin, L. Zhang, W. Yao, H. Qian, D. Ding, W. Wu, X. Jiang, Water-soluble [127] H. Kawaguchi, Functional polymer microspheres, Prog. Polym. Sci. 25 (8) (2000)
chitosan-quantum dot hybrid nanospheres toward bioimaging and biolabeling, 1171–1210.
ACS Appl. Mater. Interfaces 3 (4) (2011) 995–1002. [128] J.A. Barreto, W. O’Malley, M. Kubeil, B. Graham, H. Stephan, L. Spiccia,
[98] X.L. Sun, W. Cui, C. Haller, E.L. Chaikof, Site-specific multivalent carbohydrate Nanomaterials: applications in cancer imaging and therapy, Adv. Mater.
labeling of quantum dots and magnetic beads, Chembiochem 5 (11) (2004) (Deerfield Beach, Fla.) 23 (12) (2011) H18–40.
1593–1596. [129] J. Huang, C. Bonduelle, J. Thevenot, S. Lecommandoux, A. Heise, Biologically
[99] S.Q. Chang, B. Kang, Y.D. Dai, H.X. Zhang, D. Chen, One-step fabrication of active polymersomes from amphiphilic glycopeptides, J. Am. Chem. Soc. 134 (1)
biocompatible chitosan-coated ZnS and ZnS:Mn2+ quantum dots via a gamma- (2012) 119–122.
radiation route, Nanoscale Res. Lett. 6 (1) (2011) 591. [130] S. Vandewalle, S. Wallyn, S. Chattopadhyay, C.R. Becer, F. Du Prez,
[100] P. Babu, S. Sinha, A. Surolia, Sugar-quantum dot conjugates for a selective and Thermoresponsive hyperbranched glycopolymers: synthesis, characterization and
sensitive detection of lectins, Bioconjug. Chem. 18 (1) (2007) 146–151. lectin interaction studies, Eur. Polym. J. 69 (2015) 490–498.
[102] S.I. van Kasteren, S.J. Campbell, S. Serres, D.C. Anthony, N.R. Sibson, B.G. Davis, [131] G. Yilmaz, C.R. Becer, Precision glycopolymers and their interactions with lectins,
Glyconanoparticles allow pre-symptomatic in vivo imaging of brain disease, Proc. Eur. Polym. J. 49 (10) (2013) 3046–3051.
Natl. Acad. Sci. 106 (1) (2009) 18–23. [132] J.J. Lundquist, E.J. Toone, The cluster glycoside effect, Chem. Rev. 102 (2) (2002)
[103] K. El-Boubbou, C. Gruden, X. Huang, Magnetic glyco-nanoparticles: a unique tool 555–578.
for rapid pathogen detection, decontamination, and strain differentiation, J. Am. [133] A. Aykaç, M.C. Martos-Maldonado, J.M. Casas-Solvas, I. Quesada-Soriano,
Chem. Soc. 129 (44) (2007) 13392–13393. F. García-Maroto, L. García-Fuentes, A. Vargas-Berenguel, β-Cyclodextrin-bearing
[104] N.P. Pera, A. Kouki, S. Haataja, H.M. Branderhorst, R.M. Liskamp, G.M. Visser, gold glyconanoparticles for the development of site specific drug delivery
J. Finne, R.J. Pieters, Detection of pathogenic Streptococcus suis bacteria using systems, Langmuir 30 (1) (2013) 234–242.
magnetic glycoparticles, Org. Biomol. Chem. 8 (10) (2010) 2425–2429. [134] R. Calderon-Gonzalez, H. Terán-Navarro, I. García, M. Marradi, D. Salcines-
[105] J.S. Basuki, L. Esser, H.T. Duong, Q. Zhang, P. Wilson, M.R. Whittaker, D. Cuevas, S. Yañez-Diaz, A. Solis-Angulo, E. Frande-Cabanes, M. Fariñas, A. Garcia-
M. Haddleton, C. Boyer, T.P. Davis, Magnetic nanoparticles with diblock Castaño, Gold glyconanoparticles coupled to listeriolysin O 91–99 peptide serve
glycopolymer shells give lectin concentration-dependent MRI signals and as adjuvant therapy against melanoma, Nanoscale 9 (30) (2017) 10721–10732.
selective cell uptake, Chem. Sci. 5 (2) (2014) 715–726. [135] X. Zhang, J.Z. Xing, J. Chen, L. Ko, J. Amanie, S. Gulavita, N. Pervez, D. Yee,
[106] L.-H. Liu, H. Dietsch, P. Schurtenberger, M. Yan, Photoinitiated coupling of R. Moore, W. Roa, Enhanced radiation sensitivity in prostate cancer by gold-
unmodified monosaccharides to iron oxide nanoparticles for sensing proteins and nanoparticles, Clin. Investig. Med. (2008) E160–E167.
bacteria, Bioconjug. Chem. 20 (7) (2009) 1349–1355.

39
H. Khan et al. Seminars in Cancer Biology 69 (2021) 24–42

[136] J. Conde, F. Tian, Y. Hernandez, C. Bao, P.V. Baptista, D. Cui, T. Stoeger, M. Jesus, [162] C.F. Brewer, M.C. Miceli, L.G. Baum, Clusters, bundles, arrays and lattices: novel
RNAi-based glyconanoparticles trigger apoptotic pathways for in vitro and in vivo mechanisms for lectin-saccharide-mediated cellular interactions, Curr. Opin.
enhanced cancer-cell killing, Nanoscale 7 (19) (2015) 9083–9091. Struct. Biol. 12 (5) (2002) 616–623.
[137] C. Hu, M. Niestroj, D. Yuan, S. Chang, J. Chen, Treating cancer stem cells and [163] E.K. Woller, E.D. Walter, J.R. Morgan, D.J. Singel, M.J. Cloninger, Altering the
cancer metastasis using glucose-coated gold nanoparticles, Int. J. Nanomed. 10 strength of lectin binding interactions and controlling the amount of lectin
(2015) 2065. clustering using mannose/hydroxyl-functionalized dendrimers, J. Am. Chem. Soc.
[138] A.L. Parry, N.A. Clemson, J. Ellis, S.S. Bernhard, B.G. Davis, N.R. Cameron, 125 (29) (2003) 8820–8826.
‘Multicopy multivalent’glycopolymer-stabilized gold nanoparticles as potential [164] Q.A. Besford, M. Wojnilowicz, T. Suma, Lactosylated glycogen nanoparticles for
synthetic cancer vaccines, J. Am. Chem. Soc. 135 (25) (2013) 9362–9365. targeting prostate Cancer cells, ACS Appl. Mater. Interfaces 9 (20) (2017)
[139] R.C. Qian, J. Lv, H.W. Li, Y.T. Long, Sugar-coated nanobullet: growth inhibition of 16869–16879.
cancer cells induced by metformin-loaded glyconanoparticles, ChemMedChem 12 [165] B. Lepenies, J. Lee, S. Sonkaria, Targeting C-type lectin receptors with multivalent
(22) (2017) 1823–1827. carbohydrate ligands, Adv. Drug Deliv. Rev. 65 (9) (2013) 1271–1281.
[140] G. Yilmaz, E. Guler, C. Geyik, B. Demir, M. Ozkan, D.O. Demirkol, S. Ozcelik, [166] E.I. Rigopoulou, D. Roggenbuck, D.S. Smyk, C. Liaskos, M.G. Mytilinaiou, E. Feist,
S. Timur, C.R. Becer, pH responsive glycopolymer nanoparticles for targeted K. Conrad, D.P. Bogdanos, Asialoglycoprotein receptor (ASGPR) as target
delivery of anti-cancer drugs, Mol. Syst. Des. Eng. 3 (1) (2018) 150–158. autoantigen in liver autoimmunity: lost and found, Autoimmun. Rev. 12 (2)
[141] Q.A. Besford, M. Wojnilowicz, T. Suma, N. Bertleff-Zieschang, F. Caruso, (2012) 260–269.
F. Cavalieri, Lactosylated glycogen nanoparticles for targeting prostate cancer [167] L. Fu, C. Sun, L. Yan, Galactose targeted pH-responsive copolymer conjugated
cells, ACS Appl. Mater. Interfaces 9 (20) (2017) 16869–16879. with near infrared fluorescence probe for imaging of intelligent drug delivery,
[142] C. Shao, K. Shang, H. Xu, Y. Zhang, Z. Pei, Y. Pei, Facile fabrication of hypericin- ACS Appl. Mater. Interfaces 7 (3) (2015) 2104–2115.
entrapped glyconanoparticles for targeted photodynamic therapy, Int. J. [168] J. Du, L.A. Lane, S. Nie, Stimuli-responsive nanoparticles for targeting the tumor
Nanomedicine 13 (2018) 4319. microenvironment, J. Control. Release 219 (2015) 205–214.
[143] T. Zhang, Y. Li, W. Hong, Z. Chen, P. Peng, S. Yuan, J. Qu, M. Xiao, L. Xu, Glucose [169] D.W. Dong, B. Xiang, W. Gao, Z.Z. Yang, J.Q. Li, X.R. Qi, pH-responsive
oxidase and polydopamine functionalized iron oxide nanoparticles: combination complexes using prefunctionalized polymers for synchronous delivery of
of the photothermal effect and reactive oxygen species generation for dual- doxorubicin and siRNA to cancer cells, Biomaterials 34 (20) (2013) 4849–4859.
modality selective cancer therapy, J. Mater. Chem. B 7 (13) (2019) 2190–2200. [170] G.L. Semenza, Hypoxia-inducible factors: coupling glucose metabolism and redox
[144] K. El-Boubbou, D.C. Zhu, C. Vasileiou, B. Borhan, D. Prosperi, W. Li, X. Huang, regulation with induction of the breast cancer stem cell phenotype, EMBO J. 36
Magnetic glyco-nanoparticles: a tool to detect, differentiate, and unlock the glyco- (3) (2017) 252–259.
codes of cancer via magnetic resonance imaging, J. Am. Chem. Soc. 132 (12) [171] K. Dutta, D. Hu, B. Zhao, A.E. Ribbe, Templated self-assembly of a covalent
(2010) 4490–4499. polymer network for intracellular protein delivery and traceless release, J. Am.
[145] J. Zhou, N. Hao, T. De Zoyza, M. Yan, O. Ramström, Lectin-gated, mesoporous, Chem. Soc. 139 (16) (2017) 5676–5679.
photofunctionalized glyconanoparticles for glutathione-responsive drug delivery, [172] W.H. Chen, G.F. Luo, Q. Lei, H.Z. Jia, S. Hong, Q.R. Wang, R.X. Zhuo, X.Z. Zhang,
Chem. Commun. 51 (48) (2015) 9833–9836. MMP-2 responsive polymeric micelles for cancer-targeted intracellular drug
[146] J. Wu, J. Yuan, B. Ye, Y. Wu, Z. Xu, J. Chen, J. Chen, Dual-responsive core delivery, Chem. Commun. (Cambridge, England) 51 (3) (2015) 465–468.
crosslinking glycopolymer-drug conjugates nanoparticles for precise [173] J. Shi, P.W. Kantoff, R. Wooster, O.C. Farokhzad, Cancer nanomedicine: progress,
hepatocarcinoma therapy, Front. Pharmacol. 9 (2018) 663. challenges and opportunities, Nature reviews, Cancer 17 (1) (2017) 20–37.
[147] J. Li, F.-K. Ma, Q.-F. Dang, X.-G. Liang, X.-G. Chen, Glucose-conjugated chitosan [174] H. Xu, W. Cao, X. Zhang, Selenium-containing polymers: promising biomaterials
nanoparticles for targeted drug delivery and their specific interaction with tumor for controlled release and enzyme mimics, Acc. Chem. Res. 46 (7) (2013)
cells, Front. Mater. Sci. 8 (4) (2014) 363–372. 1647–1658.
[148] J.M. de laF uente, D. Alcantara, P. Eaton, P. Crespo, T.C. Rojas, A. Fernandez, [175] W. Xu, J. Ding, Reduction-responsive polypeptide micelles for intracellular
A. Hernando, S. Penades, Gold and gold-iron oxide magnetic glyconanoparticles: delivery of antineoplastic agent, Biomacromolecules 18 (10) (2017) 3291–3301.
synthesis, characterization and magnetic properties, J. Phys. Chem. B 110 (26) [176] G. Feng, B. Kong, J. Xing, J. Chen, Enhancing multimodality functional and
(2006) 13021–13028. molecular imaging using glucose-coated gold nanoparticles, Clin. Radiol. 69 (11)
[149] J.M. de la Fuente, S. Penades, Glyconanoparticles: types, synthesis and (2014) 1105–1111.
applications in glycoscience, biomedicine and material science, Biochimica et [177] J. Wu, J. Yuan, B. Ye, Y. Wu, Z. Xu, J. Chen, J. Chen, Dual-responsive core
biophysica acta 1760 (4) (2006) 636–651. crosslinking glycopolymer-drug conjugates nanoparticles for precise
[150] M. Reynolds, M. Marradi, A. Imberty, S. Penadés, S. Pérez, Multivalent gold hepatocarcinoma therapy, Front. Pharmacol. 9 (2018) 663.
glycoclusters: high affinity molecular recognition by bacterial lectin PA-IL, Chem. [178] A.A. D’Souza, P.V. Devarajan, Asialoglycoprotein receptor mediated hepatocyte
A Eur. J. 18 (14) (2012) 4264–4273. targeting - strategies and applications, J. Control. Release 203 (2015) 126–139.
[151] J.M. de la Fuente, P. Eaton, A.G. Barrientos, M. Menendez, S. Penades, [179] J.J. Lundquist, E.J. Toone, The cluster glycoside effect, Chem. Rev. 102 (2) (2002)
Thermodynamic evidence for Ca2+-mediated self-aggregation of Lewis X gold 555–578.
glyconanoparticles. A model for cell adhesion via carbohydrate-carbohydrate [180] J. Roy, T.X. Nguyen, A.K. Kanduluru, C. Venkatesh, W. Lv, P.V. Reddy, P.S. Low,
interaction, J. Am. Chem. Soc. 127 (17) (2005) 6192–6197. M. Cushman, DUPA conjugation of a cytotoxic indenoisoquinoline topoisomerase
[152] C.L. Schofield, A.H. Haines, R.A. Field, D.A. Russell, Silver and gold I inhibitor for selective prostate cancer cell targeting, J. Med. Chem. 58 (7) (2015)
glyconanoparticles for colorimetric bioassays, Langmuir 22 (15) (2006) 3094–3103.
6707–6711. [181] J. Wang, X. Sun, W. Mao, W. Sun, J. Tang, M. Sui, Y. Shen, Z. Gu, Tumor redox
[153] J. Conde, F. Tian, Y. Hernandez, C. Bao, P.V. Baptista, D. Cui, T. Stoeger, J.M. de heterogeneity-responsive prodrug nanocapsules for cancer chemotherapy, Adv.
la Fuente, RNAi-based glyconanoparticles trigger apoptotic pathways for in vitro Mater. (Deerfield Beach, Fla.) 25 (27) (2013) 3670–3676.
and in vivo enhanced cancer-cell killing, Nanoscale 7 (19) (2015) 9083–9091. [182] R.S. Brown, R.L. Wahl, Overexpression of Glut-1 glucose transporter in human
[154] T.J. Dougherty, C.J. Gomer, B.W. Henderson, G. Jori, D. Kessel, M. Korbelik, breast cancer. An immunohistochemical study, Cancer 72 (10) (1993)
J. Moan, Q. Peng, Photodynamic therapy, J. Natl. Cancer Inst. 90 (12) (1998) 2979–2985.
889–905. [183] M.C. Daniel, D. Astruc, Gold nanoparticles: assembly, supramolecular chemistry,
[155] V. Huntosova, Z. Nadova, L. Dzurova, V. Jakusova, F. Sureau, P. Miskovsky, Cell quantum-size-related properties, and applications toward biology, catalysis, and
death response of U87 glioma cells on hypericin photoactivation is mediated by nanotechnology, Chem. Rev. 104 (1) (2004) 293–346.
dynamics of hypericin subcellular distribution and its aggregation in cellular [184] S. Su, X. Zuo, D. Pan, H. Pei, L. Wang, C. Fan, W. Huang, Design and applications
organelles, Photochem. Photobiol. Sci. 11 (9) (2012) 1428–1436. of gold nanoparticle conjugates by exploiting biomolecule-gold nanoparticle
[156] C. Shao, X. Li, Z. Pei, D. Liu, L. Wang, H. Dong, Y. Pei, Facile fabrication of interactions, Nanoscale 5 (7) (2013) 2589–2599.
glycopolymer-based iron oxide nanoparticles and their applications in the [185] X. Zhang, J.Z. Xing, J. Chen, L. Ko, J. Amanie, S. Gulavita, N. Pervez, D. Yee,
carbohydrate–lectin interaction and targeted cell imaging, Polym. Chem. 7 (6) R. Moore, W. Roa, Enhanced radiation sensitivity in prostate cancer by gold-
(2016) 1337–1344. nanoparticles, Clinical and investigative medicine, Medecine clinique et
[157] C.R. Becer, The glycopolymer code: synthesis of glycopolymers and multivalent experimentale 31 (3) (2008) E160–7.
carbohydrate–lectin interactions, Macromol. Rapid Commun. 33 (9) (2012) [186] T. Kong, J. Zeng, X. Wang, X. Yang, J. Yang, S. McQuarrie, A. McEwan, W. Roa,
742–752. J. Chen, J.Z. Xing, Enhancement of radiation cytotoxicity in breast-cancer cells by
[158] N. Jayaraman, Multivalent ligand presentation as a central concept to study localized attachment of gold nanoparticles, Small (Weinheim an der Bergstrasse,
intricate carbohydrate-protein interactions, Chem. Soc. Rev. 38 (12) (2009) Germany) 4 (9) (2008) 1537–1543.
3463–3483. [187] S. Jain, J.A. Coulter, K.T. Butterworth, A.R. Hounsell, S.J. McMahon, W.
[159] A. Martinez, C.O. Mellet, J.M.G. Fernández, Cyclodextrin-based multivalent B. Hyland, M.F. Muir, G.R. Dickson, K.M. Prise, F.J. Currell, D.G. Hirst, J.
glycodisplays: covalent and supramolecular conjugates to assess M. O’Sullivan, Gold nanoparticle cellular uptake, toxicity and radiosensitisation
carbohydrate–protein interactions, Chem. Soc. Rev. 42 (11) (2013) 4746–4773. in hypoxic conditions, Radiother. Oncol. 110 (2) (2014) 342–347.
[160] T.K. Dam, C.F. Brewer, Multivalent lectin-carbohydrate interactions energetics [188] M.B. Calvo, A. Figueroa, E.G. Pulido, R.G. Campelo, L.A. Aparicio, Potential role
and mechanisms of binding, Adv. Carbohydr. Chem. Biochem. 63 (2010) of sugar transporters in cancer and their relationship with anticancer therapy, Int.
139–164. J. Endocrinol. 2010 (2010).
[161] D. Deniaud, K. Julienne, S.G. Gouin, Insights in the rational design of synthetic [189] M.L. Macheda, S. Rogers, J.D. Best, Molecular and cellular regulation of glucose
multivalent glycoconjugates as lectin ligands, Org. Biomol. Chem. 9 (4) (2011) transporter (GLUT) proteins in cancer, J. Cell. Physiol. 202 (3) (2005) 654–662.
966–979. [190] W. Roa, X. Zhang, L. Guo, A. Shaw, X. Hu, Y. Xiong, S. Gulavita, S. Patel, X. Sun,
J. Chen, R. Moore, J.Z. Xing, Gold nanoparticle sensitize radiotherapy of prostate

40
H. Khan et al. Seminars in Cancer Biology 69 (2021) 24–42

cancer cells by regulation of the cell cycle, Nanotechnology 20 (37) (2009), [215] G. Mattheolabakis, E. Taoufik, S. Haralambous, M.L. Roberts, K. Avgoustakis, In
375101. vivo investigation of tolerance and antitumor activity of cisplatin-loaded PLGA-
[191] W. Roa, Y. Xiong, J. Chen, X. Yang, K. Song, X. Yang, B. Kong, J. Wilson, J.Z. Xing, mPEG nanoparticles, Eur. J. Pharm. Biopharm. 71 (2) (2009) 190–195.
Pharmacokinetic and toxicological evaluation of multi-functional thiol-6-fluoro- [216] E.C. Gryparis, M. Hatziapostolou, E. Papadimitriou, K. Avgoustakis, Anticancer
6-deoxy-D-glucose gold nanoparticles in vivo, Nanotechnology 23 (37) (2012), activity of cisplatin-loaded PLGA-mPEG nanoparticles on LNCaP prostate cancer
375101. cells, Eur. J. Pharm. Biopharm. 67 (1) (2007) 1–8.
[192] C. Hu, M. Niestroj, D. Yuan, S. Chang, J. Chen, Treating cancer stem cells and [217] Y. Dong, S.-S. Feng, D. Poly, L-lactide-co-glycolide)(PLGA) nanoparticles prepared
cancer metastasis using glucose-coated gold nanoparticles, Int. J. Nanomedicine by high pressure homogenization for paclitaxel chemotherapy, Int. J. Pharm. 342
10 (2015) 2065–2077. (1-2) (2007) 208–214.
[193] G. Picco, S. Julien, I. Brockhausen, R. Beatson, A. Antonopoulos, S. Haslam, [218] X. He, J. Ma, A.E. Mercado, W. Xu, E. Jabbari, Cytotoxicity of paclitaxel in
U. Mandel, A. Dell, S. Pinder, J. Taylor-Papadimitriou, J. Burchell, Over- biodegradable self-assembled core-shell poly (lactide-co-glycolide ethylene oxide
expression of ST3Gal-I promotes mammary tumorigenesis, Glycobiology 20 (10) fumarate) nanoparticles, Pharm. Res. 25 (7) (2008) 1552–1562.
(2010) 1241–1250. [219] K.Y. Win, S.-S. Feng, In vitro and in vivo studies on vitamin E TPGS-emulsified
[194] S.J. Danishefsky, J.R. Allen, From the laboratory to the clinic: a retrospective on poly (D, L-lactic-co-glycolic acid) nanoparticles for paclitaxel formulation,
fully synthetic carbohydrate-based anticancer vaccines frequently used Biomaterials 27 (10) (2006) 2285–2291.
abbreviations are listed in the appendix, Angew. Chem. Int. Ed. Engl. 39 (5) [220] H. Chen, Y. Zheng, G. Tian, Y. Tian, X. Zeng, G. Liu, K. Liu, L. Li, Z. Li, L. Mei, Oral
(2000) 836–863. delivery of DMAB-modified docetaxel-loaded PLGA-TPGS nanoparticles for
[195] S. Wittrock, T. Becker, H. Kunz, Synthetic vaccines of tumor-associated cancer chemotherapy, Nanoscale Res. Lett. 6 (1) (2011) 4.
glycopeptide antigens by immune-compatible thioether linkage to bovine serum [221] Y. Ma, Y. Zheng, K. Liu, G. Tian, Y. Tian, L. Xu, F. Yan, L. Huang, L. Mei,
albumin, Angew. Chem. Int. Ed. Engl. 46 (27) (2007) 5226–5230. Nanoparticles of poly (lactide-co-glycolide)-da-tocopheryl polyethylene glycol
[196] S. Keil, A. Kaiser, F. Syed, H. Kunz, Dendrimers of vaccines consisting of tumor- 1000 succinate random copolymer for cancer treatment, Nanoscale Res. Lett. 5
associated glycopeptide antigens and T cell epitope peptides, Synthesis 2009 (08) (7) (2010) 1161.
(2009) 1355–1369. [222] E. Ricci-Júnior, J.M. Marchetti, Preparation, characterization, photocytotoxicity
[197] A. Kaiser, N. Gaidzik, T. Becker, C. Menge, K. Groh, H. Cai, Y.M. Li, B. Gerlitzki, assay of PLGA nanoparticles containing zinc (II) phthalocyanine for
E. Schmitt, H. Kunz, Fully synthetic vaccines consisting of tumor-associated photodynamic therapy use, J. Microencapsul. 23 (5) (2006) 523–538.
MUC1 glycopeptides and a lipopeptide ligand of the Toll-like receptor 2, Angew. [223] N. Shah, K. Chaudhari, P. Dantuluri, R. Murthy, S. Das, Paclitaxel-loaded PLGA
Chem. Int. Ed. Engl. 49 (21) (2010) 3688–3692. nanoparticles surface modified with transferrin and Pluronic® P85, an in vitro
[198] I. Jeon, D. Lee, I.J. Krauss, S.J. Danishefsky, A new model for the presentation of cell line and in vivo biodistribution studies on rat model, J. Drug Target. 17 (7)
tumor-associated antigens and the quest for an anticancer vaccine: a solution to (2009) 533–542.
the synthesis challenge via ring-closing metathesis, J. Am. Chem. Soc. 131 (40) [224] X.R. Song, Z. Cai, Y. Zheng, G. He, F.Y. Cui, D.Q. Gong, S.X. Hou, S.J. Xiong, X.
(2009) 14337–14344. J. Lei, Y.Q. Wei, Reversion of multidrug resistance by co-encapsulation of
[199] P.J. Sabbatini, G. Ragupathi, C. Hood, C.A. Aghajanian, M. Juretzka, A. Iasonos, vincristine and verapamil in PLGA nanoparticles, Eur. J. Pharm. Sci. 37 (3-4)
M.L. Hensley, M.K. Spassova, O. Ouerfelli, D.R. Spriggs, W.P. Tew, J. Konner, (2009) 300–305.
H. Clausen, N. Abu Rustum, S.J. Dansihefsky, P.O. Livingston, Pilot study of a [225] M. Zeisser-Labouèbe, N. Lange, R. Gurny, F. Delie, Hypericin-loaded
heptavalent vaccine-keyhole limpet hemocyanin conjugate plus QS21 in patients nanoparticles for the photodynamic treatment of ovarian cancer, Int. J. Pharm.
with epithelial ovarian, fallopian tube, or peritoneal cancer, Clin. Cancer Res. 13 326 (1-2) (2006) 174–181.
(14) (2007) 4170–4177. [226] F. Yan, C. Zhang, Y. Zheng, L. Mei, L. Tang, C. Song, H. Sun, L. Huang, The effect
[200] R.P. Brinas, A. Sundgren, P. Sahoo, S. Morey, K. Rittenhouse-Olson, G.E. Wilding, of poloxamer 188 on nanoparticle morphology, size, cancer cell uptake, and
W. Deng, J.J. Barchi Jr., Design and synthesis of multifunctional gold cytotoxicity, Nanomedicine: nanotechnology, Biol. Med. 6 (1) (2010) 170–178.
nanoparticles bearing tumor-associated glycopeptide antigens as potential cancer [227] M.M. Yallapu, B.K. Gupta, M. Jaggi, S.C. Chauhan, Fabrication of curcumin
vaccines, Bioconjug. Chem. 23 (8) (2012) 1513–1523. encapsulated PLGA nanoparticles for improved therapeutic effects in metastatic
[201] M. Marradi, P. Di Gianvincenzo, P.M. Enriquez-Navas, O.M. Martinez-Avila, cancer cells, J. Colloid Interface Sci. 351 (1) (2010) 19–29.
F. Chiodo, E. Yuste, J. Angulo, S. Penades, Gold nanoparticles coated with [228] A. Mukerjee, J.K. Vishwanatha, Formulation, characterization and evaluation of
oligomannosides of HIV-1 glycoprotein gp120 mimic the carbohydrate epitope of curcumin-loaded PLGA nanospheres for cancer therapy, Anticancer Res. 29 (10)
antibody 2G12, J. Mol. Biol. 410 (5) (2011) 798–810. (2009) 3867–3875.
[202] J. Rojo, V. Diaz, J.M. de la Fuente, I. Segura, A.G. Barrientos, H.H. Riese, [229] T. Betancourt, B. Brown, L. Brannon-Peppas, Doxorubicin-loaded PLGA
A. Bernad, S. Penades, Gold glyconanoparticles as new tools in antiadhesive Nanoparticles by Nanoprecipitation: Preparation, Characterization and In Vitro
therapy, Chembiochem 5 (3) (2004) 291–297. Evaluation, 2, 2007, pp. 219–232.
[203] A. Sundgren, J.J. Barchi Jr., Varied presentation of the Thomsen-Friedenreich [230] G. Mattheolabakis, E. Taoufik, S. Haralambous, M.L. Roberts, K. Avgoustakis, In
disaccharide tumor-associated carbohydrate antigen on gold nanoparticles, vivo investigation of tolerance and antitumor activity of cisplatin-loaded PLGA-
Carbohydr. Res. 343 (10-11) (2008) 1594–1604. mPEG nanoparticles, Eur. J. Pharm. Biopharm. 71 (2) (2009) 190–195.
[204] A.L. Parry, N.A. Clemson, J. Ellis, S.S. Bernhard, B.G. Davis, N.R. Cameron, [231] E.C. Gryparis, M. Hatziapostolou, E. Papadimitriou, K. Avgoustakis, Anticancer
’MUlticopy multivalent’ glycopolymer-stabilized gold nanoparticles as potential activity of cisplatin-loaded PLGA-mPEG nanoparticles on LNCaP prostate cancer
synthetic cancer vaccines, J. Am. Chem. Soc. 135 (25) (2013) 9362–9365. cells, Eur. J. Pharm. Biopharm. 67 (1) (2007) 1–8.
[205] R. Dinarvand, N. Sepehri, S. Manoochehri, H. Rouhani, F. Atyabi, Polylactide-co- [232] F. Esmaeili, F. Atyabi, R. Dinarvand, Preparation of PLGA nanoparticles using
glycolide nanoparticles for controlled delivery of anticancer agents, Int. J. TPGS in the spontaneous emulsification solvent diffusion method, J. Exp.
Nanomedicine 6 (2011) 877–895. Nanosci. 2 (3) (2007) 183–192.
[206] J.-M. Lü, X. Wang, C. Marin-Muller, H. Wang, P.H. Lin, Q. Yao, C. Chen, Current [233] C. Fonseca, S. Simoes, R. Gaspar, Paclitaxel-loaded PLGA nanoparticles:
advances in research and clinical applications of PLGA-based nanotechnology, preparation, physicochemical characterization and in vitro anti-tumoral activity,
Expert Rev. Mol. Diagn. 9 (4) (2009) 325–341. J. Control. Release 83 (2) (2002) 273–286.
[207] X. Liu, V. Novosad, E.A. Rozhkova, H. Chen, V. Yefremenko, J. Pearson, M. Torno, [234] J. Xie, C.-H. Wang, Self-assembled biodegradable nanoparticles developed by
S.D. Bader, A.J. Rosengart, Surface functionalized biocompatible magnetic direct dialysis for the delivery of paclitaxel, Pharm. Res. 22 (12) (2005)
nanospheres for cancer hyperthermia, IEEE Trans. Magn. 43 (6) (2007) 2079–2090.
2462–2464. [235] F. Danhier, N. Lecouturier, B. Vroman, C. Jérôme, J. Marchand-Brynaert,
[208] F. Esmaeili, R. Dinarvand, M.H. Ghahremani, S.N. Ostad, H. Esmaily, F. Atyabi, O. Feron, V. Préat, Paclitaxel-loaded PEGylated PLGA-based nanoparticles: in
Cellular cytotoxicity and in-vivo biodistribution of docetaxel poly(lactide-co- vitro and in vivo evaluation, J. Control. Release 133 (1) (2009) 11–17.
glycolide) nanoparticles, Anticancer Drugs 21 (1) (2010) 43–52. [236] L. Romero-Ramirez, I. Garcia-Alvarez, J. Casas, M.A. Barreda-Manso, N. Yanguas-
[209] T. Musumeci, C.A. Ventura, I. Giannone, B. Ruozi, L. Montenegro, R. Pignatello, Casas, M. Nieto-Sampedro, A. Fernandez-Mayoralas, New oleyl glycoside as anti-
G. Puglisi, PLA/PLGA nanoparticles for sustained release of docetaxel, Int. J. cancer agent that targets on neutral sphingomyelinase, Biochem. Pharmacol. 97
Pharm. 325 (1-2) (2006) 172–179. (2) (2015) 158–172.
[210] J. Cheng, B.A. Teply, I. Sherifi, J. Sung, G. Luther, F.X. Gu, E. Levy-Nissenbaum, [237] I. Garcia-Alvarez, G. Corrales, E. Doncel-Perez, A. Munoz, M. Nieto-Sampedro,
A.F. Radovic-Moreno, R. Langer, O.C. Farokhzad, Formulation of functionalized A. Fernandez-Mayoralas, Design and synthesis of glycoside inhibitors of glioma
PLGA-PEG nanoparticles for in vivo targeted drug delivery, Biomaterials 28 (5) and melanoma growth, J. Med. Chem. 50 (2) (2007) 364–373.
(2007) 869–876. [238] E. Moreno-Clavijo, A.T. Carmona, Y. Vera-Ayoso, A.J. Moreno-Vargas, C. Bello,
[211] T. Betancourt, B. Brown, L. Brannon-Peppas, Doxorubicin-loaded PLGA P. Vogel, I. Robina, Synthesis of novel pyrrolidine 3,4-diol derivatives as
nanoparticles by nanoprecipitation: preparation, characterization and in vitro inhibitors of alpha-L-fucosidases, Org. Biomol. Chem. 7 (6) (2009) 1192–1202.
evaluation, Nanomedicine (London, England) 2 (2) (2007) 219–232. [239] M. Bektas, S. Spiegel, Glycosphingolipids and cell death, Glycoconj. J. 20 (1)
[212] J. van der Zee, Heating the patient: a promising approach? Ann. Oncol. 13 (8) (2004) 39–47.
(2002) 1173–1184. [240] I. Garcia-Alvarez, M. Egido-Gabas, L. Romero-Ramirez, E. Doncel-Perez, M. Nieto-
[213] P. Wust, B. Hildebrandt, G. Sreenivasa, B. Rau, J. Gellermann, H. Riess, R. Felix, Sampedro, J. Casas, A. Fernandez-Mayoralas, Lipid and ganglioside alterations in
P.M. Schlag, Hyperthermia in combined treatment of cancer, Lancet Oncol. 3 (8) tumor cells treated with antimitotic oleyl glycoside, Mol. Biosyst. 7 (1) (2011)
(2002) 487–497. 129–138.
[214] R. Manchanda, A. Fernandez-Fernandez, A. Nagesetti, A.J. McGoron, Preparation [241] S. Lahiri, A.H. Futerman, The metabolism and function of sphingolipids and
and characterization of a polymeric (PLGA) nanoparticulate drug delivery system glycosphingolipids, Cell. Mol. Life Sci. 64 (17) (2007) 2270–2284.
with simultaneous incorporation of chemotherapeutic and thermo-optical agents, [242] I. Garcia-Alvarez, H. Groult, J. Casas, M.A. Barreda-Manso, N. Yanguas-Casas,
Colloids Surf. B Biointerfaces 75 (1) (2010) 260–267. M. Nieto-Sampedro, L. Romero-Ramirez, A. Fernandez-Mayoralas, Synthesis of

41
H. Khan et al. Seminars in Cancer Biology 69 (2021) 24–42

antimitotic thioglycosides: in vitro and in vivo evaluation of their anticancer [253] J.M. Stern, J. Stanfield, W. Kabbani, J.T. Hsieh, J.A. Cadeddu, Selective prostate
activity, J. Med. Chem. 54 (19) (2011) 6949–6955. cancer thermal ablation with laser activated gold nanoshells, J. Urol. 179 (2)
[243] S. Chojnowska, A. Kepka, S.D. Szajda, N. Waszkiewicz, M. Bierc, K. Zwierz, (2008) 748–753.
Exoglycosidase markers of diseases, Biochem. Soc. Trans. 39 (1) (2011) 406–409. [254] J.A. Schwartz, A.M. Shetty, R.E. Price, R.J. Stafford, J.C. Wang, R.K. Uthamanthil,
[244] H. Driguez, J. Thiem, J. Beau, Glycoscience: Synthesis of Substrate Analogs and K. Pham, R.J. McNichols, C.L. Coleman, J.D. Payne, Feasibility study of particle-
Mimetics, Springer, 1999. assisted laser ablation of brain tumors in orthotopic canine model, Cancer Res. 69
[245] O. Veiseh, J.W. Gunn, M. Zhang, Design and fabrication of magnetic (4) (2009) 1659–1667.
nanoparticles for targeted drug delivery and imaging, Adv. Drug Deliv. Rev. 62 [255] R.R. Love, H. Leventhal, D.V. Easterling, D.R. Nerenz, Side effects and emotional
(3) (2010) 284–304. distress during cancer chemotherapy, Cancer 63 (3) (1989) 604–612.
[246] F. Herranz, B. Salinas, H. Groult, J. Pellico, A.V. Lechuga-Vieco, R. Bhavesh, [256] D.P. Dearnaley, V.S. Khoo, A.R. Norman, L. Meyer, A. Nahum, D. Tait, J. Yarnold,
J. Ruiz-Cabello, Superparamagnetic nanoparticles for atherosclerosis imaging, A. Horwich, Comparison of radiation side-effects of conformal and conventional
Nanomaterials (Basel, Switzerland) 4 (2) (2014) 408–438. radiotherapy in prostate cancer: a randomised trial, Lancet (London, England)
[247] R. Hao, R. Xing, Z. Xu, Y. Hou, S. Gao, S. Sun, Synthesis, functionalization, and 353 (9149) (1999) 267–272.
biomedical applications of multifunctional magnetic nanoparticles, Adv. Mater. [257] A.W. Lin, N.A. Lewinski, J.L. West, N.J. Halas, R.A. Drezek, Optically tunable
(Deerfield Beach, Fla.) 22 (25) (2010) 2729–2742. nanoparticle contrast agents for early cancer detection: model-based analysis of
[248] S. Laurent, D. Forge, M. Port, A. Roch, C. Robic, L. Vander Elst, R.N. Muller, gold nanoshells, J. Biomed. Opt. 10 (6) (2005), 064035.
Magnetic iron oxide nanoparticles: synthesis, stabilization, vectorization, [258] G.P. Goodrich, L. Bao, K. Gill-Sharp, K.L. Sang, J. Wang, J.D. Payne, Photothermal
physicochemical characterizations, and biological applications, Chem. Rev. 108 therapy in a murine colon cancer model using near-infrared absorbing gold
(6) (2008) 2064–2110. nanorods, J. Biomed. Opt. 15 (1) (2010), 018001.
[249] H. Groult, I. Garcia-Alvarez, Micellar Iron oxide nanoparticles coated with anti- [259] U. Prabhakar, H. Maeda, R.K. Jain, E.M. Sevick-Muraca, W. Zamboni, O.
tumor glycosides, Nanomaterials (Basel) 8 (8) (2018). C. Farokhzad, S.T. Barry, A. Gabizon, P. Grodzinski, D.C. Blakey, Challenges and
[250] A.C. Anselmo, S. Mitragotri, A review of clinical translation of inorganic key considerations of the enhanced permeability and retention effect for
nanoparticles, AAPS J. 17 (5) (2015) 1041–1054. nanomedicine drug delivery in oncology, Cancer Res. 73 (8) (2013) 2412–2417.
[251] R.H. Flocks, H.D. Kerr, H.B. Elkins, D. Culp, Treatment of carcinoma of the [260] W.J. Gradishar, S. Tjulandin, N. Davidson, H. Shaw, N. Desai, P. Bhar,
prostate by interstitial radiation with radio-active gold (Au 198): a preliminary M. Hawkins, J. O’Shaughnessy, Phase III trial of nanoparticle albumin-bound
report, J. Urol. 68 (2) (1952) 510–522. paclitaxel compared with polyethylated castor oil-based paclitaxel in women with
[252] L.R. Hirsch, R.J. Stafford, J.A. Bankson, S.R. Sershen, B. Rivera, R.E. Price, J. breast cancer, J. Clin. Oncol. 23 (31) (2005) 7794–7803.
D. Hazle, N.J. Halas, J.L. West, Nanoshell-mediated near-infrared thermal therapy [261] W. Gradishar, T. Vishalpura, M. Franklin, T. Bramley, Cost-effectiveness of
of tumors under magnetic resonance guidance, Proc. Natl. Acad. Sci. U. S. A. 100 nanoparticle albumin-bound paclitaxel versus docetaxel in the treatment of
(23) (2003) 13549–13554. metastatic breast cancer, Breast Cancer Res. Treat. 94 (2005).

42

You might also like