ACS Publications. Most Trusted. Most Cited. Most Read
My Activity
CONTENT TYPES

Figure 1Loading Img

Self-Therapeutic Nanomaterials for Cancer Therapy: A Review

  • Muhammad Adeel*
    Muhammad Adeel
    PhD School in Science and Technology of Bio and Nanomaterials, Ca’ Foscari University of Venice, Venice 30170, Italy
    Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, Venice 30170, Italy
    Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano 33081, Italy
    *Email: [email protected]. Phone: (+39)0412348910. Fax: (+39)0434659370 (M.A.).
  • Fahriye Duzagac
    Fahriye Duzagac
    Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, Venice 30170, Italy
  • Vincenzo Canzonieri
    Vincenzo Canzonieri
    Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano 33081, Italy
    Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste 34127, Italy
  • , and 
  • Flavio Rizzolio*
    Flavio Rizzolio
    Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, Venice 30170, Italy
    Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano 33081, Italy
    *Email: [email protected] Phone: (+39)0412348910 Fax: (+39)0434659370 (F.R.).
Cite this: ACS Appl. Nano Mater. 2020, 3, 6, 4962–4971
Publication Date (Web):June 3, 2020
https://doi.org/10.1021/acsanm.0c00762

Copyright © 2022 The Authors. Published by American Chemical Society. This publication is licensed under

CC-BY 4.0.
  • Open Access

Article Views

4051

Altmetric

-

Citations

LEARN ABOUT THESE METRICS
PDF (7 MB)

Abstract

Cancer is a commonly lethal disease that causes many deaths every year around the world. Many strategies have been applied to treat cancer, such as surgery, radiation, and chemotherapy, but all of these therapeutic approaches are limited. Nanotechnology could provide a tremendous platform to boost the efficacy of therapeutic systems from the bench to clinical applications. The current trend of using nanomaterials for therapeutic applications is limited to drug delivery and external stimuli-responsive systems. However, several nanomaterials can reduce the growth of aggressive tumors through their self-therapeutic properties. In this review, we discuss the self-therapeutic nanomaterials that can kill cancer cells without the need for any external stimulation (heat, light, radiation, or a magnetic field) or the loading of any extra therapeutic compounds. These nanomaterials can produce reactive oxygen species, act as deoxygenating agents, or produce free radicals at tumor sites. Self-therapeutic peptide-based and other organic nanomaterials that are used to inhibit multidrug resistance (MDR) proteins, e.g., P-glycoprotein (P-gp), are also discussed. This review discusses the possible mechanisms of action of self-therapeutic nanomaterials for cancer inhibition, highlighting critical and future aspects.

This publication is licensed under

CC-BY 4.0.
  • cc licence
  • by licence

1. Introduction

ARTICLE SECTIONS
Jump To

Cancer is the second most common cause of death around the globe. It killed 9.6 million people in 2018. According to the WHO, one in five men and one in six women will develop cancer in their life and one in eight men and one in 11 women will die from this serious disease. (1) Several methods have been developed for treatment, but there are still no available approaches that can completely eradicate this life-threatening disease. Cancer therapy is currently mostly limited to surgery, radiation, and chemotherapy, but they each have several disadvantages and often fail to cure the condition. (2)
Recently, nanomaterials have attracted much attention from scientists interested in cancer therapy because of their versatile physical and chemical properties. (3) Many reports have focused on the use of nanomaterials as carriers of therapeutic compounds. These therapeutic systems have been extended by introducing external stimuli (e.g., light, magnetic waves and heat) to improve drug release at the tumor sites. These approaches can be further subdivided into photodynamic, photothermal, magnetic, and neutron-capturing systems. Unfortunately, the use of nanomaterials as carriers of therapeutic compounds has some flaws, which preclude these therapeutic systems from clinical applications. The major challenges are a low drug loading efficiency, low solubility in an aqueous media, poor ability to cross in vivo barriers and penetrate inside the tumor (less than 1% reach the tumor), problematic physical and chemical interactions of hydrophobic therapeutic compounds with nanomaterials, in vivo instability, a suboptimal biodistribution, low tumor targeting ability, and a suboptimal drug release profile. (4) To minimize all of these issues and bring nanomaterials from the bench to clinics, scientists are trying to develop optimized self-therapeutic nanomaterials that can work like a “magic nano bullet” without the loading of additional therapeutic compounds or external stimuli dependency to make these systems practical for clinical applications.
Most of the organic nanoparticles (liposomes, micelles, exosomes, lipids, PLA, PLGA), (5) inorganic nanoparticles (gold, silver, silica, iron, graphene, carbon quantum dots), and composites (metal–organic frameworks (MOF), transition metals dichalcogenide (TMD)) were designed as carriers in drug-delivery systems (6) or for use in external stimuli-based systems such as photodynamic therapy (PDT), (7) photothermal therapy (PTT), (8) magnetic therapy, (9) and boron neutron-capturing therapy (BNCP). (10) All of these external dependencies and low loading efficiency of therapeutic compounds are shortcomings, which has led to the failure of these systems at the clinical level. Additionally, these therapeutic systems need specific special equipment that is difficult to use and requires confining the patient in the hospital.
In this review, we will go over the available data on nanotherapeutic systems derived directly from nanomaterials through some specific mechanisms (using the intrinsic properties of the materials) and discuss the possible future uses of these systems.

2. Direct Cancer Therapy Driven by Nanomaterials

ARTICLE SECTIONS
Jump To

The application of nanotechnology to cancer therapy could extend beyond drug delivery into the creation of new therapeutics able to destroy the tumors with minimal damage to healthy tissues and organs, as well as the detection and elimination of cancer cells during the initial stage of tumorigenesis. These relatively small particles can also be functionalized with ligands, nucleic acids, peptides or antibodies that bind to specific target molecules. Because of certain intrinsic properties of nanomaterials (reaction with oxygen species, heat and hazardous gas producers) and biocompatibilities, they are quite interesting to use directly for therapeutic purposes. Herein, we summarized self-therapeutic nanomaterials that were successfully used to target and eradicate cancer cells along with their possible mechanism of action in cancer treatment.

Metalloid Boron as an Enzyme Inhibitor

Neutron-capturing therapy is usually done with compounds containing boron. Locher was the first to report on the biological effects and therapeutic possibilities of boron neutron-capturing therapy (BNCT). (11) BNCT is based on nuclear capture and fusion reactions when nonirradiated boron-10 (B10) is irradiated with low energy neutrons to form 4He+7Li+ fusion energy (E). The reaction mechanism is presented below:
These nuclei produced closely spaced ionizations (the production of alpha particles and heat) of a path length of approximately one cell dimension (5–7 μm) that is used to destroy cancer cells. (12) The neutron-capturing property is limited only to boron-containing cells and external neutron radiation bombardment. (13) Several boron therapeutic compounds such as sodium borocaptate and boronophenylalanine are in clinical trials for neutron-targeted therapy, (13) but there is still a need for external stimuli by neutron bombardment to produce 4He+7Li+E.
In the literature, there are some reports in which boron-based compounds were directly used for the growth inhibition of different cancer cells. The biological roles of boron include the regulation of gene expression, growth, and proliferation. (14) Boron is used in synthetic chemistry because of its unique characteristics that allows it to form a covalent bond with carbon. Mostly, trivalent boron reagents are electrophiles and when they get a pair of electrons from nucleophiles, they adopt a tetrahedral configuration (sp3) to satisfy the octet rule. Generally, the conversion of sp2 carbonyl carbon to sp3 tetrahedral carbon inhibits the activity of enzymes. Therefore, the substitution of carbon by boron is a good transition state analogy for the suppression of hydrolytic enzymes. Additionally, boron has a strong affinity for oxygen-forming borates that are involved in the inhibition of enzymes. (15)
Bortezomib is an FDA-approved drug for treating multiple myeloma and mantle cell lymphoma. (16) The clinical development of bortezomib (trade name Velcade) occurred after the discovery of the ability of boron to reversibly inhibit the proteolytic/chymotryptic activity of the 26S proteasome subunit in mammalian cells. (17) Suppression of the intracellular protein degradation pathway (proteasomes) changes the levels of multiple intracellular signaling and regulatory proteins in addition to altering the regulation of cellular processes that can lead to growth arrest or apoptosis. In vitro studies showed that bortezomib is oxidatively metabolized primarily through multiple cytochrome P450 enzymes. The main metabolic pathway is deboronation to create two metabolites that are hydroxylated to inactive metabolites. (18)
Recently, boron compounds have gained interest as protective and therapeutic agents for prostate cancer and other cancers. (19−22) Boric acid (BA), the predominant form of boron in plasma, shows boron-mediated anticancer mechanisms in prostate cancer by reducing intracellular calcium signals and calcium storage, decreasing enzymatic activities (serine protease, NAD-dehydrogenases, etc.) and finally inhibiting cancer cell proliferation. (23)
In 2004, Barranco et al. used boric acid for the treatment of human prostate cancer cells. (14) Boron depleted media was prepared by its treatment with boron specific ion-exchange resin, which was added to the supplemented media of the DU-145 and LNCaP prostate cancer cell lines. The growth of the cancer cell lines were inhibited in a dose-dependent manner. For nontumorigenic cells, the required dose to inhibit growth was much higher. (14)
Recently, an interesting report was published by Ciofani et al. about the synthesis of hexagonal boron nitride nanoparticles for prostate cancer therapy. The authors synthesized hexagonal boron nitride nanoparticles by a tube furnace method at a high temperature (1300 °C) while using boric acid and ammonia as precursors for boron and nitrogen sources, respectively. (24) The synthesized hexagonal boron nitride nanomaterials were degradable in an aqueous media and showed a high cellular internalization profile. The mechanism of action was explained by the production of reactive oxygen species (ROS), which results in damage to membrane lipids, protein, and DNA as critical cell components, and mitochondrial dysfunction, which activates the apoptotic processes.
In 2014, Li et al. prepared hollow boron nitride nanospheres (BNs) as a boron source for prostate cancer therapy. The authors synthesized the BNs by a traditional chemical vapor deposition (CVD) method. After the CVD reaction, the precursor of BN was collected and further heated at different temperatures (900, 1025, and 1400 °C) in an argon (Ar) atmosphere to improve its crystallinity and allow for controlled boron release. The authors examined two key damage-associated pattern protein markers: caspase-3/7 to evaluate apoptosis and lactate dehydrogenase (LDH) release to evaluate necrosis. The results indicated that apoptosis and necrosis are enhanced by BN spheres compared to boric acid in vitro. The authors also investigated the in vivo anticancer effectiveness of BA and hollow BNs in subcutaneously and orthotopically injected LNCap prostate cancer cell mouse models. It was shown that BNs and BA significantly suppressed prostate tumor growth compared with the control (Figure 1). (23) Besides, taking advantage of the hollow shape of the BNs and their maximum surface to volume ratio, paclitaxel was loaded successfully to simultaneously use the system as a traditional drug-delivery system. Significant tumor inhibition was observed with BNs loaded with paclitaxel in both in vitro and in vivo experiments.

Figure 1

Figure 1. In vitro and in vivo effects of boron nitride nanospheres (BNs). (a) Different levels of necrosis (LDH) and apoptosis (caspase 3/7) in prostate cancer cells due to the release of boron from BA or hollow BN spheres. (b) BNS, BA, and saline effects on LNCap mouse tumor models. (c) Effect of different formulations of boron on the inhibition of tumor growth and (d) tumor volume in mice models. Reproduced with permission from ref (23). Copyright 2017 Springer Nature.

Boron compounds can also be effectively used in Prostate-Specific Antigen (PSA) mediated tumor growth. PSA is a serine protease and is one of the most abundant proteins secreted with chymotrypsin-like activity in human prostate epithelium and it is a well-established marker of prostate cancer. (25) PSA is thought to cleave insulin-like growth factor-binding protein-3 (IGFBP-3) and lead to tumor growth by increasing local IGF-1 levels and its mitogenic capabilities. (26) Gallardo-Williams et al. used boron supplementation to inhibit the growth and local expression of IGF-1 in human prostate adenocarcinoma cells (LNCaP) implanted in nude mice. (25) Moreover, boron-enriched diets result in inhibition of different kinds of cancers such as lung, prostate, and cervical cancers because of boron’s role in a variety of enzymatic inhibitions such as serine proteases, NAD-dehydrogenases, and mRNA splicing, as well as receptor binding mimicry and the induction of apoptosis. (21) All of these findings together have opened up a new way to cure cancer by synthesizing biocompatible self-therapeutic boron-based nanomaterials.
The common concept in all of these reports is that the arrest of cancer growth is mediated by boron atoms. Many boron-based nanomaterials such as boron nitride nanosheets, nanotubes, nanoparticles, and rare earth boride nanostructures have been synthesized by several different methods such as CVD, hydrothermal methods and electrochemical methods. (27) Boric acid and boron nitride are the most abundant forms and could be a good source of boron atoms for therapeutic applications. Because they are highly soluble in biological fluids, (23) nanomaterials with a certain crystallinity as a boron source would be a good choice for therapeutic applications. Mateti et al. synthesized different types of boron nitride nanomaterials and tested Saos-2 osteoblast-like cells for compatibility tests. (28) The authors claim that biocompatibility strongly depends on their size, shape, structure, and surface chemical properties and that cell death is caused by the unsaturated boron atoms located at the nanosheets edge or on the particle surface. Therefore, for therapeutic applications, boron-based nanomaterials should have a structure and chemical and surface properties where plenty of unsaturated boron atoms are present. The structure should be not too amorphous because boron atoms will dissolve in biological fluids before reaching the tumor sites, and not too stable since it needs to be biodegradable and provide enough boron to arrest the tumor growth, regardless of any method of synthesis. Additionally, the replacement of other atoms such as carbon with boron could be another choice to inhibit cancer-related enzymes as previously discussed.

Oxygen-Capturing Approach

Though it is obvious, the primary tumor or metastasis can grow to a size of about 1–2 mm3 if provided a sufficient supply of oxygen and nutrients by diffusion, but tumor growth beyond this size requires vascularization, a process called angiogenesis. (29) When tumors switch to an angiogenic phenotype, which refers to a phenotypic change in an early stage of tumor development necessary for growth beyond 2–3 mm in size, an angiogenic switch is triggered and attracts the growth of blood vessels. (30)
Angiogenesis is controlled by the upregulation of stimulators and the downregulation of inhibitors, which could trigger the angiogenic transduction pathway, which plays a pivotal role in tumor growth, progression, and metastasis of cancer. (31) Antiangiogenic strategies alone might not be effective enough to eradicate tumors due to the excessive compensatory mechanisms by which blood vessels will be remodeled. (32,33) Most antiangiogenic small molecule drugs cause toxicity and require the development of new strategies in a suitable delivery system. (34) Moreover, abnormal or leaky tumor vasculature during tumor angiogenesis results in poor delivery of angiogenic inhibitors, exhibiting a poor pharmacokinetic profile within the tumor stroma. In this respect, nanomedicine plays a crucial role. (35) Because of the small size and high surface volume ratio of nanotheranostic-based antitumor agents, they can be used to more effectively target tumor endothelial cells. (36) In this scenario, several investigators demonstrated novel nanotechnology-based diagnostic and therapeutic approaches for the treatment of cancer. Mukherjee and Patra provided some examples of new nanomaterials including copper, carbon, silver, gold, silica, chitosan, and peptides conjugated with antiangiogenic properties. (37) The limits of these approaches are their dependence from external drugs and stimuli and the accumulation of toxic metals or byproducts in the human body. To overcome these hurdles, Zhang et al. developed nanomaterials that can not only stop the growth of unwanted blood vessels by blocking them with their soluble byproducts but could also deprive tumor cells of oxygen, resulting in the inhibition of tumor growth. (38) It was also reported that magnesium silicide nanoparticles (MS NPS) could be used as deoxygenation agents for cancer therapy (Figure 2). (38)

Figure 2

Figure 2. A schematic illustration of intratumoral deoxygenation utilizing magnesium silicide nanoparticles (MS NPS): Reproduced with permission from ref (38). Copyright 2017 Springer Nature.

In this report, the authors synthesized injectable MS NPs by self-propagating high-temperature synthesis in an O2/Ar mixed-gas atmosphere. The excess of O2 exothermally reacted with the excess of Mg followed by a subsequent combination reaction between Mg and Si, yielding Mg2Si/MgO mixed particles with a core–shell morphology (Mg2Si core within a thin outer MgO shell). The product (Mg2Si/MgO) was purified to obtain Mg2Si and remove the MgO particles (because it could inhibit the unwanted growth of grains) and was further modified with polyvinylpyrrolidone (PVP) to rapidly absorb molecular oxygen from cancer cells by acting as a deoxygenation agent (DOA) and to block tumor capillaries from being reoxygenated. In a mouse model, tumor growth was significantly inhibited. It was claimed that the byproducts (waste) of the nanomaterials were used to cover the blood capillaries that provided oxygen to the tumor. The possible mechanism of action is presented below:
Here, the Mg2Si nanoparticles capture oxygen from the tumor and leave the bioproduct SiO2, which is useful to cover up the ends of unwanted tumor vessels and maintain intratumoral hypoxia without any long-term toxic effects. Hence, synthesizing these kinds of materials with specific properties such as oxygen capture together with the blockage of extra blood vessels by their byproducts could bring about advances in antiangiogenesis therapy.
The synthesis of other biocompatible nanomaterials for use as deoxygenation agents based on SiO2 and manganese-based materials is demanding in the current therapeutic systems. Any technique adopted to synthesize these type of materials as deoxygenation agents is worthwhile provided that the byproducts are biocompatible or safely eliminated from the body and the material (composite) has the ability to absorb certain levels of oxygen from the tumor microenvironment (one mole of material in its composite byproduct form could have the ability to consume two moles of oxygen molecules or even more). (38)

Highly Toxic Hydroxyl Radical Ions for Cancer Cell Death

The Fenton chemical reaction mechanism is utilized in cancer by converting H2O2 (present at a much higher concentration than in normal cells, ranging from 100 μM to 1 mM in the tumoral microenvironment) into highly toxic hydroxyl free radicals (OH·) to efficiently kill cancer cells and suppress tumor growth. (39−43) Several research groups have published the synthesis of different nanomaterials such as Fe2O3, MnFe2O4, (44) FeOx-MSNs, (45) silver, (46) gold, (47) and α-Fe2O3 (48) that take advantage of the Fenton reaction mechanism to kill cancer cells.
Zhang et al. developed amorphous iron nanoparticles (AFeNPs) as cancer theragnostics while taking advantage of overproduced H2O2 in the tumor using the Fenton chemical process. The AFeNPs were synthesized by a hubble-bubble technique where Fe3+ was designed to occur in a bubble film constructed from an amphiphilic block copolymer F-127 (Pluronic). The precursor solution was confined in the small space between the F-127 bilayers to limit the long-range diffusion of iron atoms and to suppress the nucleation and growth of the crystalline phase of FeNPs. The addition of PVP to increase the viscosity and a citrate chelator to decrease the concentration of free iron ions further controlled the formation of FeNPs. Finally, a freeze-drying technique was used to obtain fragments of the bubble film.
The amorphous structure and the ionization of AFeNPs enabled the on-demand release of a ferrous ion into the tumor, which led to the production of an excessive amount of OH· free radicals in the tumor microenvironment. (49) The Fenton chemical reaction with iron ions (Fe2+, Fe3+) in the presence of H2O2 to produce free radicles (OH·) is shown below.
Ranji-Burachaloo et al. synthesized a reduced iron-based metal–organic framework attached to folic acid (rMOF-FA) as a controlled released source of iron in an acidic environment (characteristic of tumor sites). The rMOF-FA was synthesized by a simple hydrothermal method where Pluronic F-127 as a surfactant and ferric chloride (FeCl3·6H2O) as precursor solutions were mixed followed by the addition of acetic acid and the precipitate was transferred into an autoclave. For the reduction of iron-based MOF, it was treated with hydroquinone that was further functionalized with folic acid (FA) by EDC/NH chemistry to obtain rMOF-FA and used for the controlled release of iron in acidic conditions. The released iron reacted with H2O2 in tumor sites to produce OH· ions that further oxidized protein lipids and DNA to decrease cell viability. The in vitro experiments showed that rMOF-FA is highly toxic to cancer cells (HeLa) because of the production of free radicals (OH·), whereas they are much less toxic for normal cells (NIH-3T3) (Figure 3). (39)

Figure 3

Figure 3. Fenton chemical approach to OH· ion therapeutic systems. (a) Schematic illustration of rMOF-FA nanoparticles for cancer therapy. (b) rMOF-FA effects on HeLa and NIH-3T3 cells. (c) Peroxidase-like activity of rMOF-FA nanoparticles. (d) Synthesis scheme of rMOF-FA nanoparticles. Reproduced with permission from ref (39). Copyright 2017 American Chemical Society.

Because the Fenton mechanism suffers from low-level production of free radicals OH· ions, many research groups have improved this approach by combining it with other therapeutic systems such as sono-fenton, (50) photofenton, (51) photothermal therapy (PTT), and chemotherapy. (52) Several other research groups also proposed different compounds besides iron, which could produce Fenton-like reactions using cations such as Mn2+, Cu1+, Cr4+, and V2+. (53) The overall compound should have the capacity to release cations gently to start a Fenton-like reaction and the material should be biodegradable enough to provide cations in the tumor microenvironment.

Self-Therapeutic Organic Nanomaterials

Organic materials such as peptide-based nanomaterials are used in medicine for therapeutic applications. The major advantage of peptide-based therapeutic systems is that they could target cancer cells with lower toxicity to normal tissues. (54) The (KLAKLAK)2 (KLAK) amphiphilic peptide is a well-known antitumor peptide widely used in cancer therapy in vitro and in vivo that suppresses the growth of aggressive tumors by damaging the membrane of mitochondria. (55) Wang et al. prepared a library of KLAK- based nanomaterials for cancer therapy by in situ polymerization and optimized the nanomaterials with different ratios and components. (56) The beclin1 peptide is another tumor suppressor peptide; however, the in vivo application of this peptide is limited because of its low stability. The stability of this peptide was improved by a supramolecular approach, conjugating poly(β-amino ester)s with the beclin1 peptide (P-Bec1), which significantly enhanced its in vivo stability, extending its circulation time and EPR effect. The (P-Bec1) nanoparticles were tested in MCF-7 breast cancer cells and showed enhanced cytotoxicity and in vivo therapeutic effects. (57)
MDM2 is an E3 ubiquitin ligase that can target p53 for polyubiquitination and proteasomal degradation. (58) For restoring p53 tumor suppression activity, the disruption of interactions between the MDM2 and p53 axis is a promising strategy. PMI, a potent inhibitor of MDM2-p53 interactions, can efficiently activate p53 and the downstream BH3-only family proteins (such as Bim, PUMA, and NOXA) to promote apoptosis by sequestering antiapoptotic Bcl-2 family proteins. However, because of its low proteolytic stability and poor cell-membrane penetration, the tumor-inhibiting activity of PMI is severely limited. (59) Yan et al. suggested combining the BIM peptide, which forms the BH3 domain of the pro-apoptotic Bim protein, with PMI in a nanoplatform-based peptide delivery system to achieve potent antitumor activity. The peptide–lanthanide clustered nanosystems (LDC-PMI-BIM-iNGR) were synthesized through thiol–poly(ethylene glycol)–thiol (SH-PEG-SH) polymer-induced molecular assembly of three peptides (PMI, BIM, and a cyclic peptide iNGR targeted to cancer cells) with biocompatible lanthanide-doped fluorescent nanoparticles (LDN). LDC-PMI-BIM-iNGR were tested in vitro and in vivo and showed lower systemic toxicity and off-target effects than doxorubicin, a commonly used chemotherapeutic agent, achieving a safe and reliable antitumor efficacy (Figure 4). (60)

Figure 4

Figure 4. Schematic diagram of peptide-based nanoclusters and their anticancer activity: (a) Graphical illustration for the construction of lanthanide-doped nanoclusters (LDC) and their cancer cell killing activity. (b) Schematic diagram of the production of small nanoparticles by the disintegration of large size nanocluster in the reducing intracellular environment. Reproduced with the permission of ref (60). Copyright 2018 American Chemical Society.

Several other self-assembled peptide-based nanomaterials for therapeutic applications have been well summarized by Qi et al. (55) and Wu et al. (54)
MDR is one of the major problems for traditional chemotherapeutic systems and one of its mechanisms is the overexpression of drug efflux pumps, such as P-glycoprotein (P-gp), which expel different agents out of the tumor cells. Several P-gp inhibitors have been developed. Tuguntaev et al. synthesized vitamin E derivatives by combining P-gp inhibitors to overcome MDR. The authors developed a nanomicellar drug-delivery system with a thin-film hydration technique as a carrier for doxorubicin in which d-α-tocopheryl polyethylene glycol 1000 succinate was used as a P-gp inhibitor. The system showed excellent cytotoxicity for in vitro and in vivo experiments. (61) In another report, Dai et al. prepared self-assembling polyethylene glycol-phosphoethanolamine-based copolymers (PEG-pp-PE) as a targeted delivery system and as an inhibitor of P-gp (Figure 5). (62) Kou et al. summarized several other examples of P-gp inhibitory nanomaterials and their applications as drug-delivery systems. (63)

Figure 5

Figure 5. P-gp inhibition by PEG-pp-PE micelles. Reproduced with permission from ref (62). Copyright 2016 American Chemical Society.

The synthesis of these organic nanomaterials always depends on considering their action to inhibit cancer tumors, e.g., peptide-based nanoclusters are used to enhance their in vivo stability and penetration abilities inside tumors. Similarly, P-gp inhibitors are conjugated to increase their in vivo stability and enhance their cytotoxicity. However, the overall synthesis of the materials should follow the same standards as defined in the literature for any drug-delivery systems such as size, shape, and surface properties, with safe elimination of byproducts.

3. Other Self-Therapeutic Nanomaterials

ARTICLE SECTIONS
Jump To

In recent years, other reports have been published about the anticancer abilities of nanomaterials and their byproducts. Recently, Li et al. published a report about the biodegradation of graphene-based nanomaterials in blood plasma and their in vitro and in vivo antitumor abilities. After incubating graphene (G) and graphene oxide (GO) in blood plasma at 37 °C for several days, the resulting nanomaterials were tested for their antitumor abilities in in vitro and in vivo experiments. (64) The results showed that the pristine graphene-based materials caused secondary structure damage to proteins and disturbances of cellular metabolic pathways while the bio transformed materials (degraded) were biocompatible and influenced specific organ therapies. The materials were further used as traditional drug-delivery systems by the loading of doxorubicin. The bio-transformed materials exhibited high efficiencies of drug delivery and more pronounced targeting and tumor-killing abilities. In another report, Law et al. used a panel of cobalt tris (bipyridine) systems having an anticancer effect with collateral sensitivity toward multidrug resistance (MDR) cancers. The authors synthesized cobalt complexes (1–6) by a simple hydrothermal method from cobaltous chloride and ammonium hexafluorophosphate as precursors. (65) To confirm the anticancer abilities of the cobalt complexes in MDR cells, the authors utilized a panel of different MDR cells with different origins. The results showed that the cobalt complexes were able to specifically induce collateral sensitivity in Taxol-resistant and p53-deficient cancer cells.
A very interesting report has also been published by Jiang et al. using sodium chloride nanoparticles (SCNPs) synthesized from sodium oleate, molybdenum chloride, and oleylamine as a surfactant. (66) Mammalian cells maintain low ratios of intracellular to extracellular sodium and chloride and high ratios of potassium and these asymmetric ionic gradients are involved in the regulation and control of cell function. (67) Reducing and increasing extracellular sodium and chloride concentrations can cause cytoskeleton destruction, cell cycle arrest, and cell lysis. In light of all this information, the authors hypothesized that NaCl nanoparticles could be used as a Trojan horse strategy to deliver ions to cells and cause a disruption of ion homeostasis. (68) The NaCl nanoparticles enter the cell through endocytosis, bypassing the physiological ion-transport system, and release Na+ and Cl ions inside cancer cells because of their high solubility, causing a surge of osmolarity and rapid cell lysis. (66)
All of these reports could support the efforts of scientists to develop self-therapeutic nanomaterials with future applications in cancer therapy.

4. Challenges and Future Perspectives

ARTICLE SECTIONS
Jump To

There is an urgent need for researchers to develop self-therapeutic nanomaterials and successfully transition them from the bench to clinics. Since these are single-step therapeutic systems, there is no need to load any extra therapeutic compounds, such as the therapeutic systems in which nanomaterials are used only as drug carriers.
Although self-therapeutic nanomaterials still contain many drawbacks of the classical nanomaterials utilized for drug-delivery systems such as bioincompatibility and poor tumor-targeting ability, (69,70) their reduced complexity could assist in faster translation to the clinic. Combining the intrinsic therapeutic features of nanomaterials (71) without any external stimuli or other agents in one nanosystem could exhibit superior anticancer efficacy and reduce side effects, providing new strategies to fight cancer.
We also need to take into account that nanomaterials could possess quantum mechanical effects. According to the theory of quantum biology, cancer driver DNA mutations develop from the tunneling of protons that change the configuration of hydrogen bonding and allow hydrogen atoms to interact abnormally with DNA molecules. (72) Self-therapeutic nanomaterials could release the energy quanta (due to their wave-particle dual nature) or produce larger vibrations that cause changes in their frequencies at tumor sites and kill cancer cells. These larger vibrations or higher frequencies in the energy states of nanoparticles can produce heat in peritumoral regions or directly inside cancer cells and be effective in tumor metastasis treatment.
Together with the catalytic and deoxygenating effects described in this review, which could create unfavorable conditions such as hypoxia, high redox stress and inflammation, self-therapeutic nanomaterials could be a leading system in clinics in the near future if all of the associated problems could be minimized and act as “all in one” “magic bullet” that specifically targets the tumor and destroys it completely in a safe manner without any pain or other side effects. However, it remains to be seen if self-therapeutic nanomaterials are more potent than other therapeutic systems and able to inhibit cancer cells; otherwise it will be hard to breakdown the clinical barriers.

5. Conclusions

ARTICLE SECTIONS
Jump To

In this review, we summarized the main available reports of self-therapeutic nanomaterials, focusing on the mechanism of action of the nanomaterials in regard to the inhibition of tumor cells. Inorganic nanomaterials that are used to kill cancer cells due to specific cancer inhibition mechanisms such as enzyme inhibition, oxygen capture approaches from tumor sites, and the production of free radicals in the tumor microenvironment were described. It was also highlighted that the self-therapeutic properties of organic nanomaterials such as self-assembled peptide-based nanomaterials and P-gp (MDR1 protein) inhibitors are useful for providing new opportunities to overcome MDR in the current therapeutic systems. Although there are several challenges related to this technology, its multiple interesting properties suggest that these therapeutic systems are very promising in regard to cancer therapy in the near future.

Author Information

ARTICLE SECTIONS
Jump To

  • Corresponding Authors
    • Muhammad Adeel - PhD School in Science and Technology of Bio and Nanomaterials, Ca’ Foscari University of Venice, Venice 30170, ItalyDepartment of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, Venice 30170, ItalyPathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano 33081, ItalyOrcidhttp://orcid.org/0000-0003-0916-4151 Email: [email protected]
    • Flavio Rizzolio - Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, Venice 30170, ItalyPathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano 33081, ItalyOrcidhttp://orcid.org/0000-0002-3400-4363 Email: [email protected]
  • Authors
    • Fahriye Duzagac - Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, Venice 30170, ItalyOrcidhttp://orcid.org/0000-0002-4130-2246
    • Vincenzo Canzonieri - Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano 33081, ItalyDepartment of Medical, Surgical and Health Sciences, University of Trieste, Trieste 34127, ItalyOrcidhttp://orcid.org/0000-0001-6010-0976
  • Funding

    This work was financially supported by Fondazione AIRC per la Ricercasul Cancro (Grant AIRC IG23566) and Ministero della Salute–Ricerca Corrente (RC2020-Line 4).

  • Notes
    The authors declare no competing financial interest.

References

ARTICLE SECTIONS
Jump To

This article references 72 other publications.

  1. 1
    Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R. L.; Torre, L. A.; Jemal, A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. Ca-Cancer J. Clin. 2018, 68 (6), 394424,  DOI: 10.3322/caac.21492
  2. 2
    Rani, R.; Kumar, V.; Rizzolio, F. Fluorescent Carbon Nanoparticles in Medicine for Cancer Therapy: An Update. ACS Med. Chem. Lett. 2018, 9 (1), 45,  DOI: 10.1021/acsmedchemlett.7b00523
  3. 3
    Palazzolo, S.; Hadla, M.; Spena, C. R.; Bayda, S.; Kumar, V.; Lo Re, F.; Adeel, M.; Caligiuri, I.; Romano, F.; Corona, G.; Canzonieri, V.; Toffoli, G.; Rizzolio, F. Proof-of-Concept Multistage Biomimetic Liposomal DNA Origami Nanosystem for the Remote Loading of Doxorubicin. ACS Med. Chem. Lett. 2019, 10 (4), 517521,  DOI: 10.1021/acsmedchemlett.8b00557
  4. 4
    Li, J.; Fan, C.; Pei, H.; Shi, J.; Huang, Q. Smart Drug Delivery Nanocarriers with Self-Assembled DNA Nanostructures. Adv. Mater. 2013, 25 (32), 43864396,  DOI: 10.1002/adma.201300875
  5. 5
    Palazzolo, S.; Bayda, S.; Hadla, M.; Caligiuri, I.; Corona, G.; Toffoli, G.; Rizzolio, F. The Clinical Translation of Organic Nanomaterials for Cancer Therapy: A Focus on Polymeric Nanoparticles, Micelles, Liposomes and Exosomes. Curr. Med. Chem. 2018, 25 (34), 42244268,  DOI: 10.2174/0929867324666170830113755
  6. 6
    Gong, L.; Yan, L.; Zhou, R.; Xie, J.; Wu, W.; Gu, Z. Two-Dimensional Transition Metal Dichalcogenide Nanomaterials for Combination Cancer Therapy. J. Mater. Chem. B 2017, 5 (10), 18731895,  DOI: 10.1039/C7TB00195A
  7. 7
    Ge, J.; Lan, M.; Zhou, B.; Liu, W.; Guo, L.; Wang, H.; Jia, Q.; Niu, G.; Huang, X.; Zhou, H.; Meng, X.; Wang, P.; Lee, C.-S.; Zhang, W.; Han, X. A Graphene Quantum Dot Photodynamic Therapy Agent with High Singlet Oxygen Generation. Nat. Commun. 2014, 5, 4596,  DOI: 10.1038/ncomms5596
  8. 8
    Vines, J. B.; Yoon, J. H.; Ryu, N. E.; Lim, D. J.; Park, H. Gold Nanoparticles for Photothermal Cancer Therapy. Front. Chem. 2019, 7 (APR), 167,  DOI: 10.3389/fchem.2019.00167
  9. 9
    Zhang, H.; Liu, X. L.; Zhang, Y. F.; Gao, F.; Li, G. L.; He, Y.; Peng, M. L.; Fan, H. M. Magnetic Nanoparticles Based Cancer Therapy: Current Status and Applications. Sci. China: Life Sci. 2018, 61 (4), 400414,  DOI: 10.1007/s11427-017-9271-1
  10. 10
    Coderre, J. A.; Makar, M. S. Radiobiology of Boron Neutron Capture Therapy: Problems with the Concept of Relative Biological Effectiveness. In Progress in Neutron Capture Therapy for Cancer; Springer US, 1992; pp 435437.
  11. 11
    LOCHER, G. L. Biological Effects and Therapeutic Possibilities of Neutron. Am. J. Roentgenol. 1936, 36, 1
  12. 12
    Kawabata, S.; Matsushita, Y.; Furuse, M.; Miyatake, S.-I.; Kuroiwa, T.; Ono, K. Clinical Study on Modified Boron Neutron Capture Therapy for Newly Diagnosed Glioblastoma. In Advances in the Biology, Imaging and Therapies for Glioblastoma; InTech, 2011; pp 325338.
  13. 13
    Barth, R. F.; Coderre, J. A.; Vicente, M. G. H.; Blue, T. E. Boron Neutron Capture Therapy of Cancer: Current Status and Future Prospects. Clin. Cancer Res. 2005, 11 (11), 39874002,  DOI: 10.1158/1078-0432.CCR-05-0035
  14. 14
    Barranco, W. T.; Eckhert, C. D. Boric Acid Inhibits Human Prostate Cancer Cell Proliferation. Cancer Lett. 2004, 216 (1), 2129,  DOI: 10.1016/j.canlet.2004.06.001
  15. 15
    Das, B. C.; Thapa, P.; Karki, R.; Schinke, C.; Das, S.; Kambhampati, S.; Banerjee, S. K.; Van Veldhuizen, P.; Verma, A.; Weiss, L. M.; Evans, T. Boron Chemicals in Diagnosis and Therapeutics. Future Med. Chem. 2013, 5 (6), 653676,  DOI: 10.4155/fmc.13.38
  16. 16
    Kane, R. C.; Bross, P. F.; Farrell, A. T.; Pazdur, R. Velcade(R): U.S. FDA Approval for the Treatment of Multiple Myeloma Progressing on Prior Therapy. Oncologist 2003, 8 (6), 508513,  DOI: 10.1634/theoncologist.8-6-508
  17. 17
    Buac, D.; Shen, M.; Schmitt, S.; Rani Kona, F.; Deshmukh, R.; Zhang, Z.; Neslund-Dudas, C.; Mitra, B.; Dou, Q. P. From Bortezomib to Other Inhibitors of the Proteasome and Beyond. Curr. Pharm. Des. 2013, 19 (22), 40254038,  DOI: 10.2174/1381612811319220012
  18. 18
    Chen, D.; Frezza, M.; Schmitt, S.; Kanwar, J.; P. Dou, Q. Bortezomib as the First Proteasome Inhibitor Anticancer Drug: Current Status and Future Perspectives. Curr. Cancer Drug Targets 2011, 11 (3), 239253,  DOI: 10.2174/156800911794519752
  19. 19
    Pizzorno, L. Nothing Boring about Boron. Integr. Med. 2015, 14 (4), 3548
  20. 20
    Barranco, W. T.; Hudak, P. F.; Eckhert, C. D. Evaluation of Ecological and in Vitro Effects of Boron on Prostate Cancer Risk (United States). Cancer Causes Control 2007, 18, 7177,  DOI: 10.1007/s10552-006-0077-8
  21. 21
    I. Scorei, R.; Popa, R. Boron-Containing Compounds as Preventive and Chemotherapeutic Agents for Cancer. Anti-Cancer Agents Med. Chem. 2010, 10 (4), 346351,  DOI: 10.2174/187152010791162289
  22. 22
    Baker, S. J.; Ding, C. Z.; Akama, T.; Zhang, Y. K.; Hernandez, V.; Xia, Y. Therapeutic Potential of Boron-Containing Compounds. Future Med. Chem. 2009, 1 (7), 12751288,  DOI: 10.4155/fmc.09.71
  23. 23
    Li, X.; Wang, X.; Zhang, J.; Hanagata, N.; Wang, X.; Weng, Q.; Ito, A.; Bando, Y.; Golberg, D. Hollow Boron Nitride Nanospheres as Boron Reservoir for Prostate Cancer Treatment. Nat. Commun. 2017, 8, 13936,  DOI: 10.1038/ncomms13936
  24. 24
    Emanet Ciofani, M.; Şen, Ö.; Culha, M. Hexagonal Boron Nitride Nanoparticles for Prostate Cancer Treatment. ACS Appl. Nano Mater. 2020, 3 (3), 23642372,  DOI: 10.1021/acsanm.9b02486
  25. 25
    Gallardo-Williams, M. T.; Chapin, R. E.; King, P. E.; Moser, G. J.; Goldsworthy, T. L.; Morrison, J. P.; Maronpot, R. R. Boron Supplementation Inhibits the Growth and Local Expression of IGF-1 in Human Prostate Adenocarcinoma (LNCaP) Tumors in Nude Mice. Toxicol. Pathol. 2004, 32 (1), 7378,  DOI: 10.1080/01926230490260899
  26. 26
    Cohen, P.; Peehl, D. M.; Graves, H. C. B.; Rosenfeld, R. G. Biological Effects of Prostate Specific Antigen as an Insulin-like Growth Factor Binding Protein-3 Protease. J. Endocrinol. 1994, 142 (3), 407415,  DOI: 10.1677/joe.0.1420407
  27. 27
    Tian, Y.; Guo, Z.; Zhang, T.; Lin, H.; Li, Z.; Chen, J.; Deng, S.; Liu, F. Inorganic Boron-Based Nanostructures: Synthesis, Optoelectronic Properties, and Prospective Applications. Nanomaterials 2019, 9 (4), 538,  DOI: 10.3390/nano9040538
  28. 28
    Mateti, S.; Wong, C. S.; Liu, Z.; Yang, W.; Li, Y.; Li, L. H.; Chen, Y. Biocompatibility of Boron Nitride Nanosheets. Nano Res. 2018, 11 (1), 334342,  DOI: 10.1007/s12274-017-1635-y
  29. 29
    Orme, M. E.; Chaplain, M. A. J. Two-Dimensional Models of Tumour Angiogenesis and Anti-Angiogenesis Strategies. IMA J. Math. Appl. Med. Biol. 1997, 14 (3), 189205,  DOI: 10.1093/imammb/14.3.189
  30. 30
    Hillen, F.; Griffioen, A. W. Tumour Vascularization: Sprouting Angiogenesis and Beyond. Cancer Metastasis Rev. 2007, 26 (3–4), 489502,  DOI: 10.1007/s10555-007-9094-7
  31. 31
    Nishida, N.; Yano, H.; Nishida, T.; Kamura, T.; Kojiro, M. Angiogenesis in Cancer. Vasc. Health Risk Manag. 2006, 2 (3), 213219,  DOI: 10.2147/vhrm.2006.2.3.213
  32. 32
    Abunahla, H.; Mohammad, B.; Alazzam, A.; Jaoude, M. A.; Al-Qutayri, M.; Abdul Hadi, S.; Al-Sarawi, S. F. MOMSense: Metal-Oxide-Metal Elementary Glucose Sensor. Sci. Rep. 2019, 9 (1), 5524,  DOI: 10.1038/s41598-019-41892-w
  33. 33
    Kerbel, R. S.; Kamen, B. A. The Anti-Angiogenic Basis of Metronomic Chemotherapy. Nat. Rev. Cancer 2004, 4 (6), 423436,  DOI: 10.1038/nrc1369
  34. 34
    Yoncheva, K.; Momekov, G. Antiangiogenic Anticancer Strategy Based on Nanoparticulate Systems. Expert Opin. Drug Delivery 2011, 8 (8), 10411056,  DOI: 10.1517/17425247.2011.585155
  35. 35
    Ma, J.; Pulfer, S.; Li, S.; Chu, J.; Reed, K.; Gallo, J. M. Pharmacodynamic-Mediated Reduction of Temozolomide Tumor Concentrations by the Angiogenesis Inhibitor TNP-470. Cancer Res. 2001, 61 (14), 54915498
  36. 36
    Desai, N. Challenges in Development of Nanoparticle-Based Therapeutics. AAPS J. 2012, 14 (2), 282295,  DOI: 10.1208/s12248-012-9339-4
  37. 37
    Mukherjee, S.; Patra, C. R. Therapeutic Application of Anti-Angiogenic Nanomaterials in Cancers. Nanoscale 2016, 8 (25), 1244412470,  DOI: 10.1039/C5NR07887C
  38. 38
    Zhang, C.; Ni, D.; Liu, Y.; Yao, H.; Bu, W.; Shi, J. Magnesium Silicide Nanoparticles as a Deoxygenation Agent for Cancer Starvation Therapy. Nat. Nanotechnol. 2017, 12 (4), 378386,  DOI: 10.1038/nnano.2016.280
  39. 39
    Ranji-Burachaloo, H.; Karimi, F.; Xie, K.; Fu, Q.; Gurr, P. A.; Dunstan, D. E.; Qiao, G. G. MOF-Mediated Destruction of Cancer Using the Cell’s Own Hydrogen Peroxide. ACS Appl. Mater. Interfaces 2017, 9 (39), 3359933608,  DOI: 10.1021/acsami.7b07981
  40. 40
    Qian, X.; Zhang, J.; Gu, Z.; Chen, Y. Nanocatalysts-Augmented Fenton Chemical Reaction for Nanocatalytic Tumor Therapy. Biomaterials 2019, 211, 113,  DOI: 10.1016/j.biomaterials.2019.04.023
  41. 41
    Halliwell, B.; Clement, M. V.; Long, L. H. Hydrogen Peroxide in the Human Body. FEBS Lett. 2000, 486 (1), 1013,  DOI: 10.1016/S0014-5793(00)02197-9
  42. 42
    Szatrowski, T. P.; Nathan, C. F. Production of Large Amounts of Hydrogen Peroxide by Human Tumor Cells. Cancer Res. 1991, 51 (3), 794798
  43. 43
    Kim, J.; Cho, H. R.; Jeon, H.; Kim, D.; Song, C.; Lee, N.; Choi, S. H.; Hyeon, T. Continuous O2-Evolving MnFe2O4 Nanoparticle-Anchored Mesoporous Silica Nanoparticles for Efficient Photodynamic Therapy in Hypoxic Cancer. J. Am. Chem. Soc. 2017, 139 (32), 1099210995,  DOI: 10.1021/jacs.7b05559
  44. 44
    Peng, Y.; Wang, Z.; Liu, W.; Zhang, H.; Zuo, W.; Tang, H.; Chen, F.; Wang, B. Size- and Shape-Dependent Peroxidase-like Catalytic Activity of MnFe2O4 Nanoparticles and Their Applications in Highly Efficient Colorimetric Detection of Target Cancer Cells. Dalt. Trans. 2015, 44 (28), 1287112877,  DOI: 10.1039/C5DT01585E
  45. 45
    Fu, J.; Shao, Y.; Wang, L.; Zhu, Y. Lysosome-Controlled Efficient ROS Overproduction against Cancer Cells with a High PH-Responsive Catalytic Nanosystem. Nanoscale 2015, 7 (16), 72757283,  DOI: 10.1039/C5NR00706B
  46. 46
    He, W.; Zhou, Y. T.; Wamer, W. G.; Boudreau, M. D.; Yin, J. J. Mechanisms of the PH Dependent Generation of Hydroxyl Radicals and Oxygen Induced by Ag Nanoparticles. Biomaterials 2012, 33 (30), 75477555,  DOI: 10.1016/j.biomaterials.2012.06.076
  47. 47
    Maji, S. K.; Mandal, A. K.; Nguyen, K. T.; Borah, P.; Zhao, Y. Cancer Cell Detection and Therapeutics Using Peroxidase-Active Nanohybrid of Gold Nanoparticle-Loaded Mesoporous Silica-Coated Graphene. ACS Appl. Mater. Interfaces 2015, 7 (18), 98079816,  DOI: 10.1021/acsami.5b01758
  48. 48
    Chen, Z.; Yin, J. J.; Zhou, Y. T.; Zhang, Y.; Song, L.; Song, M.; Hu, S.; Gu, N. Dual Enzyme-like Activities of Iron Oxide Nanoparticles and Their Implication for Diminishing Cytotoxicity. ACS Nano 2012, 6 (5), 40014012,  DOI: 10.1021/nn300291r
  49. 49
    Zhang, C.; Bu, W.; Ni, D.; Zhang, S.; Li, Q.; Yao, Z.; Zhang, J.; Yao, H.; Wang, Z.; Shi, J. Synthesis of Iron Nanometallic Glasses and Their Application in Cancer Therapy by a Localized Fenton Reaction. Angew. Chem., Int. Ed. 2016, 55 (6), 21012106,  DOI: 10.1002/anie.201510031
  50. 50
    Li, W. P.; Su, C. H.; Chang, Y. C.; Lin, Y. J.; Yeh, C. S. Ultrasound-Induced Reactive Oxygen Species Mediated Therapy and Imaging Using a Fenton Reaction Activable Polymersome. ACS Nano 2016, 10 (2), 20172027,  DOI: 10.1021/acsnano.5b06175
  51. 51
    He, Y.; Del Valle, A.; Qian, Y.; Huang, Y. F. Near Infrared Light-Mediated Enhancement of Reactive Oxygen Species Generation through Electron Transfer from Graphene Oxide to Iron Hydroxide/Oxide. Nanoscale 2017, 9 (4), 15591566,  DOI: 10.1039/C6NR08784A
  52. 52
    Ranji-Burachaloo, H.; Gurr, P. A.; Dunstan, D. E.; Qiao, G. G. Cancer Treatment through Nanoparticle-Facilitated Fenton Reaction. ACS Nano 2018, 12 (12), 1181911837,  DOI: 10.1021/acsnano.8b07635
  53. 53
    Bokare, A. D.; Choi, W. Review of Iron-Free Fenton-like Systems for Activating H2O2 in Advanced Oxidation Processes. J. Hazard. Mater. 2014, 275, 121135,  DOI: 10.1016/j.jhazmat.2014.04.054
  54. 54
    Wu, D.; Gao, Y.; Qi, Y.; Chen, L.; Ma, Y.; Li, Y. Peptide-Based Cancer Therapy: Opportunity and Challenge. Cancer Lett. 2014, 351 (1), 1322,  DOI: 10.1016/j.canlet.2014.05.002
  55. 55
    Qi, G.-B.; Gao, Y.-J.; Wang, L.; Wang, H. Self-Assembled Peptide-Based Nanomaterials for Biomedical Imaging and Therapy. Adv. Mater. 2018, 30 (22), 1703444,  DOI: 10.1002/adma.201703444
  56. 56
    Qiao, Z. Y.; Lin, Y. X.; Lai, W. J.; Hou, C. Y.; Wang, Y.; Qiao, S. L.; Zhang, D.; Fang, Q. J.; Wang, H. A General Strategy for Facile Synthesis and in Situ Screening of Self-Assembled Polymer-Peptide Nanomaterials. Adv. Mater. 2016, 28 (9), 18591867,  DOI: 10.1002/adma.201504564
  57. 57
    Wang, Y.; Lin, Y.-X.; Qiao, Z.-Y.; An, H.-W.; Qiao, S.-L.; Wang, L.; Rajapaksha, R. P. Y. J.; Wang, H. Self-Assembled Autophagy-Inducing Polymeric Nanoparticles for Breast Cancer Interference In-Vivo. Adv. Mater. 2015, 27 (16), 26272634,  DOI: 10.1002/adma.201405926
  58. 58
    Prives, C. Signaling to P53: Breaking Minireview the MDM2–P53 Circuit. Cell 1998, 95 (1), 58,  DOI: 10.1016/S0092-8674(00)81774-2
  59. 59
    Frappier, V.; Duran, M.; Keating, A. E. PixelDB: Protein–Peptide Complexes Annotated with Structural Conservation of the Peptide Binding Mode. Protein Sci. 2018, 27 (1), 276285,  DOI: 10.1002/pro.3320
  60. 60
    Yan, J.; He, W.; Yan, S.; Niu, F.; Liu, T.; Ma, B.; Shao, Y.; Yan, Y.; Yang, G.; Lu, W.; Du, Y.; Lei, B.; Ma, P. X. Self-Assembled Peptide-Lanthanide Nanoclusters for Safe Tumor Therapy: Overcoming and Utilizing Biological Barriers to Peptide Drug Delivery. ACS Nano 2018, 12 (2), 20172026,  DOI: 10.1021/acsnano.8b00081
  61. 61
    Tuguntaev, R. G.; Chen, S.; Eltahan, A. S.; Mozhi, A.; Jin, S.; Zhang, J.; Li, C.; Wang, P. C.; Liang, X.-J. P-Gp Inhibition and Mitochondrial Impairment by Dual-Functional Nanostructure Based on Vitamin E Derivatives To Overcome Multidrug Resistance. ACS Appl. Mater. Interfaces 2017, 9 (20), 1690016912,  DOI: 10.1021/acsami.7b03877
  62. 62
    Dai, Z.; Yao, Q.; Zhu, L. MMP2-Sensitive PEG-Lipid Copolymers: A New Type of Tumor-Targeted P-Glycoprotein Inhibitor. ACS Appl. Mater. Interfaces 2016, 8 (20), 1266112673,  DOI: 10.1021/acsami.6b03064
  63. 63
    Kou, L.; Sun, R.; Bhutia, Y. D.; Yao, Q.; Chen, R. Emerging Advances in P-Glycoprotein Inhibitory Nanomaterials for Drug Delivery. Expert Opin. Drug Delivery 2018, 15 (9), 869879,  DOI: 10.1080/17425247.2018.1517749
  64. 64
    Li, D.; Hu, X.; Zhang, S. Biodegradation of Graphene-Based Nanomaterials in Blood Plasma Affects Their Biocompatibility, Drug Delivery, Targeted Organs and Antitumor Ability. Biomaterials 2019, 202, 1225,  DOI: 10.1016/j.biomaterials.2019.02.020
  65. 65
    Law, B. Y. K.; Qu, Y. Q.; Mok, S. W. F.; Liu, H.; Zeng, W.; Han, Y.; Gordillo-Martinez, F.; Chan, W. K.; Wong, K. M. C.; Wong, V. K. W. New Perspectives of Cobalt Tris(Bipyridine) System: Anti-Cancer Effect and Its Collateral Sensitivity towards Multidrug-Resistant (MDR) Cancers. Oncotarget 2017, 8 (33), 5500355021,  DOI: 10.18632/oncotarget.18991
  66. 66
    Jiang, W.; Yin, L.; Chen, H.; Paschall, A. V.; Zhang, L.; Fu, W.; Zhang, W.; Todd, T.; Yu, K. S.; Zhou, S.; Zhen, Z.; Butler, M.; Yao, L.; Zhang, F.; Shen, Y.; Li, Z.; Yin, A.; Yin, H.; Wang, X.; Avci, F. Y.; Yu, X.; Xie, J. NaCl Nanoparticles as a Cancer Therapeutic. Adv. Mater. 2019, 31 (46), 1904058,  DOI: 10.1002/adma.201904058
  67. 67
    Nagy, I. Z.; Lustyik, G.; Nagy, V. Z.; Zarándi, B.; Bertoni-Freddari, C. Intracellular Na+:K+ Ratios in Human Cancer Cells as Revealed by Energy Dispersive X-Ray Microanalysis. J. Cell Biol. 1981, 90 (3), 769777,  DOI: 10.1083/jcb.90.3.769
  68. 68
    Galvez, A.; Morales, M. P.; Eltit, J. M.; Ocaranza, P.; Carrasco, L.; Campos, X.; Sapag-Hagar, M.; Díaz-Araya, G.; Lavandero, S. A Rapid and Strong Apoptotic Process Is Triggered by Hyperosmotic Stress in Cultured Rat Cardiac Myocytes. Cell Tissue Res. 2001, 304 (2), 279285,  DOI: 10.1007/s004410100358
  69. 69
    Poon, W.; Zhang, Y.-N.; Ouyang, B.; Kingston, B. R.; Wu, J. L. Y.; Wilhelm, S.; Chan, W. C. W. Elimination Pathways of Nanoparticles. ACS Nano 2019, 13 (5), 57855798,  DOI: 10.1021/acsnano.9b01383
  70. 70
    Blanco, E.; Shen, H.; Ferrari, M. Principles of Nanoparticle Design for Overcoming Biological Barriers to Drug Delivery. Nat. Biotechnol. 2015, 33 (9), 941951,  DOI: 10.1038/nbt.3330
  71. 71
    Manoharan, D.; Li, W. P.; Yeh, C. S. Advances in Controlled Gas-Releasing Nanomaterials for Therapeutic Applications. Nanoscale Horizons 2019, 4 (3), 557578,  DOI: 10.1039/C8NH00191J
  72. 72
    Löwdin, P. O. Proton Tunneling in DNA and Its Biological Implications. Rev. Mod. Phys. 1963, 35 (3), 724732,  DOI: 10.1103/RevModPhys.35.724

Cited By

This article is cited by 34 publications.

  1. Maharshi Debnath, James Forster, III, Anujan Ramesh, Ashish Kulkarni. Protein Corona Formation on Lipid Nanoparticles Negatively Affects the NLRP3 Inflammasome Activation. Bioconjugate Chemistry 2023, Article ASAP.
  2. Muhammad Adeel, Salvatore Parisi, Matteo Mauceri, Kanwal Asif, Michele Bartoletti, Fabio Puglisi, Isabella Caligiuri, Md. Mahbubur Rahman, Vincenzo Canzonieri, Flavio Rizzolio. Self-Therapeutic Cobalt Hydroxide Nanosheets (Co(OH)2 NS) for Ovarian Cancer Therapy. ACS Omega 2021, 6 (43) , 28611-28619. https://doi.org/10.1021/acsomega.1c03010
  3. Shabnam Pordel, Rachael N. Pickens, Jessica K. White. Release of CO and Production of 1O2 from a Mn-BODIPY Photoactivated CO Releasing Molecule with Visible Light. Organometallics 2021, 40 (17) , 2983-2994. https://doi.org/10.1021/acs.organomet.1c00331
  4. Abolghasem Abbasi Kajani, Shaghayegh Haghjooy Javanmard, Mohsen Asadnia, Amir Razmjou. Recent Advances in Nanomaterials Development for Nanomedicine and Cancer. ACS Applied Bio Materials 2021, 4 (8) , 5908-5925. https://doi.org/10.1021/acsabm.1c00591
  5. Shanka Walia, Arun Richard Chandrasekaran, Banani Chakraborty, Dhiraj Bhatia. Aptamer-Programmed DNA Nanodevices for Advanced, Targeted Cancer Theranostics. ACS Applied Bio Materials 2021, 4 (7) , 5392-5404. https://doi.org/10.1021/acsabm.1c00413
  6. André Perez-Potti, Manuel Rodríguez-Pérez, Ester Polo, Beatriz Pelaz, Pablo del Pino. Nanoparticle-based immunotherapeutics: From the properties of nanocores to the differential effects of administration routes. Advanced Drug Delivery Reviews 2023, 197 , 114829. https://doi.org/10.1016/j.addr.2023.114829
  7. Ruifang Han, Yu Xiao, Qianqian Bai, Chung Hang Jonathan Choi. Self-therapeutic metal-based nanoparticles for treating inflammatory diseases. Acta Pharmaceutica Sinica B 2023, 13 (5) , 1847-1865. https://doi.org/10.1016/j.apsb.2022.07.009
  8. Fatemeh Mirzaee Rad, Farzaneh Tafvizi, Hassan Noorbazargan, Alireza Iranbakhsh. Ag-doped ZnO nanoparticles synthesized through green method using Artemisia turcomanica extract induce cytotoxicity and apoptotic activities against AGS cancer cells: an in vitro study. Journal of Nanostructure in Chemistry 2023, 7 https://doi.org/10.1007/s40097-023-00528-2
  9. Zia Ul Haq Khan, Taj Malook Khan, Amjad Khan, Noor Samad Shah, Nawshad Muhammad, Kamran Tahir, Jibran Iqbal, Abdur Rahim, Syed Khasim, Iftikhar Ahmad, Khadija Shabbir, Noor Shad Gul, Jianbo Wu. Brief review: Applications of nanocomposite in electrochemical sensor and drugs delivery. Frontiers in Chemistry 2023, 11 https://doi.org/10.3389/fchem.2023.1152217
  10. Bhoomi S. Shah, Jolly B. Raval, Deepak Kumar, Sunil H. Chaki, M.P. Deshpande. A review on ternary CuFeS2 compound: Fabrication strategies and applications. Journal of Alloys and Compounds 2023, 938 , 168566. https://doi.org/10.1016/j.jallcom.2022.168566
  11. Kanwal Asif, Muhammad Adeel, Md. Mahbubur Rahman, Andrea Augusto Sfriso, Michele Bartoletti, Vincenzo Canzonieri, Flavio Rizzolio, Isabella Caligiuri. Silver nitroprusside as an efficient chemodynamic therapeutic agent and a peroxynitrite nanogenerator for targeted cancer therapies. Journal of Advanced Research 2023, 58 https://doi.org/10.1016/j.jare.2023.03.005
  12. Xinai Zhang, Zhenzhong Wang, Xu Li, Wei Xiao, Xiaobo Zou, Qilin Huang, Lili Zhou. Competitive electrochemical sensing for cancer cell evaluation based on thionine-interlinked signal probes. The Analyst 2023, 148 (4) , 912-918. https://doi.org/10.1039/D2AN01599D
  13. Nika Kučuk, Mateja Primožič, Željko Knez, Maja Leitgeb. Sustainable Biodegradable Biopolymer-Based Nanoparticles for Healthcare Applications. International Journal of Molecular Sciences 2023, 24 (4) , 3188. https://doi.org/10.3390/ijms24043188
  14. P K Gupta. Nanotechnology in Cancer Therapy. 2023, 143-157. https://doi.org/10.1007/978-3-031-24287-8_9
  15. Mahsa Shahriari, Prashant Kesharwani, Amirhossein Sahebkar. Aptamer-based theranostic approaches for treatment of cancer. 2023, 433-454. https://doi.org/10.1016/B978-0-323-85881-6.00016-6
  16. Ruggero Belluomo, Azin Khodaei, Saber Amin Yavari. Additively manufactured Bi-functionalized bioceramics for reconstruction of bone tumor defects. Acta Biomaterialia 2023, 156 , 234-249. https://doi.org/10.1016/j.actbio.2022.08.042
  17. Zhou Xu, Yan Zheng, Ranran Qiao, Shengrui Cao, Yuan Fang, Xiumei Bo, Huilong Zhu, Changjiang Ying, Ying Sun, Junnian Zheng. Organic fluorescent probe for concurrent imaging and apoptosis induction in cancer cells in vivo and in vitro by utilizing endogenous hydrogen sulfide. Chemical Engineering Journal 2023, 456 , 141000. https://doi.org/10.1016/j.cej.2022.141000
  18. Sadaf Hameed, Pravin Bhattarai, Zhuoran Gong, Xiaolong Liang, Xiuli Yue, Zhifei Dai. Ultrasmall porphyrin-silica core–shell dots for enhanced fluorescence imaging-guided cancer photodynamic therapy. Nanoscale Advances 2022, 5 (1) , 277-289. https://doi.org/10.1039/D2NA00704E
  19. Adrija Sinha, Faizan Zarreen Simnani, Dibyangshee Singh, Aditya Nandi, Anmol Choudhury, Paritosh Patel, Ealisha Jha, Raghuraj Singh chouhan, Nagendra Kumar Kaushik, Yogendra Kumar Mishra, Pritam Kumar Panda, Mrutyunjay Suar, Suresh K. Verma. The translational paradigm of nanobiomaterials: Biological chemistry to modern applications. Materials Today Bio 2022, 17 , 100463. https://doi.org/10.1016/j.mtbio.2022.100463
  20. Dhananjay Yadav, Minseok Kwak, Pallavi Singh Chauhan, Nidhi Puranik, Peter C.W. Lee, Jun-O Jin. Cancer immunotherapy by immune checkpoint blockade and its advanced application using bio-nanomaterials. Seminars in Cancer Biology 2022, 86 , 909-922. https://doi.org/10.1016/j.semcancer.2022.02.016
  21. Shihai Wang, Yu Zhou, Wangwang Tao, Jingyi Liu, Hongxiang Chen, Zuyi Zhao. Fe3O4-modified amphiphilic polyurethane nanoparticles with good stability as magnetic-targeted drug carriers. Polymer Bulletin 2022, 79 (10) , 8617-8631. https://doi.org/10.1007/s00289-021-03931-3
  22. Jyotsnamayee Nayak, Kumari Sunita Prajapati, Shashank Kumar, Suban K. Sahoo, Rajender Kumar. Synthesis of thiolated chlorogenic acid-capped silver nanoparticles for the effective dual action towards antimicrobial and anticancer therapy. Colloid and Polymer Science 2022, 300 (9) , 1037-1047. https://doi.org/10.1007/s00396-022-05010-z
  23. Ganesh Gollavelli, Anil V. Ghule, Yong-Chien Ling. Multimodal Imaging and Phototherapy of Cancer and Bacterial Infection by Graphene and Related Nanocomposites. Molecules 2022, 27 (17) , 5588. https://doi.org/10.3390/molecules27175588
  24. Mohammad A. Obeid, Mohammed Al Qaraghuli, Marta Ruano, Sirikwan Sangboonruang, Manal Alsaadi, Yingmanee Tragoolpua, Valerie A. Ferro. Lipid-Based Nanomaterials in Cancer Treatment and Diagnosis. 2022, 49-83. https://doi.org/10.2174/9789815051278122010005
  25. Syavash Azaraz, Ali Es‐haghi, Ali Neamati. Anti‐angiogenic and anticancer activities of the nanoemulsions synthesized from Trachyspermum ammi L. tincture against human colon adenorectal carcinoma cells. Micro & Nano Letters 2022, 17 (6) , 139-147. https://doi.org/10.1049/mna2.12115
  26. Muhammad Adeel, Gloria Saorin, Giacomo Boccalon, Andrea Augusto Sfriso, Salvatore Parisi, Isabella Moro, Stefano Palazzolo, Isabella Caligiuri, Carlotta Granchi, Giuseppe Corona, Maja Cemazar, Vincenzo Canzonieri, Tiziano Tuccinardi, Flavio Rizzolio. A carrier free delivery system of a monoacylglycerol lipase hydrophobic inhibitor. International Journal of Pharmaceutics 2022, 613 , 121374. https://doi.org/10.1016/j.ijpharm.2021.121374
  27. Vancha Harish, Devesh Tewari, Manish Gaur, Awadh Bihari Yadav, Shiv Swaroop, Mikhael Bechelany, Ahmed Barhoum. Review on Nanoparticles and Nanostructured Materials: Bioimaging, Biosensing, Drug Delivery, Tissue Engineering, Antimicrobial, and Agro-Food Applications. Nanomaterials 2022, 12 (3) , 457. https://doi.org/10.3390/nano12030457
  28. Kochurani K. Johnson, Pramod Koshy, Jia‐Lin Yang, Charles C. Sorrell. Preclinical Cancer Theranostics—From Nanomaterials to Clinic: The Missing Link. Advanced Functional Materials 2021, 31 (43) https://doi.org/10.1002/adfm.202104199
  29. Fakhara Sabir, Mahira Zeeshan, Ushna Laraib, Mahmood Barani, Abbas Rahdar, Magali Cucchiarini, Sadanand Pandey. DNA Based and Stimuli-Responsive Smart Nanocarrier for Diagnosis and Treatment of Cancer: Applications and Challenges. Cancers 2021, 13 (14) , 3396. https://doi.org/10.3390/cancers13143396
  30. Yizhou Zhang, Hongling Lyu. Application of biosensors based on nanomaterials in cancer cell detection. Journal of Physics: Conference Series 2021, 1948 (1) , 012149. https://doi.org/10.1088/1742-6596/1948/1/012149
  31. Yufei Xue, Hua Bai, Bo Peng, Bin Fang, Jonathan Baell, Lin Li, Wei Huang, Nicolas Hans Voelcker. Stimulus-cleavable chemistry in the field of controlled drug delivery. Chemical Society Reviews 2021, 50 (8) , 4872-4931. https://doi.org/10.1039/D0CS01061H
  32. Charlene Andraos, Mary Gulumian. Intracellular and extracellular targets as mechanisms of cancer therapy by nanomaterials in relation to their physicochemical properties. WIREs Nanomedicine and Nanobiotechnology 2021, 13 (2) https://doi.org/10.1002/wnan.1680
  33. Chun-Yan Shih, Pei-Ting Wang, Wu-Chou Su, Hsisheng Teng, Wei-Lun Huang. Nanomedicine-Based Strategies Assisting Photodynamic Therapy for Hypoxic Tumors: State-of-the-Art Approaches and Emerging Trends. Biomedicines 2021, 9 (2) , 137. https://doi.org/10.3390/biomedicines9020137
  34. Mingming Song, Chang Liu, Siyu Chen, Wenxiang Zhang. Nanocarrier-Based Drug Delivery for Melanoma Therapeutics. International Journal of Molecular Sciences 2021, 22 (4) , 1873. https://doi.org/10.3390/ijms22041873
  • Abstract

    Figure 1

    Figure 1. In vitro and in vivo effects of boron nitride nanospheres (BNs). (a) Different levels of necrosis (LDH) and apoptosis (caspase 3/7) in prostate cancer cells due to the release of boron from BA or hollow BN spheres. (b) BNS, BA, and saline effects on LNCap mouse tumor models. (c) Effect of different formulations of boron on the inhibition of tumor growth and (d) tumor volume in mice models. Reproduced with permission from ref (23). Copyright 2017 Springer Nature.

    Figure 2

    Figure 2. A schematic illustration of intratumoral deoxygenation utilizing magnesium silicide nanoparticles (MS NPS): Reproduced with permission from ref (38). Copyright 2017 Springer Nature.

    Figure 3

    Figure 3. Fenton chemical approach to OH· ion therapeutic systems. (a) Schematic illustration of rMOF-FA nanoparticles for cancer therapy. (b) rMOF-FA effects on HeLa and NIH-3T3 cells. (c) Peroxidase-like activity of rMOF-FA nanoparticles. (d) Synthesis scheme of rMOF-FA nanoparticles. Reproduced with permission from ref (39). Copyright 2017 American Chemical Society.

    Figure 4

    Figure 4. Schematic diagram of peptide-based nanoclusters and their anticancer activity: (a) Graphical illustration for the construction of lanthanide-doped nanoclusters (LDC) and their cancer cell killing activity. (b) Schematic diagram of the production of small nanoparticles by the disintegration of large size nanocluster in the reducing intracellular environment. Reproduced with the permission of ref (60). Copyright 2018 American Chemical Society.

    Figure 5

    Figure 5. P-gp inhibition by PEG-pp-PE micelles. Reproduced with permission from ref (62). Copyright 2016 American Chemical Society.

  • References

    ARTICLE SECTIONS
    Jump To

    This article references 72 other publications.

    1. 1
      Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R. L.; Torre, L. A.; Jemal, A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. Ca-Cancer J. Clin. 2018, 68 (6), 394424,  DOI: 10.3322/caac.21492
    2. 2
      Rani, R.; Kumar, V.; Rizzolio, F. Fluorescent Carbon Nanoparticles in Medicine for Cancer Therapy: An Update. ACS Med. Chem. Lett. 2018, 9 (1), 45,  DOI: 10.1021/acsmedchemlett.7b00523
    3. 3
      Palazzolo, S.; Hadla, M.; Spena, C. R.; Bayda, S.; Kumar, V.; Lo Re, F.; Adeel, M.; Caligiuri, I.; Romano, F.; Corona, G.; Canzonieri, V.; Toffoli, G.; Rizzolio, F. Proof-of-Concept Multistage Biomimetic Liposomal DNA Origami Nanosystem for the Remote Loading of Doxorubicin. ACS Med. Chem. Lett. 2019, 10 (4), 517521,  DOI: 10.1021/acsmedchemlett.8b00557
    4. 4
      Li, J.; Fan, C.; Pei, H.; Shi, J.; Huang, Q. Smart Drug Delivery Nanocarriers with Self-Assembled DNA Nanostructures. Adv. Mater. 2013, 25 (32), 43864396,  DOI: 10.1002/adma.201300875
    5. 5
      Palazzolo, S.; Bayda, S.; Hadla, M.; Caligiuri, I.; Corona, G.; Toffoli, G.; Rizzolio, F. The Clinical Translation of Organic Nanomaterials for Cancer Therapy: A Focus on Polymeric Nanoparticles, Micelles, Liposomes and Exosomes. Curr. Med. Chem. 2018, 25 (34), 42244268,  DOI: 10.2174/0929867324666170830113755
    6. 6
      Gong, L.; Yan, L.; Zhou, R.; Xie, J.; Wu, W.; Gu, Z. Two-Dimensional Transition Metal Dichalcogenide Nanomaterials for Combination Cancer Therapy. J. Mater. Chem. B 2017, 5 (10), 18731895,  DOI: 10.1039/C7TB00195A
    7. 7
      Ge, J.; Lan, M.; Zhou, B.; Liu, W.; Guo, L.; Wang, H.; Jia, Q.; Niu, G.; Huang, X.; Zhou, H.; Meng, X.; Wang, P.; Lee, C.-S.; Zhang, W.; Han, X. A Graphene Quantum Dot Photodynamic Therapy Agent with High Singlet Oxygen Generation. Nat. Commun. 2014, 5, 4596,  DOI: 10.1038/ncomms5596
    8. 8
      Vines, J. B.; Yoon, J. H.; Ryu, N. E.; Lim, D. J.; Park, H. Gold Nanoparticles for Photothermal Cancer Therapy. Front. Chem. 2019, 7 (APR), 167,  DOI: 10.3389/fchem.2019.00167
    9. 9
      Zhang, H.; Liu, X. L.; Zhang, Y. F.; Gao, F.; Li, G. L.; He, Y.; Peng, M. L.; Fan, H. M. Magnetic Nanoparticles Based Cancer Therapy: Current Status and Applications. Sci. China: Life Sci. 2018, 61 (4), 400414,  DOI: 10.1007/s11427-017-9271-1
    10. 10
      Coderre, J. A.; Makar, M. S. Radiobiology of Boron Neutron Capture Therapy: Problems with the Concept of Relative Biological Effectiveness. In Progress in Neutron Capture Therapy for Cancer; Springer US, 1992; pp 435437.
    11. 11
      LOCHER, G. L. Biological Effects and Therapeutic Possibilities of Neutron. Am. J. Roentgenol. 1936, 36, 1
    12. 12
      Kawabata, S.; Matsushita, Y.; Furuse, M.; Miyatake, S.-I.; Kuroiwa, T.; Ono, K. Clinical Study on Modified Boron Neutron Capture Therapy for Newly Diagnosed Glioblastoma. In Advances in the Biology, Imaging and Therapies for Glioblastoma; InTech, 2011; pp 325338.
    13. 13
      Barth, R. F.; Coderre, J. A.; Vicente, M. G. H.; Blue, T. E. Boron Neutron Capture Therapy of Cancer: Current Status and Future Prospects. Clin. Cancer Res. 2005, 11 (11), 39874002,  DOI: 10.1158/1078-0432.CCR-05-0035
    14. 14
      Barranco, W. T.; Eckhert, C. D. Boric Acid Inhibits Human Prostate Cancer Cell Proliferation. Cancer Lett. 2004, 216 (1), 2129,  DOI: 10.1016/j.canlet.2004.06.001
    15. 15
      Das, B. C.; Thapa, P.; Karki, R.; Schinke, C.; Das, S.; Kambhampati, S.; Banerjee, S. K.; Van Veldhuizen, P.; Verma, A.; Weiss, L. M.; Evans, T. Boron Chemicals in Diagnosis and Therapeutics. Future Med. Chem. 2013, 5 (6), 653676,  DOI: 10.4155/fmc.13.38
    16. 16
      Kane, R. C.; Bross, P. F.; Farrell, A. T.; Pazdur, R. Velcade(R): U.S. FDA Approval for the Treatment of Multiple Myeloma Progressing on Prior Therapy. Oncologist 2003, 8 (6), 508513,  DOI: 10.1634/theoncologist.8-6-508
    17. 17
      Buac, D.; Shen, M.; Schmitt, S.; Rani Kona, F.; Deshmukh, R.; Zhang, Z.; Neslund-Dudas, C.; Mitra, B.; Dou, Q. P. From Bortezomib to Other Inhibitors of the Proteasome and Beyond. Curr. Pharm. Des. 2013, 19 (22), 40254038,  DOI: 10.2174/1381612811319220012
    18. 18
      Chen, D.; Frezza, M.; Schmitt, S.; Kanwar, J.; P. Dou, Q. Bortezomib as the First Proteasome Inhibitor Anticancer Drug: Current Status and Future Perspectives. Curr. Cancer Drug Targets 2011, 11 (3), 239253,  DOI: 10.2174/156800911794519752
    19. 19
      Pizzorno, L. Nothing Boring about Boron. Integr. Med. 2015, 14 (4), 3548
    20. 20
      Barranco, W. T.; Hudak, P. F.; Eckhert, C. D. Evaluation of Ecological and in Vitro Effects of Boron on Prostate Cancer Risk (United States). Cancer Causes Control 2007, 18, 7177,  DOI: 10.1007/s10552-006-0077-8
    21. 21
      I. Scorei, R.; Popa, R. Boron-Containing Compounds as Preventive and Chemotherapeutic Agents for Cancer. Anti-Cancer Agents Med. Chem. 2010, 10 (4), 346351,  DOI: 10.2174/187152010791162289
    22. 22
      Baker, S. J.; Ding, C. Z.; Akama, T.; Zhang, Y. K.; Hernandez, V.; Xia, Y. Therapeutic Potential of Boron-Containing Compounds. Future Med. Chem. 2009, 1 (7), 12751288,  DOI: 10.4155/fmc.09.71
    23. 23
      Li, X.; Wang, X.; Zhang, J.; Hanagata, N.; Wang, X.; Weng, Q.; Ito, A.; Bando, Y.; Golberg, D. Hollow Boron Nitride Nanospheres as Boron Reservoir for Prostate Cancer Treatment. Nat. Commun. 2017, 8, 13936,  DOI: 10.1038/ncomms13936
    24. 24
      Emanet Ciofani, M.; Şen, Ö.; Culha, M. Hexagonal Boron Nitride Nanoparticles for Prostate Cancer Treatment. ACS Appl. Nano Mater. 2020, 3 (3), 23642372,  DOI: 10.1021/acsanm.9b02486
    25. 25
      Gallardo-Williams, M. T.; Chapin, R. E.; King, P. E.; Moser, G. J.; Goldsworthy, T. L.; Morrison, J. P.; Maronpot, R. R. Boron Supplementation Inhibits the Growth and Local Expression of IGF-1 in Human Prostate Adenocarcinoma (LNCaP) Tumors in Nude Mice. Toxicol. Pathol. 2004, 32 (1), 7378,  DOI: 10.1080/01926230490260899
    26. 26
      Cohen, P.; Peehl, D. M.; Graves, H. C. B.; Rosenfeld, R. G. Biological Effects of Prostate Specific Antigen as an Insulin-like Growth Factor Binding Protein-3 Protease. J. Endocrinol. 1994, 142 (3), 407415,  DOI: 10.1677/joe.0.1420407
    27. 27
      Tian, Y.; Guo, Z.; Zhang, T.; Lin, H.; Li, Z.; Chen, J.; Deng, S.; Liu, F. Inorganic Boron-Based Nanostructures: Synthesis, Optoelectronic Properties, and Prospective Applications. Nanomaterials 2019, 9 (4), 538,  DOI: 10.3390/nano9040538
    28. 28
      Mateti, S.; Wong, C. S.; Liu, Z.; Yang, W.; Li, Y.; Li, L. H.; Chen, Y. Biocompatibility of Boron Nitride Nanosheets. Nano Res. 2018, 11 (1), 334342,  DOI: 10.1007/s12274-017-1635-y
    29. 29
      Orme, M. E.; Chaplain, M. A. J. Two-Dimensional Models of Tumour Angiogenesis and Anti-Angiogenesis Strategies. IMA J. Math. Appl. Med. Biol. 1997, 14 (3), 189205,  DOI: 10.1093/imammb/14.3.189
    30. 30
      Hillen, F.; Griffioen, A. W. Tumour Vascularization: Sprouting Angiogenesis and Beyond. Cancer Metastasis Rev. 2007, 26 (3–4), 489502,  DOI: 10.1007/s10555-007-9094-7
    31. 31
      Nishida, N.; Yano, H.; Nishida, T.; Kamura, T.; Kojiro, M. Angiogenesis in Cancer. Vasc. Health Risk Manag. 2006, 2 (3), 213219,  DOI: 10.2147/vhrm.2006.2.3.213
    32. 32
      Abunahla, H.; Mohammad, B.; Alazzam, A.; Jaoude, M. A.; Al-Qutayri, M.; Abdul Hadi, S.; Al-Sarawi, S. F. MOMSense: Metal-Oxide-Metal Elementary Glucose Sensor. Sci. Rep. 2019, 9 (1), 5524,  DOI: 10.1038/s41598-019-41892-w
    33. 33
      Kerbel, R. S.; Kamen, B. A. The Anti-Angiogenic Basis of Metronomic Chemotherapy. Nat. Rev. Cancer 2004, 4 (6), 423436,  DOI: 10.1038/nrc1369
    34. 34
      Yoncheva, K.; Momekov, G. Antiangiogenic Anticancer Strategy Based on Nanoparticulate Systems. Expert Opin. Drug Delivery 2011, 8 (8), 10411056,  DOI: 10.1517/17425247.2011.585155
    35. 35
      Ma, J.; Pulfer, S.; Li, S.; Chu, J.; Reed, K.; Gallo, J. M. Pharmacodynamic-Mediated Reduction of Temozolomide Tumor Concentrations by the Angiogenesis Inhibitor TNP-470. Cancer Res. 2001, 61 (14), 54915498
    36. 36
      Desai, N. Challenges in Development of Nanoparticle-Based Therapeutics. AAPS J. 2012, 14 (2), 282295,  DOI: 10.1208/s12248-012-9339-4
    37. 37
      Mukherjee, S.; Patra, C. R. Therapeutic Application of Anti-Angiogenic Nanomaterials in Cancers. Nanoscale 2016, 8 (25), 1244412470,  DOI: 10.1039/C5NR07887C
    38. 38
      Zhang, C.; Ni, D.; Liu, Y.; Yao, H.; Bu, W.; Shi, J. Magnesium Silicide Nanoparticles as a Deoxygenation Agent for Cancer Starvation Therapy. Nat. Nanotechnol. 2017, 12 (4), 378386,  DOI: 10.1038/nnano.2016.280
    39. 39
      Ranji-Burachaloo, H.; Karimi, F.; Xie, K.; Fu, Q.; Gurr, P. A.; Dunstan, D. E.; Qiao, G. G. MOF-Mediated Destruction of Cancer Using the Cell’s Own Hydrogen Peroxide. ACS Appl. Mater. Interfaces 2017, 9 (39), 3359933608,  DOI: 10.1021/acsami.7b07981
    40. 40
      Qian, X.; Zhang, J.; Gu, Z.; Chen, Y. Nanocatalysts-Augmented Fenton Chemical Reaction for Nanocatalytic Tumor Therapy. Biomaterials 2019, 211, 113,  DOI: 10.1016/j.biomaterials.2019.04.023
    41. 41
      Halliwell, B.; Clement, M. V.; Long, L. H. Hydrogen Peroxide in the Human Body. FEBS Lett. 2000, 486 (1), 1013,  DOI: 10.1016/S0014-5793(00)02197-9
    42. 42
      Szatrowski, T. P.; Nathan, C. F. Production of Large Amounts of Hydrogen Peroxide by Human Tumor Cells. Cancer Res. 1991, 51 (3), 794798
    43. 43
      Kim, J.; Cho, H. R.; Jeon, H.; Kim, D.; Song, C.; Lee, N.; Choi, S. H.; Hyeon, T. Continuous O2-Evolving MnFe2O4 Nanoparticle-Anchored Mesoporous Silica Nanoparticles for Efficient Photodynamic Therapy in Hypoxic Cancer. J. Am. Chem. Soc. 2017, 139 (32), 1099210995,  DOI: 10.1021/jacs.7b05559
    44. 44
      Peng, Y.; Wang, Z.; Liu, W.; Zhang, H.; Zuo, W.; Tang, H.; Chen, F.; Wang, B. Size- and Shape-Dependent Peroxidase-like Catalytic Activity of MnFe2O4 Nanoparticles and Their Applications in Highly Efficient Colorimetric Detection of Target Cancer Cells. Dalt. Trans. 2015, 44 (28), 1287112877,  DOI: 10.1039/C5DT01585E
    45. 45
      Fu, J.; Shao, Y.; Wang, L.; Zhu, Y. Lysosome-Controlled Efficient ROS Overproduction against Cancer Cells with a High PH-Responsive Catalytic Nanosystem. Nanoscale 2015, 7 (16), 72757283,  DOI: 10.1039/C5NR00706B
    46. 46
      He, W.; Zhou, Y. T.; Wamer, W. G.; Boudreau, M. D.; Yin, J. J. Mechanisms of the PH Dependent Generation of Hydroxyl Radicals and Oxygen Induced by Ag Nanoparticles. Biomaterials 2012, 33 (30), 75477555,  DOI: 10.1016/j.biomaterials.2012.06.076
    47. 47
      Maji, S. K.; Mandal, A. K.; Nguyen, K. T.; Borah, P.; Zhao, Y. Cancer Cell Detection and Therapeutics Using Peroxidase-Active Nanohybrid of Gold Nanoparticle-Loaded Mesoporous Silica-Coated Graphene. ACS Appl. Mater. Interfaces 2015, 7 (18), 98079816,  DOI: 10.1021/acsami.5b01758
    48. 48
      Chen, Z.; Yin, J. J.; Zhou, Y. T.; Zhang, Y.; Song, L.; Song, M.; Hu, S.; Gu, N. Dual Enzyme-like Activities of Iron Oxide Nanoparticles and Their Implication for Diminishing Cytotoxicity. ACS Nano 2012, 6 (5), 40014012,  DOI: 10.1021/nn300291r
    49. 49
      Zhang, C.; Bu, W.; Ni, D.; Zhang, S.; Li, Q.; Yao, Z.; Zhang, J.; Yao, H.; Wang, Z.; Shi, J. Synthesis of Iron Nanometallic Glasses and Their Application in Cancer Therapy by a Localized Fenton Reaction. Angew. Chem., Int. Ed. 2016, 55 (6), 21012106,  DOI: 10.1002/anie.201510031
    50. 50
      Li, W. P.; Su, C. H.; Chang, Y. C.; Lin, Y. J.; Yeh, C. S. Ultrasound-Induced Reactive Oxygen Species Mediated Therapy and Imaging Using a Fenton Reaction Activable Polymersome. ACS Nano 2016, 10 (2), 20172027,  DOI: 10.1021/acsnano.5b06175
    51. 51
      He, Y.; Del Valle, A.; Qian, Y.; Huang, Y. F. Near Infrared Light-Mediated Enhancement of Reactive Oxygen Species Generation through Electron Transfer from Graphene Oxide to Iron Hydroxide/Oxide. Nanoscale 2017, 9 (4), 15591566,  DOI: 10.1039/C6NR08784A
    52. 52
      Ranji-Burachaloo, H.; Gurr, P. A.; Dunstan, D. E.; Qiao, G. G. Cancer Treatment through Nanoparticle-Facilitated Fenton Reaction. ACS Nano 2018, 12 (12), 1181911837,  DOI: 10.1021/acsnano.8b07635
    53. 53
      Bokare, A. D.; Choi, W. Review of Iron-Free Fenton-like Systems for Activating H2O2 in Advanced Oxidation Processes. J. Hazard. Mater. 2014, 275, 121135,  DOI: 10.1016/j.jhazmat.2014.04.054
    54. 54
      Wu, D.; Gao, Y.; Qi, Y.; Chen, L.; Ma, Y.; Li, Y. Peptide-Based Cancer Therapy: Opportunity and Challenge. Cancer Lett. 2014, 351 (1), 1322,  DOI: 10.1016/j.canlet.2014.05.002
    55. 55
      Qi, G.-B.; Gao, Y.-J.; Wang, L.; Wang, H. Self-Assembled Peptide-Based Nanomaterials for Biomedical Imaging and Therapy. Adv. Mater. 2018, 30 (22), 1703444,  DOI: 10.1002/adma.201703444
    56. 56
      Qiao, Z. Y.; Lin, Y. X.; Lai, W. J.; Hou, C. Y.; Wang, Y.; Qiao, S. L.; Zhang, D.; Fang, Q. J.; Wang, H. A General Strategy for Facile Synthesis and in Situ Screening of Self-Assembled Polymer-Peptide Nanomaterials. Adv. Mater. 2016, 28 (9), 18591867,  DOI: 10.1002/adma.201504564
    57. 57
      Wang, Y.; Lin, Y.-X.; Qiao, Z.-Y.; An, H.-W.; Qiao, S.-L.; Wang, L.; Rajapaksha, R. P. Y. J.; Wang, H. Self-Assembled Autophagy-Inducing Polymeric Nanoparticles for Breast Cancer Interference In-Vivo. Adv. Mater. 2015, 27 (16), 26272634,  DOI: 10.1002/adma.201405926
    58. 58
      Prives, C. Signaling to P53: Breaking Minireview the MDM2–P53 Circuit. Cell 1998, 95 (1), 58,  DOI: 10.1016/S0092-8674(00)81774-2
    59. 59
      Frappier, V.; Duran, M.; Keating, A. E. PixelDB: Protein–Peptide Complexes Annotated with Structural Conservation of the Peptide Binding Mode. Protein Sci. 2018, 27 (1), 276285,  DOI: 10.1002/pro.3320
    60. 60
      Yan, J.; He, W.; Yan, S.; Niu, F.; Liu, T.; Ma, B.; Shao, Y.; Yan, Y.; Yang, G.; Lu, W.; Du, Y.; Lei, B.; Ma, P. X. Self-Assembled Peptide-Lanthanide Nanoclusters for Safe Tumor Therapy: Overcoming and Utilizing Biological Barriers to Peptide Drug Delivery. ACS Nano 2018, 12 (2), 20172026,  DOI: 10.1021/acsnano.8b00081
    61. 61
      Tuguntaev, R. G.; Chen, S.; Eltahan, A. S.; Mozhi, A.; Jin, S.; Zhang, J.; Li, C.; Wang, P. C.; Liang, X.-J. P-Gp Inhibition and Mitochondrial Impairment by Dual-Functional Nanostructure Based on Vitamin E Derivatives To Overcome Multidrug Resistance. ACS Appl. Mater. Interfaces 2017, 9 (20), 1690016912,  DOI: 10.1021/acsami.7b03877
    62. 62
      Dai, Z.; Yao, Q.; Zhu, L. MMP2-Sensitive PEG-Lipid Copolymers: A New Type of Tumor-Targeted P-Glycoprotein Inhibitor. ACS Appl. Mater. Interfaces 2016, 8 (20), 1266112673,  DOI: 10.1021/acsami.6b03064
    63. 63
      Kou, L.; Sun, R.; Bhutia, Y. D.; Yao, Q.; Chen, R. Emerging Advances in P-Glycoprotein Inhibitory Nanomaterials for Drug Delivery. Expert Opin. Drug Delivery 2018, 15 (9), 869879,  DOI: 10.1080/17425247.2018.1517749
    64. 64
      Li, D.; Hu, X.; Zhang, S. Biodegradation of Graphene-Based Nanomaterials in Blood Plasma Affects Their Biocompatibility, Drug Delivery, Targeted Organs and Antitumor Ability. Biomaterials 2019, 202, 1225,  DOI: 10.1016/j.biomaterials.2019.02.020
    65. 65
      Law, B. Y. K.; Qu, Y. Q.; Mok, S. W. F.; Liu, H.; Zeng, W.; Han, Y.; Gordillo-Martinez, F.; Chan, W. K.; Wong, K. M. C.; Wong, V. K. W. New Perspectives of Cobalt Tris(Bipyridine) System: Anti-Cancer Effect and Its Collateral Sensitivity towards Multidrug-Resistant (MDR) Cancers. Oncotarget 2017, 8 (33), 5500355021,  DOI: 10.18632/oncotarget.18991
    66. 66
      Jiang, W.; Yin, L.; Chen, H.; Paschall, A. V.; Zhang, L.; Fu, W.; Zhang, W.; Todd, T.; Yu, K. S.; Zhou, S.; Zhen, Z.; Butler, M.; Yao, L.; Zhang, F.; Shen, Y.; Li, Z.; Yin, A.; Yin, H.; Wang, X.; Avci, F. Y.; Yu, X.; Xie, J. NaCl Nanoparticles as a Cancer Therapeutic. Adv. Mater. 2019, 31 (46), 1904058,  DOI: 10.1002/adma.201904058
    67. 67
      Nagy, I. Z.; Lustyik, G.; Nagy, V. Z.; Zarándi, B.; Bertoni-Freddari, C. Intracellular Na+:K+ Ratios in Human Cancer Cells as Revealed by Energy Dispersive X-Ray Microanalysis. J. Cell Biol. 1981, 90 (3), 769777,  DOI: 10.1083/jcb.90.3.769
    68. 68
      Galvez, A.; Morales, M. P.; Eltit, J. M.; Ocaranza, P.; Carrasco, L.; Campos, X.; Sapag-Hagar, M.; Díaz-Araya, G.; Lavandero, S. A Rapid and Strong Apoptotic Process Is Triggered by Hyperosmotic Stress in Cultured Rat Cardiac Myocytes. Cell Tissue Res. 2001, 304 (2), 279285,  DOI: 10.1007/s004410100358
    69. 69
      Poon, W.; Zhang, Y.-N.; Ouyang, B.; Kingston, B. R.; Wu, J. L. Y.; Wilhelm, S.; Chan, W. C. W. Elimination Pathways of Nanoparticles. ACS Nano 2019, 13 (5), 57855798,  DOI: 10.1021/acsnano.9b01383
    70. 70
      Blanco, E.; Shen, H.; Ferrari, M. Principles of Nanoparticle Design for Overcoming Biological Barriers to Drug Delivery. Nat. Biotechnol. 2015, 33 (9), 941951,  DOI: 10.1038/nbt.3330
    71. 71
      Manoharan, D.; Li, W. P.; Yeh, C. S. Advances in Controlled Gas-Releasing Nanomaterials for Therapeutic Applications. Nanoscale Horizons 2019, 4 (3), 557578,  DOI: 10.1039/C8NH00191J
    72. 72
      Löwdin, P. O. Proton Tunneling in DNA and Its Biological Implications. Rev. Mod. Phys. 1963, 35 (3), 724732,  DOI: 10.1103/RevModPhys.35.724

Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

You’ve supercharged your research process with ACS and Mendeley!

STEP 1:
Click to create an ACS ID

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

MENDELEY PAIRING EXPIRED
Your Mendeley pairing has expired. Please reconnect