Next Article in Journal
The Core Stem Genes SOX2, POU5F1/OCT4, and NANOG Are Expressed in Human Parathyroid Tumors and Modulated by MEN1, YAP1, and β-catenin Pathways Activation
Next Article in Special Issue
coupled Hydrodynamic Flow Focusing (cHFF) to Engineer Lipid–Polymer Nanoparticles (LiPoNs) for Multimodal Imaging and Theranostic Applications
Previous Article in Journal
Survival Bias and Crosstalk between Chronological and Behavioral Age: Age- and Genotype-Sensitivity Tests Define Behavioral Signatures in Middle-Aged, Old, and Long-Lived Mice with Normal and AD-Associated Aging
Previous Article in Special Issue
Transdermal Delivery of Lidocaine-Loaded Elastic Nano-Liposomes with Microneedle Array Pretreatment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Perspective of Nanoparticle Mediated Therapeutic Targeting in Breast Cancer: An Odyssey of Endoplasmic Reticulum Unfolded Protein Response (UPRER) and Beyond

1
Department of Botany, Munshi Singh College, BR Ambedkar Bihar University, Muzaffarpur 845401, India
2
Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Korea
3
School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Korea
4
Council of Scientific and Industrial Research–Indian Institute of Integrative Medicine (CSIR–IIIM), Jammu 180001, India
5
Department of Biological Sciences, Faculty of Science, King Abdulaziz University (KAU), P.O. Box 80141, Jeddah 21589, Saudi Arabia
6
School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri 185234, India
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomedicines 2021, 9(6), 635; https://doi.org/10.3390/biomedicines9060635
Submission received: 30 April 2021 / Revised: 26 May 2021 / Accepted: 28 May 2021 / Published: 2 June 2021
(This article belongs to the Special Issue Theranostic Drug Delivery: Prospects and Problems)

Abstract

:
Breast cancer (BC) is the second most frequent cause of death among women. Representing a complex and heterogeneous type of cancer, its occurrence is attributed by both genetic (gene mutations, e.g., BRCA1, BRCA2) and non-genetic (race, ethnicity, etc.) risk factors. The effectiveness of available treatment regimens (small molecules, cytotoxic agents, and inhibitors) decreased due to their poor penetration across biological barriers, limited targeting, and rapid body clearance along with their effect on normal resident cells of bone marrow, gastrointestinal tract, and hair follicles. This significantly reduced their clinical outcomes, which led to an unprecedented increase in the number of cases worldwide. Nanomedicine, a nano-formulation of therapeutics, emerged as a versatile delivering module for employment in achieving the effective and target specific delivery of pharmaceutical payloads. Adoption of nanotechnological approaches in delivering therapeutic molecules to target cells ensures not only reduced immune response and toxicity, but increases the stability of therapeutic entities in the systemic circulation that averts their degradation and as such increased extravasations and accumulation via enhanced permeation and the retention (EPR) effect in target tissues. Additionally, nanoparticle (NP)-induced ER stress, which enhances apoptosis and autophagy, has been utilized as a combative strategy in the treatment of cancerous cells. As nanoparticles-based avenues have been capitalized to achieve better efficacy of the new genera of therapeutics with enhanced specificity and safety, the present study is aimed at providing the fundamentals of BC, nanotechnological modules (organic, inorganic, and hybrid) employed in delivering different therapeutic molecules, and mechanistic insights of nano-ER stress induced apoptosis and autophagy with a perspective of exploring this avenue for use in the nano-toxicological studies. Furthermore, the current scenario of USA FDA approved nano-formulations and the future perspective of nanotechnological based interventions to overcome the existing challenges are also discussed.

1. Introduction

Cancer—a life threatening disease, is a major cause of global mortality and morbidity [1,2]. Irrespective of the slow momentum in the occurrence of some cancers, cancer (in particular lung, colorectal, breast, and prostate), is still leading and as such represents a major public health concern worldwide [3]. The world health organization in its report has estimated an unprecedented increase in the number of cases to 19.3 million by 2025 [4]. As estimated by the American Cancer Society, around 1.8 million (0.2 million for breast cancer) new cancer cases and 0.6 million (0.04 million for breast cancer) cancer related deaths are projected to occur in the US alone in 2020 [3]. For cancers, various treatment options are available under clinical settings, including conventional (chemotherapy, radiotherapy, and surgery) and novel modalities such as immunotherapy, phototherapy, and gene therapy. Chemotherapy has its own limitations; the prominent limitations being the non-specific distribution of chemotherapeutics in the body, the compromised effect to chemotherapeutic doses after repeated administration, and in being ineffective in the cessation of tumor growth, metastasis, and its recurrence. Under such circumstances, it becomes imperative to develop effective means of drug delivery systems that could enhance the therapeutic efficiency of drugs (chemotherapeutics singly or in combination with other treatment regimens). With the advancement in nanotechnological approaches, it has now become possible to integrate a large number of components with various customized targeting strategies, therapeutic agents, and controlled-release mechanisms within an architecture framed at nano-scale [5]. The superiority of nanoparticles (NPs) in overcoming the multi-layered stromal-cell barriers for deeper tumor infiltration and drug perfusion makes it a promising future strategy for clinical applications either as an anticancer agent carrier or contrast agents in the biomedical imaging [6,7,8]. The current review symbolizes information about types and the occurrence of breast cancer (BC), a shift in the treatment paradigm from conventional to nanotechnological approaches for target specific delivery of therapeutic entities, along with mechanistic insights on the nanoparticle-induced endoplasmic reticulum unfolded protein response (UPRER) to overcome the resistance, and as such the progression of cancer cells towards gaining an insight into the development of novel therapeutics for employment in the treatment of breast cancer.

2. Methodology

The article covers the literature available from 1991 to 2021. The information was located, selected, and extracted preferably from scientific journals, books, and reports via, library and electronic search (Pubmed). Documentation of the available information from the literature helped in drafting the different sections of the manuscript.

3. Breast Cancer

Breast cancer (BC) is a disease characterized by uncontrolled cell division that changes the topology of the breast tissue; thereby resulting in a lump or mass that arises particularly in the lobules (milk glands; lobular carcinoma) or in the ducts (ductal carcinoma) connecting the lobule to the nipple. There are two categorization stages in BC: (1) the anatomic stage (as per AJCC; American Joint Committee on Cancer) representing the extent of BC; restricted to regional (spread to surrounding tissue or nearby lymph nodes) or exhibiting distant spread (spread to different organs and/or lymph nodes), and (2) the prognostic stage (as per SEER; Surveillance, Epidemiology and End Results) with reference to the display of estrogen receptor (ER), progesterone receptor (PR), levels of human epidermal growth factor receptor 2 (HER2; a growth promoting protein), and grade (resemblance of appearance of cancer under microscope to normal breast tissue) [3,9]. Based on the ER, PR, and HER2 expression profile, BC has been further divided into five intrinsic subtypes: luminal A, luminal B, HER2-enriched, basal, and claudin-low [10,11].
BC is the second leading cause of cancer related deaths among women worldwide. Although age and hereditary factors contribute as the predisposing factors to BC, women exhibiting an increased risk (>100-fold) are more prone to breast cancer as compared to men [3,12]. Representing a complex and heterogenous type of cancer, BC is characterized by multiple genetic alterations (the prominent being the inherited alterations in the BRCA1 and BRCA2 genes, which accounts for 5 to 10% of female BC and 15 to 20% of familial BC), which are used as its diagnostic and prognostic markers [13,14]. Compared to women exhibiting about 10% risk of developing BC in general population, women with pathogenic variants of BRCA1 and BRCA2 were found exhibiting around a 70% risk of developing BC [15]. A risk factor of 35% is observed among the elderly women of 70+ age with the pathogenic variant of PALB2 (another gene complementing BRCA2 in its working) [16]. Mutations in other genes such as TP53, PTEN, CDH1, and STK11 are also found to be associated with an increased risk for the development of BC [17]. Triple negative BC (TNBC; characterized by absence of ER, PR, and HER2 on tumor cell membrane) owes 15 to 20% of invasive BC subtypes [18,19]. Additionally, non-genetic risk factors such as race, ethnicity, exposure to diethyl-stilbestrol, excessive alcohol consumption, birth control and contraceptive use, hormone replacement therapy after menopause, lack of physical activity, etc., also contribute significantly to the occurrence of the disease [3,20].

4. Nano-Based Therapeutics: A Paradigm Shift in the Treatment of BC

Treatment of BC involves the use of small molecules that serve the purpose of drugs such as anti-microtubule agents (Vincristine, Paclitaxel, etc.) and cytotoxic agents (Doxorubicin, etc.) along with other inhibitors that proved effective in reducing the occurrence of the disease ([21]; Table 1).
In the treatment regimes, the small molecules were found exerting their anti-cancerous effects on other cells residing in bone marrow, hair follicles, and the gastrointestinal (GI) tract as well; thereby compromising the function of the immune system, loss of hair, and inflammation of the GI tract [21]. Additionally, drugs exhibiting poor penetration across the biological barriers that control drug permeation, limited targeting and rapid clearance from the body, which limits the clinical outcome of the drugs [22,23]. With this, creating effective delivery modules that ensure efficient delivery of drugs to the target cells with reduced immune response and toxicity was explored, which ended with the adoption of nanotechnological approaches. These approaches ensure increased bioavailability and stability of the drugs in systemic circulation, which averts their degradation and activation of immune response along with efficient delivery to target the following increased extravasations and accumulation via, enhanced permeation and retention (EPR) effect [24,25,26,27,28,29]. Drug encapsulation and efficient delivery modules help in reducing the toxicity of drug cargoes to non-target organs.
Strategic improvement over the decades for long term success in the delivery of drugs has resulted in the development of three different nanotechnological platforms: (1) organic (polymers, liposomes, dendrimers, etc.), (2) inorganic (gold, iron oxide, silica, etc.), and hybrid (liposome filled with magnetic nanoparticles, metal-organic frameworks, etc). In the organic system, therapeutic cargo is either passively loaded or forcibly incorporated for its controlled release [30,31]. Hybrid nanotechnological platforms retain the stability and function of inorganic platforms and biocompatibility feature of the organic ones [32,33]. The information on size, synthesis, stability, and other functions of nanotechnological platforms are discussed in several excellent papers published over the years ([31], and references therein) and are hence not included here.

4.1. Polymeric-Based Nanoparticles

Organic polymers (50 nm to 10 µm) are categorized into; (1) natural polymers (alginate, chitosan, gelatin, etc.), (2) synthetic polymers (hydrogels, polyethylene glycol, polyethyleneimine, etc.), and (3) degradable polymers (collagen, polycaprolactone, etc.). The biocompatibility and reduced toxicity of the polymeric substances makes them a preferable choice for employment in the synthesis of nanoparticles [34,35,36]. Polymers of natural origin prepared by encapsulating therapeutic moieties without any modification in its makeup constitutes a preferable choice of delivering proteins, DNA, oligonucleotides, and drugs. In addition to basic methods of encapsulation such as emulsification, nanospray, and nanoprecipitation, the PRINT (particle replication in non-wetting template) method given by Xu et al. (2013) provides a new route of customizing encapsulation of different therapeutic moieties in uniformly sized polymeric nanoparticles [37]. Abraxane (a nanoparticle formulation of paclitaxel encapsulated in serum albumin) has been used in clinics for the treatment of metastatic BC [38]. Another nano-formulation of doxorubicin encapsulated in the magnetic nanoparticles with albumin coating conjugated to monoclonal antibodies to VEGF (vascular endothelial growth factor) increases the targeting of VEGF in the tumor [39]. Its magnetic core makes it possible for utilization in the diagnosis as it is detected by MRI on injection to tissues. On one side, where Poly-L-glutamic acid (PLGA; a synthetic biodegradable polymer) is employed for conjugate synthesis, polyethylene glycol (PEG) and N-(2-hydroxypropyl)-methacryl amide copolymer (HPMA) are the most widely used non-biodegradable polymers of synthetic origin [40,41,42,43]. PLGA (Poly-(lactic-co-glycolic acid) nanoencapsulated with Callistemon citrinus phenolics exhibited anticancer properties against BC cell lines viz. MDA-MB231, MCF-7 and MCF 10A [44]. PK1 (conjugate of doxorubicin with HPMA) is undergoing clinical trials for its possible employment in the treatment of cancer [45]. Folic acid conjugated β-lactoglobulin nanoparticles loaded with doxorubicin were found effective against MCF-7 and MDA-MB 231 [46]. Folic acid conjugated β-lactoglobulin nanoparticles prevent the release of doxorubicin into healthy cells as well as in the systemic circulation [46]. The release of doxorubicin occurs at the tumor site in response to an acidic environment; thereby presenting a higher toxic profile against BC cell lines even after 72 h compared with the free doxorubicin. Polymeric micelles-loaded drugs serve the purpose of delivering hydrophobic drugs by accommodating them in the hydrophobic core, while its hydrophilic shell renders it with the water solubility exploited for IV administration of the drug [47,48]. Genexol-PM (PEG-poly (D,L-lactide)-paclitaxel) is the first micelle formulation of drug paclitaxel [49]. Succinobucol with P188 (poloxamer) combination exhibiting 13-fold better bioavailability and the potential to inhibit vascular cell adhesion molecule -1 (VCAM-1), emerged as the best oral treatment for BC [50]. Polymeric nanoparticle PLGA-b-PEG based delivery of antisense-miR-21 and -miR10b and siRNA against multidrug resistant protein with doxorubicin cargo caused a significant reduction in the tumor growth and volume [51,52]. In triple negative BC (TNBC) models, RGD (Arg-Gly-Asp) ligand was found facilitating either target-specific drug delivery or inhibiting cancer invasion. RGD-conjugated polymer lipid nanoparticle with cargo of doxorubicin and mitomycin C (RGD-DMPLN) displayed enhanced cytotoxicity in the TNBC mouse models [53,54].

4.2. Liposomal Nanoparticles

Liposomes (400 nm) are lipid-based (phospholipids, triglycerides, and cholesterol) nanocarriers capable of carrying the therapeutic payload either embedded in the lipid bilayer or compartmentalized in the aqueous core. Liposome nanoparticles are prepared by reverse phase evaporation, solvent injection, and by extrusion methods [55]. Their biodegradable nature and compatibility with the biological environment and less toxicity compared with other delivery modules make them idealistic for delivery of the drug [56,57,58,59]. As tumor environments are acidic, the development of pH sensitive liposomes that remain stable at a physiological pH but collapse at acidic pH helps in the target-specific delivery of therapeutic payloads such as neoplastic drugs, recombinant proteins, etc. [60,61,62,63]. Liposome encapsulated doxorubicin/Paclitaxel exhibit remarkable enhancement in its anticancer activity compared to treatment with free drug molecule [64,65,66]. The development of liposomes with the capacity to accommodate two drug molecules simultaneously was found more effective in exerting anti-cancerous activity. Meng et al. (2016) observed that dual drug (reservatol and paclitaxel) developed liposomes were found effective in combating the resistance of cancer [67]. Adoption of the same strategy in encapsulating doxorubicin together with gadoteridol were used in developing long-circulating liposomes that release their payload in a target specific manner on applying ultrasound [68,69]. This strategy successfully achieved target-specific delivery of drugs with complete tumor regression in a breast tumor model. On testing the effect of nano-emulsion of doxorubicin with bromotetratrandrine (P-glycoprotein inhibitor) on MCF-7 cell line, increased doxorubicin uptake and accumulation with reduced cardiac toxicity was observed [70]. Doxorubicin loaded on to ER targeting long-circulating liposomes improved its cellular uptake and accumulation of drug in the tumor; thereby improving the drug release profile of doxorubicin in the treatment of BC [71]. Dai et al. (2014) exploited integrin α3 (a highly expressed receptor in TNBC cell line MDA MB-23) in treating TNBC with dual drug, doxorubicin, and rapamycin encapsulated in long-circulating liposomes with an attachment of cyclic octapeptide and observed enhanced antitumor activity compared to free drug [72]. Similar effects were observed for liposomes encapsulating a combination of doxorubicin and sorafenib [73]. Co-delivery of antagomir-10b and PTX were found reducing tumor growth and as such lung metastasis of BC [74]. A significant reduction (~80%) in the tumor growth was observed for PEG coated PTX-nanocrystal formulation in nude mice and lung tumor metastasis model [75]. Additionally, a reduction (~87%) in the tumor growth was observed for lipid conjugated estrogenic (bioactive form) nanoparticles having therapeutic load of cisplatin [22].

4.3. Dendrimer Nanoparticles

Dendrimers (10–100 nm) are synthetic macromolecules of multi-branched polymers prepared from the repeated monomer units following a convergent or divergent synthesis protocol, with a hydrophobic core that accommodates for therapeutic cargo surrounded by hydrophilic periphery [76,77]. The peripheral layer consists of multivalent functional groups that interact with the charged polar molecules, while its interior boundary encapsulates uncharged non-polar molecules through multiple interactions. Hydrophobic drugs (Paclitaxel, Doxorubicin) are frequently encapsulated in dendrimers through covalent interactions [78,79]. The outer functional layer controls the release of drug molecules in response to a change in the pH or after encountering certain specific enzymes. Wang et al. employed them in delivering antisense oligo (AODNs) conjugated to polyamidoamine dendrimers via, utilization of VEGF as AODNs receptor and observed a significant reduction in the tumor vascularization in TNBC xenograft mouse model [80]. Acting as vehicles for siRNA delivery, Finlay et al. used siRNA conjugated polyamidoamine dendrimer and observed a down regulation in the TWIST transcription factor in the TNBC model [81]. Linear dendrimeric copolymers blocks of polyamidoamine dendrimer and PEG with or without galactose and amphiphilic copolymer deblock of hydrophilic PEG and polymethacrylate hydrophobic block with acid labile side chains exhibited excellent biodistribution and high biocompatibility; and are thereby capable of achieving an enhanced permeation and retention (EPR) effect with greater efficiency to penetrate solid tumors for achieving their effect [82,83].

4.4. Inorganic Nanoparticles

Inorganic nanoparticles with optical, thermal, electrical, and magnetic properties at the expense of biocompatibility are preferred over organic form for desired clinical outcomes. Inorganic nanoparticles include a wide range of materials; (1) Metals such as gold, silver, zinc oxide, etc.; (2) Magnetic materials such as iron oxide, etc.; (3) Quantum dots; and (4) carbon-based materials. Metallic nanoparticles adopt diverse mechanisms in combating the growth of cancerous cells prominent in (1) the intracellular production of reactive oxygen species (ROS); (2) the enhancement in oxidative stress; (3) the progression of tumor cells to apoptotic pathway; and (4) the induction of hyperthermia (property of transition metals, i.e., conversion of electromagnetic radiation to heat) [84]. Of the metallic materials, gold, being relatively inert, is extensively used in the synthesis of versatile nano-vehicles such as nanorods (AuNR), nanoshells (AuNS) and nanocages (AuNC) with wide biological applications in the delivery of anticancer drugs such as paclitaxel [85,86,87,88,89]. The release of encapsulated therapeutic molecules such as proteins, DNA or RNA delivered as gold nanoparticles at q target site are made on the basis of their photo-physical properties. PEGylation of metallic nanoparticles has been found to increase their stability in the systemic circulation [90]. PEGylated gold nanoparticles together with ionizing radiation increase the survival in mouse models of breast cancer [91]. The use of gold nanoparticle based therapeutic modules with radiation therapy was found to be associated with the induction of radiosensitization, a phenomenon of increasing the susceptibility of tumor cells to radiation (discussed in detail in the studies of Turnbull et al. [92], Penninckx et al. [93], and Schuemann et al. [94]). The studies have reported that dose enhancement of the ionizing radiations could excite or ionize surrounding atoms, thereby leading to a series of ionizing events with a possible formation of the free radicals capable of damaging biological macromolecules, such as the double strand breaks (DSBs) in DNA [93,95,96,97]. Additionally, NPs induce alterations in the regulation of cellular genes including the genes of DNA damage repair such as thymidylate synthase (an important enzyme in the DNA damage repair), contribute to cell sensitization, which ultimately progresses with the killing of the cells [98,99,100]. Similarly, serum coated gold nanoparticles were found to inhibit migration and therefore the invasion of the cancer cells under both in vivo and in vitro conditions via down-regulation of the genes associated with energy generation. Cisplatin-loaded gold nanoparticles guided by laser leads to the inhibition of TNBC tumor and its metastasis to the nearby tissues [22].
Silver nanoparticles mediated enhancement of oxidative stress via an increase in ROS production proceeds with the cellular damage, which exerts pressure on the cell for progression to apoptosis [101,102]. These nanoparticles limit metastasis via inhibition of the VEGF. In gliomas, treatment with silver nanoparticles followed by radiotherapy proved effective in reducing the tumor [103]. Similar to silver nanoparticles, zinc oxide nanoparticles increase progression of the cell to apoptotic pathway [104]. With reduced cell toxicity, zinc oxide nanoparticles encapsulating cisplatin and paclitaxel increases the efficacy of these drugs in the BC cell line [105]. In another study, zinc oxide nanoparticles encapsulating paclitaxel and doxorubicin in conjugation with asparaginase were found effective in the target-specific delivery of these drugs [106]. Magnetic materials such as iron oxide (magnetite) exist as another variant that can be adopted in the synthesis of nanoparticles for the delivery of drugs in a target specific manner [107,108]. With less intrinsic magnetization unless applied with a magnetic field, ferromagnetic nanoparticles with therapeutic payload exhibiting less aggregation in colloidal suspension have been used in the treatment of cancer [109]. Quantum dots (semiconductor material with broad excitation spectrum) have also been administered as a delivery vehicle in the target specific delivery of therapeutic molecules [110,111,112]. Inorganic nanoparticles capable of responding to changes in pH have been worked to facilitate the enhancement in the stability of drugs in systemic circulation and their delivery to target tissues [113,114,115]. Zinc oxide quantum dots nanoparticles conjugated to folic acid with a doxorubicin payload remains stable at physiological pH [116]. With less toxicity, the release of the doxorubicin payload occurs in response to acidic condition at the tumor site, ensures stability and tumor-specific delivery of the drug. Carbon-based materials such as carbon nanotubes, etc., which employ its structural property in getting penetrated in the cell, have also been employed in achieving stability, prolonged distribution and target specific delivery of small molecules (antisense oligonucleotides, short interfering RNA) and drugs (doxorubicin, paclitaxel) [109]. A list of different types of nanoparticles inducing ER stress in breast cancer and other cancer cell line is given in Table 2.

4.5. Hybrid Nanoparticles

Hybrid platforms represent a combination of organic with the inorganic components that presents exciting properties compared with their individual counterparts [131]. As a multifunctional platform, it merges the biocompatibility of organic platforms with stability of structures that promotes target specific drug delivery property of the inorganic counterpart. Examples of hybrid nanotechnological platforms include metal organic frameworks [132,133], liposome based magnetic nanoparticles [134,135,136], and coordination polymer nanoparticles [137,138]. Graphene oxide hybrid with magnetic materials offer biocompatibility and controlled drug-release property. Doxorubicin loaded graphene oxide—Fe3O4 yolk shell nanoparticles exhibited high loading capacity, high biocompatibility, perfect dispersibility, and pH response drug release [139].

5. Mechanistic Insights of UPR Activation in Breast Cancer Treatment

The most prominent feature of a cancerous cell is uncontrolled cell division without obeying the normal signaling cascade. In general, cancer cells encounter increased cellular stresses, which include mitotic stress, oxidative stress, proteotoxic stress, etc. These stress pathways are the route for the survival of cancer cells. The dependence of cancer cells on these pathways for survival can be exploited in cancer therapy either as stress sensitization or stress overload [140]. Endoplasmic stress (ER) stress response is one such type of stress response in which ER launches various coping mechanisms to alleviate the damage, allowing the cell to adapt to such stresses. If the recovery of cellular adaptation is not achieved, it prolongs ER stress and thereby triggers apoptosis. Any drug targeting the ER response pathways can potentially act synergistically in inducing stress overload among cancer cells, leading to enhanced cancer cell killing or reduced side effects. Several tumor cells, including breast, gastric, and hepatocellular carcinoma cells showed altered unfolded protein response (UPR) [141,142,143]. Studies have suggested that the selective inhibitor of 26S proteasomal system, bortezomib (Velcade), approved for treating multiple myeloma and mantle cell lymphoma, results in the accumulation of ERAD (endoplasmic reticulum-associated degradation) causing ER stress, and contributes to its cytotoxic activity against cancer cells [144,145,146,147]. As cancer cells are sensitive to ER stress pathway, the selective activation of ER stress pathway by NPs exposure could serve a possible way in the treatment of different cancers.

5.1. ER Stress Response Pathway

In a cell, ER is an organelle of high functionality with respect to the folding of proteins into a stable and functionally correct conformation. Under normal conditions, a cell either restricts the misfolded proteins in the ER lumen, where molecular chaperons or enzymes assists them to get properly folded for retaining their functional state, or directs them to cytosol for degradation via a ubiquitin-proteosome system (UPS; ~80% cellular proteins follow the UPS path for degradation) or autophagy-lysosome pathway (primary degradation route for misfolded or aggregated proteins) [148]. Playing a crucial role in cellular homeostasis and cell survival, any impediment in the protein folding capacity of ER leads to enhancement in the accumulation of misfolded or aggregated proteins, a condition referred to as ER stress [149]. Characterized by a high level of misfolded or aggregated proteins, ER stress induction greatly influences the fate of cells, which ultimately progresses to the development of different diseases in humans [150,151]. Dysfunction in the functional state of ER triggers signaling across the cascade, termed as unfolded protein response (UPRER) mediated by transmembrane stress sensors resident on the ER towards restoration of the ER homeostasis [149,152,153,154]. ER stress signal is sensed by three transmembrane resident stress sensors, namely PERK (Protein Kinase RNA-like ER Kinase), IRE1α (Inositol requiring enzyme 1α) and ATF6 (Activating transcription factor 6) (Figure 1) [152,154]. Improving the folding capacity of ER proteins, these stress sensors dissociate themselves from intraluminal-bound GRP78 (glucose regulated protein; an ER resident molecular chaperone also referred to as BiP (Binding immunoglobulin protein)) once it crosses the stress threshold; thereby switching from an inactive to active state, which leads to the stimulation of the UPRER response [155]. In this stimulatory event, the activation of ATF6 occurs on its cleavage, while activation of the remaining two factors occurs by self-transphosphorylation [11,154,156,157,158].

5.1.1. PERK Lineage of UPRER Pathway

PERK—a crucial transducer of UPR, is composed of a cytosolic and kinase domain. From attachment to GRP78 in uninduced cells, its activation is marked by homo-dimerization that subsequently progresses to trans-autophosphorylation of its cytoplasmic component, which ultimately leads to the inhibition of eukaryotic cytoplasmic initiation factor 2A (eIF2A) [159,160,161,162]. Phosphorylation of the eIF2A drops the activity of eIF2A levels (eIF2A); thereby provisionally admitting association of Met-tRNAiMet to 40S ribosome and as such the architect’s ternary complex of the translational apparatus [163,164]. Although inhibition of the eIF2A restricts the translation process in the majority of proteins in ER [160,162], it fails in doing so for GRP78 and transcription factor ATF4 [165,166]. ATF4 plays an important role in regulating the expression of genes involved in apoptosis, antioxidant response, and amino acid metabolism [167,168,169]. ATF4 upregulates the expression of C/EBP homologous protein (CHOP), whose translocation to the nucleus causes an increase in the BiP level and a simultaneous decrease in the expression of Bcl2 (full form). A decrease in the Bcl2 level enables release of cytochrome C and apoptosis inducing factor (AIF), which triggers fast forward progression to the mitochondrial apoptotic pathway [165,170,171,172]. Additionally, phosphorylated PERK induces nuclear translocation of Nrf2 (an antioxidant cytoplasmic protein associated with the BTB-domain containing protein Keap1); thereby leading to upregulation of the defensive antioxidant response. Nrf2 was found inducing the expression of detoxifying enzymes under in vivo conditions [173,174], besides activating the PERK-eIF2α axis of the NF-κB pathway [175,176].

5.1.2. IRE1 Lineage of UPRER Pathway

IRE1 is a transmembrane protein with Ser/Thr activity that exists in two forms (IRE1α; ubiquitously expressed in different tissues, and IRE1β; confined to lungs and intestines) among mammals [177,178,179]. Increase in the ER stress results in dissociation of IRE1α from GRP78, which proceeds with the excision of transcription factor X-box binding protein 1 (XBP1); thereby causing a shift in the reading frame, which subsequently leads to an increase in the expression of the stable and active form of XBP1s [180,181]. All this proceeds with the transactivation of genes involved in ER expansion, maintenance and ER-associated degradation (ERAD) [180,182,183,184]. IRE1α-enabled degradation of misfolded glycoproteins is mediated by a change in the expression of Derlin-2 in association with ER degradation enhancing α-mannosidase–like protein (EDEM) and EDEM2 [185,186,187]. Interaction of IRE1α with TNF receptor-associated factor 2 (TRAF2) leads to the activation of JUN N-terminal kinase (JNK) and apoptosis signal-regulating kinase 1 (ASK1) [185,186,187]. JNK modulates the apoptotic process via, regulation in nuclear factor kappa-light-chain-enhancer of activated B cells, BIM and BCL-2 functioning [188,189,190], while ASK1 together with JNK inhibitory kinase (JIK), and ASK1-interacting protein-1 (AIP1) activates the IRE1α-JNK cell death signaling pathway [187,191,192]. Additionally, phosphorylation of JNK causes its translocation across the mitochondrial membrane where it elevates the proapoptotic protein, BaD (Bcl-2-associated death promoter) and Bax (Bcl-2-associated X protein) levels that progresses with serious damage to the mitochondrial membrane [193].

5.1.3. ATF6 Lineage of UPRER Pathway

ATF6 is a transcriptional factor with CREB/ATF bZIP (basic leucine zipper) domain [194]. Prevalent as an inactive precursor in the ER membrane, its activation by S1P and S2P occurs after translocation to Golgi; whereby it is released as p50ATF-6 (a functional isomer of 50 Kd) [195]. Acting as a third mediator of UPRER response, its senses ER perturbations on exposure to stressful conditions through its cytosolic C-terminal region. The cytosolic N-terminal cleaved product, p50ATF6 of the full length ATF6 (p90ATF) translocates to the nucleus and binds to ER stress-response elements (ERSE-1) and ATF/cAMP response elements (CRE); thereby acting as a transcription factor associated with the upregulation of genes involved in normal ER functioning, such as GRP78, PDI (Protein Disulfide Isomerase), XBP1, and CHOP [152,196].

5.2. ER Stress Induced Apoptosis

In the event of insufficient UPRER response mediated by stress sensors (PERK, IRE1α and ATF6), the cells progress to the apoptosis (intrinsic as well as the extrinsic pathways) [157,197,198,199]. Major players involved in the apoptotic event include; (i) PERK/eIF2α-dependent induction of the pro-apoptotic transcriptional factor CHOP; (ii) IRE1-mediated activation of TRAF2 stimulating ASK1 and JNK cascade, and (iii) Bax/Bcl2-regulated Ca2+ release from the ER. CHOP/GADD153 (growth arrest/DNA damage) is recognized as one of the most important mediators of ER stress-induced apoptosis. In the intrinsic lineage of apoptotic pathway, a signaling platform comprising of BAX or BAK (BCL2 family members) promotes mitochondrial outer membrane permeabilization (MOMP) by assembling themselves on the mitochondrion; thereby triggering apoptosis by promoting a release of various apoptogenic factors as well as cytochrome C [200]. An extrinsic lineage of the apoptotic pathway commences with extracellular stimuli on the transmembrane receptors, the prominent being the TNF-receptor gene superfamily [201,202]. Reports suggest activation of JNK by exogenous stimuli during ER stress [186]. Activation of JNK transduces the signal along the TRAF2 bound to IRE1 (at cytoplasmic component), which invariably promotes activation of caspase-12 [186]. On the other hand, activation of ASK1 was found stimulating p38, MAPK and JNK [187].

5.3. ER Stress Induced Autophagy

Autophagy (lysosome-mediated bulk degradation pathway) emerged as a protective mechanism for the elimination of damaged organelles, protein aggregates, and worn-out proteins. To accommodate a high protein load, ER components proliferate, which proceeds with ER-associated degradation (ERAD) of misfolded or unfolded proteins. The ERAD mechanism of protein degradation includes (1) ubiquitin-proteasome-dependent ERAD (autophagy pathway targeting soluble form of misfolded protein) and (2) autophagy lysosome dependent ERAD (autophagy pathway targeting misfolded protein of both soluble and insoluble nature) [203]. Similar to UPRER, autophagy induces cell death under stress conditions. Induction of autophagy proceeds through the release of Ca2+ via, inositol 1, 4, 5-trisphosphate receptor channel (IP3R), which results in the phosphorylation of CaMKKβ (Ca2+-calmodulin-dependent kinase β) and activation of AMPK (AMP-activated kinase); indirectly associated with inactivation of the ULK1 (autophagy-activating kinase 1) complex through the inhibition of mTOR (Mechanistic target of rapamycin) [204]. The release of Ca2+ also results in the activation of DAPK (death-associated protein kinase) which phosphorylates Beclin1 and Bcl2 (B-cell lymphoma-2), which ultimately leads to induction of autophagy via activation of JNK1 [204]. Additionally, XBP1 (spliced) enhance the formation of the microtubule associated protein1 light chain 3 (LC3-I/II), which results in decreasing the activity of Fork head box O1 (FoxO1); thereby triggering Beclin-1. Another arm of UPR activated PERK induced autophagy via expression of Autophagy Protein, ATG16L, DNA-damage inducible transcript 3(DDIT3), ATG12, by ATF4 transcription factor. Similarly, CHOP activates tribbles-related protein 3 (TRIB3), which suppresses the activity of Akt/mTOR pathway and induces autophagy. The ATF6α arm of UPR induces the autophagy through inhibition of phosphorylation in mTOR and Akt pathway.
The extensive exploitation of ER module by cancerous cells turns them immortal and thereby prevents them from undergoing the apoptotic event. There are several findings that demonstrate the prominent role of UPR in cancer cells progression. For instance, in normal cells, GRP78 (an ER-resident molecular chaperone) binds to and inactivates the ER stress sensor, however, in BC or other malignancies, GRP78 is found to be overexpressed [198,205]. The overexpression of GRP78 is attributed for cell survival as peptides specifically targets GRP78 suppressed tumor growth [206].

6. FDA Approved Nanotechnological Formulations for BC

Nanotechnology is a technology employed in improving the diagnosis and therapeutics of a disease. Focused on the delivery of the substances in the nanoscale range, the structures developed possess incredible potential to encapsulate a wide range of therapeutic moieties such as protein-based drugs, peptides, nucleic acids, etc. With improved solubility and stability in the biological environment, their release in a controlled manner helps in the maintenance of drug concentration in systemic circulation; thereby enhancing the stability of the drug in circulation and its distribution to the target tissues. Efficient delivery of drugs to target tissues helps in reducing the toxicity of drug cargoes to non-target organs. Nanotechnology based drug formulation approved by the FDA include: Trastuzumab (ADC, antibody-drug conjugate targeting HER2 in HER2 positive BC), Abraxane (nab-paclitaxel, albumin bound nanoparticle bound to paclitaxel), Onivyde (irinotecan liposome injection), and more recently Onpattro (patisiran; first RNAi drug) [38,207,208,209]. These nanomedicines have demonstrated increased bioavailability, enhanced stability, active tumor targeting, and high drug loading, being successfully brought to the market.

7. Conclusions and Future Perspectives

Erroneous efforts to develop methods of delivering therapeutic cargo with improved efficacy and reduced toxicity have led to the production of biomaterials capable of delivering therapeutics in a target-specific manner. Despite significant advances, there is still a problem of premature release of the therapeutic payload, which decreases the treatment outcome and often becomes a cause in inducing systemic toxicity. As safety remains the principal concern for efficient translation of nanoparticle-based therapeutics from bench to bedside, exploration of efficient formulations of nanomaterial that prolong the stability of therapeutics in systemic circulation for optimum delivery at the target site with minimum potential off-target effects of the therapeutic cargo loaded on to nanoparticles. Anticipating improvement in delivering therapeutic cargo, camouflaging the nanoparticles with a surface covering the biocompatible material or cell membrane conjugated to a target-specific ligand for improving the stability, and as such selective delivery of the therapeutics to tumors in the response of stimuli, would avoid a non-specific or pre-mature release of the therapeutics. Taking into consideration the surface composition of the nanoparticles correlating with the stability of therapeutics in systemic circulation for the selective and efficient release of the therapeutics, engineering of nanoparticles bearing these properties would remove the impeding obstacles of nanoparticles for use in clinical applications towards improving patient outcome.

Author Contributions

Conceptualization, I.A.R. and A.T.J.; resources, S.R., I.A.R. and A.T.J.; data curation, S.R. and A.T.J.; writing—original draft preparation, S.R., V.K., A.K., T.S.A. and A.T.J.; compilation of data for tables, I.A.R. and A.K.; writing—review and editing, S.R., V.K., T.S.A. and A.T.J.; project administration, A.T.J.; funding acquisition, A.T.J. All authors have read and agreed to the published version of the manuscript.

Funding

The work was supported by Department of Science and Technology, India under Science and Engineering Research Board (DST-SERB) grant no. CRG/2019/004106.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Authors acknowledge colleagues especially Mohammad Hassan Baig, whose suggestions helped in improving the contents of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Nagai, H.; Kim, Y.H. Cancer prevention from the perspective of global cancer burden patterns. J. Thorac Dis 2017, 9, 448–451. [Google Scholar] [CrossRef] [PubMed]
  2. Ma, X.; Yu, H. Global burden of cancer. Yale J. Biol. Med. 2006, 79, 85–94. [Google Scholar] [PubMed]
  3. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2020. CA A Cancer J. Clin. 2020, 70, 7–30. [Google Scholar] [CrossRef]
  4. Gulland, A. Global cancer prevalence is growing at “alarming pace,” says WHO. BMJ 2014, 348, g1338. [Google Scholar] [CrossRef]
  5. Lou, J.; Zhang, L.; Zheng, G. Advancing Cancer Immunotherapies with Nanotechnology. Adv. Therap. 2019, 2, 1800128. [Google Scholar] [CrossRef]
  6. Siddique, S.; Chow, J.C.L. Gold Nanoparticles for Drug Delivery and Cancer Therapy. Appl. Sci. 2020, 10, 3824. [Google Scholar] [CrossRef]
  7. Siddique, S.; Chow, J.C.L. Application of Nanomaterials in Biomedical Imaging and Cancer Therapy. Nanomaterials 2020, 10. [Google Scholar] [CrossRef]
  8. Moore, J.A.; Chow, J.C.L. Recent progress and applications of gold nanotechnology in medical biophysics using artificial intelligence and mathematical modeling. Nano Ex 2021, 2, 022001. [Google Scholar] [CrossRef]
  9. Amin, M.B.; Greene, F.L.; Edge, S.B.; Compton, C.C.; Gershenwald, J.E.; Brookland, R.K.; Meyer, L.; Gress, D.M.; Byrd, D.R.; Winchester, D.P. The Eighth Edition AJCC Cancer Staging Manual: Continuing to build a bridge from a population-based to a more “personalized” approach to cancer staging. CA Cancer J. Clin. 2017, 67, 93–99. [Google Scholar] [CrossRef]
  10. Perou, C.M.; Sorlie, T.; Eisen, M.B.; van de Rijn, M.; Jeffrey, S.S.; Rees, C.A.; Pollack, J.R.; Ross, D.T.; Johnsen, H.; Akslen, L.A.; et al. Molecular portraits of human breast tumours. Nature 2000, 406, 747–752. [Google Scholar] [CrossRef]
  11. Rahman, S.; Archana, A.; Dutta, D.; Kumar, V.; Kim, J.; Jan, A.T.; Minakshi, R. The onus of cannabinoids in interrupting the molecular odyssey of breast cancer: A critical perspective on UPR(ER) and beyond. Saudi Pharm. J. 2019, 27, 437–445. [Google Scholar] [CrossRef] [PubMed]
  12. Howlader, N.; Noone, A.M.; Krapcho, M.; Miller, D.; Brest, A.; Yu, M.; Ruhl, J.; Tatalovich, Z.; Mariotto, A.; Lewis, D.R.; et al. SEER Cancer Statistics Review, 1975-2018. Natl. Cancer Inst. 2021. Available online: https://seer.cancer.gov/csr/1975_2018/ (accessed on 15 April 2021); based on November 2020 SEER data submission, posted to the SEER web site.
  13. Turnbull, C.; Rahman, N. Genetic Predisposition to Breast Cancer: Past, Present, and Future. Annu Rev. Genom. Hum. Genet. 2008, 9, 321–345. [Google Scholar] [CrossRef] [PubMed]
  14. Tung, N.; Lin, N.U.; Kidd, J.; Allen, B.A.; Singh, N.; Wenstrup, R.J.; Hartman, A.R.; Winer, E.P.; Garber, J.E. Frequency of Germline Mutations in 25 Cancer Susceptibility Genes in a Sequential Series of Patients with Breast Cancer. J. Clin. Oncol. 2016, 34, 1460–1468. [Google Scholar] [CrossRef] [Green Version]
  15. Kuchenbaecker, K.B.; Hopper, J.L.; Barnes, D.R.; Phillips, K.A.; Mooij, T.M.; Roos-Blom, M.J.; Jervis, S.; van Leeuwen, F.E.; Milne, R.L.; Andrieu, N.; et al. Risks of Breast, Ovarian, and Contralateral Breast Cancer for BRCA1 and BRCA2 Mutation Carriers. JAMA 2017, 317, 2402–2416. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Antoniou, A.C.; Casadei, S.; Heikkinen, T.; Barrowdale, D.; Pylkas, K.; Roberts, J.; Lee, A.; Subramanian, D.; De Leeneer, K.; Fostira, F.; et al. Breast-cancer risk in families with mutations in PALB2. N. Engl. J. Med. 2014, 371, 497–506. [Google Scholar] [CrossRef] [Green Version]
  17. Ferreira, M.A.; Gamazon, E.R.; Al-Ejeh, F.; Aittomaki, K.; Andrulis, I.L.; Anton-Culver, H.; Arason, A.; Arndt, V.; Aronson, K.J.; Arun, B.K.; et al. Genome-wide association and transcriptome studies identify target genes and risk loci for breast cancer. Nat. Commun. 2019, 10, 1741. [Google Scholar] [CrossRef] [Green Version]
  18. Pietri, E.; Conteduca, V.; Andreis, D.; Massa, I.; Melegari, E.; Sarti, S.; Cecconetto, L.; Schirone, A.; Bravaccini, S.; Serra, P. Androgen receptor signaling pathways as a target for breast cancer treatment. Endocr. Relat. Cancer 2016, 23, R485–R498. [Google Scholar] [CrossRef] [Green Version]
  19. He, Y.; Jiang, Z.; Chen, C.; Wang, X. Classification of triple-negative breast cancers based on Immunogenomic profiling. J. Exp. Clin. Cancer Res. 2018, 37, 327. [Google Scholar] [CrossRef]
  20. Feng, Y.; Spezia, M.; Huang, S.; Yuan, C.; Zeng, Z.; Zhang, L.; Ji, X.; Liu, W.; Huang, B.; Luo, W.; et al. Breast cancer development and progression: Risk factors, cancer stem cells, signaling pathways, genomics, and molecular pathogenesis. Genes Dis. 2018, 5, 77–106. [Google Scholar] [CrossRef]
  21. Dawidczyk, C.M.; Russell, L.M.; Searson, P.C. Nanomedicines for cancer therapy: State-of-the-art and limitations to pre-clinical studies that hinder future developments. Front. Chem. 2014, 2. [Google Scholar] [CrossRef] [Green Version]
  22. Andey, T.; Sudhakar, G.; Marepally, S.; Patel, A.; Banerjee, R.; Singh, M. Lipid nanocarriers of a lipid-conjugated estrogenic derivative inhibit tumor growth and enhance cisplatin activity against triple-negative breast cancer: Pharmacokinetic and efficacy evaluation. Mol. Pharm. 2015, 12, 1105–1120. [Google Scholar] [CrossRef] [PubMed]
  23. Sharma, A.; Goyal, A.K.; Rath, G. Recent advances in metal nanoparticles in cancer therapy. J. Drug Target. 2018, 26, 617–632. [Google Scholar] [CrossRef] [PubMed]
  24. Li, M.; Pei, J.; Ma, Z.; Fu, J.; Chen, F.; Du, S. Docetaxel-loaded ultrasmall nanostructured lipid carriers for cancer therapy: In vitro and in vivo evaluation. Cancer Chemother. Pharmacol. 2020, 85, 731–739. [Google Scholar] [CrossRef]
  25. Dallavalle, S.; Dobricic, V.; Lazzarato, L.; Gazzano, E.; Machuqueiro, M.; Pajeva, I.; Tsakovska, I.; Zidar, N.; Fruttero, R. Improvement of conventional anti-cancer drugs as new tools against multidrug resistant tumors. Drug Resist. Updat 2020, 50, 100682. [Google Scholar] [CrossRef]
  26. Rodallec, A.; Sicard, G.; Giacometti, S.; Carre, M.; Maia, T.; Valette, M.; Bouquet, F.; Savina, A.; Lacarelle, B.; Ciccolini, J.; et al. Tumor uptake and associated greater efficacy of anti-Her2 immunoliposome does not rely on Her2 expression status: Study of a docetaxel-trastuzumab immunoliposome on Her2+ breast cancer model (SKBR3). Anticancer Drugs 2019. [Google Scholar] [CrossRef]
  27. Lee, S.H.; Park, O.K.; Kim, J.; Shin, K.; Pack, C.G.; Kim, K.; Ko, G.; Lee, N.; Kwon, S.H.; Hyeon, T. Deep Tumor Penetration of Drug-Loaded Nanoparticles by Click Reaction-Assisted Immune Cell Targeting Strategy. J. Am. Chem. Soc. 2019, 141, 13829–13840. [Google Scholar] [CrossRef]
  28. Tahmasbi Rad, A.; Chen, C.W.; Aresh, W.; Xia, Y.; Lai, P.S.; Nieh, M.P. Combinational Effects of Active Targeting, Shape, and Enhanced Permeability and Retention for Cancer Theranostic Nanocarriers. ACS Appl. Mater. Interfaces 2019, 11, 10505–10519. [Google Scholar] [CrossRef]
  29. Naidu, P.S.R.; Norret, M.; Dunlop, S.A.; Fitzgerald, M.; Clemons, T.D.; Iyer, K.S. Novel Hydrophilic Copolymer-Based Nanoparticle Enhances the Therapeutic Efficiency of Doxorubicin in Cultured MCF-7 Cells. ACS Omega 2019, 4, 17083–17089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Kumar, S.S.; Mahesh, A.; Mahadevan, S.; Mandal, A.B. Synthesis and characterization of curcumin loaded polymer/lipid based nanoparticles and evaluation of their antitumor effects on MCF-7 cells. Biochim. Biophys. Acta 2014, 1840, 1913–1922. [Google Scholar] [CrossRef] [PubMed]
  31. Liyanage, P.Y.; Hettiarachchi, S.D.; Zhou, Y.; Ouhtit, A.; Seven, E.S.; Oztan, C.Y.; Celik, E.; Leblanc, R.M. Nanoparticle-mediated targeted drug delivery for breast cancer treatment. Biochim. Biophys. Acta 2019, 1871, 419–433. [Google Scholar] [CrossRef] [PubMed]
  32. Powell, D.; Chandra, S.; Dodson, K.; Shaheen, F.; Wiltz, K.; Ireland, S.; Syed, M.; Dash, S.; Wiese, T.; Mandal, T.; et al. Aptamer-functionalized hybrid nanoparticle for the treatment of breast cancer. Eur. J. Pharm. Biopharm. 2017, 114, 108–118. [Google Scholar] [CrossRef] [Green Version]
  33. Jadon, R.S.; Sharma, M. Docetaxel-loaded lipid-polymer hybrid nanoparticles for breast cancer therapeutics. J. Drug Deliv. Sci. Technol. 2019, 51, 475–484. [Google Scholar] [CrossRef]
  34. Liu, X.Y.; Nothias, J.M.; Scavone, A.; Garfinkel, M.; Millis, J.M. Biocompatibility Investigation of Polyethylene Glycol and Alginate-Poly-l-Lysine for Islet Encapsulation. ASAIO J. 2010, 56, 241–245. [Google Scholar] [CrossRef] [PubMed]
  35. Rodrigues, S.; Dionisio, M.; Lopez, C.R.; Grenha, A. Biocompatibility of chitosan carriers with application in drug delivery. J. Funct. Biomater. 2012, 3, 615–641. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Salgado, C.L.; Sanchez, E.M.; Zavaglia, C.A.; Granja, P.L. Biocompatibility and biodegradation of polycaprolactone-sebacic acid blended gels. J. Biomed. Mater. Res. A 2012, 100, 243–251. [Google Scholar] [CrossRef]
  37. Xu, J.; Luft, J.C.; Yi, X.; Tian, S.; Owens, G.; Wang, J.; Johnson, A.; Berglund, P.; Smith, J.; Napier, M.E.; et al. RNA Replicon Delivery via Lipid-Complexed PRINT Protein Particles. Mol. Pharm. 2013, 10, 3366–3374. [Google Scholar] [CrossRef] [Green Version]
  38. Miele, E.; Spinelli, G.P.; Tomao, F.; Tomao, S. Albumin-bound formulation of paclitaxel (Abraxane ABI-007) in the treatment of breast cancer. Int. J. Nanomed. 2009, 4, 99–105. [Google Scholar]
  39. Semkina, A.S.; Abakumov, M.A.; Skorikov, A.S.; Abakumova, T.O.; Melnikov, P.A.; Grinenko, N.F.; Cherepanov, S.A.; Vishnevskiy, D.A.; Naumenko, V.A.; Ionova, K.P.; et al. Multimodal doxorubicin loaded magnetic nanoparticles for VEGF targeted theranostics of breast cancer. Nanomedicine 2018, 14, 1733–1742. [Google Scholar] [CrossRef] [PubMed]
  40. Mir, M.; Ahmed, N.; Rehman, A. Recent applications of PLGA based nanostructures in drug delivery. Colloids Surf. B Biointerfaces 2017, 159, 217–231. [Google Scholar] [CrossRef]
  41. Astete, C.E.; Sabliov, C.M. Synthesis and characterization of PLGA nanoparticles. J. Biomater. Sci. Polym. Ed. 2006, 17, 247–289. [Google Scholar] [CrossRef]
  42. Zimel, M.N.; Horowitz, C.B.; Rajasekhar, V.K.; Christ, A.B.; Wei, X.; Wu, J.; Wojnarowicz, P.M.; Wang, D.; Goldring, S.R.; Purdue, P.E.; et al. HPMA-Copolymer Nanocarrier Targets Tumor-Associated Macrophages in Primary and Metastatic Breast Cancer. Mol. Cancer Ther. 2017, 16, 2701–2710. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Simpson, J.D.; Ediriweera, G.R.; Howard, C.B.; Fletcher, N.L.; Bell, C.A.; Thurecht, K.J. Polymer design and component selection contribute to uptake, distribution & trafficking behaviours of polyethylene glycol hyperbranched polymers in live MDA-MB-468 breast cancer cells. Biomater. Sci. 2019, 7, 4661–4674. [Google Scholar]
  44. Ahmed, R.; Tariq, M.; Ahmad, I.S.; Fouly, H.; Hasan, A.; Kushad, M. Poly (lactic-co-glycolic acid) Nanoparticles Loaded with Callistemon citrinus Phenolics Exhibited Anticancer Properties against Three Breast Cancer Cell Lines. J. Food Qual. 2019, 2019. [Google Scholar] [CrossRef] [Green Version]
  45. Seymour, L.W.; Ferry, D.R.; Kerr, D.J.; Rea, D.; Whitlock, M.; Poyner, R.; Boivin, C.; Hesslewood, S.; Twelves, C.; Blackie, R. Phase II studies of polymer-doxorubicin (PK1, FCE28068) in the treatment of breast, lung and colorectal cancer. Int. J. Oncol. 2009, 34, 1629–1636. [Google Scholar] [CrossRef] [Green Version]
  46. Kayani, Z.; Bordbar, A.K.; Firuzi, O. Novel folic acid-conjugated doxorubicin loaded beta-lactoglobulin nanoparticles induce apoptosis in breast cancer cells. Biomed. Pharm. 2018, 107, 945–956. [Google Scholar] [CrossRef] [PubMed]
  47. Shin, H.C.; Alani, A.W.; Rao, D.A.; Rockich, N.C.; Kwon, G.S. Multi-drug loaded polymeric micelles for simultaneous delivery of poorly soluble anticancer drugs. J. Control. Release 2009, 140, 294–300. [Google Scholar] [CrossRef] [Green Version]
  48. Wang, N.; Wang, Z.; Nie, S.; Song, L.; He, T.; Yang, S.; Yang, X.; Yi, C.; Wu, Q.; Gong, C. Biodegradable polymeric micelles coencapsulating paclitaxel and honokiol: A strategy for breast cancer therapy in vitro and in vivo. Int. J. Nanomed. 2017, 12, 1499–1514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Kim, S.O.; Lee, Y.H.; Jeong, S.W.; In, C.H.; Seo, M.H.; Lee, S.W.; Oh, H.S. Genexol®-PM, a polymeric micellar paclitaxel formulation, suppresses primary tumor growth and metastasis in a green fluorescent protein orthotopic model of breast and pancreatic cancers. Cancer Res. 2006, 66, 723. [Google Scholar]
  50. Cao, H.; Zhang, Z.; Zhao, S.; He, X.; Yu, H.; Yin, Q.; Zhang, Z.; Gu, W.; Chen, L.; Li, Y. Hydrophobic interaction mediating self-assembled nanoparticles of succinobucol suppress lung metastasis of breast cancer by inhibition of VCAM-1 expression. J. Control. Release 2015, 205, 162–171. [Google Scholar] [CrossRef] [PubMed]
  51. Devulapally, R.; Sekar, N.M.; Sekar, T.V.; Foygel, K.; Massoud, T.F.; Willmann, J.K.; Paulmurugan, R. Polymer Nanoparticles Mediated Codelivery of AntimiR-10b and AntimiR-21 for Achieving Triple Negative Breast Cancer Therapy. ACS Nano 2015, 9, 2290–2302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Deng, Z.J.; Morton, S.W.; Ben-Akiva, E.; Dreaden, E.C.; Shopsowitz, K.E.; Hammond, P.T. Layer-by-Layer Nanoparticles for Systemic Codelivery of an Anticancer Drug and siRNA for Potential Triple-Negative Breast Cancer Treatment. ACS Nano 2013, 7, 9571–9584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Shan, D.; Li, J.C.; Cai, P.; Prasad, P.; Liu, F.; Rauth, A.M.; Wu, X.Y. RGD-conjugated solid lipid nanoparticles inhibit adhesion and invasion of αvβ3 integrin-overexpressing breast cancer cells. Drug Deliv. Transl. Res. 2015, 5, 15–26. [Google Scholar] [CrossRef] [PubMed]
  54. Zhang, T.; Prasad, P.; Cai, P.; He, C.; Shan, D.; Rauth, A.M.; Wu, X.Y. Dual-targeted hybrid nanoparticles of synergistic drugs for treating lung metastases of triple negative breast cancer in mice. Acta Pharmacol. Sin. 2017, 38, 835–847. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Thakur, V.; Kutty, R.V. Recent advances in nanotheranostics for triple negative breast cancer treatment. J. Exp. Clin. Cancer Res. 2019, 38, 430. [Google Scholar] [CrossRef] [Green Version]
  56. Fenske, D.B.; Cullis, P.R. Liposomal nanomedicines. Expert Opin. Drug Deliv. 2008, 5, 25–44. [Google Scholar] [CrossRef]
  57. Torchilin, V.P. Recent advances with liposomes as pharmaceutical carriers. Nat. Rev. Drug Discov. 2005, 4, 145–160. [Google Scholar] [CrossRef]
  58. Allen, T.M.; Cullis, P.R. Drug delivery systems: Entering the mainstream. Science 2004, 303, 1818–1822. [Google Scholar] [CrossRef] [Green Version]
  59. Drummond, D.C.; Meyer, O.; Hong, K.; Kirpotin, D.B.; Papahadjopoulos, D. Optimizing liposomes for delivery of chemotherapeutic agents to solid tumors. Pharmacol. Rev. 1999, 51, 691–743. [Google Scholar]
  60. Zhou, F.; Huang, L. Liposome-mediated cytoplasmic delivery of proteins: An effective means of accessing the MHC class I-restricted antigen presentation pathway. Immunomethods 1994, 4, 229–235. [Google Scholar] [CrossRef]
  61. Reddy, R.; Zhou, F.; Huang, L.; Carbone, F.; Bevan, M.; Rouse, B.T. pH sensitive liposomes provide an efficient means of sensitizing target cells to class I restricted CTL recognition of a soluble protein. J. Immunol. Methods 1991, 141, 157–163. [Google Scholar] [CrossRef]
  62. Yuba, E.; Kojima, C.; Harada, A.; Watarai, S.; Kono, K. pH-Sensitive fusogenic polymer-modified liposomes as a carrier of antigenic proteins for activation of cellular immunity. Biomaterials 2010, 31, 943–951. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Tachibana, R.; Harashima, H.; Shono, M.; Azumano, M.; Niwa, M.; Futaki, S.; Kiwada, H. Intracellular regulation ofmacromolecules using pH-sensitive liposomes and nuclear localization signal: Qualitative and quantitative evaluation of intracellular trafficking. Biochem. Biophys. Res. Commun. 1998, 251, 538–544. [Google Scholar] [CrossRef]
  64. Hardiansyah, A.; Huang, L.Y.; Yang, M.C.; Liu, T.Y.; Tsai, S.C.; Yang, C.Y.; Kuo, C.Y.; Chan, T.Y.; Zou, H.M.; Lian, W.N.; et al. Magnetic liposomes for colorectal cancer cells therapy by high-frequency magnetic field treatment. Nanoscale Res. Lett. 2014, 9, 497. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Yan, F.; Wang, S.; Yang, W.; Goldberg, S.N.; Wu, H.; Duan, W.L.; Deng, Z.T.; Han, H.B.; Zheng, H.R. Tumor-penetrating Peptide-integrated Thermally Sensitive Liposomal Doxorubicin Enhances Efficacy of Radiofrequency Ablation in Liver Tumors. Radiology 2017, 285, 462–471. [Google Scholar] [CrossRef]
  66. Ramadass, S.K.; Anantharaman, N.V.; Subramanian, S.; Sivasubramanian, S.; Madhan, B. Paclitaxel/epigallocatechin gallate coloaded liposome: A synergistic delivery to control the invasiveness of MDA-MB-231 breast cancer cells. Colloids Surf. B Biointerfaces 2015, 125, 65–72. [Google Scholar] [CrossRef]
  67. Meng, J.; Guo, F.; Xu, H.; Liang, W.; Wang, C.; Yang, X.D. Combination Therapy using Co-encapsulated Resveratrol and Paclitaxel in Liposomes for Drug Resistance Reversal in Breast Cancer Cells in vivo. Sci. Rep. 2016, 6, 22390. [Google Scholar] [CrossRef] [PubMed]
  68. Rizzitelli, S.; Giustetto, P.; Cutrin, J.C.; Delli Castelli, D.; Boffa, C.; Ruzza, M.; Menchise, V.; Molinari, F.; Aime, S.; Terreno, E. Sonosensitive theranostic liposomes for preclinical in vivo MRI-guided visualization of doxorubicin release stimulated by pulsed low intensity non-focused ultrasound. J. Control. Release 2015, 202, 21–30. [Google Scholar] [CrossRef]
  69. Rizzitelli, S.; Giustetto, P.; Faletto, D.; Delli Castelli, D.; Aime, S.; Terreno, E. The release of Doxorubicin from liposomes monitored by MRI and triggered by a combination of US stimuli led to a complete tumor regression in a breast cancer mouse model. J. Control. Release 2016, 230, 57–63. [Google Scholar] [CrossRef] [Green Version]
  70. Cao, X.; Luo, J.; Gong, T.; Zhang, Z.R.; Sun, X.; Fu, Y. Coencapsulated doxorubicin and bromotetrandrine lipid nanoemulsions in reversing multidrug resistance in breast cancer in vitro and in vivo. Mol. Pharm. 2015, 12, 274–286. [Google Scholar] [CrossRef] [PubMed]
  71. Ma, M.; Lei, M.; Tan, X.; Tan, F.; Li, N. Theranostic liposomes containing conjugated polymer dots and doxorubicin for bio-imaging and targeted therapeutic delivery. RSC Adv. 2016, 6, 1945–1957. [Google Scholar] [CrossRef]
  72. Dai, W.; Yang, F.; Ma, L.; Fan, Y.; He, B.; He, Q.; Wang, X.; Zhang, H.; Zhang, Q. Combined mTOR inhibitor rapamycin and doxorubicin-loaded cyclic octapeptide modified liposomes for targeting integrin α3 in triple-negative breast cancer. Biomaterials 2014, 35, 5347–5358. [Google Scholar] [CrossRef] [PubMed]
  73. Lee, S.M.; Ahn, R.W.; Chen, F.; Fought, A.J.; O’Halloran, T.V.; Cryns, V.L.; Nguyen, S.T. Biological Evaluation of pH-Responsive Polymer-Caged Nanobins for Breast Cancer Therapy. ACS Nano 2010, 4, 4971–4978. [Google Scholar] [CrossRef] [PubMed]
  74. Sun, Y.; Zou, W.; Bian, S.; Huang, Y.; Tan, Y.; Liang, J.; Fan, Y.; Zhang, X. Bioreducible PAA-g-PEG graft micelles with high doxorubicin loading for targeted antitumor effect against mouse breast carcinoma. Biomaterials 2013, 34, 6818–6828. [Google Scholar] [CrossRef] [PubMed]
  75. Zhang, H.; Hu, H.; Dai, W.; Wang, X.; Zhang, Q. Effects of PEGylated paclitaxel nanocrystals on breast cancer and its lung metastasis. Nanoscale 2015, 7, 10790–10800. [Google Scholar] [CrossRef]
  76. Svenson, S.; Tomalia, D.A. Dendrimers in biomedical applications—Reflections on the field. Adv. Drug Deliv. Rev. 2005, 57, 2106–2129. [Google Scholar] [CrossRef] [PubMed]
  77. Bawarski, W.E.; Chidlowsky, E.; Bharali, D.J.; Mousa, S.A. Emerging nanopharmaceuticals. Nanomedicine 2008, 4, 273–282. [Google Scholar] [CrossRef]
  78. Hu, C.M.; Zhang, L. Nanoparticle-based combination therapy toward overcoming drug resistance in cancer. Biochem. Pharmacol. 2012, 83, 1104–1111. [Google Scholar] [CrossRef]
  79. Lim, J.; Simanek, E.E. Triazine dendrimers as drug delivery systems: From synthesis to therapy. Adv. Drug Deliv. Rev. 2012, 64, 826–835. [Google Scholar] [CrossRef]
  80. Wang, P.; Zhao, X.H.; Wang, Z.Y.; Meng, M.; Li, X.; Ning, Q. Generation 4 polyamidoamine dendrimers is a novel candidate of nano-carrier for gene delivery agents in breast cancer treatment. Cancer Lett. 2010, 298, 34–49. [Google Scholar] [CrossRef]
  81. Finlay, J.; Roberts, C.M.; Lowe, G.; Loeza, J.; Rossi, J.J.; Glackin, C.A. RNA-based TWIST1 inhibition via dendrimer complex to reduce breast cancer cell metastasis. Biomed. Res. Int. 2015, 2015, 382745. [Google Scholar] [CrossRef]
  82. Hui, H.; Xiao-dong, F.; Zhong-lin, C. Thermo-and pH-sensitive dendrimer derivatives with a shell of poly (N, N-dimethylaminoethyl methacrylate) and study of their controlled drug release behavior. Polymer 2005, 46, 9514–9522. [Google Scholar] [CrossRef]
  83. Lai, P.S.; Lou, P.J.; Peng, C.L.; Pai, C.L.; Yen, W.N.; Huang, M.Y.; Young, T.H.; Shieh, M.J. Doxorubicin delivery by polyamidoamine dendrimer conjugation and photochemical internalization for cancer therapy. J. Control. Release 2007, 122, 39–46. [Google Scholar] [CrossRef]
  84. Su, X.Y.; Liu, P.D.; Wu, H.; Gu, N. Enhancement of radiosensitization by metal-based nanoparticles in cancer radiation therapy. Cancer Biol. Med. 2014, 11, 86–91. [Google Scholar]
  85. Zhang, W.; Ji, Y.; Wu, X.; Xu, H. Trafficking of gold nanorods in breast cancer cells: Uptake, lysosome maturation, and elimination. ACS Appl. Mater. Interfaces 2013, 5, 9856–9865. [Google Scholar] [CrossRef]
  86. Paciotti, G.F.; Zhao, J.; Cao, S.; Brodie, P.J.; Tamarkin, L.; Huhta, M.; Myer, L.D.; Friedman, J.; Kingston, D.G. Synthesis and Evaluation of Paclitaxel-Loaded Gold Nanoparticles for Tumor-Targeted Drug Delivery. Bioconjug. Chem. 2016, 27, 2646–2657. [Google Scholar] [CrossRef]
  87. Peralta, D.V.; Heidari, Z.; Dash, S.; Tarr, M.A. Hybrid paclitaxel and gold nanorod-loaded human serum albumin nanoparticles for simultaneous chemotherapeutic and photothermal therapy on 4T1 breast cancer cells. ACS Appl. Mater. Interfaces 2015, 7, 7101–7111. [Google Scholar] [CrossRef]
  88. Gibson, J.D.; Khanal, B.P.; Zubarev, E.R. Paclitaxel-functionalized gold nanoparticles. J. Am. Chem Soc. 2007, 129, 11653–11661. [Google Scholar] [CrossRef]
  89. Singh, P.; Pandit, S.; Mokkapati, V.; Garg, A.; Ravikumar, V.; Mijakovic, I. Gold Nanoparticles in Diagnostics and Therapeutics for Human Cancer. Int. J. Mol. Sci. 2018, 19, 1979. [Google Scholar] [CrossRef] [PubMed]
  90. Webster, D.M.; Sundaram, P.; Byrne, M.E. Injectable nanomaterials for drug delivery: Carriers, targeting moieties, and therapeutics. Eur. J. Pharm. Biopharm. 2013, 84, 1–20. [Google Scholar] [CrossRef]
  91. Kong, T.; Zeng, J.; Wang, X.; Yang, X.; Yang, J.; McQuarrie, S.; McEwan, A.; Roa, W.; Chen, J.; Xing, J.Z. Enhancement of radiation cytotoxicity in breast-cancer cells by localized attachment of gold nanoparticles. Small 2008, 4, 1537–1543. [Google Scholar] [CrossRef] [PubMed]
  92. Turnbull, T.; Douglass, M.; Williamson, N.H.; Howard, D.; Bhardwaj, R.; Lawrence, M.; Paterson, D.J.; Bezak, E.; Thierry, B.; Kempson, I.M. Cross-correlative single-cell analysis reveals biological mechanisms of nanoparticle radiosensitization. ACS Nano 2019, 13, 5077–5090. [Google Scholar] [CrossRef]
  93. Penninckx, S.; Heuskin, A.-C.; Michiels, C.; Lucas, S. Gold Nanoparticles as a Potent Radiosensitizer: A Transdisciplinary Approach from Physics to Patient. Cancers 2020, 12, 2021. [Google Scholar] [CrossRef]
  94. Schuemann, J.; Bagley, A.F.; Berbeco, R.; Bromma, K.; Butterworth, K.T.; Byrne, H.L.; Chithrani, B.D.; Cho, S.H.; Cook, J.R.; Favaudon, V.; et al. Roadmap for metal nanoparticles in radiation therapy: Current status, translational challenges, and future directions. Phys. Med. Biol. 2020, 65, 21RM02. [Google Scholar] [CrossRef]
  95. Mesbahi, A. A review on gold nanoparticles radiosensitization effect in radiation therapy of cancer. Rep. Pract. Oncol. Radiother. 2010, 15, 176–180. [Google Scholar] [CrossRef] [Green Version]
  96. Jain, S.; Coulter, J.A.; Hounsell, A.R.; Butterworth, K.T.; McMahon, S.J.; Hyland, W.B.; Muir, M.F.; Dickson, G.R.; Prise, K.M.; Currell, F.J.; et al. Cell-specific radiosensitization by gold nanoparticles at megavoltage radiation energies. Int. J. Radiat. Oncol. Biol. Phys. 2011, 79, 531–539. [Google Scholar] [CrossRef] [Green Version]
  97. Stefancikova, L.; Lacombe, S.; Salado, D.; Porcel, E.; Pagáčová, E.; Tillement, O.; Lux, F.; Depes, D.; Kozubek, S.; Falk, M. Effect of Gadolinium-Based Nanoparticles on Nuclear DNA Damage and Repair in Glioblastoma Tumor Cells. J. Nanobiotechnol. 2016, 14, 63. [Google Scholar] [CrossRef] [Green Version]
  98. Liang, R.Y.; Tu, H.F.; Tan, X.; Yeh, Y.S.; Chueh, P.J.; Chuang, S.M. A gene signature for gold nanoparticle-exposed human cell lines. Toxicol. Res. 2015, 4, 365–375. [Google Scholar] [CrossRef]
  99. He, C.; Chow, J.C.L. Gold nanoparticle DNA damage in radiotherapy: A Monte Carlo study. AIMS Bioeng. 2016, 3, 352. [Google Scholar] [CrossRef]
  100. Chandran, P.; Riviere, J.E.; Monteiro-Riviere, N.A. Surface Chemistry of Gold Nanoparticles Determines the Biocorona Composition Impacting Cellular Uptake, Toxicity and Gene Expression Profiles in Human Endothelial Cells. Nanotoxicology 2017, 11, 507–519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Gurunathan, S.; Han, J.W.; Eppakayala, V.; Jeyaraj, M.; Kim, J.H. Cytotoxicity of biologically synthesized silver nanoparticles in MDA-MB-231 human breast cancer cells. Biomed. Res. Int. 2013, 2013, 535796. [Google Scholar] [CrossRef] [Green Version]
  102. Liu, P.; Yang, H.; Chen, W.; Zhao, J.; Li, D. Silver nanotriangles and chemotherapy drugs synergistically induce apoptosis in breast cancer cells via production of reactive oxygen species. J. Nanopart. Res. 2019, 21, 250. [Google Scholar] [CrossRef]
  103. Liu, P.; Huang, Z.; Chen, Z.; Xu, R.; Wu, H.; Zang, F.; Wang, C.; Gu, N. Silver nanoparticles: A novel radiation sensitizer for glioma? Nanoscale 2013, 5, 11829–11836. [Google Scholar] [CrossRef] [PubMed]
  104. Wahab, R.; Siddiqui, M.A.; Saquib, Q.; Dwivedi, S.; Ahmad, J.; Musarrat, J.; Al-Khedhairy, A.A.; Shin, H.S. ZnO nanoparticles induced oxidative stress and apoptosis in HepG2 and MCF-7 cancer cells and their antibacterial activity. Colloids Surf. B Biointerfaces 2014, 117, 267–276. [Google Scholar] [CrossRef] [PubMed]
  105. Hackenberg, S.; Scherzed, A.; Harnisch, W.; Froelich, K.; Ginzkey, C.; Koehler, C.; Hagen, R.; Kleinsasser, N. Antitumor activity of photo-stimulated zinc oxide nanoparticles combined with paclitaxel or cisplatin in HNSCC cell lines. J. Photochem. Photobiol. B 2012, 114, 87–93. [Google Scholar] [CrossRef]
  106. Baskar, G.; Chandhuru, J.; Sheraz Fahad, K.; Praveen, A.S.; Chamundeeswari, M.; Muthukumar, T. Anticancer activity of fungal L-asparaginase conjugated with zinc oxide nanoparticles. J. Mater. Sci Mater. Med. 2015, 26, 5380. [Google Scholar] [CrossRef] [PubMed]
  107. Farzin, A.; Etesami, S.A.; Quint, J.; Memic, A.; Tamayol, A. Magnetic Nanoparticles in Cancer Therapy and Diagnosis. Adv. Healthc. Mater. 2020, e1901058. [Google Scholar] [CrossRef]
  108. Arruebo, M.; Fernández-Pacheco, R.; Ibarra, M.R.; Santamaría, J. Magnetic nanoparticles for drug delivery. Nano Today 2007, 2, 22–32. [Google Scholar] [CrossRef]
  109. Yu, B.; Mao, Y.; Bai, L.Y.; Herman, S.E.; Wang, X.; Ramanunni, A.; Jin, Y.; Mo, X.; Cheney, C.; Chan, K.K.; et al. Targeted nanoparticle delivery overcomes off-target immunostimulatory effects of oligonucleotides and improves therapeutic efficacy in chronic lymphocytic leukemia. Blood 2013, 121, 136–147. [Google Scholar] [CrossRef]
  110. Umrao, S.; Maurya, A.K.; Shukla, V.; Grigoriev, A.; Ahuja, R.; Vinayak, M.; Srivastava, R.R.; Saxena, P.S.; Oh, I.K.; Srivastava, A. Anticarcinogenic activity of blue fluorescent hexagonal boron nitride quantum dots: As an effective enhancer for DNA cleavage activity of anticancer drug doxorubicin. Mater. Today Bio 2019, 1, 100001. [Google Scholar] [CrossRef]
  111. Freitas, M.; Neves, M.; Nouws, H.P.A.; Delerue-Matos, C. Quantum dots as nanolabels for breast cancer biomarker HER2-ECD analysis in human serum. Talanta 2020, 208, 120430. [Google Scholar] [CrossRef] [PubMed]
  112. Medintz, I.L.; Uyeda, H.T.; Goldman, E.R.; Mattoussi, H. Quantum dot bioconjugates for imaging, labelling and sensing. Nat. Mater. 2005, 4, 435–446. [Google Scholar] [CrossRef]
  113. Norberg, N.S.; Gamelin, D.R. Influence of surface modification on the luminescence of colloidal ZnO nanocrystals. J. Phys. Chem. B 2005, 109, 20810–20816. [Google Scholar] [CrossRef]
  114. Bang, J.; Yang, H.; Holloway, P.H. Enhanced and stable green emission of ZnO nanoparticles by surface segregation of Mg. Nanotechnology 2006, 17, 973–978. [Google Scholar] [CrossRef] [PubMed]
  115. Rakshit, S.; Vasudevan, S. Trap-State Dynamics in Visible-Light-Emitting ZnO:MgO Nanocrystals. J. Phys. Chem. C 2008, 112, 4531–4537. [Google Scholar] [CrossRef]
  116. Muhammad, F.; Guo, M.; Guo, Y.; Qi, W.; Qu, F.; Sun, F.; Zhao, H.; Zhu, G. Acid degradable ZnO quantum dots as a platform for targeted delivery of an anticancer drug. J. Mater. Chem. 2011, 21, 13406–13412. [Google Scholar] [CrossRef]
  117. Zhou, Z.; Yan, Y.; Wang, L.; Zhang, Q.; Cheng, Y. Melanin-like nanoparticles decorated with an autophagy-inducing peptide for efficient targeted photothermal therapy. Biomaterials 2019, 203, 63–72. [Google Scholar] [CrossRef] [PubMed]
  118. Zhang, X.; Zhang, H.; Liang, X.; Zhang, J.; Tao, W.; Zhu, X.; Chang, D.; Zeng, X.; Liu, G.; Mei, L. Iron Oxide Nanoparticles Induce Autophagosome Accumulation through Multiple Mechanisms: Lysosome Impairment, Mitochondrial Damage, and ER Stress. Mol. Pharm. 2016, 13, 2578–2587. [Google Scholar] [CrossRef] [PubMed]
  119. Yuan, L.; Zhang, F.; Qi, X.; Yang, Y.; Yan, C.; Jiang, J.; Deng, J. Chiral polymer modified nanoparticles selectively induce autophagy of cancer cells for tumor ablation. J. Nanobiotechnol. 2018, 16, 55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Wang, H.; Liu, Z.; Gou, Y.; Qin, Y.; Xu, Y.; Liu, J.; Wu, J.Z. Apoptosis and necrosis induced by novel realgar quantum dots in human endometrial cancer cells via endoplasmic reticulum stress signaling pathway. Int. J. Nanomed. 2015, 10, 5505–5512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Wang, Y.; Kaur, G.; Chen, Y.; Santos, A.; Losic, D.; Evdokiou, A. Bioinert Anodic Alumina Nanotubes for Targeting of Endoplasmic Reticulum Stress and Autophagic Signaling: A Combinatorial Nanotube-Based Drug Delivery System for Enhancing Cancer Therapy. ACS Appl Mater. Interfaces 2015, 7, 27140–27151. [Google Scholar] [CrossRef]
  122. Simard, J.C.; Durocher, I.; Girard, D. Silver nanoparticles induce irremediable endoplasmic reticulum stress leading to unfolded protein response dependent apoptosis in breast cancer cells. Apoptosis 2016, 21, 1279–1290. [Google Scholar] [CrossRef]
  123. Gopisetty, M.K.; Kovács, D.; Igaz, N.; Rónavári, A.; Bélteky, P.; Rázga, Z.; Venglovecz, V.; Csoboz, B.; Boros, I.M.; Kónya, Z.; et al. Endoplasmic reticulum stress: Major player in size-dependent inhibition of P-glycoprotein by silver nanoparticles in multidrug-resistant breast cancer cells. J. Nanobiotechnol. 2019, 17, 9. [Google Scholar] [CrossRef] [PubMed]
  124. Swanner, J.; Fahrenholtz, C.D.; Tenvooren, I.; Bernish, B.W.; Sears, J.J.; Hooker, A.; Furdui, C.M.; Alli, E.; Li, W.; Donati, G.L.; et al. Silver nanoparticles selectively treat triple-negative breast cancer cells without affecting non-malignant breast epithelial cells in vitro and in vivo. FASEB Bioadv. 2019, 1, 639–660. [Google Scholar] [CrossRef] [PubMed]
  125. Buttacavoli, M.; Albanese, N.N.; Di Cara, G.; Alduina, R.; Faleri, C.; Gallo, M.; Pizzolanti, G.; Gallo, G.; Feo, S.; Baldi, F.; et al. Anticancer activity of biogenerated silver nanoparticles: An integrated proteomic investigation. Oncotarget 2018, 9, 9685–9705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Hou, C.C.; Tsai, T.L.; Su, W.P.; Hsieh, H.P.; Yeh, C.S.; Shieh, D.B.; Su, W.C. Pronounced induction of endoplasmic reticulum stress and tumor suppression by surfactant-free poly(lactic-co-glycolic acid) nanoparticles via modulation of the PI3K signaling pathway. Int. J. Nanomed. 2013, 8, 2689–2707. [Google Scholar] [CrossRef] [Green Version]
  127. Wang, X.; Yang, L.; Zhang, H.; Tian, B.; Li, R.; Hou, X.; Wei, F. Fluorescent magnetic PEI-PLGA nanoparticles loaded with paclitaxel for concurrent cell imaging, enhanced apoptosis and autophagy in human brain cancer. Colloids Surf. B Biointerfaces 2018, 172, 708–717. [Google Scholar] [CrossRef] [PubMed]
  128. Wang, J.; Fang, X.; Liang, W. Pegylated phospholipid micelles induce endoplasmic reticulum-dependent apoptosis of cancer cells but not normal cells. ACS Nano 2012, 6, 5018–5030. [Google Scholar] [CrossRef]
  129. Yasui, H.; Takeuchi, R.; Nagane, M.; Meike, S.; Nakamura, Y.; Yamamori, T.; Ikenaka, Y.; Kon, Y.; Murotani, H.; Oishi, M.; et al. Radiosensitization of tumor cells through endoplasmic reticulum stress induced by PEGylated nanogel containing gold nanoparticles. Cancer Lett. 2014, 347, 151–158. [Google Scholar] [CrossRef] [PubMed]
  130. Cui, D.; Ma, J.; Liang, T.; Sun, L.; Meng, L.; Li, Q. Selenium nanoparticles fabricated in laminarin polysaccharides solutions exert their cytotoxicities in HepG2 cells by inhibiting autophagy and promoting apoptosis. Int. J. Biol. Macromol. 2019, 137, 829–835. [Google Scholar] [CrossRef] [PubMed]
  131. Wang, J.; Chen, B.; Zhao, D.; Peng, Y.; Zhuo, R.X.; Cheng, S.X. Peptide decorated calcium phosphate/carboxymethyl chitosan hybrid nanoparticles with improved drug delivery efficiency. Int. J. Pharm. 2013, 446, 205–210. [Google Scholar] [CrossRef]
  132. Yang, J.; Yang, Y.W. Metal-Organic Frameworks for Biomedical Applications. Small 2020, 16, e1906846. [Google Scholar] [CrossRef]
  133. Horcajada, P.; Gref, R.; Baati, T.; Allan, P.K.; Maurin, G.; Couvreur, P.; Ferey, G.; Morris, R.E.; Serre, C. Metal-organic frameworks in biomedicine. Chem. Rev. 2012, 112, 1232–1268. [Google Scholar] [CrossRef] [PubMed]
  134. Sailor, M.J.; Park, J.H. Hybrid nanoparticles for detection and treatment of cancer. Adv. Mater. 2012, 24, 3779–3802. [Google Scholar] [CrossRef] [PubMed]
  135. Wang, C.; Han, M.; Liu, X.; Chen, S.; Hu, F.; Sun, J.; Yuan, H. Mitoxantrone-preloaded water-responsive phospholipid-amorphous calcium carbonate hybrid nanoparticles for targeted and effective cancer therapy. Int. J. Nanomed. 2019, 14, 1503–1517. [Google Scholar] [CrossRef] [Green Version]
  136. Hardiansyah, A.; Yang, M.C.; Liu, T.Y.; Kuo, C.Y.; Huang, L.Y.; Chan, T.Y. Hydrophobic Drug-Loaded PEGylated Magnetic Liposomes for Drug-Controlled Release. Nanoscale Res. Lett. 2017, 12, 355. [Google Scholar] [CrossRef] [PubMed]
  137. Giliopoulos, D.; Zamboulis, A.; Giannakoudakis, D.; Bikiaris, D.; Triantafyllidis, K. Polymer/Metal Organic Framework (MOF) Nanocomposites for Biomedical Applications. Molecules 2020, 25, 185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Mu, X.; Yan, C.; Tian, Q.; Lin, J.; Yang, S. BSA-assisted synthesis of ultrasmall gallic acid-Fe(III) coordination polymer nanoparticles for cancer theranostics. Int. J. Nanomed. 2017, 12, 7207–7223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Li, S.; Zheng, J.; Chen, D.; Wu, Y.; Zhang, W.; Zheng, F.; Cao, J.; Ma, H.; Liu, Y. Yolk-shell hybrid nanoparticles with magnetic and pH-sensitive properties for controlled anticancer drug delivery. Nanoscale 2013, 5, 11718–11724. [Google Scholar] [CrossRef] [PubMed]
  140. Luo, J.; Solimini, N.L.; Elledge, S.J. Principles of cancer therapy: Oncogene and non-oncogene addiction. Cell 2009, 136, 823–837. [Google Scholar] [CrossRef] [Green Version]
  141. Sisinni, L.; Pietrafesa, M.; Lepore, S.; Maddalena, F.; Condelli, V.; Esposito, F.; Landriscina, M. Endoplasmic Reticulum Stress and Unfolded Protein Response in Breast Cancer: The Balance between Apoptosis and Autophagy and Its Role in Drug Resistance. Int. J. Mol. Sci. 2019, 20, 857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Liu, X.; Green, R.M. Endoplasmic reticulum stress and liver diseases. Liver Res. 2019, 3, 55–64. [Google Scholar] [CrossRef]
  143. Wang, Y.; Wang, K.; Jin, Y.; Sheng, X. Endoplasmic reticulum proteostasis control and gastric cancer. Cancer Lett. 2019, 449, 263–271. [Google Scholar] [CrossRef]
  144. Chauhan, D.; Hideshima, T.; Anderson, K.C. Proteasome inhibition in multiple myeloma: Therapeutic implication. Annu. Rev. Pharmacol. Toxicol. 2005, 45, 465–476. [Google Scholar] [CrossRef]
  145. Adams, J.; Kauffman, M. Development of the Proteasome Inhibitor Velcade™ (Bortezomib). Cancer Investig. 2004, 22, 304–311. [Google Scholar] [CrossRef] [PubMed]
  146. Fribley, A.; Zeng, Q.; Wang, C.Y. Proteasome inhibitor PS-341 induces apoptosis through induction of endoplasmic reticulum stress-reactive oxygen species in head and neck squamous cell carcinoma cells. Mol. Cell Biol. 2004, 24, 9695–9704. [Google Scholar] [CrossRef] [Green Version]
  147. Obeng, E.A.; Carlson, L.M.; Gutman, D.M.; Harrington, W.J., Jr.; Lee, K.P.; Boise, L.H. Proteasome inhibitors induce a terminal unfolded protein response in multiple myeloma cells. Blood 2006, 107, 4907–4916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Wan, H.Y.; Chen, J.L.; Zhu, X.; Liu, L.; Wang, J.; Zhu, X.M. Titania-Coated Gold Nano-Bipyramids for Blocking Autophagy Flux and Sensitizing Cancer Cells to Proteasome Inhibitor-Induced Death. Adv. Sci 2018, 5, 1700585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Ellgaard, L.; Helenius, A. Quality control in the endoplasmic reticulum. Nat. Rev. Mol. Cell Biol. 2003, 4, 181–191. [Google Scholar] [CrossRef]
  150. Xu, J.; Reumers, J.; Couceiro, J.R.; De Smet, F.; Gallardo, R.; Rudyak, S.; Cornelis, A.; Rozenski, J.; Zwolinska, A.; Marine, J.C.; et al. Gain of function of mutant p53 by coaggregation with multiple tumor suppressors. Nat. Chem. Biol. 2011, 7, 285–295. [Google Scholar] [CrossRef] [Green Version]
  151. Rodriguez-Gonzalez, A.; Lin, T.; Ikeda, A.K.; Simms-Waldrip, T.; Fu, C.; Sakamoto, K.M. Role of the aggresome pathway in cancer: Targeting histone deacetylase 6-dependent protein degradation. Cancer Res. 2008, 68, 2557–2560. [Google Scholar] [CrossRef] [Green Version]
  152. Walter, P.; Ron, D. The unfolded protein response: From stress pathway to homeostatic regulation. Science 2011, 334, 1081–1086. [Google Scholar] [CrossRef] [Green Version]
  153. Mori, K. Signalling pathways in the unfolded protein response: Development from yeast to mammals. J. Biochem. 2009, 146, 743–750. [Google Scholar] [CrossRef]
  154. Schroder, M.; Kaufman, R.J. The mammalian unfolded protein response. Annu. Rev. Biochem. 2005, 74, 739–789. [Google Scholar] [CrossRef] [PubMed]
  155. Zhu, G.; Lee, A.S. Role of the Unfolded Protein Response, GRP78 and GRP94 in Organ Homeostasis. J. Cell. Physiol. 2015, 230, 1413–1420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Rahman, S.; Archana, A.; Jan, A.T.; Dutta, D.; Shankar, A.; Kim, J.; Minakshi, R. Molecular Insights into the Relationship Between Autoimmune Thyroid Diseases and Breast Cancer: A Critical Perspective on Autoimmunity and ER Stress. Front. Immunol. 2019, 10, 344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Rahman, S.; Archana, A.; Jan, A.T.; Minakshi, R. Dissecting Endoplasmic Reticulum Unfolded Protein Response (UPR(ER)) in Managing Clandestine Modus Operandi of Alzheimer’s Disease. Front. Aging Neurosci. 2018, 10, 30. [Google Scholar] [CrossRef] [Green Version]
  158. Rahman, S.; Jan, A.T.; Ayyagari, A.; Kim, J.; Kim, J.; Minakshi, R. Entanglement of UPRER in Aging Driven Neurodegenerative Diseases. Front. Aging Neurosci. 2017, 9. [Google Scholar] [CrossRef] [Green Version]
  159. Bertolotti, A.; Zhang, Y.; Hendershot, L.M.; Harding, H.P.; Ron, D. Dynamic interaction of BiP and ER stress transducers in the unfolded-protein response. Nat. Cell Biol. 2000, 2, 326–332. [Google Scholar] [CrossRef] [PubMed]
  160. Ma, K.; Vattem, K.M.; Wek, R.C. Dimerization and release of molecular chaperone inhibition facilitate activation of eukaryotic initiation factor-2 kinase in response to endoplasmic reticulum stress. J. Biol. Chem. 2002, 277, 18728–18735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  161. Marciniak, S.J.; Garcia-Bonilla, L.; Hu, J.; Harding, H.P.; Ron, D. Activation-dependent substrate recruitment by the eukaryotic translation initiation factor 2 kinase PERK. J. Cell. Biol. 2006, 172, 201–209. [Google Scholar] [CrossRef]
  162. Harding, H.P.; Zhang, Y.; Ron, D. Protein translation and folding are coupled by an endoplasmic-reticulum-resident kinase. Nature 1999, 397, 271–274. [Google Scholar] [CrossRef]
  163. Dever, T.E. Gene-specific regulation by general translation factors. Cell 2002, 108, 545–556. [Google Scholar] [CrossRef] [Green Version]
  164. Hinnebusch, A.G. 5 Mechanism and Regulation of Initiator Methionyl-tRNA Binding to Ribosomes; Cold Spring Harbor Laboratory Press: Cold Spring Harbor, NY, USA, 2000. [Google Scholar]
  165. Harding, H.P.; Novoa, I.; Zhang, Y.; Zeng, H.; Wek, R.; Schapira, M.; Ron, D. Regulated translation initiation controls stress-induced gene expression in mammalian cells. Mol. Cell 2000, 6, 1099–1108. [Google Scholar] [CrossRef]
  166. Baumeister, P.; Luo, S.; Skarnes, W.C.; Sui, G.; Seto, E.; Shi, Y.; Lee, A.S. Endoplasmic Reticulum Stress Induction of the Grp78/BiP Promoter: Activating Mechanisms Mediated by YY1 and Its Interactive Chromatin Modifiers. Mol. Cell. Biol. 2005, 25, 4529–4540. [Google Scholar] [CrossRef] [Green Version]
  167. Vattem, K.M.; Wek, R.C. Reinitiation involving upstream ORFs regulates ATF4 mRNA translation in mammalian cells. Proc. Natl. Acad. Sci. USA 2004, 101, 11269–11274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Ron, D. Translational control in the endoplasmic reticulum stress response. J. Clin. Investig. 2002, 110, 1383–1388. [Google Scholar] [CrossRef]
  169. Harding, H.P.; Zhang, Y.; Zeng, H.; Novoa, I.; Lu, P.D.; Calfon, M.; Sadri, N.; Yun, C.; Popko, B.; Paules, R.; et al. An integrated stress response regulates amino acid metabolism and resistance to oxidative stress. Mol. Cell 2003, 11, 619–633. [Google Scholar] [CrossRef]
  170. Palam, L.R.; Baird, T.D.; Wek, R.C. Phosphorylation of eIF2 facilitates ribosomal bypass of an inhibitory upstream ORF to enhance CHOP translation. J. Biol. Chem. 2011, 286, 10939–10949. [Google Scholar] [CrossRef] [Green Version]
  171. Marciniak, S.J.; Yun, C.Y.; Oyadomari, S.; Novoa, I.; Zhang, Y.; Jungreis, R.; Nagata, K.; Harding, H.P.; Ron, D. CHOP induces death by promoting protein synthesis and oxidation in the stressed endoplasmic reticulum. Genes Dev. 2004, 18, 3066–3077. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Song, B.; Scheuner, D.; Ron, D.; Pennathur, S.; Kaufman, R.J. Chop deletion reduces oxidative stress, improves beta cell function, and promotes cell survival in multiple mouse models of diabetes. J. Clin. Investig. 2008, 118, 3378–3389. [Google Scholar] [CrossRef] [Green Version]
  173. Cullinan, S.B.; Diehl, J.A. PERK-dependent activation of Nrf2 contributes to redox homeostasis and cell survival following endoplasmic reticulum stress. J. Biol. Chem. 2004, 279, 20108–20117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Cullinan, S.B.; Zhang, D.; Hannink, M.; Arvisais, E.; Kaufman, R.J.; Diehl, J.A. Nrf2 is a direct PERK substrate and effector of PERK-dependent cell survival. Mol. Cell Biol. 2003, 23, 7198–7209. [Google Scholar] [CrossRef] [Green Version]
  175. Deng, J.; Lu, P.D.; Zhang, Y.; Scheuner, D.; Kaufman, R.J.; Sonenberg, N.; Harding, H.P.; Ron, D. Translational repression mediates activation of nuclear factor kappa B by phosphorylated translation initiation factor 2. Mol. Cell Biol. 2004, 24, 10161–10168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Jiang, H.Y.; Wek, S.A.; McGrath, B.C.; Scheuner, D.; Kaufman, R.J.; Cavener, D.R.; Wek, R.C. Phosphorylation of the alpha subunit of eukaryotic initiation factor 2 is required for activation of NF-kappaB in response to diverse cellular stresses. Mol. Cell Biol. 2003, 23, 5651–5663. [Google Scholar] [CrossRef] [Green Version]
  177. Tirasophon, W.; Welihinda, A.A.; Kaufman, R.J. A stress response pathway from the endoplasmic reticulum to the nucleus requires a novel bifunctional protein kinase/endoribonuclease (Ire1p) in mammalian cells. Genes Dev. 1998, 12, 1812–1824. [Google Scholar] [CrossRef] [Green Version]
  178. Bertolotti, A.; Wang, X.; Novoa, I.; Jungreis, R.; Schlessinger, K.; Cho, J.H.; West, A.B.; Ron, D. Increased sensitivity to dextran sodium sulfate colitis in IRE1beta-deficient mice. J. Clin. Investig. 2001, 107, 585–593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Martino, M.B.; Jones, L.; Brighton, B.; Ehre, C.; Abdulah, L.; Davis, C.W.; Ron, D.; O’Neal, W.K.; Ribeiro, C.M. The ER stress transducer IRE1beta is required for airway epithelial mucin production. Mucosal Immunol. 2013, 6, 639–654. [Google Scholar] [CrossRef] [Green Version]
  180. Yoshida, H.; Matsui, T.; Yamamoto, A.; Okada, T.; Mori, K. XBP1 mRNA is induced by ATF6 and spliced by IRE1 in response to ER stress to produce a highly active transcription factor. Cell 2001, 107, 881–891. [Google Scholar] [CrossRef] [Green Version]
  181. Calfon, M.; Zeng, H.; Urano, F.; Till, J.H.; Hubbard, S.R.; Harding, H.P.; Clark, S.G.; Ron, D. IRE1 couples endoplasmic reticulum load to secretory capacity by processing the XBP-1 mRNA. Nature 2002, 415, 92–96. [Google Scholar] [CrossRef]
  182. McCracken, A.A.; Brodsky, J.L. Assembly of ER-associated protein degradation in vitro: Dependence on cytosol, calnexin, and ATP. J. Cell Biol. 1996, 132, 291–298. [Google Scholar] [CrossRef] [Green Version]
  183. Haynes, C.M.; Caldwell, S.; Cooper, A.A. An HRD/DER-independent ER quality control mechanism involves Rsp5p-dependent ubiquitination and ER-Golgi transport. J. Cell Biol. 2002, 158, 91–101. [Google Scholar] [CrossRef]
  184. Yoshida, H.; Matsui, T.; Hosokawa, N.; Kaufman, R.J.; Nagata, K.; Mori, K. A time-dependent phase shift in the mammalian unfolded protein response. Dev. Cell 2003, 4, 265–271. [Google Scholar] [CrossRef] [Green Version]
  185. Zeng, T.; Peng, L.; Chao, H.; Xi, H.; Fu, B.; Wang, Y.; Zhu, Z.; Wang, G. IRE1alpha-TRAF2-ASK1 complex-mediated endoplasmic reticulum stress and mitochondrial dysfunction contribute to CXC195-induced apoptosis in human bladder carcinoma T24 cells. Biochem. Biophys. Res. Commun. 2015, 460, 530–536. [Google Scholar] [CrossRef]
  186. Urano, F.; Wang, X.; Bertolotti, A.; Zhang, Y.; Chung, P.; Harding, H.P.; Ron, D. Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science 2000, 287, 664–666. [Google Scholar] [CrossRef] [Green Version]
  187. Nishitoh, H.; Matsuzawa, A.; Tobiume, K.; Saegusa, K.; Takeda, K.; Inoue, K.; Hori, S.; Kakizuka, A.; Ichijo, H. ASK1 is essential for endoplasmic reticulum stress-induced neuronal cell death triggered by expanded polyglutamine repeats. Genes Dev. 2002, 16, 1345–1355. [Google Scholar] [CrossRef] [Green Version]
  188. Deng, X.; Xiao, L.; Lang, W.; Gao, F.; Ruvolo, P.; May, W.S., Jr. Novel role for JNK as a stress-activated Bcl2 kinase. J. Biol. Chem. 2001, 276, 23681–23688. [Google Scholar] [CrossRef] [Green Version]
  189. Lei, K.; Davis, R.J. JNK phosphorylation of Bim-related members of the Bcl2 family induces Bax-dependent apoptosis. Proc. Natl. Acad. Sci. USA 2003, 100, 2432–2437. [Google Scholar] [CrossRef] [Green Version]
  190. Ron, D.; Hubbard, S.R. How IRE1 reacts to ER stress. Cell 2008, 132, 24–26. [Google Scholar] [CrossRef] [Green Version]
  191. Yoneda, T.; Imaizumi, K.; Oono, K.; Yui, D.; Gomi, F.; Katayama, T.; Tohyama, M. Activation of caspase-12, an endoplastic reticulum (ER) resident caspase, through tumor necrosis factor receptor-associated factor 2-dependent mechanism in response to the ER stress. J. Biol. Chem. 2001, 276, 13935–13940. [Google Scholar] [CrossRef] [Green Version]
  192. Luo, D.; He, Y.; Zhang, H.; Yu, L.; Chen, H.; Xu, Z.; Tang, S.; Urano, F.; Min, W. AIP1 is critical in transducing IRE1-mediated endoplasmic reticulum stress response. J. Biol. Chem. 2008, 283, 11905–11912. [Google Scholar] [CrossRef] [Green Version]
  193. Junjappa, R.P.; Patil, P.; Bhattarai, K.R.; Kim, H.R.; Chae, H.J. IRE1alpha Implications in Endoplasmic Reticulum Stress-Mediated Development and Pathogenesis of Autoimmune Diseases. Front. Immunol. 2018, 9, 1289. [Google Scholar] [CrossRef] [Green Version]
  194. Yoshida, H.; Haze, K.; Yanagi, H.; Yura, T.; Mori, K. Identification of the cis-acting endoplasmic reticulum stress response element responsible for transcriptional induction of mammalian glucose-regulated proteins. Involvement of basic leucine zipper transcription factors. J. Biol. Chem. 1998, 273, 33741–33749. [Google Scholar] [CrossRef] [Green Version]
  195. Ye, J.; Rawson, R.B.; Komuro, R.; Chen, X.; Dave, U.P.; Prywes, R.; Brown, M.S.; Goldstein, J.L. ER stress induces cleavage of membrane-bound ATF6 by the same proteases that process SREBPs. Mol. Cell 2000, 6, 1355–1364. [Google Scholar] [CrossRef]
  196. Haze, K.; Yoshida, H.; Yanagi, H.; Yura, T.; Mori, K. Mammalian transcription factor ATF6 is synthesized as a transmembrane protein and activated by proteolysis in response to endoplasmic reticulum stress. Mol. Biol. Cell 1999, 10, 3787–3799. [Google Scholar] [CrossRef] [Green Version]
  197. Ikeyama, S.; Wang, X.T.; Li, J.; Podlutsky, A.; Martindale, J.L.; Kokkonen, G.; van Huizen, R.; Gorospe, M.; Holbrook, N.J. Expression of the pro-apoptotic gene gadd153/chop is elevated in liver with aging and sensitizes cells to oxidant injury. J. Biol. Chem. 2003, 278, 16726–16731. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  198. Minakshi, R.; Rahman, S.; Jan, A.T.; Archana, A.; Kim, J. Implications of aging and the endoplasmic reticulum unfolded protein response on the molecular modality of breast cancer. Exp. Mol. Med. 2017, 49, e389. [Google Scholar] [CrossRef] [Green Version]
  199. Wang, M.; Kaufman, R.J. The impact of the endoplasmic reticulum protein-folding environment on cancer development. Nat. Rev. Cancer 2014, 14, 581–597. [Google Scholar] [CrossRef] [PubMed]
  200. Yadav, N.; Chandra, D. Mitochondrial and postmitochondrial survival signaling in cancer. Mitochondrion 2014, 16, 18–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  201. Granger, G.A.; Shacks, S.J.; Williams, T.W.; Kolb, W.P. Lymphocyte in vitro cytotoxicity: Specific release of lymphotoxin-like materials from tuberculin-sensitive lymphoid cells. Nature 1969, 221, 1155–1157. [Google Scholar] [CrossRef] [Green Version]
  202. Suda, T.; Takahashi, T.; Golstein, P.; Nagata, S. Molecular cloning and expression of the Fas ligand, a novel member of the tumor necrosis factor family. Cell 1993, 75, 1169–1178. [Google Scholar] [CrossRef]
  203. Fujita, E.; Kouroku, Y.; Isoai, A.; Kumagai, H.; Misutani, A.; Matsuda, C.; Hayashi, Y.K.; Momoi, T. Two endoplasmic reticulum-associated degradation (ERAD) systems for the novel variant of the mutant dysferlin: Ubiquitin/proteasome ERAD(I) and autophagy/lysosome ERAD(II). Hum. Mol. Genet. 2007, 16, 618–629. [Google Scholar] [CrossRef] [PubMed]
  204. Rashid, H.O.; Yadav, R.K.; Kim, H.R.; Chae, H.J. ER stress: Autophagy induction, inhibition and selection. Autophagy 2015, 11, 1956–1977. [Google Scholar] [CrossRef] [PubMed]
  205. Araujo, N.; Hebbar, N.; Rangnekar, V.M. GRP78 Is a Targetable Receptor on Cancer and Stromal Cells. EBioMedicine 2018, 33, 2–3. [Google Scholar] [CrossRef] [Green Version]
  206. Kao, C.; Chandna, R.; Ghode, A.; Dsouza, C.; Chen, M.; Larsson, A.; Lim, S.H.; Wang, M.; Cao, Z.; Zhu, Y.; et al. Proapoptotic Cyclic Peptide BC71 Targets Cell-Surface GRP78 and Functions as an Anticancer Therapeutic in Mice. EBioMedicine 2018, 33, 22–32. [Google Scholar] [CrossRef] [PubMed]
  207. Verma, S.; Miles, D.; Gianni, L.; Krop, I.E.; Welslau, M.; Baselga, J.; Pegram, M.; Oh, D.Y.; Diéras, V.; Guardino, E.; et al. Trastuzumab Emtansine for HER2-Positive Advanced Breast Cancer. N. Engl. J. Med. 2012, 367, 1783–1791. [Google Scholar] [CrossRef] [Green Version]
  208. Lu, D.; Burris, H.A., 3rd; Wang, B.; Dees, E.C.; Cortes, J.; Joshi, A.; Gupta, M.; Yi, J.H.; Chu, Y.W.; Shih, T.; et al. Drug interaction potential of trastuzumab emtansine (T-DM1) combined with pertuzumab in patients with HER2-positive metastatic breast cancer. Curr. Drug Metab. 2012, 13, 911–922. [Google Scholar] [CrossRef]
  209. Barenholz, Y. Doxil(R)—The first FDA-approved nano-drug: Lessons learned. J. Control. Release 2012, 160, 117–134. [Google Scholar] [CrossRef]
Figure 1. Endoplasmic reticulum stress mediated cell signaling pathway.
Figure 1. Endoplasmic reticulum stress mediated cell signaling pathway.
Biomedicines 09 00635 g001
Table 1. Information of representative inhibitors currently undergoing clinical trials for TNBC.
Table 1. Information of representative inhibitors currently undergoing clinical trials for TNBC.
Inhibitor NameClinical ImplicationsPhaseNCT Number
Epidermal Growth Factor (EGF) receptor inhibitors
Afatinib Pan-HER tyrosine kinase inhibitorPhase IINCT02511847
DasatinibPan-Src tyrosine kinase inhibitor Phase IINCT02720185
Vascular Endothelial Growth Factor (VEGF) receptor inhibitors
SunitinibInhibition of proliferation, invasion, and apoptosis resistance in TNBC cell linesPhase I/IINCT00887575
ApatinibTyrosine kinase inhibitor targetting VEGFR2Phase II NCT01176669
Poly ADP-Ribose Polymerase (PARP) inhibitor
IniparibNon-competitive PARP inhibitorPhase IINCT01045304
VeliparibPARP1/2 inhibitorPhase IINCT01306032
Fibroblast Growth Factor (FGF) receptor inhibitor
DovitinibFGFR, VEGFR and PDGFR inhibitorPhase IINCT01528345
LucitanibFGFR1/2/3 and VEGFR1/2/3 inhibitorPhase IINCT02202746
Androgen Receptor (AR) inhibitors
BicalutamideAndrogen antagonistPhase IIINCT03055312
SeviteronelCytochrome P450c17a inhibitorPhase I/IINCT02580448
Source: http://ClinicalTrials.gov (Accessed date, 26 May 2021).
Table 2. Activation and modulation of ER stress, unfolded protein response, and autophagy in treating breast and other cancer cells/models.
Table 2. Activation and modulation of ER stress, unfolded protein response, and autophagy in treating breast and other cancer cells/models.
Formulations of NPsModel-TypeRemarksReference
Dual peptide-decorated melanin-like NPsIn vitro: breast cancer Hela, and MDA-MB-231 cells
In vivo: BALB/c nude mice
Beclin 1 promoted autophagy, but activated minimal in vitro cytotoxicity, sensitized cancer cells to photothermal therapy thereby enhancing significant cell killing, RGD- mediated tumor targeting increased the tumor targeting for well-regulated inhibition of tumor growth at a mild temperature [117]
Fe3O4 NPs In vitro: breast cancer
In vivo: mice
Causes ER stress, which includes autophagy and apoptosis
Extensive accumulation of autophagosome in the kidney and spleen
[118]
PAV-AuNPsIn vitro: triple-negative breast cancer cells) MDA-MB-231 cells Activation of autophagy[119]
RQDsIn vitro: endometrium carcinoma JEC cellsInduction of apoptosis and necrosis via, ER stress[120]
AANTs-TG-3MAIn vitro: Breast cancer cell MDA-MB-231-TXSA, HFF, and THP-1 cellsAANTs showed good biocompatibility, 3MA, at a non-toxic dose, reduced the autophagy-level, and improved the cell killing effect of AANTs-TG, TG and 3MA in combination enhanced the ER stress signaling[121]
AgNP (2 and 15 nm) In vitro: human MCF-7 and T-47D breast cancer cellsInduced upregulation of the transcription factors ATF-4 and GADD153/CHOP and induction of apoptosis.[122,123]
AgNPs (75 nm)In vitro: human MCF-7 and T-47D breast cancer cells
In vivo: TNBC tumor xenografts mice.
MCF-7 and T-47D are more sensitive to MCF-10A cells and Induced ER stress in TNBC cells
Effective at non-toxic doses for reducing the growth
[124]
AgNPs-EPSae (AgNPs-specific polysaccharide)In vitro: human breast (SKBR3 and 8701-BC)ER stress, oxidative stress and mitochondrial impairment triggering cell death trough apoptosis and/or autophagy activation.[125]
AgNPs-EPSae (AgNPs-specific polysaccharide)In vitro: human breast (SKBR3 and 8701-BC)ER stress, oxidative stress, and mitochondrial impairment triggering cell death trough apoptosis and/or autophagy activation.[125]
PLGA NPs loaded with LY294002In vitro: lungs cancer cells H157, H460, H1650 In vivo: Xenograft animal model (female athymic nude mice)Accumulation and pronounced induction of ER stress, activation of JNK and prominent in vivo antitumor effect[126]
FTIC labeled-PEI-PLGA-PTX-MNPsIn vitro: human brain glioblastoma U251 cellsNPs effectively endocytosed by targeted U251 cells, induction of cell apoptosis, and autophagy[127]
PEG-PE micellesIn vitro: A549 lungs cancer cells Accumulation and induction of ER stress via disturbing ER lipid homeostasis, high expression of CHOP, and proapoptotic Bax/Bak in cancer cells[128]
PEGylated nanogels containing AuNPsIn vitro: SCCVII (murine) or A549 (human lung) cells Enhance cell radiosensitivity, activation of ER stress, JNK activation, and induction of apoptosis, [129]
LP-SeNPsIn vitro: human liver carcinoma HepG2 cellsInhibition of autophagy and activation of mitochondria pathway for the induction of apoptosis [130]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Rahman, S.; Kumar, V.; Kumar, A.; Abdullah, T.S.; Rather, I.A.; Jan, A.T. Molecular Perspective of Nanoparticle Mediated Therapeutic Targeting in Breast Cancer: An Odyssey of Endoplasmic Reticulum Unfolded Protein Response (UPRER) and Beyond. Biomedicines 2021, 9, 635. https://doi.org/10.3390/biomedicines9060635

AMA Style

Rahman S, Kumar V, Kumar A, Abdullah TS, Rather IA, Jan AT. Molecular Perspective of Nanoparticle Mediated Therapeutic Targeting in Breast Cancer: An Odyssey of Endoplasmic Reticulum Unfolded Protein Response (UPRER) and Beyond. Biomedicines. 2021; 9(6):635. https://doi.org/10.3390/biomedicines9060635

Chicago/Turabian Style

Rahman, Safikur, Vijay Kumar, Anuj Kumar, Tasduq S. Abdullah, Irfan A. Rather, and Arif Tasleem Jan. 2021. "Molecular Perspective of Nanoparticle Mediated Therapeutic Targeting in Breast Cancer: An Odyssey of Endoplasmic Reticulum Unfolded Protein Response (UPRER) and Beyond" Biomedicines 9, no. 6: 635. https://doi.org/10.3390/biomedicines9060635

APA Style

Rahman, S., Kumar, V., Kumar, A., Abdullah, T. S., Rather, I. A., & Jan, A. T. (2021). Molecular Perspective of Nanoparticle Mediated Therapeutic Targeting in Breast Cancer: An Odyssey of Endoplasmic Reticulum Unfolded Protein Response (UPRER) and Beyond. Biomedicines, 9(6), 635. https://doi.org/10.3390/biomedicines9060635

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop