Cancer stem cells (CSCs): the blockage of metastatic and stemness properties by metal nanoparticles

Document Type : Review Paper

Authors

1 affiliation: Research Assistant, Department of Research and Academic Affairs, Larkin Community Hospital, Miami, FL

2 M.D.,Internist,Assistant professor ,department of Internal Medicine ,Faculty of Medicine ,Mashhad University of Medical Sciences,Mashhad,lran

3 Obstetric&gynecologist specialist,Isfahan university of medical sciences

4 Comparative Anatomy and Embryology, School of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz-Iran

Abstract

Cancer stem cells (CSCs) are comprised of hierarchically-organized subpopulations of cells with  distinguished phenotypes and tumorigenic capabilities that concrete to metastasis and cancer recurrence. According to related studies, their presence stands as the main reason of cancer associated fatalities. The fundamental feature of these cells is their ability to provide resistance towards conventional treatments or facilitate escaping routes, which include the overexpression of multifunctional ATP-binding cassette (ABC) efflux transporter gene family, metabolism reprogramming, and activation of survivance pathways . Conventional therapies are mainly capable of annihilating cancer cells, while lacking the ability to remove vital CSCs. The recurrence of tumors can be impeded through the targeting of CSCs by different therapies. Nanoparticles with unique properties have emerged as a promising approach for combating stem cancer cells. Therefore, the exertion of nanoparticles, especially metal nanoparticles ‐ based drug delivery systems in cancer imaging and remedial treatment, can surpass the obstacles of conventional treatments. Therefore, the possibility of achieving nonspecific toxicities through the administration of lower but more accurate targeted doses can be provided by the  production of theranostic metal nanoparticles and the incorporation of payload drugs into metal nanoparticles carriers, which requires a particular focus on the significance of biomarker targeting for remedial purposes and the unique contrast‐enhancing features of theranostic metal nanoparticles for facilitating image‐guided delivery. Despite the benefits of using nanoparticles for treating cancer stem cells, yet it is necessary to surpass the numerous challenges and further conduct comprehensive researches.

Keywords

Main Subjects


INTRODUCTION

According to the strong proofs on the heterogeneous form of most of the malignant cancers, it is affirmed that they contain a populace of cancer stem cells (CSCs) and differentiated cancer cells. Generally, the Heterogeneity of applied cancer stems are recruited to the tumor from various cell types, as well as genetic and/or epigenetic dissimilarities among the cancer cells. In conformity to the discovered proofs in regards to cancer cells, the represented plasticity by tumors is indicative of two classifications of tumor cell population that include CSCs and non-CSCs. There are numerous arguments on the topic of resemblances and diversities among normal tissue stem cells and cancer stem cells (CSCs). The significant traits of normal stem cells and CSCs throughout the quiescent stage are known to be self-renewal and maintenance. Considering the lack of a complete comprehension on the origination of CSCs, yet the related data are indicative of their emanation from normal stem, progenitor cells, or possibly other cancer cells. Known as tumor-initiating cells, CSCs are abled to exhibit self-renewal, proliferation, and differentiation proficiency in novel cancer cells. In comparison to non-stem cancer cells, cancer stem cells contain various functionalities and phenotypic traits . According to studies, apparently CSCs are the basis of every villainous that are hidden far from the surface of tumor tissues, while eluding the drugs and anticipating for the proper time to devastate the organ [1-8]. The statues of metastasis, drug resistance, tumor progression, and tumor recurrence condition rely on the functionality of these cells [9-11]. CSCs contain some vital qualities that can significantly affect the process of metastasis and tumor recurrence, which include multidrug-resistant, the overexpression of multifunctional ATP-binding cassette (ABC) efflux transporter gene family as a similar phenotypic property, metabolism reprogramming, and activation of survival pathways. Different elements such as tumor microenvironment, which is consisted of numerous varieties of proteins that contain growth factors and cytokines, can activate CSC survival pathways and possibly fill the functionality of chemo- or radiotherapy for improving the stemness feature through the aid of transforming cancer cells into CSCs. Moreover, the inducement of local and distant metastasis is usually initiated by CSCs through the epithelial-mesenchymal transition (EMT) program (EMT is a property of embryogenesis). In cancers, EMT is implicated throughout the development of tumors into a metastatic phenotype, which is marked by the inactivation of epithelial properties and the upregulation of mesenchymal features. [6, 12-25]. Next to their numerous embryonic or tissue stem cell traits, different cellular signaling pathways are involved in the management of CSC cellular physiology throughout their tumor microenvironment. The arrangement of these signaling routes, which include Notch, Wnt, and Hedgehog, has a substantial functionality in regularizing self-renewal, proliferation, and differentiation traits of stem cells. The mutation or abnormal activation of these pathways’ genes can disturb their regulation. There are researches that confirmed the potential of these three pathways in inducing tumorigenesis, driving tumor progression and facilitating epithelial to mesenchymal transition in malignant cells, ​causing the growth of CSC, forcing metastasis, maintaining the stemness of stem cells, and initiating drug-resistant behaviors in the course of cancer treatment [26-29]. CSCs are considered as the cause of tumor resistance due to their innate resistance to standard therapies such as chemo- and radiotherapy. In this regard, the available treatments can be enhanced and the obstacle of cancer drug resistance can be surpassed in longterm through the application of novel therapeutic strategies with the ability to target CSCs markers. As a result, it is crucial for clinical implications to develop new methods for improving the sensitivity of CSC markers. Nanotechnology provides the possibility of performing targeted and effective drug delivery to desired locations, as well as decreasing the rate of induced side effects on normal cells, facilitating the production of personal medicine, providing simultaneous diagnosis and treatment, and creating a suitable platform to overcome the existing obstacles. The potential of several nanomaterials, such as liposomes, nanoemulsion, polymeric micelles, and metal  nanoparticles , in functioning as the carriers of therapeutic agents were examined for the treatment of CSCs [30-32]. A large number of research has been conducted on the exertion of metal nanoparticles throughout CSCs therapies. Choi et al. reported the application of Graphene Oxide – Silver Nanocomposite for improving the Cytotoxic and Apoptotic Potential of Salinomycin in Human Ovarian Cancer Stem Cells (OvCSCs).  Their results displayed the inducement of a notable toxicity in both ovarian cancer cells and OvCSCs. Apparently, the applied nanocomposite showed toxicity towards OvCSCs and decreased the cell viability through the mediation of generated reactive oxygen species, causing the leakage of lactate dehydrogenase, decreasing the potential of mitochondrial membrane, and improving the expression of apoptotic genes, which leads to the inducement of mitochondrial dysfunction and possibly initiates the occurrence of apoptosis [33]. In the work of Hembram and colleagues, Quinacrine Based Gold Hybrid was applied in Nanoparticles, CSCs model SCC-9 oral cancer cells, to achieve QAuNP, which displayed a satisfying anti-CSC growth potential in opposition to SCC-9-cancer stem like, while down-regulating the agents of CSC marker. The observance of an extended G2 / M population and apoptosis to SCC-9-CSC like cells were considered as the signs of S-phase arrest and the generated re-replication that occurred as a result of QAuNP lengthened exposure. In general, a irreversible replication fork movement was caused by the QAuNP treatment. Additionally, MRE-11 may have caused a degradation in  the stalled replication fork that ultimately leads to the occurrence of apoptosis and CSCs annihilation [34]. In the current review, next to presenting the highlights of recent major progresses in metal nano particle-based techniques in regards to CSC-specific markers and/or related signalling pathways, we also explored the application prospects and discussed the related issues, approaches, and challenges.

 

CSC ISOLATION AND CHARACTERIZATION

There are similarities and differences between the functionality and phenotypic properties of normal tissue stem cells and cancer stem cells (CSCs), which resulted in the expansion of various assays for isolating and distinguishing the CSCs. Certain substantial features, including self-renewal and lineage capacity, can facilitate the recognition of CSCs [35-38]. Moreover, they can be also distinguished through more specific qualities that include phenotypic surface markers similar to CD34+/CD38− in leukemia cells, CD44+/CD24− in solid tumors, CD133+ in other tumors, and EpCAM. In the form of a transmembrane protein, there are reports on the overexpression of CD44 on varying cancer cells, which include breast, prostate, gastric, pancreas, ovary, colorectal, bladder, hepatocellular, head and neck, and leukemia CSCs. The existing glycosaminoglycan (hyaluronic acid) in extracellular matrix forms a binding with this protein to facilitate the attachment of CSC, as well as contribute to the proliferation and migration of stem cells. Known as a cell surface glycoprotein, the major expression of CD133 (Prominin-1) can be observed on certain types of CSCs of solid tumors that are implicated in glioma, lung, and breast cancer. In addition, reports are indicative of its highly expression on the CSCs of various cancers throughout varying tissue origins that augmented drug resistance. CD24 is a 27-amino-acid single-chain protein that can form a binding to the extracellular matrix and is widely exerted as a cancer stem cell marker. Its overexpression has been observed in numerous cancer cases including nasopharyngeal carcinoma, ovarian cancer, and pancreatic cancer. The hallmark of haematopoietic stem cells is known to be CD34 transmembrane protein. The population of CD34 cell within bone marrow is composed of haematopoietic stem cells and progenitor cells, while being capable of functioning throughout reconstruction progresses in humans and certain primates. According to related reports, CD34 can maintain the self-renewal, bipotency, and tumorigenicity properties of CSCs. In the form of a I trans-membrane glycoprotein, EpCAM is composed of 314 amino acids of extracellular, trans-membrane, and cytoplasmic domains . This protein can function in various roles such as cell-cell adhesion migration, proliferation, cell cycle metabolism, cell signaling, cell differentiation, metastasis, regeneration, and organogenesis [39-46]. CSCs can be chiefly isolated through the exertion of (fluorescence-activated cell sorting) technique. This uncomplicated procedure involves fluorescent activated cell sorting and is exerted for the purification and isolation of CSC. FACS is contingent on the expression of various particular cell surface markers including CD24, CD34, CD44, and CD133, EpCAM [47-50] . One of the standard methods for isolating CSC is MACS (magnetic-activated cell sorting), which is build upon the implication of specific stem cell markers and can provide the isolation of high-quality cells from a heterogeneous population cell. In this technology, the cell surface markers are initially tagged with monoclonal antibody (mAb) or magnetic microbeads to perform a complete isolation. Then, positive selection is conducted to remove the unmarked cells and sequester the marked cells, as well as to impressively isolate the objective cells from a cell suspension [51-54]. CFU( colony-forming unit assay) is  recognized as a quantitative and high-throughput procedure, which is reported to be analogous for in vivo transplantation. The utilization of CFU assay helps to examine the pattern of CSC proliferation and differentiation through their quality of producing colonies within a semisolid medium. A peculiar number of input cells are required to create these colonies in order to provide vital data on the proliferation and differentiation potential of CSCs. Briefly, in a non-adhesive manner, CSCs are cultured within a serum-free medium that had been supplied with growth factors for the purpose of developing into tumorspheres. As the cancer cells are subjected to anoikis (a suspension-induced apoptosis) throughout the arranged conditions, CSCs have the ability to remain alive and produce tumorspheres on the colony basis. The ability of this technique in isolating highly pure CFU can facilitate the achievement of accurate cellular and molecular characterization of existing cell populations [55-57]. An overexpressed situation of drug can efflux transporters, particularly BCRP or ABCG2, and consequently function as the basis of CSCs isolation. For instance, the cell populations with the ability to efflux Hoechst 33342 dye can maintain the properties of CSC throughout different types of cancer,; considering these facts, this procedure stands as the most popular route for isolating CSCs [19, 58-61].

 

COMMON TREATMENTS IN CANCER STEM CELL THERAPY

Due to the many disadvantages and restrictions of the common therapeutic tactics for cancer, including chemo- and radiotherapy, the applied treatments are often defeated and result in the recurrence of cancer in patients. The available treatments are incapable of particularly targeting CSCs and consequently cause toxicity in normal tissues, which heightens the risk of illness recurrence in patients[62-64]. Recently, a number of developed approaches with the particular goal of eliminating CSCs and varying their niche were studied due to the significance of CSC omission for deflecting cancer recurrence. The effectiveness of a therapy relies on its ability in targeting both CSCs and non-CSCs. Current researches attempted to consecrate the multiple modern remedial approaches  for extinguishing CSCs. The alterations in signaling pathways ( Notch, Wnt, and Hedgehog) and surface marker differences are alluring  remedial purposes for CSC therapy. The securing of EMT and acquisition of CSC phenotype, which assists the metastasis potential of CSS, are the factors of establishing the direct link of signaling pathways. The focus of many studies was centered on the surface marker differences and dysregulation of signaling pathways in CSCs in order to discover enhanced techniques to successfully treat cancer patients. According to related researches,  the application of surface markers as significant targets can be considered for therapies,  which include  CD133 ,CD44,CD24 and  etc . The selected ligands or antibodies are applied as the surface markers in order to be implicated in chemotherapy, radiotherapy, and  surgery . As a very significant factor, the enhancement of monoclonal antibody is emphasized in the process of targeting CSCs [13, 26, 65-78] . Moreover, some studies reported the certain CSC remedial targets with a higher potential such as ABC transporter-binding protein and microenvironment niche. Next to the expression of high levels of ABC transporter proteins in CSCs, these proteins can facilitate the preserving of CSCs from therapeutic agents. Therefore, the downregulation of these proteins can stand as a applicable method for conquering the inducement of drug resistance to common Traditional cancer therapies and prevent the occurrence of recurred conditions. Tumors are composed of cancer cells and intricate organs that contain a large number of other recruited cells, which may be in correlation to the transformed cells. Tumors microenvironment (TME) is consisted of interplays that exist among cancer and non-transformed cells. The tumor microenvironment (The cells of the immune system, Proteins, peptides, growth factors, cytokines, the lymphatics, endothelial cells, extracellular matrix,the tumor vasculature, pericytes, fibroblasts, and adipocytes and etc) aids to defend the CSCs from outside toxic agents  [79-90]. In addition, next to CSC survival and chemo-resistance, tumor angiogenesis is a vital factor that is triggered by VEGF. According to many studies, targeting VEGF with certain antibodies, such as  bevacizumab, can normalize the tumor vasculature and cause a decrease in tumor stem cell number [91-94] . Generally, the current treatments in Cancer Stem Cell Therapy that are build on various targeting methods (Table 1) include tumor microenvironment, surface marker expression, deregulated signal cascades, and ABC transporters, which facilitate the prevention of relapsed conditions.

Abbreviations in this table are defined as the following: TRXT :Tarextumab, McAb: monoclonal antibody , PTX: paclitaxel, FAP :fibroblast activation protein, Smo: smoothened, Hh: Hedgehog, LGR5: encoding an R-spondin (RSPO) receptor, FZD: Frizzled, CS : Chondroitin sulfate[ is founded in extracellular matrix], VEGF : Vascular endothelial growth factor, EpCAM : epithelial cellular adhesion molecule, small-molecule porcupine inhibitors (Signaling pathwayWNT: ETC-159, WNT-C59 and WNT974), tankyrase inhibitors (Signaling pathwayWNT: AZ1366, G007-LK, NVP-TNKS656 and XAV939),CD44: Cluster of differentiation 44, CD24: Cluster of Differentiation 24‎, CD34: Cluster of Differentiation34, miRNAs: microRNAs , ABCB1: P‐glycoprotein/P‐gp; multidrug resistance 1/MDR1,ABCC5: multidrug resistance protein 5, ABCG2: ATP-binding cassette sub-family G member 2, MDR : multidrug resistant, ABCA2:ATP -binding cassette transporters A2,  ABCA5:ATP -binding cassette transporters A5 , ABCB5: ATP-binding cassette sub-family B member 5,  ABCC5: Multidrug resistance-associated protein 5, ABCG5: ATP-binding cassette sub-family G member 5,  ABCB4 : ATP Binding Cassette Subfamily B Member 4.

 

METAL NANOPARTICLESBASED DELIVERY SYSTEM FOR CANCER STEM CELL THERAPY

Currently, there are several effective therapeutic agents available in clinics for cancer patients that generally include surgery, chemo- or radiotherapy drugs, therapeutic nucleic acids, targeted monoclonal antibodies, small molecular inhibitors, and their combinations. These treatments are mainly capable of annihilating the cancer cells and can not remove the CSCs that exist throughout the population of tumor cell, which effectively getaway by applying certain resistance processes. According to the recent concept of CSC, the recurring condition is fundamentally assisted by the innate and earned resistance technique from the existing CSCs population in cancer cell mass. The potency of CSC in eluding the regular therapeutic orders is caused by their slow-cycling phenotype, the upregulated expression of efflux pumps (ABC), antiapoptotic proteins, competent DNA response, and repair machinery. Apparently, the therapeutic potential of these agents faced a reduction in clinical trials as a result of varying restrictions such as very weak stability, weak water solubility, lenient biodistribution, terse circulation time, or off-target impacts. Moreover, CSCs can inhabit throughout low oxygen regions (Hypoxia) away from vascularized area and consequently hinder the effectiveness of remedial agents delivery [202-222] . The results of some studies indicated the feasible functionality of chemo- or radiotherapy in augmenting the stemness feature through the conversion of cancer cells into CSCs. According to recent reports, the irradiation of breast cancer cells can result in augmenting a portion of CSCs population, while other discoveries pointed out the ability of some noncancerous cells in gaining the phenotype feature of CSC. Furthermore, traditional viewpoints claim that cancer cells initiate the progress of a small cancer cell population with drug resistance behaviors as a result of repeated chemotherapeutic remedy, which can lead to the inactivation of drugs, changing drug targets, and decreased drug aggregation within the cancer cells. The performed investigations on the monoclonal antibodies that are exerted for targeting CSC marker inculcated their potency in impeding the progress of  tumors. In fact, there are many successful studies on CSC targeting antibodies that were permeated to propel on to the clinical trial stage. Nevertheless, a great number of antibodies lacked the sufficient efficacy for treating patients and caused the recurrence of tumors due to the drug-resistant behavior of cancer cells [223-227] . This survival ability of CSC result in illness recurrence with the creation of more malignant and highly invasive tumors that display resistance to chemo- and radiotherapy. In this regard, the remedy of tumors with conventional methods ends up in the increment of CSC fraction that cause the tumor cells survival and induce metastasis at distant positions. Generally, a complete treatment requires the annihilation of CSC along with the removal of non-CSCs. Therefore, tumor-recurrence conditions can be ruled out by targeting CSCS with diverse remedial modalities. According to previous data, the solo targeted elimination of CSC can not thoroughly cure a cancer disease due to the plasticity and heterogeneity of cancer cells that evert their phenotype into CSCs.  Considerably, it is necessary to focus on the enhancement of modern remedial procedures capable of performing the simultaneous elimination of both multiple drug-resistant CSCs and bulk malignant tumor cells. Therefore, nanomedicine-assisted drug delivery systems succeeded in attaining the interest of many for conquering these obstacles . Nanotechnology has made different considerable developments throughout biomedical science such as the design of nanoparticle-based drug delivery systems, including liposomes, dendrimers, metal oxide nanoparticles, polymeric nanomicelles, and carbon nanotubes that attained the attention of many researchers. The loading of nano-medicines with high payload of single or multiple drugs requires control over their size and surface feature. Therefore, the enhancement of pharmacokinetic and pharmacodynamic features of nanomedicines became possible through the reduction of their side effects on normal cells. The amazing potential of nanomedicine-based procedures was  conformed due to providing a multipronged route of selectivity and more profound bioavailability. Moreover, the optimization of biocompatibility and pharmacokinetic features of these nanodrug carriers is achieved by modifying the surface of nanoparticles. The previously mentioned restrictions can be hopefully resolved through the distinct qualities of nanomedicines, which include having control over size, tunable surface features, surface-to-volume ratios, interesting surface functional groups for bioconjugation, reduced rate of nonspecific biological distribution, and fewer side effects [18, 227-245] .  The simple passage of nanomedicines through blood capillaries for contacting the target site is facilitated by their smaller size (~200 nm). Various multifunctional nanoparticles formed their cancer therapeutic usages in available settings under the specific goal of targeting CSC. In recent years, some novel procedures were formulated to successfully target CSS, such as the design of nanoparticles that implicate a targeting ligand particularly for CSC that accommodate an anticancer drug molecule for omitting the combined CSS with a chemosensitizer to conquer drug resistance (such as an ABC  transporter inhibitor) and an imaging agent to assist the tumor.  Such combination procedures may be capable of performing a more impressive anti-tumor impact along with a reduced rate of side effects, while simplifying the accurate recognition of primary tumor localization and its metastases as well. The most significant benefit of nanocarriers is their ability to conduct the simultaneous delivery of multiple drugs . The exertion of varying kinds of nanomaterials, such as polymeric nanoparticles, metal-based nanoparticles, carbon nanotube, magnetic nanoparticles, and liposome, were considered for preparing targeted nano-drug carriers in order to target CSC by the application of chemo-drugs, antibiotics, nucleic acids, peptides, and proteins.  The mentioned remedial agent modalities are capable of targeting downstream cellular signaling pathways, CSC survival-associated genes, cell surface markers, and metabolic pathways [246-249].  The interest of many has been invested in designing and developing a multifunctional and stimuli‐responsive metal nanoparticles ‐based drug delivery system in regards to the diagnosis and therapy of cancer stem cells. Considering the quick progress of nanomedicine, the unique physical and chemical features of metal nanoparticles (gold, iron, silver, copper, tetanium,cobalt, nickel), including their high surface areas ,size, shape and surface construction, as well as their distinct optical , electronic , chemical and photoelectrochemical qualities ,size-dependent physicochemical features  and etc, outshined the other options as a theranostic tool in biomedical implementations such as diagnostic imaging, drug delivery , gene therapy, novel therapeutics, magnetic resonance imaging , cell mechanics , hyperthermia , tumor advancement, in vivo tracking of stem cells, and cell detachment [234, 250-260]. The constructed Surface‐functionalized metal nanoparticles by the utilization of engineered surface ligands provided useful approaches for the application of metal nanoparticles‐based drug delivery systems in cancer imaging and remedial treatment for the objective of surpassing the difficulties of conventional treatments. The design and development of stimuli‐responsive ligands were incorporated with the engineering of multiple physicochemical properties into metal nanoparticles for enhancing the efficiency of metal nanoparticles‐based delivery system (Fig. 1). As a result , the production of theranostic metal nanoparticles and incorporation of payload drugs into metal nanoparticles carriers can offer a chance of achieving nonspecific toxicities through the application of lower but more accurate targeted doses, which required a particular focus on the significance of biomarker targeting in regards to remedial purposes along with the distinct contrast‐amplifying features of theranostic metal nanoparticles that provide image‐guided delivery  [261-269] . Recently, researchers attempted to formulate several metal nanoparticles ‐based drug delivery system for CSC therapy (Table 2).

Abbreviations in this table are defined as the following: Glu-NP: Glucose-installed nanoparticle, siRNA :small interfering RNA, AuNPs: Au nanoparticle, PEG: Poly (ethylene glycol), HA: Hyaluronic acid, DOX :doxorubicin , G5- PAMAM: fifth-generation polyamidoamine dendrimer, Fe3O4@SiNPs:core/shell construction that the silica shell encapsulating Fe3O4 nanoparticles as the magnetic core, HSPI: heat shock protein inhibitor, CD20: Cluster of Differentiation 20, SPIONPs: super-paramagnetic iron oxide nanoparticles, aptamer CSC1:aptamers selected against DU145 prostate cancer cells, aptamer CSC13:subpopulation of prostate cancer stem cells, PDC:polydiallyldimethylammonium chloride, ABCG2: ATP-binding cassette sub-family G member 2, PTX: paclitaxel, EGFR: epidermal growth factor receptor, Dtxl:docetaxel, PLGA :poly(D,L-lactic-co-glycolic acid), PAH: poly(allylamine hydrochloride).

 

CHALLENGES OF TARGETED BIO-CONJUGATED NANOPARTICLE CANCER STEM CELL THERAPY

The promising stance of targeted nanoparticles design for CSC therapy is due to the ability of nanoparticles in enhancing the drug concentration in CSCs for the occurrence of omission throughout the tumor mass. In this regard, the remedial effectiveness of anti-CSC drugs for clinical trials can be increased by synthesizing targeted nanoparticles, which would also decrease the required time of treatment and result in achieving better outcomes from the patients; nevertheless, the form and optimizing process of impressive nanodrug carriers needs further investigations [294-297] .In recent years, next to the outshining progress in assessing the design of Targeted  Nanoparticle  bioconjugates to function as effective chemotherapeutic agents, however, the achievement of applicable and impressive cancer treatment is predicted to be very distant. The conjugation of nanoparticles (NPs) with different ligands can lead to the production of very selective products in binding to the target, which would consequently enhance their efficacy and lower the induced toxicity. It is necessary to consider the challenges that rely on different parameters which determine the success and effectiveness of these products [100, 298, 299] . One of the most substantial obstacles  is  the existence of an interplay among the nature and size of NP and ligand. The traffic of NP throughout the body is controlled by their size as a significant parameter. Next to the befitting ability of small NPs in passively targeting tumors, however, they can be easily cleared by kidneys which is quiet problematic. On the other hand, the availability of larger NPs is restricted by their size which is considered as a disadvantage [300-304] . Another challenge that needs to be addressed is the modification of targeting moiety that is required to obtain a higher therapeutic efficacy and leads to the inducement of several issues such as expanded complexity, regulatory barriers, and extra cost. In addition, numerous questions were generated by the performed practices in regards to nanoparticle targeting and drug aggregation throughout the appointed tumor and CSC subpopulation. Considering this scheme, it is assumed by a substantial paradox that the appending of targeting moiety onto the surface of nanoparticles concedes with the stealth quality of nanoparticles, while intensifying their clearance by reticuloendothelial system from host body. The benefit of nanoparticles high avidity is recognizes as one of their conundrums, however, this quality  causes a reduction in the infiltration ability of targeted nanoparticles into the tumor core. As another major challenge, observations were indicative of the existence of some CSC populations in the necrotic region of tumors, which is quiet difficult to be reached by targeted nanocarriers. Overall, the treatment of cancers that implicate accessible CSCs such as leukemia diseases can benefit from the application of targeted nanocarriers. Since a large number of CSC markers are utilized throughout the enhancement of targeted nanocarriers, the inducement of unwanted toxicity is expected due to their reported expression on normal stem cells; consequently, the discovery of highly CSC-specific ligands remains as a ambitious and difficult assignment [305-312]. Another major challenge related to CSC is to succeed in the particular targeting of slow-cycling cancer stem cells as one of the fundamental causes of recurrence condition. Moreover, there are other confrontations such as therapeutics targeting brain-related cancers that are limited by the blood-brain barrier, which makes it very difficult for targeting NP-bioconjugated to reach such tumors[3, 313, 314] . Furthermore, the high surface area and free surface energy of NPs stand as a crucial obstacle that requires attention since they can impact the obtained colloidal stability. There are inquiries for the exertion  of surfactants, polymers, and proteins to cause improvement in the colloidal stability . Nevertheless, it is of outmost importance to discover a method for the oral delivery of bio-conjugated NPs by crossing the intravenous route and completing the procedure, which would benefit a tremendous number of patients [315-320] . Another fundamental challenge that has concerned many is related to the adverse biological impacts of NPs at cellular, tissue, organ, and organism levels due to the possibility of resulting in the inducement of nanotoxicity.  Certain biophysical properties, including size and surface features, can influence these products in vivo distribution, and consequently affect the signaling pathways and biological functionalities. different researches reported the negative effect of varying NPs on the liver, kidney, and skin through the upregulation of inflammatory pathway[321-324], Some studies indicated the potential ability of nanoparticles in heightening the rate of epigenetic alterations, which implicate histone posttranslational modifications and DNA methylation [325, 326].

 

CONCLUSION

cancer stem cells (CSCs) are known to contain different mechanisms for escaping conventional treatments, which leads to tumor recurrence and relapse. Moreover, the therapeutic efficacy of conventional agents faced a reduction as a result of different limitations such as weaker stability, weak water solubility, lenient biodistribution, terse circulation time, or off-target effects. A complete treatment requires the omitting of CSCs without destroying  non-CSCs. Therefore, it is essential to develop modern remedial procedures with the ability to perform the simultaneous elimination of both multiple drug-resistant CSCs and bulk malignant tumor cells. The potential capabilities of nanomedicine can overcome the resulting therapies from conventional methods. nanomedicine proved to be a promising tool for conquering aforementioned limitations due to containing specific properties, which include controllable size, tunable surface features, surface-to-volume ratios, appealing surface functional groups for bioconjugation, less nonspecific Biological distribution, and the lowest rate of side effects .The design and development of a multifunctional and stimuli‐responsive metal nanoparticles ‐based drug delivery system with distinct physical and chemical qualities can be efficient throughout the treatment of cancer and the elimination of multiple drug-resistant CSCs and bulk malignant tumor cells. The form and development of stimuli‐responsive ligands were incorporated with the engineering of multiple physicochemical properties into metal nanoparticles in order to improve the efficiency of metal nanoparticles‐based delivery systems. As a result , the theranostic metal nanoparticles with the incorporation of loading drugs into metal nanoparticles carriers and  contrast  agent  imaging may offer the possibility of achieving nonspecific toxicities through the administration of lower but more accurate targeted doses. Despite the efforts and advances in targeted bio-conjugated nanoparticle cancer stem cell therapy, there are still many challenges in this area that require solutions and till then, the achievement of an effective and impressive cancer treatment is predicted to be very distant.

 

ACKNOWLEDGEMENT

The authors thank Biornder website for designing the figure

 

CONFLICT OF INTEREST

The authors declare no conflicts of interest

 

1.    Batlle, E. and H. Clevers, Cancer stem cells revisited. Nature medicine, 2017. 23(10): p. 1124.
2.    Chang, W.H. and A.G. Lai, Aberrations in Notch-Hedgehog signalling reveal cancer stem cells harbouring conserved oncogenic properties associated with hypoxia and immunoevasion. British journal of cancer, 2019. 121(8): p. 666-678.
3.    Moore, N. and S. Lyle, Quiescent, slow-cycling stem cell populations in cancer: a review of the evidence and discussion of significance. Journal of oncology, 2011. 2011.
4.    Najafi, M., B. Farhood, and K. Mortezaee, Cancer stem cells (CSCs) in cancer progression and therapy. Journal of cellular physiology, 2019. 234(6): p. 8381-8395.
5.    Osman, A., et al., Revisiting cancer stem cells as the origin of cancer-associated cells in the tumor microenvironment: a hypothetical view from the potential of iPSCs. Cancers, 2020. 12(4): p. 879.
6.    Phi, L.T.H., et al., Cancer stem cells (CSCs) in drug resistance and their therapeutic implications in cancer treatment. Stem cells international, 2018. 2018.
7.    Zhu, Z., et al., Targeting self-renewal in high-grade brain tumors leads to loss of brain tumor stem cells and prolonged survival. Cell stem cell, 2014. 15(2): p. 185-198.
8.    Wang, L., et al., Cervical Cancer Cell Growth, Drug Resistance, and Epithelial-Mesenchymal Transition Are Suppressed by γ-Secretase Inhibitor RO4929097. Medical science monitor: international medical journal of experimental and clinical research, 2018. 24: p. 4046.
9.    Doherty, M.R., et al., The opposing effects of interferon-beta and oncostatin-M as regulators of cancer stem cell plasticity in triple-negative breast cancer. Breast Cancer Research, 2019. 21(1): p. 1-12.
10.    Liao, T.T. and M.H. Yang, Revisiting epithelial‐mesenchymal transition in cancer metastasis: the connection between epithelial plasticity and stemness. Molecular oncology, 2017. 11(7): p. 792-804.
11.    Mashouri, L., et al., Exosomes: composition, biogenesis, and mechanisms in cancer metastasis and drug resistance. Molecular cancer, 2019. 18(1): p. 75.
12.    Atena, M., A.M. Reza, and G. Mehran, A review on the biology of cancer stem cells. Stem Cell Discov, 2014. 4(04): p. 83-89.
13.    Cai, Z., et al., Signalling mechanism (s) of epithelial–mesenchymal transition and cancer stem cells in tumour therapeutic resistance. Clinica Chimica Acta, 2018. 483: p. 156-163.
14.    Celià-Terrassa, T. and M.K. Jolly, Cancer stem cells and epithelial-to-mesenchymal transition in cancer metastasis. Cold Spring Harbor perspectives in medicine, 2020. 10(7): p. a036905.
15.    Chae, Y.C. and J.H. Kim, Cancer stem cell metabolism: target for cancer therapy. BMB reports, 2018. 51(7): p. 319.
16.    Chaurasiya, S., N.G. Chen, and S.G. Warner, Oncolytic virotherapy versus cancer stem cells: A review of approaches and mechanisms. Cancers, 2018. 10(4): p. 124.
17.    Dong, B., et al., MiRNA-mediated EMT and CSCs in cancer chemoresistance. Experimental Hematology & Oncology, 2021. 10(1): p. 1-12.
18.    Lytle, N.K., A.G. Barber, and T. Reya, Stem cell fate in cancer growth, progression and therapy resistance. Nature Reviews Cancer, 2018. 18(11): p. 669-680.
19.    McIntosh, K., C. Balch, and A.K. Tiwari, Tackling multidrug resistance mediated by efflux transporters in tumor-initiating cells. Expert opinion on drug metabolism & toxicology, 2016. 12(6): p. 633-644.
20.    Prieto-Vila, M., et al., Drug resistance driven by cancer stem cells and their niche. International journal of molecular sciences, 2017. 18(12): p. 2574.
21.    Richard, V., T.S. Kumar, and R.M. Pillai, Transitional dynamics of cancer stem cells in invasion and metastasis. Translational Oncology, 2021. 14(1): p. 100909.
22.    Sato, M., et al., Spheroid cancer stem cells display reprogrammed metabolism and obtain energy by actively running the tricarboxylic acid (TCA) cycle. Oncotarget, 2016. 7(22): p. 33297.
23.    Toden, S., et al., Epigallocatechin-3-gallate targets cancer stem-like cells and enhances 5-fluorouracil chemosensitivity in colorectal cancer. Oncotarget, 2016. 7(13): p. 16158.
24.    Zhang, T., et al., Core signaling pathways in ovarian cancer stem cell revealed by integrative analysis of multi-marker genomics data. PLoS One, 2018. 13(5): p. e0196351.
25.    Zhao, H., et al., Differentiated cancer cell-originated lactate promotes the self-renewal of cancer stem cells in patient-derived colorectal cancer organoids. Cancer Letters, 2020. 493: p. 236-244.
26.    Clara, J.A., et al., Targeting signalling pathways and the immune microenvironment of cancer stem cells—A clinical update. Nature Reviews Clinical Oncology, 2020. 17(4): p. 204-232.
27.    Kamdje, A.H.N., et al., Developmental pathways associated with cancer metastasis: Notch, Wnt, and Hedgehog. Cancer biology & medicine, 2017. 14(2): p. 109.
28.    Koury, J., L. Zhong, and J. Hao, Targeting signaling pathways in cancer stem cells for cancer treatment. Stem cells international, 2017. 2017.
29.    Palermo, R., et al., Natural products inspired modulators of cancer stem cells-specific signaling pathways Notch and Hedgehog. Current pharmaceutical design, 2018. 24(36): p. 4251-4269.
30.    Dong, Y., et al., Nanotechnology shaping stem cell therapy: Recent advances, application, challenges, and future outlook. Biomedicine & Pharmacotherapy, 2021. 137: p. 111236.
31.    Rahman, M., et al., The future of glioma treatment: stem cells, nanotechnology and personalized medicine. Future oncology, 2012. 8(9): p. 1149-1156.
32.    Ranji, P., et al., Targeting cancer stem cell-specific markers and/or associated signaling pathways for overcoming cancer drug resistance. Tumor Biology, 2016. 37(10): p. 13059-13075.
33.    Choi, Y.-J., S. Gurunathan, and J.-H. Kim, Graphene oxide–silver nanocomposite enhances cytotoxic and apoptotic potential of salinomycin in human ovarian cancer stem cells (OvCSCs): A novel approach for cancer therapy. International journal of molecular sciences, 2018. 19(3): p. 710.
34.    Hembram, K.C., et al., Quinacrine based gold hybrid nanoparticles caused apoptosis through modulating replication fork in oral cancer stem cells. Molecular pharmaceutics, 2020. 17(7): p. 2463-2472.
35.    Abbaszadegan, M.R., et al., Isolation, identification, and characterization of cancer stem cells: A review. Journal of cellular physiology, 2017. 232(8): p. 2008-2018.
36.    Celià-Terrassa, T., Mammary stem cells and breast cancer stem cells: molecular connections and clinical implications. Biomedicines, 2018. 6(2): p. 50.
37.    Hardavella, G., R. George, and T. Sethi, Lung cancer stem cells—characteristics, phenotype. Translational lung cancer research, 2016. 5(3): p. 272.
38.    Yang, M., P. Liu, and P. Huang, Cancer stem cells, metabolism, and therapeutic significance. Tumor Biology, 2016. 37(5): p. 5735-5742.
39.    Alvarado-Ortiz, E., M.Á. Sarabia-Sánchez, and A. García-Carrancá, Molecular mechanisms underlying the functions of cellular markers associated with the phenotype of cancer stem cells. Current stem cell research & therapy, 2019. 14(5): p. 405-420.
40.    Gerber, J.M., et al., A clinically relevant population of leukemic CD34+ CD38− cells in acute myeloid leukemia. Blood, The Journal of the American Society of Hematology, 2012. 119(15): p. 3571-3577.
41.    Güler, G., U. Guven, and G. Oktem, Characterization of CD133+/CD44+ human prostate cancer stem cells with ATR-FTIR spectroscopy. Analyst, 2019. 144(6): p. 2138-2149.
42.    Herrmann, H., et al., Delineation of target expression profiles in CD34+/CD38− and CD34+/CD38+ stem and progenitor cells in AML and CML. Blood advances, 2020. 4(20): p. 5118-5132.
43.    Kapucuoğlu, N., et al., The clinicopathological and prognostic significance of CD24, CD44, CD133, ALDH1 expressions in invasive ductal carcinoma of the breast: CD44/CD24 expression in breast cancer. Pathology-Research and Practice, 2015. 211(10): p. 740-747.
44.    Park, D.J., et al., EpCAM-high liver cancer stem cells resist natural killer cell–mediated cytotoxicity by upregulating CEACAM1. Journal for immunotherapy of cancer, 2020. 8(1).
45.    Park, S.C., et al., Clonogenically culturing and expanding CD34+ liver cancer stem cells in vitro. Stem cells and development, 2015. 24(13): p. 1506-1514.
46.    Taniguchi, H., et al., Cancer stem cells in human gastrointestinal cancer. Cancer science, 2016. 107(11): p. 1556-1562.
47.    Hu, Y., et al., Cancer stem cells: A shifting subpopulation of cells with stemness? Medical hypotheses, 2013. 80(5): p. 649-655.
48.    Masciale, V., et al., Isolation and identification of cancer stem-like cells in adenocarcinoma and squamous cell carcinoma of the lung: a pilot study. Frontiers in oncology, 2019. 9: p. 1394.
49.    Michishita, M., et al., Identification of tumor-initiating cells in a canine hepatocellular carcinoma cell line. Research in veterinary science, 2014. 96(2): p. 315-322.
50.    Wang, C., et al., Evaluation of CD44 and CD133 as cancer stem cell markers for colorectal cancer. Oncology reports, 2012. 28(4): p. 1301-1308.
51.    He, Y.-F., et al., An efficient method of sorting liver stem cells by using immuno-magnetic microbeads. World Journal of Gastroenterology: WJG, 2006. 12(19): p. 3050.
52.    Jiang, P., et al., Epigallocatechin‑3‑gallate inhibits self‑renewal ability of lung cancer stem‑like cells through inhibition of CLOCK. International Journal of Molecular Medicine, 2020. 46(6): p. 2216-2224.
53.    Poitevin, Y., et al., Magnetic sorting of membrane associated IgG for phenotype-based selection of stable antibody producing cells. Journal of immunological methods, 2017. 444: p. 1-6.
54.    Shen, M.-J., et al., Magnetic-Activated Cell Sorting Using Coiled-Coil Peptides: An Alternative Strategy for Isolating Cells with High Efficiency and Specificity. ACS applied materials & interfaces, 2021. 13(10): p. 11621-11630.
55.    Huang, X.-T., et al., Intracerebroventricular transplantation of ex vivo expanded endothelial colony-forming cells restores blood–brain barrier integrity and promotes angiogenesis of mice with traumatic brain injury. Journal of neurotrauma, 2013. 30(24): p. 2080-2088.
56.    Lei, M., et al., Mesenchymal stem cell characteristics of dental pulp and periodontal ligament stem cells after in vivo transplantation. Biomaterials, 2014. 35(24): p. 6332-6343.
57.    Saheera, S. and R.R. Nair, Accelerated decline in cardiac stem cell efficiency in Spontaneously hypertensive rat compared to normotensive Wistar rat. PloS one, 2017. 12(12): p. e0189129.
58.    Jia, Y., et al., Aberrantly elevated redox sensing factor Nrf2 promotes cancer stem cell survival via enhanced transcriptional regulation of ABCG2 and Bcl-2/Bmi-1 genes. Oncology reports, 2015. 34(5): p. 2296-2304.
59.    Khan, M.I., et al., Current approaches in identification and isolation of human renal cell carcinoma cancer stem cells. Stem cell research & therapy, 2015. 6(1): p. 1-11.
60.    Moghbeli, M., et al., Cancer stem cell detection and isolation. Medical Oncology, 2014. 31(9): p. 69.
61.    Roundhill, E.A., S. Jabri, and S.A. Burchill, ABCG1 and Pgp identify drug resistant, self-renewing osteosarcoma cells. Cancer letters, 2019. 453: p. 142-157.
62.    Lindegaard, A.M., et al., Outcome in patients with isolated regional recurrence after primary radiotherapy for head and neck cancer. Head & Neck, 2020. 42(11): p. 3161-3170.
63.    Liu, Y., et al., Radiotherapy targeting cancer stem cells “awakens” them to induce tumour relapse and metastasis in oral cancer. International journal of oral science, 2020. 12(1): p. 1-12.
64.    Najafi, M., et al., Mechanisms of inflammatory responses to radiation and normal tissues toxicity: clinical implications. International journal of radiation biology, 2018. 94(4): p. 335-356.
65.    Barzaman, K., et al., Breast cancer: Biology, biomarkers, and treatments. International immunopharmacology, 2020. 84: p. 106535.
66.    Brabletz, T., et al., EMT in cancer. Nature Reviews Cancer, 2018. 18(2): p. 128-134.
67.    Chang, L., et al., Acquisition of epithelial–mesenchymal transition and cancer stem cell phenotypes is associated with activation of the PI3K/Akt/mTOR pathway in prostate cancer radioresistance. Cell death & disease, 2013. 4(10): p. e875-e875.
68.    Cruz, M., et al., The stemness phenotype model. International Scholarly Research Notices, 2012. 2012.
69.    Doherty, M.R. and M.W. Jackson, The critical, clinical role of interferon-beta in regulating cancer stem cell properties in triple-negative breast cancer. DNA and cell biology, 2018. 37(6): p. 513-516.
70.    Hii, L.-W., et al., Sphingosine kinase 1 regulates the survival of breast cancer stem cells and non-stem breast cancer cells by suppression of STAT1. Cells, 2020. 9(4): p. 886.
71.    Khelwatty, S.A., et al., Co-expression and prognostic significance of putative CSC markers CD44, CD133, wild-type EGFR and EGFRvIII in metastatic colorectal cancer. Oncotarget, 2019. 10(18): p. 1704.
72.    Lee, Y.-J., et al., A rational approach for cancer stem-like cell isolation and characterization using CD44 and prominin-1 (CD133) as selection markers. Oncotarget, 2016. 7(48): p. 78499.
73.    Leon, G., et al., Cancer stem cells in drug resistant lung cancer: Targeting cell surface markers and signaling pathways. Pharmacology & therapeutics, 2016. 158: p. 71-90.
74.    Li, H. and F. Li, Exosomes from BM-MSCs increase the population of CSCs via transfer of miR-142-3p. British journal of cancer, 2018. 119(6): p. 744-755.
75.    McCubrey, J.A., et al., Effects of mutations in Wnt/β-catenin, hedgehog, Notch and PI3K pathways on GSK-3 activity—Diverse effects on cell growth, metabolism and cancer. Biochimica et Biophysica Acta (BBA)-Molecular Cell Research, 2016. 1863(12): p. 2942-2976.
76.    Morrison, R., et al., Targeting the mechanisms of resistance to chemotherapy and radiotherapy with the cancer stem cell hypothesis. Journal of oncology, 2011. 2011.
77.    Sahlberg, S.H., et al., Evaluation of cancer stem cell markers CD133, CD44, CD24: association with AKT isoforms and radiation resistance in colon cancer cells. PloS one, 2014. 9(4): p. e94621.
78.    Tiwari, A., et al., Evaluation of piperine against cancer stem cells (CSCs) of hepatocellular carcinoma: Insights into epithelial-mesenchymal transition (EMT). Bioorganic Chemistry, 2021. 110: p. 104776.
79.    An, Y., et al., Molecular insights into cancer drug resistance from a proteomics perspective. Expert review of proteomics, 2019. 16(5): p. 413-429.
80.    Carnero, A., et al., The cancer stem-cell signaling network and resistance to therapy. Cancer treatment reviews, 2016. 49: p. 25-36.
81.    Comito, G., et al., Nutritional Exchanges Within Tumor Microenvironment: Impact for Cancer Aggressiveness. Frontiers in oncology, 2020. 10.
82.    Egeblad, M., E.S. Nakasone, and Z. Werb, Tumors as organs: complex tissues that interface with the entire organism. Developmental cell, 2010. 18(6): p. 884-901.
83.    Relation, T., M. Dominici, and E.M. Horwitz, Concise review: an (im) penetrable shield: how the tumor microenvironment protects cancer stem cells. Stem Cells, 2017. 35(5): p. 1123-1130.
84.    Ricketts, C.J., et al., The cancer genome atlas comprehensive molecular characterization of renal cell carcinoma. Cell reports, 2018. 23(1): p. 313-326. e5.
85.    Robey, R.W., et al., Revisiting the role of ABC transporters in multidrug-resistant cancer. Nature Reviews Cancer, 2018. 18(7): p. 452-464.
86.    Senthebane, D.A., et al., The role of tumor microenvironment in chemoresistance: to survive, keep your enemies closer. International journal of molecular sciences, 2017. 18(7): p. 1586.
87.    Tabassum, N., et al., Nanomedicine in cancer stem cell therapy: from fringe to forefront. Cell and tissue research, 2018. 374(3): p. 427-438.
88.    Varas-Godoy, M., G. Rice, and S.E. Illanes, The crosstalk between ovarian cancer stem cell niche and the tumor microenvironment. Stem Cells International, 2017. 2017.
89.    Wang, S.-M., et al., CCAAT/Enhancer-binding protein delta mediates glioma stem-like cell enrichment and ATP-binding cassette transporter ABCA1 activation for temozolomide resistance in glioblastoma. Cell Death Discovery, 2021. 7(1): p. 1-11.
90.    Yi, S.-Y., et al., Cancer stem cells niche: a target for novel cancer therapeutics. Cancer treatment reviews, 2013. 39(3): p. 290-296.
91.    Ji, S., et al., Clinical efficacy of anti-VEGF medications for central serous chorioretinopathy: a meta-analysis. International journal of clinical pharmacy, 2017. 39(3): p. 514-521.
92.    Tang, J.-M., et al., VEGF/SDF-1 promotes cardiac stem cell mobilization and myocardial repair in the infarcted heart. Cardiovascular research, 2011. 91(3): p. 402-411.
93.    Viallard, C. and B. Larrivée, Tumor angiogenesis and vascular normalization: alternative therapeutic targets. Angiogenesis, 2017. 20(4): p. 409-426.
94.    Wang, R., et al., Glioblastoma stem-like cells give rise to tumour endothelium. Nature, 2010. 468(7325): p. 829-833.
95.    Hoffman, L.M., et al., Phase I trial of weekly MK-0752 in children with refractory central nervous system malignancies: a pediatric brain tumor consortium study. Child’s Nervous System, 2015. 31(8): p. 1283-1289.
96.    Wang, D., et al., IL6 blockade potentiates the anti-tumor effects of γ-secretase inhibitors in Notch3-expressing breast cancer. Cell Death & Differentiation, 2018. 25(2): p. 330-339.
97.    Dhar, D., K. Raina, and R. Agarwal, Mechanisms and drug targets for pancreatic cancer chemoprevention. Current medicinal chemistry, 2018. 25(22): p. 2545-2565.
98.    Gordon, W.R., et al., Mechanical allostery: evidence for a force requirement in the proteolytic activation of Notch. Developmental cell, 2015. 33(6): p. 729-736.
99.    Ferrarotto, R., et al., A phase I dose-escalation and dose-expansion study of brontictuzumab in subjects with selected solid tumors. Annals of Oncology, 2018. 29(7): p. 1561-1568.
100.    Falk, R., et al., Generation of anti-Notch antibodies and their application in blocking Notch signalling in neural stem cells. Methods, 2012. 58(1): p. 69-78.
101.    Tang, J.-Y., et al., HPV 16 E6/E7 Promote the Glucose Uptake of GLUT1 in Lung Cancer Through Downregulation of TXNIP Due to Inhibition of PTEN Phosphorylation. Frontiers in Oncology, 2020. 10: p. 2470.
102.    Xiang, D.-B., et al., Curcumin: From a controversial “panacea” to effective antineoplastic products. Medicine, 2020. 99(2).
103.    Huang, D., et al., In Vitro Evaluation of Clinical Candidates of γ-Secretase Inhibitors: Effects on Notch Inhibition and Promoting Beige Adipogenesis and Mitochondrial Biogenesis. Pharmaceutical Research, 2020. 37(10): p. 1-13.
104.    O’Reilly, E.M., et al., Results of a randomized phase II trial of an anti-notch 2/3, tarextumab (OMP-59R5, TRXT, anti-Notch2/3), in combination with nab-paclitaxel and gemcitabine (Nab-P+ Gem) in patients (pts) with untreated metastatic pancreatic cancer (mPC). J Clin Oncol, 2017. 35(Suppl 4): p. 279.
105.    Takebe, N., et al., Targeting Notch, Hedgehog, and Wnt pathways in cancer stem cells: clinical update. Nature reviews Clinical oncology, 2015. 12(8): p. 445.
106.    Martinas, I. and E.T. Parkin, Notch Signalling as a Therapeutic Target in Cancer. International Journal of Cancer Therapy and Oncology, 2016. 4(4).
107.    Venkatesh, V., et al., Targeting Notch signalling pathway of cancer stem cells. Stem cell investigation, 2018. 5.
108.    Harb, J., P.-J. Lin, and J. Hao, Recent development of Wnt signaling pathway inhibitors for cancer therapeutics. Current oncology reports, 2019. 21(2): p. 12.
109.    Tuysuz, E.C., et al., Distinctive role of dysregulated miRNAs in chordoma cancer stem-like cell maintenance. Experimental cell research, 2019. 380(1): p. 9-19.
110.    Diamond, J.R., et al., Phase Ib clinical trial of the anti-frizzled antibody vantictumab (OMP-18R5) plus paclitaxel in patients with locally advanced or metastatic HER2-negative breast cancer. Breast Cancer Research and Treatment, 2020. 184(1): p. 53-62.
111.    Tamiro, F., A.P. Weng, and V. Giambra, Targeting Leukemia-Initiating Cells in Acute Lymphoblastic Leukemia. Cancer Research, 2021.
112.    Marcucci, F., et al., Antibody-Drug Conjugates (ADC) Against Cancer Stem-Like Cells (CSC)—Is There Still Room for Optimism? Frontiers in oncology, 2019. 9: p. 167.
113.    Karvonen, H., et al., Molecular mechanisms associated with ROR1-mediated drug resistance: crosstalk with Hippo-YAP/TAZ and BMI-1 pathways. Cells, 2019. 8(8): p. 812.
114.    van Schie, E.H. and R. van Amerongen, Aberrant WNT/CTNNB1 signaling as a therapeutic target in human breast Cancer: weighing the evidence. Frontiers in cell and developmental biology, 2020. 8: p. 25.
115.    Cheng, Y., et al., Wnt-C59 arrests stemness and suppresses growth of nasopharyngeal carcinoma in mice by inhibiting the Wnt pathway in the tumor microenvironment. Oncotarget, 2015. 6(16): p. 14428.
116.    Yang, Y., et al., Emerging agents that target signaling pathways in cancer stem cells. Journal of Hematology & Oncology, 2020. 13: p. 1-18.
117.    Katoh, M., Multi‑layered prevention and treatment of chronic inflammation, organ fibrosis and cancer associated with canonical WNT/β‑catenin signaling activation. International journal of molecular medicine, 2018. 42(2): p. 713-725.
118.    Nakagawa, Y., et al., Multiple myeloma cells adapted to long-exposure of hypoxia exhibit stem cell characters with TGF-β/Smad pathway activation. Biochemical and biophysical research communications, 2018. 496(2): p. 490-496.
119.    Kierulf-Vieira, K.S., et al., A small-molecule tankyrase inhibitor reduces glioma stem cell proliferation and sphere formation. Cancers, 2020. 12(6): p. 1630.
120.    Pouyafar, A., et al., Treatment of cancer stem cells from human colon adenocarcinoma cell line HT-29 with resveratrol and sulindac induced mesenchymal-endothelial transition rate. Cell and tissue research, 2019. 376(3): p. 377-388.
121.    Schatoff, E.M., B.I. Leach, and L.E. Dow, Wnt signaling and colorectal cancer. Current colorectal cancer reports, 2017. 13(2): p. 101-110.
122.    Stakheev, D., et al., The WNT/β-catenin signaling inhibitor XAV939 enhances the elimination of LNCaP and PC-3 prostate cancer cells by prostate cancer patient lymphocytes in vitro. Scientific reports, 2019. 9(1): p. 1-14.
123.    Dai, Y., X. Guo, and C. Yang, Effect of bortezomib on proliferation and apoptosis of myeloma cells by activating Wnt/β‑catenin signaling pathway. Oncology Letters, 2020. 20(2): p. 1295-1299.
124.    Li, Y., et al., Overexpression of G3BP1 facilitates the progression of colon cancer by activating β‑catenin signaling. Molecular Medicine Reports, 2020. 22(5): p. 4403-4411.
125.    Manegold, P., et al., Differentiation therapy targeting the β-catenin/CBP interaction in pancreatic cancer. Cancers, 2018. 10(4): p. 95.
126.    Schmidtova, S., et al., Targeting of Deregulated Wnt/β-Catenin Signaling by PRI-724 and LGK974 Inhibitors in Germ Cell Tumor Cell Lines. International journal of molecular sciences, 2021. 22(8): p. 4263.
127.    Vallée, A. and Y. Lecarpentier, Crosstalk between peroxisome proliferator-activated receptor gamma and the canonical WNT/β-catenin pathway in chronic inflammation and oxidative stress during carcinogenesis. Frontiers in immunology, 2018. 9: p. 745.
128.    Zhang, X., L. Wang, and Y. Qu, Targeting the β-catenin signaling for cancer therapy. Pharmacological research, 2020: p. 104794.
129.    Liu, B., et al., Celecoxib enhances anticancer effect of cisplatin and induces anoikis in osteosarcoma via PI3K/Akt pathway. Cancer cell international, 2017. 17(1): p. 1-8.
130.    Harada, K., et al., Hedgehog Signal Inhibitor GANT61 Inhibits the Malignant Behavior of Undifferentiated Hepatocellular Carcinoma Cells by Targeting Non-Canonical GLI Signaling. International journal of molecular sciences, 2020. 21(9): p. 3126.
131.    Israel, B.e.B., et al., Phytochemicals: Current strategies for treating breast cancer. Oncology letters, 2018. 15(5): p. 7471-7478.
132.    Reyes-Ramos, A.M., et al., Mesenchymal Cells Support the Oncogenicity and Therapeutic Response of the Hedgehog Pathway in Triple-Negative Breast Cancer. Cancers, 2019. 11(10): p. 1522.
133.    Sharma, N., et al., PI3K/AKT/mTOR and sonic hedgehog pathways cooperate together to inhibit human pancreatic cancer stem cell characteristics and tumor growth. Oncotarget, 2015. 6(31): p. 32039.
134.    Takebe, N., et al., Targeting cancer stem cells by inhibiting Wnt, Notch, and Hedgehog pathways. Nature reviews Clinical oncology, 2011. 8(2): p. 97-106.
135.    Zhang, X., et al., Development of anticancer agents targeting the Hedgehog signaling. Cellular and Molecular Life Sciences, 2017. 74(15): p. 2773-2782.
136.    Mistretta, C.M. and A. Kumari, Hedgehog signaling regulates taste organs and oral sensation: distinctive roles in the epithelium, stroma, and innervation. International journal of molecular sciences, 2019. 20(6): p. 1341.
137.    Sukrithan, V., et al., Emerging drugs for the treatment of Adrenocortical Carcinoma. Expert opinion on emerging drugs, 2021(just-accepted).
138.    Lian, S., et al., Checkpoint CD47 function on tumor metastasis and immune therapy. OncoTargets and therapy, 2019. 12: p. 9105.
139.    Watanabe, I., et al., Isolation and characterization of monoclonal antibodies specific for chondroitin sulfate E. Glycobiology, 2015. 25(9): p. 953-962.
140.    Hof, D.J., et al., A versatile salt-based method to immobilize glycosaminoglycans and create growth factor gradients. Glycoconjugate journal, 2019. 36(3): p. 227-236.
141.    Smetsers, T.F., et al., Human single-chain antibodies reactive with native chondroitin sulfate detect chondroitin sulfate alterations in melanoma and psoriasis. Journal of investigative dermatology, 2004. 122(3): p. 707-716.
142.    Vallen, M.J., et al., Highly sulfated chondroitin sulfates, a novel class of prognostic biomarkers in ovarian cancer tissue. Gynecologic oncology, 2012. 127(1): p. 202-209.
143.    Brossa, A., L. Buono, and B. Bussolati, Effect of the monoclonal antibody TRC105 in combination with Sunitinib on renal tumor derived endothelial cells. Oncotarget, 2018. 9(32): p. 22680.
144.    Han, C., T. Liu, and R. Yin, Biomarkers for cancer-associated fibroblasts. Biomarker Research, 2020. 8(1): p. 1-8.
145.    Lappano, R., et al., Cancer associated fibroblasts: Role in breast cancer and potential as therapeutic targets. Expert opinion on therapeutic targets, 2020. 24(6): p. 559-572.
146.    Sandberg, T.P., et al., Increased expression of cancer-associated fibroblast markers at the invasive front and its association with tumor-stroma ratio in colorectal cancer. BMC cancer, 2019. 19(1): p. 284.
147.    Jacquier, V., et al., RIP140 inhibits glycolysis-dependent proliferation of cancer cells by regulating transcriptional crosstalk between hypoxia induced factor and p53. bioRxiv, 2020.
148.    Almutairi, F.M., A.A. Abd-Rabou, and M.S. Mohamed, Raloxifene-encapsulated hyaluronic acid-decorated chitosan nanoparticles selectively induce apoptosis in lung cancer cells. Bioorganic & medicinal chemistry, 2019. 27(8): p. 1629-1638.
149.    Duggal, R., et al., Biotherapeutic approaches to target cancer stem cells. Journal of stem cells, 2013. 8(3/4): p. 135.
150.    Pęcak, A., et al., Anti-CD44 DNA Aptamers Selectively Target Cancer Cells. nucleic acid therapeutics, 2020. 30(5): p. 289-298.
151.    Levy, R., et al., Enhancement of antibody fragment secretion into the Escherichia coli periplasm by co-expression with the peptidyl prolyl isomerase, FkpA, in the cytoplasm. Journal of immunological methods, 2013. 394(1-2): p. 10-21.
152.    Liao, M.-Y., et al., An anti-EpCAM antibody EpAb2-6 for the treatment of colon cancer. Oncotarget, 2015. 6(28): p. 24947.
153.    Knödler, M., et al., Randomised phase II trial to investigate catumaxomab (anti-EpCAM× anti-CD3) for treatment of peritoneal carcinomatosis in patients with gastric cancer. British journal of cancer, 2018. 119(3): p. 296-302.
154.    Bartholomeusz, C., et al., MEK inhibitor selumetinib (AZD6244; ARRY-142886) prevents lung metastasis in a triple-negative breast cancer xenograft model. Molecular cancer therapeutics, 2015. 14(12): p. 2773-2781.
155.    Abdel-Salam, I.M., A. Abou-Bakr, and M. Ashour, Cytotoxic effect of aqueous ethanolic extract of Luffa cylindrica leaves on cancer stem cells CD44+/24-in breast cancer patients with various molecular sub-types using tissue samples in vitro. Journal of ethnopharmacology, 2019. 238: p. 111877.
156.    Obayashi, S., et al., Stathmin1 expression is associated with aggressive phenotypes and cancer stem cell marker expression in breast cancer patients. International journal of oncology, 2017. 51(3): p. 781-790.
157.    Jang, M.H., et al., Clinicopathological analysis of CD44 and CD24 expression in invasive breast cancer. Oncology letters, 2016. 12(4): p. 2728-2733.
158.    Moon, Y.W., et al., CD44/CD24 and aldehyde dehydrogenase 1 in estrogen receptor-positive early breast cancer treated with tamoxifen: CD24 positivity is a poor prognosticator. Oncotarget, 2018. 9(2): p. 2622.
159.    Loh, T.J., et al., CD44 alternative splicing and hnRNP A1 expression are associated with the metastasis of breast cancer. Oncology reports, 2015. 34(3): p. 1231-1238.
160.    Yang, Z., et al., Tenascin-C predicts poor outcomes for patients with colorectal cancer and drives cancer stemness via Hedgehog signaling pathway. Cancer cell international, 2020. 20: p. 1-11.
161.    Hu, B., et al., CD44 promotes cell proliferation in non-small cell lung cancer. Oncology letters, 2018. 15(4): p. 5627-5633.
162.    Hanke, M., et al., Differences between healthy hematopoietic progenitors and leukemia cells with respect to CD44 mediated rolling versus adherence behavior on hyaluronic acid coated surfaces. Biomaterials, 2014. 35(5): p. 1411-1419.
163.    Al Ssadh, H. and W. Al Abdulmonem, Immunophenotyping of the cluster of differentiation 74, migration inhibitory factor, and cluster of differentiation 44 expression on human breast cancer-derived cell lines. International journal of health sciences, 2019. 13(2): p. 17.
164.    Bostad, M., et al., Light-controlled endosomal escape of the novel CD133-targeting immunotoxin AC133–saporin by photochemical internalization—A minimally invasive cancer stem cell-targeting strategy. Journal of Controlled Release, 2015. 206: p. 37-48.
165.    Glumac, P.M., et al., The identification of a novel antibody for CD133 using human antibody phage display. The Prostate, 2018. 78(13): p. 981-991.
166.    Schmied, B.J., et al., An Fc-Optimized CD133 antibody for induction of natural killer cell reactivity against colorectal cancer. Cancers, 2019. 11(6): p. 789.
167.    Kato, Y., et al., Anti-CD133 monoclonal antibody CMab-43 exerts antitumor activity in a mouse xenograft model of colon cancer. Monoclonal antibodies in immunodiagnosis and immunotherapy, 2019. 38(2): p. 75-78.
168.    Yohana, Y., Pendeteksian petanda kepuncaan glioblastoma multiforme. Jurnal Biomedika dan Kesehatan, 2020. 3(1): p. 39-45.
169.    Han, Y., et al., Simultaneously target of normal and stem cells-like gastric cancer cells via cisplatin and anti-CD133 CAR-T combination therapy. Cancer Immunology, Immunotherapy, 2021: p. 1-9.
170.    Alfakir, M., et al., The temporal and spatial expression patterns of ABCG2 in the developing human heart. International journal of cardiology, 2012. 156(2): p. 133-138.
171.    Broxmeyer, H.E., S. Cooper, and M.L. Capitano, Enhanced collection of phenotypic and engrafting human cord blood hematopoietic stem cells at 4° C. Stem Cells, 2020. 38(10): p. 1326-1331.
172.    Fan, C.-Y., et al., De novo protein sequencing, humanization and in vitro effects of an antihuman CD34 mouse monoclonal antibody. Biochemistry and biophysics reports, 2017. 9: p. 51-60.
173.    Maleki, L.A., et al., Generation and characterization of anti-CD34 monoclonal antibodies that react with hematopoietic stem cells. Cell Journal (Yakhteh), 2014. 16(3): p. 361.
174.    Qian, W., et al., Development of new versions of anti-human CD34 monoclonal antibodies with potentially reduced immunogenicity. Biochemical and biophysical research communications, 2008. 367(2): p. 497-502.
175.    Kuranda, K., et al., A subpopulation of malignant CD34+ CD138+ B7-H1+ plasma cells is present in multiple myeloma patients. Experimental hematology, 2010. 38(2): p. 124-131. e4.
176.    Valipour, B., et al., Cord blood stem cell derived CD16+ NK cells eradicated acute lymphoblastic leukemia cells using with anti-CD47 antibody. Life sciences, 2020. 242: p. 117223.
177.    Sato, T., et al., A novel mode of stimulating platelet formation activity in megakaryocytes with peanut skin extract. Journal of natural medicines, 2018. 72(1): p. 211-219.
178.    Hou, S., et al., Humanization of an anti-CD34 monoclonal antibody by complementarity-determining region grafting based on computer-assisted molecular modelling. Journal of biochemistry, 2008. 144(1): p. 115-120.
179.    Berger, M.G., et al., Efficiency of nilotinib to target chronic phase-chronic myeloid leukaemia primary mature CD34− and immature CD34+ cells. Scientific reports, 2021. 11(1): p. 1-11.
180.    Capone, S., et al., Methylome of human senescent hematopoietic progenitors. Experimental hematology & oncology, 2018. 7(1): p. 1-7.
181.    Amici, S.A., et al., CD38 is robustly induced in human macrophages and monocytes in inflammatory conditions. Frontiers in immunology, 2018. 9: p. 1593.
182.    Wozniak, M., A. Mielczarek, and M. Czyz, miRNAs in melanoma: tumor suppressors and oncogenes with prognostic potential. Current medicinal chemistry, 2016. 23(28): p. 3136-3153.
183.    Motti, M.L., et al., MicroRNAs as key players in melanoma cell resistance to MAPK and immune checkpoint inhibitors. International Journal of Molecular Sciences, 2020. 21(12): p. 4544.
184.    Bruhn, O., et al., Length variants of the ABCB1 3′-UTR and loss of miRNA binding sites: possible consequences in regulation and pharmacotherapy resistance. Pharmacogenomics, 2016. 17(4): p. 327-340.
185.    Messingerova, L., et al., A decrease in cellular microRNA-27a content is involved in azacytidine-induced P-glycoprotein expression in SKM-1 cells. Toxicology in Vitro, 2016. 36: p. 81-88.
186.    Cheng, Y., et al., Knockdown of lncRNA NCK-AS1 regulates cisplatin resistance through modulating miR-137 in osteosarcoma cells. OncoTargets and therapy, 2019. 12: p. 11057.
187.    Wang, Z., et al., Circular RNA PVT1 promotes metastasis via miR-145 sponging in CRC. Biochemical and biophysical research communications, 2019. 512(4): p. 716-722.
188.    Xie, Y., et al., MicroRNA-298 reverses multidrug resistance to antiepileptic drugs by suppressing MDR1/P-gp expression in vitro. Frontiers in neuroscience, 2018. 12: p. 602.
189.    do Imperio, G.E., et al., Chorioamnionitis induces a specific signature of placental ABC transporters associated with an increase of miR-331-5p in the human preterm placenta. Cellular Physiology and Biochemistry, 2018. 45(2): p. 591-604.
190.    Soltani, I., et al., Downregulation of miR-451 in Tunisian chronic myeloid leukemia patients: potential implication in imatinib resistance. Hematology, 2017. 22(4): p. 201-207.
191.    Arkorful, M.A., et al., MicroRNA-1253 Regulation of WASF2 (WAVE2) and its Relevance to Racial Health Disparities. Genes, 2020. 11(5): p. 572.
192.    Xie, Y., et al., Long Non-coding RNA KCNQ1OT1 Contributes to antiepileptic drug resistance through the miR-138-5p/ABCB1 axis in vitro. Frontiers in neuroscience, 2019. 13: p. 1358.
193.    Feng, W., et al., ncRNAs associated with drug resistance and the therapy of digestive system neoplasms. Journal of cellular physiology, 2019. 234(11): p. 19143-19157.
194.    Rodrigues, A.C., et al., Atorvastatin attenuation of ABCB1 expression is mediated by microRNA miR-491-3p in Caco-2 cells. European Journal of Pharmaceutical Sciences, 2016. 93: p. 431-436.
195.    Li, Y., et al., [Corrigendum] miR-9 regulates the multidrug resistance of chronic myelogenous leukemia by targeting ABCB1. oncology reports, 2019. 41(5): p. 3148-3148.
196.    Zhang, H., et al., Sensitivity of non-small cell lung cancer to erlotinib is regulated by the Notch/miR-223/FBXW7 pathway. Bioscience reports, 2017. 37(3).
197.    Feng, J. and T. Wang, MicroRNA‑873 serves a critical role in human cervical cancer proliferation and metastasis via regulating glioma‑associated oncogene homolog 1. Experimental and therapeutic medicine, 2020. 19(2): p. 1243-1250.
198.    Al-Momany, B.Z., H.M. Hammad, and M. Ahram, Regulation of chemoresponsiveness in triple-negative breast cancer: androgen receptor, ABCG2, and microRNA. Jordan Journal of Biological Sciences, 2020. 13(3).
199.    Hsieh, M.-J., et al., Effects of miR-34b/miR-892a upregulation and inhibition of ABCB1/ABCB4 on melatonin-induced apoptosis in VCR-resistant oral cancer cells. Molecular Therapy-Nucleic Acids, 2020. 19: p. 877-889.
200.    Hu, W.-X., H. Li, and J.-Z. Jiang, MiR-491-3p is down-regulated in postmenopausal osteoporosis and affects growth, differentiation and apoptosis of hFOB1. 19 cells through targeting CTSS. Folia histochemica et cytobiologica, 2020. 1(1): p. 9-16.
201.    Qin, Q., et al., ZNF295-AS1 inhibits autophagy via the ZNF295-AS1/miR-508-5p/ATG7 axis in AS. Eur Rev Med Pharmacol Sci, 2020. 24(12): p. 7024-7032.
202.    Bonde, G.V., et al., Lapatinib nano-delivery systems: a promising future for breast cancer treatment. Expert opinion on drug delivery, 2018. 15(5): p. 495-507.
203.    Guha, D., et al., Reactive oxygen species: friends or foes of lung cancer?, in Oxidative stress in lung diseases. 2020, Springer. p. 331-352.
204.    Hajari, M.A., S.B. Islami, and X. Chen, A numerical study on tumor-on-chip performance and its optimization for nanodrug-based combination therapy. Biomechanics and Modeling in Mechanobiology, 2021: p. 1-20.
205.    Karpuz, M., M. Silindir-Gunay, and A.Y. Ozer, Current and future approaches for effective cancer imaging and treatment. Cancer biotherapy & radiopharmaceuticals, 2018. 33(2): p. 39-51.
206.    Konrad, C.V., et al., The role of cancer stem cells in tumor heterogeneity and resistance to therapy. Canadian journal of physiology and pharmacology, 2017. 95(1): p. 1-15.
207.    Li, J., et al., Adsorption of lysozyme by alginate/graphene oxide composite beads with enhanced stability and mechanical property. Materials Science and Engineering: C, 2018. 89: p. 25-32.
208.    López-Gil, J.C., et al., The CXCL12 Crossroads in Cancer Stem Cells and Their Niche. Cancers, 2021. 13(3): p. 469.
209.    Louka, M., et al., DNA damage response/repair in cancer stem cells—potential vs. controversies. Advances in DNA Repair, 2015. 10: p. 61355.
210.    Marzagalli, M., et al., Cancer Stem Cells—Key Players in Tumor Relapse. Cancers, 2021. 13(3): p. 376.
211.    Montaseri, H., C.A. Kruger, and H. Abrahamse, Organic nanoparticle based active targeting for photodynamic therapy treatment of breast cancer cells. Oncotarget, 2020. 11(22): p. 2120.
212.    Muriithi, W., et al., ABC transporters and the hallmarks of cancer: roles in cancer aggressiveness beyond multidrug resistance. Cancer Biology & Medicine, 2020. 17(2): p. 253.
213.    Nejad, A.E., et al., The role of hypoxia in the tumor microenvironment and development of cancer stem cell: a novel approach to developing treatment. Cancer Cell International, 2021. 21(1): p. 1-26.
214.    Niero, E.L., et al., The multiple facets of drug resistance: one history, different approaches. Journal of Experimental & Clinical Cancer Research, 2014. 33(1): p. 1-14.
215.    Prabhu, K.S., et al., Non-coding RNAs as regulators and markers for targeting of breast cancer and cancer stem cells. Cancers, 2020. 12(2): p. 351.
216.    Roca, M.S., E. Di Gennaro, and A. Budillon, Implication for cancer stem cells in solid cancer chemo-resistance: promising therapeutic strategies based on the use of HDAC inhibitors. Journal of clinical medicine, 2019. 8(7): p. 912.
217.    Sahu, A., W.I. Choi, and G. Tae, Recent Progress in the Design of Hypoxia‐Specific Nano Drug Delivery Systems for Cancer Therapy. Advanced Therapeutics, 2018. 1(4): p. 1800026.
218.    Shen, S., J.-X. Xia, and J. Wang, Nanomedicine-mediated cancer stem cell therapy. Biomaterials, 2016. 74: p. 1-18.
219.    Tarone, L., et al., Naturally occurring cancers in pet dogs as pre-clinical models for cancer immunotherapy. Cancer Immunology, Immunotherapy, 2019. 68(11): p. 1839-1853.
220.    Wang, N., et al., Nanoparticulate carriers used as vaccine adjuvant delivery systems. Critical Reviews™ in Therapeutic Drug Carrier Systems, 2019. 36(5).
221.    Xiang, D., et al., Transforming doxorubicin into a cancer stem cell killer via EpCAM aptamer-mediated delivery. Theranostics, 2017. 7(17): p. 4071.
222.    Xiao, Y., et al., Combinational dual drug delivery system to enhance the care and treatment of gastric cancer patients. Drug Delivery, 2020. 27(1): p. 1491-1500.
223.    Chen, K., Y.-h. Huang, and J.-l. Chen, Understanding and targeting cancer stem cells: therapeutic implications and challenges. Acta Pharmacologica Sinica, 2013. 34(6): p. 732-740.
224.    Leary, M., et al., Sensitization of drug resistant cancer cells: a matter of combination therapy. Cancers, 2018. 10(12): p. 483.
225.    Walker, S., et al., Extracellular vesicle-based drug delivery systems for cancer treatment. Theranostics, 2019. 9(26): p. 8001.
226.    Wang, S., et al., Effect and mechanism of resveratrol on drug resistance in human bladder cancer cells. Molecular medicine reports, 2017. 15(3): p. 1179-1187.
227.    Ye, B., et al., Pulsatilla saponin A Induces Apoptosis and Differentiation of Myeloma Cells. Anti-Cancer Agents in Medicinal Chemistry (Formerly Current Medicinal Chemistry-Anti-Cancer Agents), 2021. 21(7): p. 919-926.
228.    Dalpiaz, A., et al., Cancer stem cells and nanomedicine: new opportunities to combat multidrug resistance? Drug Discovery Today, 2020.
229.    Das, M.K., A. Sarma, and T. Deka, Polydopamine-based simple and versatile surface modification of polymeric nano drug carriers, in Surface modification of nanoparticles for targeted drug delivery. 2019, Springer. p. 369-389.
230.    Elzahhar, P., et al., Bioconjugation in drug delivery: practical perspectives and future perceptions. Pharmaceutical Nanotechnology, 2019: p. 125-182.
231.    Gener, P., et al., The potential of nanomedicine to alter cancer stem cell dynamics: the impact of extracellular vesicles. Nanomedicine, 2020. 15(29): p. 2785-2800.
232.    Gong, P., et al., Nano-sized paramagnetic and fluorescent fluorinated carbon fiber with high NIR absorbance for cancer chemo-photothermal therapy. Journal of Materials Chemistry B, 2018. 6(19): p. 3068-3077.
233.    Haeri, A., et al., Nanomedicine approaches for sirolimus delivery: a review of pharmaceutical properties and preclinical studies. Artificial cells, nanomedicine, and biotechnology, 2018. 46(sup1): p. 1-14.
234.    Huang, L., et al., Nanomedicine–a promising therapy for hematological malignancies. Biomaterials science, 2020. 8(9): p. 2376-2393.
235.    Leung, H.-W., et al., The natural agent 4-vinylphenol targets metastasis and stemness features in breast cancer stem-like cells. Cancer chemotherapy and pharmacology, 2018. 82(2): p. 185-197.
236.    Majumder, J. and T. Minko, Multifunctional and stimuli-responsive nanocarriers for targeted therapeutic delivery. Expert Opinion on Drug Delivery, 2020: p. 1-23.
237.    Molina-Peña, R., J.C. Tudon-Martinez, and O. Aquines-Gutiérrez, A mathematical model of average dynamics in a stem cell hierarchy suggests the combinatorial targeting of cancer stem cells and progenitor cells as a potential strategy against tumor growth. Cancers, 2020. 12(9): p. 2590.
238.    Pasban, S. and H. Raissi, New insights into Hexakis macrocycles as a novel nano-carrier for highly potent anti-cancer treatment: A new challenge in drug delivery. Colloids and Surfaces B: Biointerfaces, 2021. 197: p. 111402.
239.    Pichler, R., et al., PD-L1 expression in bladder cancer and metastasis and its influence on oncologic outcome after cystectomy. Oncotarget, 2017. 8(40): p. 66849.
240.    Raj, S., et al. Specific targeting cancer cells with nanoparticles and drug delivery in cancer therapy. in Seminars in cancer biology. 2021. Elsevier.
241.    Schulz, A., et al., Cancer stem cells and radioresistance: DNA repair and beyond. Cancers, 2019. 11(6): p. 862.
242.    Tsolou, A., et al., Folate and Pegylated Aliphatic Polyester Nanoparticles for Targeted Anticancer Drug Delivery. International Journal of Nanomedicine, 2020. 15: p. 4899.
243.    Vaze, N., et al., A nano-carrier platform for the targeted delivery of nature-inspired antimicrobials using Engineered Water Nanostructures for food safety applications. Food control, 2019. 96: p. 365-374.
244.    Yang, Z., et al., Hybrid nanoparticles coated with hyaluronic acid lipoid for targeted co-delivery of paclitaxel and curcumin to synergistically eliminate breast cancer stem cells. Journal of Materials Chemistry B, 2017. 5(33): p. 6762-6775.
245.    Younus, N., et al., Comparative effects of dietary micro-and nano-scale chitosan on the growth performance, non-specific immunity, and resistance of silver carp Hypophthalmichthys molitrix against Staphylococcus aureus infection. Aquaculture International, 2020. 28(6): p. 2363-2378.
246.    Gupta, P.K., et al., Nanomedicine in Cancer Stem Cell Therapy, in NanoBioMedicine. 2020, Springer. p. 67-105.
247.    Jena, L.N., et al., Exploiting the anticancer effects of a nitrogen bisphosphonate nanomedicine for glioblastoma multiforme. Journal of nanobiotechnology, 2021. 19(1): p. 1-18.
248.    Prasad, S.R., A. Jayakrishnan, and T.S. Kumar, Combinational delivery of anticancer drugs for osteosarcoma treatment using electrosprayed core shell nanocarriers. Journal of Materials Science: Materials in Medicine, 2020. 31(5): p. 1-11.
249.    Wang, H., et al., Carbon nano-onion-mediated dual targeting of P-selectin and P-glycoprotein to overcome cancer drug resistance. Nature communications, 2021. 12(1): p. 1-14.
250.    Bao, Z., et al., Nanoscale metal–organic framework composites for phototherapy and synergistic therapy of cancer. Materials Chemistry Frontiers, 2021. 5(4): p. 1632-1654.
251.    Cheng, D.-B., et al., Recent advances of morphology adaptive nanomaterials for anti-cancer drug delivery. Progress in Natural Science: Materials International, 2020.
252.    Gao, D., et al., Multifunctional phototheranostic nanomedicine for cancer imaging and treatment. Materials Today Bio, 2020. 5: p. 100035.
253.    Kim, S.H., et al., NIR fluorescence for monitoring in vivo scaffold degradation along with stem cell tracking in bone tissue engineering. Biomaterials, 2020. 258: p. 120267.
254.    Liu, W., et al., Nanomedicine Enables Drug-Potency Activation with Tumor Sensitivity and Hyperthermia Synergy in the Second Near-Infrared Biowindow. ACS nano, 2021. 15(4): p. 6457-6470.
255.    Meng, J., et al., Advances in metal–organic framework coatings: versatile synthesis and broad applications. Chemical Society Reviews, 2020. 49(10): p. 3142-3186.
256.    Roma‐Rodrigues, C., et al., Combined cancer therapeutics—Tackling the complexity of the tumor microenvironment. Wiley Interdisciplinary Reviews: Nanomedicine and Nanobiotechnology, 2021: p. e1704.
257.    Saifullah, S., et al., Surface functionalized magnetic nanoparticles for targeted cancer therapy and diagnosis, in Metal Nanoparticles for Drug Delivery and Diagnostic Applications. 2020, Elsevier. p. 215-236.
258.    Shi, Z., et al., Inorganic nano-carriers based smart drug delivery systems for tumor therapy. Smart Materials in Medicine, 2020.
259.    Tan, Y.Y., et al., Perspectives and advancements in the design of nanomaterials for targeted cancer theranostics. Chemico-biological interactions, 2020: p. 109221.
260.    Zhao, X., S.-Q. Zang, and X. Chen, Stereospecific interactions between chiral inorganic nanomaterials and biological systems. Chemical Society Reviews, 2020. 49(8): p. 2481-2503.
261.    Carrillo-Carrión, C., Nanoscale metal–organic frameworks as key players in the context of drug delivery: evolution toward theranostic platforms. Analytical and bioanalytical chemistry, 2020. 412(1): p. 37-54.
262.    Kang, T., et al., Surface design of magnetic nanoparticles for stimuli-responsive cancer imaging and therapy. Biomaterials, 2017. 136: p. 98-114.
263.    Liu, W., et al., Tumor-targeted pH-low insertion peptide delivery of theranostic gadolinium nanoparticles for image-guided nanoparticle-enhanced radiation therapy. Translational Oncology, 2020. 13(11): p. 100839.
264.    Mi, P., Stimuli-responsive nanocarriers for drug delivery, tumor imaging, therapy and theranostics. Theranostics, 2020. 10(10): p. 4557.
265.    Rai, M., et al., Bioactivity of noble metal nanoparticles decorated with biopolymers and their application in drug delivery. International journal of pharmaceutics, 2015. 496(2): p. 159-172.
266.    Sharma, A., A.K. Goyal, and G. Rath, Recent advances in metal nanoparticles in cancer therapy. Journal of drug targeting, 2018. 26(8): p. 617-632.
267.    Tran, S., et al., Cancer nanomedicine: a review of recent success in drug delivery. Clinical and translational medicine, 2017. 6(1): p. 1-21.
268.    Turan, O., et al., Effect of dose and selection of two different ligands on the deposition and antitumor efficacy of targeted nanoparticles in brain tumors. Molecular pharmaceutics, 2019. 16(10): p. 4352-4360.
269.    Ye, J., et al., Low-density lipoprotein decorated silica nanoparticles co-delivering sorafenib and doxorubicin for effective treatment of hepatocellular carcinoma. Drug delivery, 2018. 25(1): p. 2007-2014.
270.    Kunoh, T., et al., Use of DNA-generated gold nanoparticles to radiosensitize and eradicate radioresistant glioma stem cells. Nanotechnology, 2018. 30(5): p. 055101.
271.    Yi, Y., et al., Glucose-linked sub-50-nm unimer polyion complex-assembled gold nanoparticles for targeted siRNA delivery to glucose transporter 1-overexpressing breast cancer stem-like cells. Journal of Controlled Release, 2019. 295: p. 268-277.
272.    Mehrnia, S.S., et al., Radiosensitization of breast cancer cells using AS1411 aptamer-conjugated gold nanoparticles. Radiation Oncology, 2021. 16(1): p. 1-12.
273.    Huai, Y., et al., Gold Nanoparticles sensitize pancreatic cancer cells to gemcitabine. Cell Stress, 2019. 3(8): p. 267.
274.    Kaushik, N.K., et al., Low doses of PEG-coated gold nanoparticles sensitize solid tumors to cold plasma by blocking the PI3K/AKT-driven signaling axis to suppress cellular transformation by inhibiting growth and EMT. Biomaterials, 2016. 87: p. 118-130.
275.    Zhao, N., et al., An ultra pH‐sensitive and aptamer‐equipped nanoscale drug‐delivery system for selective killing of tumor cells. Small, 2013. 9(20): p. 3477-3484.
276.    Li, Y.-F., H.-T. Zhang, and L. Xin, Hyaluronic acid-modified polyamidoamine dendrimer G5-entrapped gold nanoparticles delivering METase gene inhibits gastric tumor growth via targeting CD44+ gastric cancer cells. Journal of cancer research and clinical oncology, 2018. 144(8): p. 1463-1473.
277.    Cho, J.-H., et al., Development of a novel imaging agent using peptide-coated gold nanoparticles toward brain glioma stem cell marker CD133. Acta biomaterialia, 2017. 47: p. 182-192.
278.    Shamsian, A., et al., Targeting Tumorigenicity of Breast Cancer Stem Cells Using SAHA/Wnt-b Catenin Antagonist Loaded Onto Protein Corona of Gold Nanoparticles. International Journal of Nanomedicine, 2020. 15: p. 4063.
279.    Han, J.W., et al., Dual functions of silver nanoparticles in F9 teratocarcinoma stem cells, a suitable model for evaluating cytotoxicity-and differentiation-mediated cancer therapy. International journal of nanomedicine, 2017. 12: p. 7529.
280.    Choi, Y.-J., et al., Differential cytotoxic potential of silver nanoparticles in human ovarian cancer cells and ovarian cancer stem cells. International journal of molecular sciences, 2016. 17(12): p. 2077.
281.    Bu, L.L., et al., Cancer stem cell‐platelet hybrid membrane‐coated magnetic nanoparticles for enhanced photothermal therapy of head and neck squamous cell carcinoma. Advanced Functional Materials, 2019. 29(10): p. 1807733.
282.    Liu, D., et al., Targeted destruction of cancer stem cells using multifunctional magnetic nanoparticles that enable combined hyperthermia and chemotherapy. Theranostics, 2020. 10(3): p. 1181.
283.    Su, Z., et al., CD44-targeted magnetic nanoparticles kill head and neck squamous cell carcinoma stem cells in an alternating magnetic field. International journal of nanomedicine, 2019. 14: p. 7549.
284.    Chen, L., et al., Neuropilin-1 (NRP-1) and magnetic nanoparticles, a potential combination for diagnosis and therapy of gliomas. Current pharmaceutical design, 2015. 21(37): p. 5434-5449.
285.    Aires, A., et al., Multifunctionalized iron oxide nanoparticles for selective drug delivery to CD44-positive cancer cells. Nanotechnology, 2016. 27(6): p. 065103.
286.    Dalzon, B., et al., Utility of macrophages in an antitumor strategy based on the vectorization of iron oxide nanoparticles. Nanoscale, 2019. 11(19): p. 9341-9352.
287.    Wang, J., et al., Aptamer‐conjugated nanorods for targeted photothermal therapy of prostate cancer stem cells. Chemistry–An Asian Journal, 2013. 8(10): p. 2417-2422.
288.    Sun, T.-M., et al., Cancer stem cell therapy using doxorubicin conjugated to gold nanoparticles via hydrazone bonds. Biomaterials, 2014. 35(2): p. 836-845.
289.    Yang, C., et al., Target therapy of multiple myeloma by PTX-NPs and ABCG2 antibody in a mouse xenograft model. Oncotarget, 2015. 6(29): p. 27714.
290.    Kaluzova, M., et al., Targeted therapy of glioblastoma stem-like cells and tumor non-stem cells using cetuximab-conjugated iron-oxide nanoparticles. Oncotarget, 2015. 6(11): p. 8788.
291.    Gao, X., et al., Prostate stem cell antigen-targeted nanoparticles with dual functional properties: in vivo imaging and cancer chemotherapy. International journal of nanomedicine, 2012. 7: p. 4037.
292.    Sadhukha, T., et al., Effective elimination of cancer stem cells by magnetic hyperthermia. Molecular pharmaceutics, 2013. 10(4): p. 1432-1441.
293.    Patra, C.R., et al., Targeted delivery of gemcitabine to pancreatic adenocarcinoma using cetuximab as a targeting agent. Cancer research, 2008. 68(6): p. 1970-1978.
294.    Bonvalot, S., et al., NBTXR3, a first-in-class radioenhancer hafnium oxide nanoparticle, plus radiotherapy versus radiotherapy alone in patients with locally advanced soft-tissue sarcoma (Act. In. Sarc): a multicentre, phase 2–3, randomised, controlled trial. The Lancet Oncology, 2019. 20(8): p. 1148-1159.
295.    El Hout, M., et al., Crosstalk between autophagy and metabolic regulation of cancer stem cells. Molecular cancer, 2020. 19(1): p. 27.
296.    Han, J., et al., Cancer stem cell-targeted bio-imaging and chemotherapeutic perspective. Chemical Society Reviews, 2020. 49(22): p. 7856-7878.
297.    Northcote-Smith, J., et al., Breast Cancer Stem Cell Active Copper (II) Complexes with Naphthol Schiff Base and Polypyridyl Ligands. Inorganics, 2021. 9(1): p. 5.
298.    Beg, S., et al. Nanomedicinal strategies as efficient therapeutic interventions for delivery of cancer vaccines. in Seminars in cancer biology. 2021. Elsevier.
299.    Zhang, M., et al., Charge-reversal nanocarriers: An emerging paradigm for smart cancer nanomedicine. Journal of Controlled Release, 2020. 319: p. 46-62.
300.    Bort, G., et al., EPR-mediated tumor targeting using ultrasmall-hybrid nanoparticles: From animal to human with theranostic AGuIX nanoparticles. Theranostics, 2020. 10(3): p. 1319.
301.    Howard, G.P., et al., Critical size limit of biodegradable nanoparticles for enhanced lymph node trafficking and paracortex penetration. Nano research, 2019. 12(4): p. 837-844.
302.    Kirkby, C.A., et al., Nutrient availability limits carbon sequestration in arable soils. Soil Biology and Biochemistry, 2014. 68: p. 402-409.
303.    Nguyen, V., et al., Femtosecond laser-assisted synthesis of highly photoluminescent carbon nanodots for Fe 3+ detection with high sensitivity and selectivity. Optical Materials Express, 2016. 6(2): p. 312-320.
304.    Ni, D., et al., Molybdenum-based nanoclusters act as antioxidants and ameliorate acute kidney injury in mice. Nature communications, 2018. 9(1): p. 1-11.
305.    Guo, J., et al., The potential for clinical translation of antibody-targeted nanoparticles in the treatment of acute myeloid leukaemia. Journal of controlled release, 2018. 286: p. 154-166.
306.    Hubert, C.G., et al., A three-dimensional organoid culture system derived from human glioblastomas recapitulates the hypoxic gradients and cancer stem cell heterogeneity of tumors found in vivo. Cancer research, 2016. 76(8): p. 2465-2477.
307.    Karsten, U. and S. Goletz, What makes cancer stem cell markers different? Springerplus, 2013. 2(1): p. 1-8.
308.    Lee, H., et al., The effects of particle size and molecular targeting on the intratumoral and subcellular distribution of polymeric nanoparticles. Molecular pharmaceutics, 2010. 7(4): p. 1195-1208.
309.    Olivares-Urbano, M.A., et al., CSC radioresistance: a therapeutic challenge to improve radiotherapy effectiveness in cancer. Cells, 2020. 9(7): p. 1651.
310.    Rattan, R., et al., Nanoparticle-macrophage interactions: A balance between clearance and cell-specific targeting. Bioorganic & medicinal chemistry, 2017. 25(16): p. 4487-4496.
311.    Xia, P., Surface markers of cancer stem cells in solid tumors. Current stem cell research & therapy, 2014. 9(2): p. 102-111.
312.    Zhou, G., et al., Aptamer-based therapeutic approaches to target cancer stem cells. Theranostics, 2017. 7(16): p. 3948.
313.    Aghaalikhani, N., et al., Cancer stem cells as a therapeutic target in bladder cancer. Journal of cellular physiology, 2019. 234(4): p. 3197-3206.
314.    Tang, X., et al., The use of nanoparticulates to treat breast cancer. Nanomedicine, 2017. 12(19): p. 2367-2388.
315.    Alba‑Molina, D., J.J. Giner‑Casares, and M. Cano, Bioconjugated plasmonic nanoparticles for enhanced skin penetration. Surface-modified Nanobiomaterials for Electrochemical and Biomedicine Applications, 2020: p. 219-235.
316.    Azeez, F., et al., The effect of surface charge on photocatalytic degradation of methylene blue dye using chargeable titania nanoparticles. Scientific reports, 2018. 8(1): p. 1-9.
317.    Fukuoka, Y., et al., Combination strategy with complexation hydrogels and cell-penetrating peptides for oral delivery of insulin. Biological and Pharmaceutical Bulletin, 2018. 41(5): p. 811-814.
318.    Lee, W., et al., High colloidal stability ZnO nanoparticles independent on solvent polarity and their application in polymer solar cells. Scientific reports, 2020. 10(1): p. 1-10.
319.    Mekseriwattana, W., et al., The impact of serum proteins and surface chemistry on magnetic nanoparticle colloidal stability and cellular uptake in breast cancer cells. AAPS PharmSciTech, 2019. 20(2): p. 1-9.
320.    Nielsen, H.M. and L. Jorgensen, Challenges in delivery of biopharmaceuticals; the need for advanced delivery systems. Delivery Technologies for Biopharmaceuticals: John Wiley & Sons, Ltd, 2009: p. 1-8.
321.    Hassanen, E.I., et al., Pomegranate juice diminishes the mitochondria-dependent cell death and NF-kB signaling pathway induced by copper oxide nanoparticles on liver and kidneys of rats. International journal of nanomedicine, 2019. 14: p. 8905.
322.    Hong, F., et al., Nanosized titanium dioxide resulted in the activation of TGF‐β/S mads/p38 MAPK pathway in renal inflammation and fibration of mice. Journal of Biomedical Materials Research Part A, 2016. 104(6): p. 1452-1461.
323.    Kanwal, Z., et al., A comparative assessment of nanotoxicity induced by metal (silver, nickel) and metal oxide (cobalt, chromium) nanoparticles in Labeo rohita. Nanomaterials, 2019. 9(2): p. 309.
324.    Lacave, J.M., et al., Waterborne exposure of adult zebrafish to silver nanoparticles and to ionic silver results in differential silver accumulation and effects at cellular and molecular levels. Science of the total environment, 2018. 642: p. 1209-1220.
325.    Patil, N.A., W. Gade, and D.D. Deobagkar, Epigenetic modulation upon exposure of lung fibroblasts to TiO2 and ZnO nanoparticles: alterations in DNA methylation. International journal of nanomedicine, 2016. 11: p. 4509.
326.    Rajaee Behbahani, S., et al., Red elemental selenium nanoparticles mediated substantial variations in growth, tissue differentiation, metabolism, gene transcription, epigenetic cytosine DNA methylation, and callogenesis in bittermelon (Momordica charantia); an in vitro experiment. PloS one, 2020. 15(7): p. e0235556.