Next Article in Journal
Combinatory Effect of Pequi Oil (Caryocar brasiliense)-Based Nanoemulsions Associated to Docetaxel and Anacardic Acid (Anacardium occidentale) in Triple-Negative Breast Cancer Cells In Vitro
Previous Article in Journal
RETRACTED: Alhakamy et al. Ceftriaxone and Melittin Synergistically Promote Wound Healing in Diabetic Rats. Pharmaceutics 2021, 13, 1622
Previous Article in Special Issue
Synergetic Effect of β-Cyclodextrin and Its Simple Carbohydrate Substituents on Complexation of Folic Acid and Its Structural Analog Methotrexate
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Application of Nanotechnology and Phytochemicals in Anticancer Therapy

by
Jin Hee Kim
1,*,
Boluwatife Olamide Dareowolabi
1,
Rekha Thiruvengadam
2 and
Eun-Yi Moon
1
1
Department of Integrative Bioscience & Biotechnology, Sejong University, Seoul 05006, Republic of Korea
2
Center for Global Health Research, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha Medical College, Saveetha University, Chennai 600077, India
*
Author to whom correspondence should be addressed.
Pharmaceutics 2024, 16(9), 1169; https://doi.org/10.3390/pharmaceutics16091169
Submission received: 30 July 2024 / Revised: 22 August 2024 / Accepted: 31 August 2024 / Published: 5 September 2024
(This article belongs to the Special Issue Anti-Cancer Drug Delivery Systems)

Abstract

:
Cancer is well recognized as a leading cause of mortality. Although surgery tends to be the primary treatment option for many solid cancers, cancer surgery is still a risk factor for metastatic diseases and recurrence. For this reason, a variety of medications has been adopted for the postsurgical care of patients with cancer. However, conventional medicines have shown major challenges such as drug resistance, a high level of drug toxicity, and different drug responses, due to tumor heterogeneity. Nanotechnology-based therapeutic formulations could effectively overcome the challenges faced by conventional treatment methods. In particular, the combined use of nanomedicine with natural phytochemicals can enhance tumor targeting and increase the efficacy of anticancer agents with better solubility and bioavailability and reduced side effects. However, there is limited evidence in relation to the application of phytochemicals in cancer treatment, particularly focusing on nanotechnology. Therefore, in this review, first, we introduce the drug carriers used in advanced nanotechnology and their strengths and limitations. Second, we provide an update on well-studied nanotechnology-based anticancer therapies related to the carcinogenesis process, including signaling pathways related to transforming growth factor-β (TGF-β), mitogen-activated protein kinase (MAPK), phosphatidylinositol 3 kinase (PI3K), Wnt, poly(ADP-ribose) polymerase (PARP), Notch, and Hedgehog (HH). Third, we introduce approved nanomedicines currently available for anticancer therapy. Fourth, we discuss the potential roles of natural phytochemicals as anticancer drugs. Fifth, we also discuss the synergistic effect of nanocarriers and phytochemicals in anticancer therapy.

1. Introduction

Cancer is well recognized as a leading cause of mortality. Approximately 19.3 million new cancer cases and 10 million cancer-caused deaths in 2020 worldwide were reported [1,2]. Among the new cases, breast cancer was the most frequent, followed by lung, colon, prostate, skin, and stomach cancers [2]. Although surgery tends to be the primary treatment option for many solid cancers, cancer surgery has been well documented to be a risk factor for metastatic diseases and recurrence in many clinical and experimental studies [3]. The perioperative phase of cancer surgery offers a treatment window against lingering malignant illness and is critical for assessing the risk of postoperative metastatic diseases [3]. For this reason, a variety of medications has been adopted for the postsurgical care of patients with cancer [4]. However, conventional medicines have shown major challenges such as drug resistance, a high level of drug toxicity, and different drug responses due to tumor heterogeneity [5,6]. Recently, a nanomedicine-based therapeutic strategy was suggested to be a promising alternative to improve the efficiency and selectivity of anticancer drugs in anticancer therapy [7]. Because nanomedicine-based therapeutic drugs help target tumor sites, these drugs can control the local and systemic releases of medicines, resulting in enhanced therapy efficacy, reduced toxicity, and improved patient outcomes [5,7,8]. In particular, tumor-targeted nanoparticle (NP)-based anticancer therapy is considered an extensive and favorable era in cancer biology [9].
Many proteins are involved in the carcinogenesis process, including signaling pathways related to transforming growth factor-β (TGF-β), mitogen-activated protein kinase (MAPK), phosphatidylinositol 3 kinase (PI3K), Wnt, poly(ADP-ribose) polymerase (PARP), Notch, and Hedgehog (HH) [10]. Because inhibitors against the proteins involved in carcinogenesis can specifically target molecular mechanisms related to the promotion of cancer growth, metastasis, and carcinogenesis-related inflammatory processes, conjugation of the inhibitors in chemotherapy can be effective in destroying cancer cells and preventing metastasis [9]. Especially, NPs based on the inhibitors of carcinogenesis-related proteins provide better anticancer drug efficacy, overcoming major constraints in conventional chemotherapy such as low bioavailability, many side effects, and poor solubility [5,9].
Phytochemicals are considered anticancer agents because of their inhibitory roles against inflammation and postsurgical recurrence of cancer and metastasis [11,12,13]. Natural phytochemicals have few side effects as well as anticancer effects [14,15]. However, there is limited evidence in relation to the application of phytochemicals in cancer treatment, particularly focusing on nanotechnology. Therefore, in this review, we discussed carcinogenesis inhibitor-based drug delivery strategies using nanotechnology in anticancer therapy and the potential role of natural phytochemicals as anticancer agents.

2. Drug Delivery Strategies Using Nanotechnology

A variety of anticancer drugs can be used in anticancer therapy. Notably, drug carriers play a pivotal role in anticancer therapy by improving the delivery and efficacy of therapeutic agents while minimizing side effects [5,9]. Many types of drug carriers, including NP, nanocapsules, nanoemulsions, and hydrogels, can be used in cancer therapy [16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47]. Moreover, these nanotechnology-based anticancer drugs can be used to prevent carcinogenesis through several signaling pathways. Therefore, in this Section, we introduce the drug carriers used in advanced nanotechnology and summarize anticancer strategies focusing on nanotechnology-based anticancer therapy by carcinogenesis signaling pathways related to TGF-β, MAPK/PI3K/Wnt, PARP, Notch/HH, and others.

2.1. Drug Carriers Used in Advanced Nanotechnology

Figure 1 shows drug carriers used in advanced nanotechnology and their strengths and limitations.
The lipid polymer hybrid NP is a new form of hybrid NP, created for the targeted transportation of chemotherapeutic medicines to tumor cells [16]. It is made up of three layers, a polymer core where the drugs are contained, a lipid monolayer that surrounds the polymeric core, and a lipid polyethylene glycol (PEG) layer on which special targeting moieties can be attached [16]. This NP has increased stability and biocompatibility, increased drug half-lives, and increased rate-limiting controlled release [17].
Layer-by-layer liposomal NPs are formulated to combine the advantages of designing multilayer structures with nanometer precision with the advantage of liposomes [18]. Because of the multilayer, the elimination time of drugs from the systemic circulation is reduced, thus promoting effective drug delivery [18]. However, this multilayer technique is time-consuming, making production on a large-scale level difficult [19].
A lipid nanocapsule is a carrier system made up of an oily hydrophobic core surrounded by a combination of PEGylated surfactants and phospholipids [20]. Lipid nanocapsules are more efficient in encapsulating drugs for delivery when compared to other conventional NPs [21]. They can also encapsulate multiple drugs at once and enhance the bioavailability of encapsulated drugs [21]. Lipid nanocapsules, however, require a high-level dose for their function [22]. Conjugation of ligands with lipid nanocapsules has also proven to be challenging [22].
Lipid-based ECO NP is a multifunctional drug carrier that is effective at mediating gene silencing [23]. The ECO structure promotes the stability of the nanocarrier [23] and is quite efficient in the delivery of genetic materials [24]. Cationic lipids, however, have several adverse effects including disturbance of nuclear and cellular membranes and releasing degrading enzymes from lysosomes [25].
A nanoemulsion is a colloidal system of 10 to 1000 nm in size [26]. It is made up of solid spheres with lipophilic and amorphous surfaces [26]. A nanoemulsion has several strengths as a carrier for drugs [26]. It is non-toxic and not energy-intensive, improves the bioavailability and solubility of the drug, and provides greater surface area for improved absorption of drugs [26]. However, it is susceptible to degradation [27].
Lipidoids are synthetic cationic lipids that have secondary and tertiary amine functions and efficient interactions with anionic siRNA molecules [28]. Lipidoid NPs are capable of delivering siRNA [29]. Ionizable cationic lipidoids allow improved encapsulation of siRNA and its intracellular release [29].
Polymeric NPs with a size ranging from 1 to 1000 nm can contain drugs within or on their surface [30]. They have several advantages that prove their potential to effectively deliver drugs. They protect the drugs they carry from biological activity in the environment, thus improving their bioavailability and drug safety [30]. They also have the ability to release drugs at a controlled rate [30]. Despite all these advantages, polymeric NPs are still limited in large-scale production due to particle aggregation, premature release of drugs, and microbial proliferation in liquid dosage forms [30].
A gold NP (gNP) can be developed with a synthetic method that involves treating hydrogen tetrachloroaurate and citric acid [31]. A gNP has a low toxicity, high biocompatibility, and large surface-to-volume ratio [32]. However, the size and surface charge of gNPs affect their biodistribution leading to aggregation in a few organs, which may promote toxicity [32].
A silver NP (sNP) can be synthesized via several methodologies including biological and chemical methods [33]. sNPs have the advantage of being non-toxic, faster to synthesize, and environmentally friendly, especially when synthesized via the biological method [33]. It is expensive and hazardous to synthesize via the chemical method [34].
Zinc oxide NPs are one of the most popular metal NPs used in anticancer medication [35]. Zinc oxide NPs can be synthesized through chemical precipitation using a highly purified zinc forerunner and a precipitator [35]. These NPs are relatively inexpensive, non-toxic, and easily absorbed by the body [35]. However, their major limitation is that they can easily build up in the body, resulting in organ toxicity [36].
Iron oxide NPs are made from iron oxide via physical, chemical, or biological techniques [37]. Iron oxide NPs are one of the most preferred NPs for drug delivery because they possess a number of advantages. They cause minimal toxicity, are stable in aqueous solutions, possess superparamagnetism, and are biocompatible [38]. However, they are hard to produce; in particular, the physical technique requires expensive and complex machines [37]. This makes it difficult to control the size of the NPs [37]. Moreover, the chemical technique leads to it being easily contaminated by external materials as well as requiring very high temperatures and complex conditions for its production [37].
Mesoporous silica NPs are inorganic NPs with a size that ranges from 30 to 300 nm [39]. They possess several advantages including a large surface area and large pore volume, and they are quite stable and biocompatible [40]. They are quite difficult to produce in an industrial setting due to their high cost [40].
Planetary ball milling used for the synthesis of NPs involves rotating a vial in a planet-like motion to reduce the size of large crystals [41]. This technique mixes drug powder with a dispersion medium and a stabilizer, which helps to prevent drug aggregation [42]. Planetary ball-milled NPs (PBM-NPs) are sustainable and environmentally friendly as planetary ball milling adopts sustainable materials as precursors for NPs [41]. The planetary ball-milling process, however, is both time and energy-consuming [42].
Exosomes are natural vesicular structures released from cells and have sizes ranging from 30 to 150 nm [43]. Exosome-based NPs may have extensive biodistribution and reduced accumulation in organs, resulting in reduced toxicity [43]. However, there are some difficulties in differentiating exosomes based on biochemical and biophysical characteristics [43]. Large-scale production of exosome-based NPs is expensive as it requires a large number of manufacturing devices [43].
A bioresponsive gel is a class of highly hydrated biomaterials [44]. It provides an environment that is semi-wet and suitable for biological interactions on a molecular level [44]. It also provides an inert surface that prevents the adsorption of non-specific proteins [44]. It can be designed to change properties in response to external materials [44]. Its major limitation is that it needs extensive testing to explore the effects of its component parts in the body [44,45]. This could be time and money-intensive [45].
An amino-functionalized polystyrene NP is a class of NPs based on a polymer backbone with amino moieties [46]. Polystyrene NPs are relatively thermally stable; however, they have high toxicity potentials, and are known to pollute and cause harm to aquatic animals [47].

2.2. TGF-β Signaling-Based Nanotherapies

The TGF-β superfamily consists of ligand proteins, such as bone morphogenetic proteins, activins, and proteins related to their associated receptors, which allow the translocation of Smad proteins into the nucleus for transcription of tumor progression and metastasis-related genes [48]. The TGF-β signaling pathway plays a pivotal part in various processes, including the proliferation and migration of cells [49]. The anticancer effects of TGF-β signaling-based nanotherapies are listed in Table 1.
Small interfering RNA (siRNA) to integrin β3 via lipid ECO-based NPs (ECO/siβ3) can effectively silence integrin β3 expression, restore TGF-β-mediated cytostasis, decrease TGF-β-mediated epithelial–mesenchymal transition (EMT) and invasion, and suppress three-dimensional organoid growth in triple-negative breast cancer (TNBC) [50]. Thus, it acts as a promising therapeutic regimen to combat TNBC [50]. Poly-N-(2-hydroxypropyl) methacrylamide (pHPMA)-coated hybrid NPs with modified lipid polymer co-loaded with cryptotanshinone (S/C-pW-LPNs) and silibinin have been evaluated to identify their anti-metastasis efficacy in a mouse model with breast cancer [51]. These NPs effectively decreased microenvironment biomarkers such as TGF-β1, matrix metalloprotease 9 (MMP-9), and platelet and endothelial cell adhesion molecule 1 (PECAM1, also called CD31) related to metastasis, indicating that the NPs are effective nanocarriers of an oral drug to inhibit metastasis of breast cancer to the lung [51]. Zinc oxide NPs can effectively decrease cell proliferation and migration, enhance apoptotic bodies, alter cell cycle distribution, and decrease the synthesis of MMP-9 and TGF-β in murine photoreceptor-derived cells [52]. The codelivery of TGF-β1 with silver NPs can effectively modulate the immune response and significantly reduce the severity of inflammatory diseases such as autoimmune encephalomyelitis and multiple sclerosis by programming antigen-presenting cells to induce a more efficient tolerance [53]. In TGF-β-stimulated fibroblasts, the NPs releasing siRNAs targeting heat shock protein 47 (HSP47) effectively reduced profibrotic markers such as NADPH oxidase 4, collagen type I, and alpha-smooth muscle actin in a fibrosis model [54]. This evidence supports that TGF-β signaling-based nanotherapies can improve the anticancer effect.

2.3. MAPK/PI3K/Wnt Signaling-Based Nanotherapies

The MAPK pathway is composed of several important signaling cascades, including rat sarcoma (RAS), rapidly accelerated fibrosarcoma (RAF), mitogen-activated protein kinase (MEK), and extracellular signal-regulated kinase (ERK called MAPK) [55]. Extracellular signals, such as growth factors and cytokines, activate tyrosine kinase receptors, inducing MAPK signaling [55]. The ERK is stimulated by various inflammatory mediators, including cytokines, chemokines, and lipopolysaccharides [56] and activated ERK stimulates proinflammatory cytokines, indicators of carcinogenesis [57]. The PI3K pathway is also dysregulated in approximately 30% of cancers [58]. PI3K, a heterodimer, consists of catalytic and regulatory subunits [58]. Activated PI3K phosphorylates phosphatidylinositol 4,5-bisphosphate (PIP2) to make phosphatidylinositol 3,4,5-trisphosphate (PIP3), which can activate pyruvate dehydrogenase kinase 1 and sequentially phosphorylate AKT serine/threonine kinase 1 (AKT), which inhibit the transcription of tumor suppressor genes [58]. Dysregulation of the canonical Wnt pathway signaled by Wnt and β-catenin is also crucial for cancer progression [59]. The anticancer effects of MAPK/PI3K/Wnt signaling-based nanotherapies are listed in Table 2.
The siRNA to RAF or AKT could be loaded in cationic nanoliposomes, and the siRNA-loaded nanoliposomes selectively target melanoma tumor cells as well as early melanocytic lesions, resulting in the prevention of melanoma metastasis [60]. Sorafenib is an inhibitor of the receptor tyrosine kinase that plays a pivotal role in the MAPK/PI3K signaling cascade for tumor development and metastasis in various human cancers, including metastatic liver, gastrointestinal stromal, hypernephroma, and colorectal cancers [61]. Sorafenib-loaded lipid-based nanosuspensions have greater aqueous solubility, higher encapsulation efficiency, and improved bioavailability compared with free Sorafenib, which results in better treatment efficacy by reducing proliferation of tumor cells and enhancing reorganization of the MAPK cascade in glioblastoma therapy [61].
The anticarcinogenic effect of amino-functionalized polystyrene (NH2-PS) NPs was compared with that of amino-functionalized silica (NH2-Si) or hydroxyl-functionalized silica (OH-Si) NPs in hepatocellular carcinoma (HCC) Huh7 and HepG2 cell lines [62]. At the molecular level, NH2-PS NPs obstructed mammalian target of rapamycin (mTOR) signaling, damaged the mitochondrial membrane, and enhanced lysosomes that precede cell death [62]. Generally, the NH2-PS NPs were more effective than the NH2-Si NPs [62]. Iron oxide can stimulate lysosome dysfunction and change the subcellular localization of p53 and mTOR, which can affect the autophagic flux [63]. The treatment of the chemotherapeutic drug cisplatin inhibiting DNA replication with anti-human epidermal growth factor receptor 2 (HER2) antibody-conjugated and autophagy inhibitory microRNA (MIR376B)-loaded superparamagnetic iron oxide NPs enhanced anticancer treatment efficiency in both xenograft nude mice with breast cancer and HER2-positive breast cancer cells [64]. Everolimus, a mTOR inhibitor, is used as an immune suppressor [65]. When p53-encoding synthetic mRNA was delivered using NP technology, it effectively restored tumor suppressor p53 in tumor sites and resulted in tumor cells sensitive to everolimus [66]. Thus, co-targeting p53 and the mTOR signaling pathway can effectively exhibit an antitumor effect in HCC and non-small cell lung cancer (NSCLC) [66]. Bioreducible polymer was used to encapsulate siRNA inhibiting mTOR and it exhibited strong potential to deliver siRNA to lung cancer cells [7]. PI3K inhibitors entrapped in supramolecular nanoassemblies with 1,2-distearoyl-sn-glycero-3-phosphoethanolamine (DSPE)-PEG and L-α-phosphatidylcholine could induce an anticancer effect by increasing the phosphorylation of mTOR and AKT, thus resulting in increased antitumor efficacy and longevity [67]. The overactivation of PI3K/mTOR signaling has been observed in non-Hodgkin’s lymphoma [68]. BEZ235, a dual PI3K and mTOR inhibitor, has been found to be an effective suppressor of lung cancer [69]. When dibenzocyclooctyne-functionalized anti-Lym1 and anti-CD20 antibodies were used in an NP-based drug delivery system for delivering BEZ235 to lymphoma cells, this system improved antitumor activity of BEZ235 both in vivo and in vitro via inhibiting the PI3K/mTOR signaling pathway [68]. When AZD6244 (selumetinib, an allosteric inhibitor of MEK1/2) and PX-866 (a PI3K inhibitor) were layer-by-layer co-encapsulated in a cancer-targeting nanoscale therapeutic formulation, they effectively blocked lobular carcinoma in xenograft-bearing NCR nude mice [70]. Chrysophanol gNPs have been evaluated against human LNCaP prostate cancer cells [71]. Chrysophanol gNPs were found to reduce histone deacetylase activities and halt the cell cycle in the sub-G phase via the inactivation of AKT and upregulation of AMP-activated protein kinase (AMPK) and sequentially controlling the activity of mTOR, and finally inhibit prostate cancer cell growth [71,72]. PH-427, an AKT/pyruvate dehydrogenase kinase 1 (PDK1) inhibitor, is encapsulated into poly(lactic-co-glycolic acid) (PLGA) NPs (PH-427-PLGA-NPs), and treatment with PH-427-PLGA-NPs reduced the tumor size in a MiaPaCa-2 pancreatic cancer model, indicating that NPs can be efficient drug carriers targeting pancreatic cancer that harbors RAS mutations [73].
Sorafenib-loaded PEG-PLGA NPs modified with the antibody hGC33 to glypican-3 (GPC3) can effectively target GPC3-positive HCC cells by inhibiting the Wnt pathway, downregulating cyclin D1 expression, inhibiting EMT, and inactivating the RAS and RAF on MAPK signaling pathway [74]. All the above-mentioned evidence supports that MAPK/PI3K/Wnt signaling-based nanotherapies can improve the anticancer effect.
Table 2. MAPK/PI3K/Wnt signaling-based nanotherapies.
Table 2. MAPK/PI3K/Wnt signaling-based nanotherapies.
Nanomedicine NameDrug in NanomedicineDelivery SystemTarget CancerExperimental ModelEffect of Nanomedicine on CancerRef.
Nanoliposomal siRNASmall interfering RNA targeting B-Raf with V600E and AKT3Cationic nanoliposomesMelanomaHuman melanoma cell lines; human fibroblastsDecreased expression of B-Raf with V600E and AKT; decreased melanoma by 65%[60]
SFN-LNCSorafenibLipid nanocapsulesGlioblastomaHuman U87MG glioblastoma cell lines; mice with orthotopic U87MG human glioblastoma xenograftsInhibited in vitro angiogenesis; decreased glioblastoma cell viability; decreased proliferating cells in tumor[61]
NH2-PS and NH2-Si NPAmino-functionalized polystyrene and biodegradable silicaAmino-functionalized polystyrene NPs and amino-functionalized silica NPs HCCHCC cell linesNH2-PS NPs trigger death of Huh7 and HepG2 cells by obstructing mTOR signaling and inducing lysosomal destabilization; NH2-Si enhances cell proliferation by activating mTOR signaling[62]
Iron oxide-based NPsMagnetite core coated with carboxymethyldextran shellGreen fluorescent labeled iron oxide NPs (nano-screenMAG-CMX) and non-fluorescent magnetic particles (fluidMAG-MX)HepatoblastomaHepatic cell line (HepG2)Induced lysosomal dysfunction; altered subcellular localizations of pmTOR and p53 proteins[63]
SPION NPsMicroRNA (MIR376B)AGO2 conjugated and anti-HER2 labeled SPIONs (SP-AH)Breast cancerHER2-positive breast cancer cell lines; xenograft nude mice model of breast cancerBlocked autophagy; increased the efficacy of anticancer treatment[64]
Supramolecular NPsPI103 and PI828Supramolecular nano-assembly using L-α-phosphatidylcholine, and DSPE-PEG [1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(polyethylene glycol)]Breast and ovarian cancers4T1 breast cancer and K-Ras (LSL/+)/PTEN (fl/fl) ovarian cancer modelsTemporally sustained inhibition of phosphorylation of AKT, mTOR, S6K, and 4EBP in vivo; increased antitumor efficacy; abrogated insulin resistance[67]
NP-based pre-targeted system for the therapeutic delivery of BEZ235BEZ235Azide-functionalized BEZ235-encapsulated NPsNon-Hodgkin’s lymphomaLymphoma cell linesImproved in vivo and in vitro antitumor activity of BEZ235 by inhibiting the PI3K/mTOR pathway[68]
Lbl NPAZD6244; PX-866Tumor targeting nanoscale drug formulation (layer-by-layer NPs)TNBC; RAS-mutant lung tumorCancer cell lines (MDA-MB-231, Hep G2, KP7B, and OVCAR-3 cells)Caused cytotoxicity in both the TNBC cell line and RAS-mutant lung tumor cell line; blocked tumor-specific phosphorylation of ERK and AKT[70]
Gold-chrysophanol NPsChrysophanolPLGA NPsProstate cancerLNCap prostate cancer cellsInduced apoptosis; increased ROS production; caused DNA damage; expressed differentially pro- and anti-apoptotic proteins; reduced tumor volume and weight[71]
PLGA NPsPH-427PLGA NPsPancreatic cancerMiaPaCa-2 pancreatic cancer model with mutant K-rasImproved drug delivery and therapeutic efficacy against pancreatic cancer with mutant K-ras[73]
hGC33-modified NPs (hGC33-SFB-NP)SorafenibPolyethylene glycol-b-PLGA polymer NPsHCCIn vivo model of liver cancerInhibited growth and progression of liver cancer by targeting GPC3+ HCC cells; attenuated HCC cell migration; inhibited epithelial–mesenchymal transition[74]
AKT, AKT serine/threonine kinase 1; GPC3, glypican-3; HCC, hepatocellular carcinoma; HER2, human epidermal growth factor receptor 2; HSP47, heat shock protein 47; NH2-PS, amino-functionalized polystyrene; NH2-Si, amino-functionalized silica; NP, nanoparticle; PLGA, poly(lactic-co-glycolic acid); TGF-β, transforming growth factor-beta; TNBC, triple-negative breast cancer.

2.4. PARP Signaling-Based Nanotherapies

PARP is a family of proteins crucial for DNA repair and apoptosis [75]. Upon sensing damage, PARP is activated via its phosphorylation by AKT to recruit the machinery required for DNA repair [76,77]. Because activated PARP reduces the death of cancer cells in cancer therapy, the inhibition of PARP activation can be very important in cancer therapy. The anticancer effects of PARP signaling-based nanotherapies are listed in Table 3. A PARP inhibitor (PARPi), Talazoparib, has been encapsulated in the bilayer of a nanoliposome to develop nanoTalazoparib [78]. In BRCA-deficient breast cancer mice, nanoTalazoparib enhanced the survival of mice, induced DNA damage, led to cell cycle arrest, and inhibited cell proliferation by modulating the immune cells [78]. When radiation-resistant cells and tumors derived from a p53/phosphatase and tensin homolog (PTEN)-deficient mouse model of advanced prostate cancer were treated with a lipid-based nanoformulation of Olaparib (nanoOlaparib), it made radiation-resistant tumors without BRCA mutations radiosensitive [79]. The newly developed fluorescence-labeled PARPi-encapsulated nanoemulsion (PARPi-FL) had a prolonged circulation time [80]. The longer half-life indicates the pharmacokinetic benefits of nanoemulsions as nanocarriers, thereby confirming the importance of PARPi-FL as an imaging agent targeting PARP in small cell lung cancer [80]. The overexpression of the Rad6 protein in breast cancer during the aggressive stage contributes to DNA damage tolerance [81]. The inhibitor for Rad6 (SMI#9)-conjugated gNP (SMI#9-gNP) can be endocytosed by mesenchymal TNBC cells, resulting in cytotoxicity [82]. In addition, the co-administration of SMI#9-gNP with cisplatin demonstrated a synergistic effect by enhancing cisplatin sensitivity without causing damage to normal breast cells [82]. SMI#9 released from gNPs causes cell death by inducing mitochondrial dysfunction and PARP1 hyperactivation, finally acting as an effective formulation that specifically targets chemo-resistant TNBC cells [82]. Liposomal NPs co-loaded with PARPi and cisplatin have been developed layer-by-layer using electrostatics with a hyaluronic acid layer at the terminal, which facilitates targeting the CD44 receptor, and thus could selectively target ovarian cancer [83]. The liposomal NPs showed increased blood circulation time with significantly lower systemic toxicity, minimized tumor metastasis, and increased the survival of CD44-expressing female nude mice, thus improving their therapeutic efficacy against high-grade serous ovarian cancer [83]. NP-mediated delivery of siRNA targeting PARP1 in mouse ovarian cancer models significantly inhibited cell proliferation, induced apoptosis, and prolonged the survival of mice with tumors [84]. In one study, Veliparib (a PARPi) and methylene blue (a photosensitizer) co-encapsulated in PLGA NPs presented reduced cytotoxicity of normal cells in the dark with reduced viability of cancer cells [85]. This result indicates that the co-encapsulation of Veliparib and methylene blue could be an important strategy to improve photodynamic therapy [85]. Linalool is a monoterpene compound, and when gNPs conjugated with linalool and the CALNN peptide were treated to ovarian SKOV-3 cancer cells, apoptosis was induced by activating p53 and caspase-8 [86]. This indicates that ovarian cancer could be suppressed by NPs by inducing apoptosis via extrinsic and intrinsic pathways [86]. This evidence supports that PARP signaling-based nanotherapies can improve the anticancer effect.

2.5. Notch/HH Signaling-Based Nanotherapies

Notch signaling is also an oncogenic signaling pathway crucial for cancer invasiveness and progression [87]. It is also involved in EMT stimulation [88]. The HH signaling is also known to be pivotal for cancer stem cell maintenance, chemoresistance, and radioresistance via inducing transcription of oncogenes [89]. Anticancer effects of Notch/HH signaling-based nanotherapies are listed in Table 4.
Mesoporous silica NPs with glucose moieties and γ-secretase inhibitors, strong interceptors to Notch signaling, have been tested against breast cancer cells [90]. These NPs were effectively internalized and decreased the population of malignant stem cells [90]. α-Mangostin-encapsulated PLGA NPs (Mang-PLGA-NPs) have been developed and assessed in colorectal cancer cells [91]. Mang-PLGA-NPs inhibited colorectal cancer cell viability, colony formation, and EMT, enhanced programmed cell death, and inhibited the cancer stem-like cell population by suppressing Notch signaling components (such as Notch-1, Notch-2, DLL4, and Jagged 1), Hes-1, and γ-secretase complex protein, indicating that Mang-PLGA-NPs could be used to treat and prevent colorectal cancer [91].
A PBM-NP has been developed using thymoquinone (TQ), a natural polysaccharide, and A10, an RNA aptamer that is bound to prostate-specific membrane antigen [92]. When the A10-coated PBM-NP with TQ (A10-TQ-PBM-NP) was used to treat two prostate cancer cell lines LNCaP-R and C4-2B-R, which are resistant to docetaxel with high HH expression, the A10-TQ-PBM-NP was highly effective in inhibiting the HH signaling pathway and sequentially suppressing prostate cancer progression [92]. Another study engineered a nano-size molecule HH signaling inhibitor (nanoHHi)-containing polymeric NPs using PLGA conjugated with PEG [93]. Encapsulated nanoHHi effectively decreased pancreatic cancer cell proliferation, and the use of nanoHHi together with gemcitabine impeded the growth of orthotopic xenografted Pa03C pancreatic cancer better than use of gemcitabine alone [93]. In addition, engineered dual-targeting biomimetic NPs containing LDE225 (an inhibitor to Sonic hedgehog (Shh)) and apolipoprotein A1 (an anti-CD15) functioned as an effective and stable drug carrier [94]. They were able to cross the blood–brain barrier (BBB) and deliver drugs to the cancer stem-like cells of medulloblastoma with a high Shh level, indicating that the NP could be used as a potent and effective nanomedicine to treat medulloblastoma with a high Shh level [94]. The medulloblastoma with a high Shh level can be also targeted by biomimetic high-density lipoprotein (HDL) NPs that bind to the HDL receptor (scavenger receptor type B-1, SCARB1), resulting in the depletion of cholesterol levels in cancer cells and thus effectively blocking the proliferation of medulloblastoma cells and colony formation [95]. Spherical nucleic acid NPs wrapped by a polyethylenimine shell target the transcription factor Gli1, which plays a role in the HH signaling pathway required for glioma stem cell maintenance [96]. These Gli1-targeted NPs bind to scavenger receptors on glioblastoma cells and induce dynamin-dependent and caveolae-mediated endocytosis [96]. The Gli1-targeted NPs can inhibit the tumor-promoting HH pathway and its downstream target genes, thereby alleviating drug resistance and glioblastoma recurrence [96]. The effect of nanoHHi alone or in combination with Sorafenib has been tested in HCC cell lines, and it significantly inhibited the proliferation, invasion, systemic metastasis, as well as tumor growth, of HCC and reduced the population of CD133-expressing HCC cells compared with Sorafenib treatment alone, thereby providing a new treatment regime for patients with HCC [97]. All the above-mentioned evidence supports that Notch/HH signaling-based nanotherapies can improve the anticancer effect.
Table 4. Anticancer effect of Notch and HH signaling-based nanotherapies.
Table 4. Anticancer effect of Notch and HH signaling-based nanotherapies.
Nanomedicine NameDrug in NanomedicineDelivery SystemTarget CancerExperimental ModelEffect of Nanomedicine on CancerRef.
Silica NPsγ-Secretase inhibitorMesoporous silica NPs functionalized with glucose moietiesBreast cancerHuman MCF7 and MDA-MB-231 breast cancer cell linesReduced cancer stem cell population[90]
PLGA NPsα-MangostinPLGA NPsColorectal cancerHuman colorectal cancer (HCT116 and HT29) cell linesInhibited EMT, colony formation, cell viability, and induced apoptosis; suppressed Notch signaling pathway leading to inhibition of cancer stem-like cell population and self-renewal capacity[91]
Planetary ball-milled NPsThymoquinePlanetary ball-milled NPs coated with an RNA aptamer, A10Prostate cancerDocetaxel-resistant C4-2B-R and LNCaP-R cells with high expression of HH signaling moleculesInhibited HH signaling pathway, thereby suppressing prostate cancer progression[92]
NanoHHiHPI-1Polymeric NP (PLGA-PEG) encapsulating HPI-1MedulloblastomaAllografts derived from Ptch (−/+); p53 (−/−) mouse medulloblastomas; orthotopic Pa03C pancreatic cancer xenograftsInhibited tumor growth; downregulated mGli1 and HH target genes[93]
High-density lipoprotein-mimetic NPs (eHNPs)LDE225Apolipoprotein A1 and anti-CD15 incorporated eHNPsShh subtype of medulloblastomaDAOY human medulloblastoma cells and PZp53 cellsReduced cholesterol in Shh MB cells[94]
Biomimetic high-density lipoprotein NPsSynthetic HDL NPsHigh-density lipoprotein NPsMedulloblastomaIn vitro studies using medulloblastoma cell linesDepleted cholesterol in cancer cells; inhibited proliferation and colony formation; depleted cancer stem cell population[95]
PEI-SNAssiRNA targeting Gli1Polyethylenimine-wrapped spherical nucleic acid NPsGlioblastomaGlioblastoma U87-MG cell linesSilenced tumor-promoting HH pathway genes; decreased glioblastoma cell proliferation; promoted glioblastoma cell senescence; decreased metabolic activity and self-renewal ability of glioblastoma cells; promoted apoptosis[96]
NanoHHiGli1Polymeric NP-encapsulated delivery systemHCCIn vitro HCC cell lines; in vivo subcutaneous and orthotopic HCC xenografts nude miceInhibited invasion and proliferation of HCC cells; suppressed in vivo tumor growth; reduced systemic metastases[97]
EMT, epithelial–mesenchymal transition; HCC, hepatocellular carcinoma; HH, Hedgehog; NP, nanoparticle; PLGA, poly(lactic-co-glycolic acid); Shh, Sonic hedgehog.

2.6. Other Signaling-Based Nanotherapies

Other signaling-based nanotherapies could affect anticancer effects as well. Anticancer effects of other signaling-based nanotherapies are listed in Table 5. Novel elongated-type peanut-shaped gNPs have been tested to evaluate their cytotoxic potential against ovarian SKOV-3 cancer cells [98]. The results revealed that cell viability and the proliferation capability of ovarian cancer cells were decreased because of increased cell apoptosis and autophagy as well as increased reactive oxygen species (ROS) production [98]. RNA NPs containing RNA aptamers binding to the CD133 receptor and inhibiting microRNA-21 have been developed and delivered to breast cancer stem cells [99]. RNA NPs effectively inhibited cancer cell movement and microRNA-21 expression, enhancing the expression of tumor suppressors PTEN and PDCD4 with greater specificity and efficacy [99]. Erlotinib (an inhibitor to epidermal growth factor receptor, EGFR) has been delivered using phospholipase A (PLA)-based NPs [100]. To prevent against the interplay of EGFR with Notch signaling for carcinogenesis, an γ-secretase inhibitor was also enclosed in the core of the NPs with Erlotinib, resulting in effective inhibition of Notch signaling [100]. Because tumors have an acidic microenvironment, NPs can easily target cancer cells by controlling the interleukins (ILs) related to cancer cell resistance at an acidic pH [101]. Because the overexpression of EGFR is noted in 50% of patients with lung cancer and the inhibition of the mitotic regulator polo-like kinase 1 (PLK1) can enhance radiation sensitivity, EGFR-positive NSCLC cells were targeted by the siPLK1-NP [102]. This resulted in the reduced expression of PLK1, which led to cell death, tumor growth reduction, G2/M cell cycle arrest, and extended survival [102]. This indicated that siPLK1-NPs could be an effective targeted therapy that can function as a radiation sensitizer in NSCLC [102].
Previous studies have demonstrated that cytosolic PLA2 is an effective therapeutic molecular target in several human metastatic cancers, including leukemia and breast, prostate, and ovarian cancers [103]. Cytosolic PLA2 has diverse functions, including the biosynthesis of eicosanoids such as prostaglandins and leukotrienes, which are mainly involved in the cytochrome c oxidase (COX) and lipoxygenase pathways [102,104]. Gowda et al. [105] developed a novel PEGylated nanoliposomal delivery system that targeted the cytosolic PLA2 inhibitor arachidonyl trifluoromethyl ketone (ATK). Therefore, this nanoliposomal ATK delivery system can increase circulation time, enhance drug stability, and avoid clearance by the reticuloendothelial system [105]. Because it is less toxic to normal cells than to melanoma cancer cells, nanoliposomal ATK delivery has proven to be remarkable in treating melanoma in preclinical trials [105].
An exosome-based nanoformulation loaded with aspirin can be used as an effective anticancer therapy against breast and colorectal cancer cells [106]. This novel nanoexosome-based drug delivery system improved tumor cell cytotoxicity [106]. It exhibits a higher encapsulation capacity and a better dissolution rate than water-soluble drugs [106]. Nanotherapy with immune checkpoint inhibitors could be a good solution for NSCLC treatment because of its enhanced survival rate, reduced side effects, and stimulation of immune responses against malignancies in patients with NSCLC [107]. Immune checkpoint inhibitors are small molecules that disturb the immune checkpoint signaling pathways, thereby impeding the tumor suppression of immune cells [108]. Zhao et al. [109] showed that Cu-doped gold nanoclusters (CuAuNCs) could be useful for C-X-C motif chemokine receptor 4 (CXCR4)-targeting positron emission tomography imaging as an alternative diagnostic method in cancer biology. Nanomaterials and nanoclusters loaded with chemotherapeutic drugs can provide a new avenue in cancer biology for theragnostic applications because of their advantages, such as accurate and early detection of cancer cells and targeted specificity toward tumor cells [110].
Anti-programmed death ligand 1 (anti-PDL1) therapy reduces locally recurrent and distant cancers [111]. Based on the natural targeting potential of platelet to circulating tumor cells, researchers developed an in situ sprayable chemo-immunotherapy gel that acts as a drug reservoir and releases both anti-PDL1 monoclonal antibody and platelet-derived tiny extracellular vesicles combined with doxorubicin (PexD), an anticancer drug that prevents post-surgery tumor recurrence and spread [112]. Anti-PDL1 antibody and PexD co-encapsulated in a fibrin gel can be sprayed using a dual-cartridge sprayer [113]. Because the released anti-PDL1 antibody effectively blocks the PD1/PDL1 pathway while PexD efficiently stimulates the antitumor immune response by inducing tumor immunogenic cell death, entering the systemic circulation through damaged blood vessels, and attaching to circulating tumor cells, the combined use of anti-PDL1 antibodies and PexD triggers strong T cell immunogenic responses, which ultimately initiate the host’s immunogenic response by inhibiting both metastatic potential postsurgery as well as local tumor recurrence [113]. A radioimmunostimulant and PI3Kγ inhibitor, IPI549, can specifically target myeloid cells and act as a catalase to convert endogenous hydrogen peroxide into oxygen to achieve hypoxia-relieved postoperative radiotherapy [114]. Combined use of IPI549 with anti-PDL1 antibodies increased susceptibility to anti-PDL1 therapy and enhanced radiotherapy-mediated immunogenic cell death by reprogramming the tumor microenvironment into an immunogenic phenotype [114]. Ultimately, this acts as a simple and effective therapeutic strategy to inhibit postsurgical cancer recurrence and metastasis.
Table 5. Anticancer effect of other signaling-based nanotherapies.
Table 5. Anticancer effect of other signaling-based nanotherapies.
Nanomedicine NameDrug in NanomedicineDelivery SystemTarget CancerExperimental ModelEffect of Nanomedicine on CancerRef.
AuP NPsNanogoldPeanut-shaped gNPsOvarian cancerIn vitro study using SKOV-3 cellsDecreased proliferation and viability of ovarian cancer cells; induced autophagy and apoptosis; increased oxidative stress of cancer cells[98]
RNA NPsAnti-miR21Chemically and thermodynamically stable RNA NPsTNBCIn vivo and in vitro studies using TNBC and breast cancer stem-like cellsReduced migration of cancer cells; inhibited miR21 expression; upregulated expression of tumor suppressors; efficiently inhibited tumor growth[99]
CF-EB/DART-dual-loaded NPsErlotinib (EB) and gamma-secretase inhibitor (GSI)-DAPTPLA-based nano-platformTNBCIn vitro studies using MDA-MB-231 cell line Enhanced tumor penetration ability of drug; reduced side effects of drugs[100]
Nanographene sheets and SPION@silica nanospheresSPIONNanographene sheets and SPION@silica nanospheresBreast cancerIn vitro study using MDA-MB 231 cancer cellsEnhanced apoptosis, necrosis, and oxidative stress induction in cancer cells; disrupted cell cycle phases; increased the levels of anticarcinogenic interleukins[101]
C-siPLK1-NPSmall interfering RNA (siRNA) against PLK1Cetuximab-conjugated NPNSCLCIn vitro and in vivo studies using EGFR and NSCLC cells, A549 flank tumors, and an orthotopic lung tumor modelReduced PLK1 expression; caused cell cycle arrest; induced reduction in tumor growth and cell death[102]
NanoATKArachidonyl trifluoromethyl ketone (ATK)Nanoliposomal delivery systemMelanomaXenograft tumor modelDecreased cellular proliferation, triggered apoptosis, and inhibited melanoma xenograft tumor growth without animal weight loss; inhibited the STAT3, AKT, and cPLA2 pathways[105]
Nano-amorphous aspirin-loaded exosomesAspirinExosomesBreast and colorectal cancersHuman colorectal adenocarcinoma HT29 cell line and human metastatic breast cancer MDA-MB-231 cell lineEnhanced cellular uptake, improved cytotoxicity of aspirin, increased apoptosis and autophagy, eradication of cancer stem cells, efficient delivery to in vivo tumors[106]
CuAuNCsAMD3100 (also known as Plerixafor)Gold nanoclustersBreast cancer and lung metastasisMouse 4T1 orthotopic breast cancer modelSensitive and accurate detection of CXCR4 in early-stage cancers; accurate imaging for early detection of breast cancer[109]
PexDDoxorubicin and Adp-L1Sprayable bioresponsive gel MelanomaB16-F10 tumor-bearing miceInhibited local tumor recurrence and metastasis, induced tumor immunogenic cell death, promoted antitumor immune response, tracked and eliminated circulating tumor cells, impaired PD-1/PD-L1 pathway, restored the tumor-killing effect of cytotoxic T cells, improved tumor immune microenvironment[113]
IPI549@HMPIPI549 (PI3Kγ inhibitor)PEGylated HMnO2 (HMP)-bridged radioimmunotherapy nanoplatformCancer recurrence after surgeryExperimental model demonstrating the genomic landscape shaped by surgical resection and the effects on the tumor microenvironmentSuppressed/eradicated local residual and distant tumors and elicited strong immune memory effects to resist tumor rechallenge[114]
AKT, AKT serine/threonine kinase 1; gNP, gold NP; NP, nanoparticle; NSCLC, non-small cell lung cancer; PexD, platelet-derived tiny extracellular vesicles combined with doxorubicin; PLA, phospholipase A; PLK1, polo-like kinase 1; TNBC, triple-negative breast cancer.

3. Approved Nanomedicines Currently Available for Anticancer Therapy

Approved nanomedicines currently available for anticancer therapy are listed in Table 6. Several types of nanomedicines have been approved by official regulatory institutions including the Food and Drug Administration (FDA) for cancer treatment, leveraging the unique properties of NPs to enhance drug efficacy and delivery [115,116,117]. Doxil® (Doxorubicin Liposome) is a liposomal formulation of doxorubicin, used in treating various cancers including ovarian cancer and multiple myeloma. The liposome helps to reduce the cardiotoxicity associated with doxorubicin. Abraxane® (Paclitaxel Albumin-bound) uses albumin NPs in delivering paclitaxel, a chemotherapy drug. It is used to treat breast cancer, NSCLC, and pancreatic cancer. Irinotecan Liposome Onivyde® is used to treat metastatic pancreatic cancer. The liposome helps to improve the drug’s pharmacokinetics and reduce side effects. Vyxeos® (Daunorubicin and Cytarabine Liposome) contains daunorubicin and cytarabine in a liposomal formulation and it is used to treat acute myeloid leukemia. The liposome allows for a more controlled release of the drugs. These nanomedicines represent significant advancements in cancer therapy, offering improved targeting and reduced toxicity compared to traditional formulations.

4. Phytochemicals Used in Anticancer Therapy

Patients with cancer who have undergone chemotherapy had higher levels of inflammation compared with their healthy counterparts [118]. For this reason, the development of anticancer drugs with anti-inflammatory properties and low toxicity could be a potential therapeutic strategy in treating patients with cancer [14]. Because phytochemicals could be used for anti-inflammatory purposes to aid anticancer application [11,12,13], natural phytochemicals that could be used as anticancer drugs are summarized in this Section. Natural phytochemicals used in anticancer therapy are listed in Table 7.

4.1. Phytochemicals against Inflammatory Microenvironment in Cancer

Scutellarin, a flavone glucuronide, has been extracted from Erigeron breviscapus, a traditional Chinese medicine plant [119]. It can inhibit the production of proinflammatory mediators by inhibiting the MAPK and I-kappaB kinase (IKK)-dependent nuclear factor-kappa B (NFκB) signaling pathway [112]. TQ is an important constituent of black cumin seed oil from Nigella sativa [120]. TQ can inhibit NFκB-dependent neuroinflammation in BV2 microglia via activating the antioxidant response element (ARE)/nuclear erythroid 2 related factor 2 (Nrf2) antioxidant pathway [120]. Oxyresveratrol is a polyphenolic molecule present in various plants, including Artocarpus lakoocha [121]. It exerts anti-inflammatory effects in IL-1β-induced human microglial clone 3 cells by inhibiting ERKs on MAPK signaling cascades and the AKT on PI3K signaling cascades, indicating that oxyresveratrol could be an effective pharmacologic agent to treat neuroinflammation in microglia [121]. Terpenoids extracted from Abies holophylla exert neuroprotective and anti-inflammatory effects via increasing nerve growth factor production and decreasing nitrite production through the inhibition of JNK phosphorylation, thereby inhibiting the secretion of proinflammatory cytokines such as IL-1β, IL-6, tumor necrosis factor (TNF), and prostaglandin E2, and effectively decreasing neuroinflammation in microglial cells [122]. Curcumin, a phytochemical extracted from Curcuma longa, possesses antioxidant, anticancer, and anti-inflammatory effects [11]. Curcumin can decrease neuroinflammation post-subarachnoid hemorrhage by inhibiting the toll-like receptor/NFκB signaling pathway and sequentially a shift of microglia M1 phenotype to M2, which promotes tumor survival [123,124]. Moringin, isolated from Moringa oleifera seeds, effectively normalized Wnt/β-catenin signaling in mice with autoimmune encephalomyelitis [125]. Moringin can upregulate β-catenin and inhibit glycogen synthase kinase-3, which regulates FoxP3 and CD4 expression in T cell activation, inhibiting COX-2, IL-6, and IL-1β, decreasing apoptosis, and increasing expression of antioxidant Nrf2 in mice with autoimmune encephalomyelitis [125]. Hesperetin, a phytochemical, can effectively inhibit nitric oxide, decrease expression of IL-1β, IL-6, and MAPK, downregulate ERK1/2 phosphorylation, suppress astrocyte and microglial cell activation, and ultimately decrease neuroinflammation in BV-2 microglial cells [126].

4.2. Phytochemicals against Postsurgical Recurrence of Cancer and Metastasis

Rottlerin is a natural polyphenol compound [127]. It can inhibit metastasis-related MMPs by inhibiting protein kinase C (PKC)-mediated ROS, inactivating ERK1/2, and suppressing the AP-1/c-Fos signaling pathway, which suppresses astrocyte migration in phorbol-12-myristate-13-acetate-induced rats [127]. Genistein (a soy-derived isoflavone) could be used to boost the inhibitory role of cisplatin widely used to treat HCC to protect against tumor recurrence and metastasis following curative hepatectomy [12,13]. Some experimental evidence has shown that the combined use of genistein with cisplatin might lower the dose requirement of cisplatin as well as improve anticancer activity in various malignancies, including lung, prostate, pancreatic, and breast cancers [128]. Furthermore, various combinations of drugs showed greater inhibitory effects against cancers than the use of individual drugs alone [13].

5. Application of Nanotechnology and Phytochemicals in Clinical Trials for Anticancer Therapy

Because of the previously mentioned strengths of nanotechnology and phytochemicals, the combined use of nanotechnology with phytochemicals has been applied to clinical trials for anticancer therapy, as shown in Table 8. Patients treated with the Nano Swarna Bhasma (NSB) drug showed 100% clinical benefit compared to patients treated without NSB, indicating the clinical role of NSB [129]. CRLX101, a cyclodextrin-containing polymer NP loaded with camptothecin, was prescribed to patients with esophageal or gastrointestinal cancers [15,130]. In a phase II clinical trial of CRLX101, it downregulated tumor biomarkers in gastric, gastroesophageal, and esophageal cancers [130]. The safety of camptothecin was checked in patients with advanced rectal carcinoma [15]. This phytochemical not only resulted in the downstaging of rectal cancer but did not induce any severe side effects among the patients treated [15]. A cyclodextrin-containing polymer loaded with docetaxel led to the stable condition of patients with prostate or breast adenocarcinoma with a 19.4% clinical benefit rate [131]. Moreover, this nanoformulated drug exhibited some pharmacokinetic advantages over docetaxel, including longer retention of drug in plasma, slower clearance, and controlled release rate of docetaxel from the carriers [131]. When another drug Pm-Pac, polymeric micellar NPs conjugated with phytochemical paclitaxel, was used in treating patients with advanced NSCLC, it significantly increased the overall and progression-free survivals of NSCLC patients without pleural metastasis [132]. When another phytochemical, ursolic acid, was loaded into nanoliposomes (UANL), it did not accumulate in the body and showed no adverse effects when it was treated at 37 mg/m2 of UANL [133]. Overall, the combined application of nanotechnology and phytochemicals can give many benefits in relation to higher treatment efficacy, lower side effects, a more stable condition, and prolonged overall survival after treatment of patients with cancer.

6. Current Challenges and Opportunities for Future Nanotherapeutic Strategies

Generally, cancer shows uncontrolled cell development because of the deactivation of tumor suppressors or activation of oncogenes, dysregulated cell cycling, and metastatic properties, and it is a main cause of mortality worldwide [134]. Although surgery tends to be the primary treatment option for many solid cancers, cancer surgery is still a risk factor for metastatic diseases and recurrence [3]. Although a variety of medications has been adopted for the postsurgical care of patients with cancer [4], conventional medicines have shown major challenges such as drug resistance, a high level of drug toxicity, and different drug responses due to tumor heterogeneity [5,6]. Nanocarriers in nanomedicine could be modulated and thus nanotechnology-based therapeutic formulations could effectively overcome the challenges faced by conventional treatment methods [50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114]. In relation to anticancer drugs in nanomedicine, the proteins against several carcinogenesis signaling mechanisms, including the TGF-β, MAPK, PI3K, Wnt, PARP, Notch, and HH signaling pathways, should be considered in anticancer therapy [10]. Because natural phytochemicals can support reducing carcinogenesis-related inflammation, they should also be considered in the development of anticancer drugs [11,12,13]. As proven in anticancer nanodrugs approved by official regulatory institutions such as the FDA, nanomedicine can provide better anticancer drug efficacy, overcoming major constraints in conventional chemotherapy such as poor solubility, many side effects, and low bioavailability [5,9,115,116,117]. In addition, during anticancer therapy, nanomedicine has a better treatment potential because it can deliver chemotherapeutic agents to specific tumor sites better [6]. Moreover, nanomedicine helps conventional medicines overcome their major challenges such as drug resistance, a high level of drug toxicity, and different drug responses [6]. In particular, the combined use of nanotechnology with natural phytochemicals can enhance tumor targeting and increase the efficacy of anticancer agents with better solubility and bioavailability and reduced side effects [135,136]. Furthermore, nanomedicine can transfer multiple materials, including DNA, RNA, fluorescence agents, and so on, as well as drugs, to tumor sites specifically in a controlled manner [137]. The control of the continuous secretion of anticancer drugs using NPs by regulated light intensity and the prevention against phagocytic clearance of NPs by their surface modifications could give better benefits in the treatment of patients with cancer [138,139]. In addition, NPs allow imaging for the detection, diagnosis, and monitoring of treatment outcomes, as well as delivery of therapy [140]. Therefore, in the future, the anticancer effect of various NPs should be evaluated in clinical trials to consider their safety.

Author Contributions

Conceptualization, J.H.K. and R.T.; writing—original draft preparation, J.H.K., B.O.D. and R.T.; writing—review and editing, J.H.K. and E.-Y.M.; visualization, B.O.D.; supervision, J.H.K.; project administration, J.H.K.; funding acquisition, J.H.K. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the Ministry of Education, Republic of Korea (grant number 2019R1I1A2A01050001).

Acknowledgments

We created all figures with the help of BioRender.com.

Conflicts of Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

References

  1. Ferlay, J.; Colombet, M.; Soerjomataram, I.; Parkin, D.M.; Piñeros, M.; Znaor, A.; Bray, F. Cancer statistics for the year 2020: An overview. Int. J. Cancer 2021, 149, 778–789. [Google Scholar] [CrossRef] [PubMed]
  2. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  3. Tohme, S.; Simmons, R.L.; Tsung, A. Surgery for cancer: A trigger for metastases. Cancer Res. 2017, 77, 1548–1552. [Google Scholar] [CrossRef]
  4. Bu, L.L.; Yan, J.; Wang, Z.; Ruan, H.; Chen, Q.; Gunadhi, V.; Bell, R.B.; Zhen, G. Advances in drug delivery for post-surgical cancer treatment. Biomaterials 2019, 219, 119182. [Google Scholar] [CrossRef]
  5. Gavas, S.; Quazi, S.; Karpinski, T.M. Nanoparticles for cancer therapy: Current progress and challenges. Nanoscale Res. Lett. 2021, 16, 173. [Google Scholar] [CrossRef]
  6. Yao, Y.; Zhou, Y.; Liu, L.; Xu, Y.; Chen, Q.; Wang, Y.; Wu, S.; Deng, Y.; Zhang, J.; Shao, A. Nanoparticle-based drug delivery in cancer therapy and its role in overcoming drug resistance. Front. Mol. Biosci. 2020, 7, 193. [Google Scholar] [CrossRef]
  7. Gandhi, N.S.; Godeshala, S.; Koomoa-Lange, D.T.; Miryala, B.; Rege, K.; Chougule, M.B. Bioreducible poly(amino ethers) based mTOR siRNA delivery for lung cancer. Pharm. Res. 2018, 35, 188. [Google Scholar] [CrossRef]
  8. Riley, R.S.; June, C.H.; Langer, R.; Mitchell, M.J. Delivery technologies for cancer immunotherapy. Nat. Rev. Drug Discov. 2019, 18, 175–176. [Google Scholar] [CrossRef]
  9. Anand, U.; Dey, A.; Chandel, A.K.S.; Sanyal, R.; Mishra, A.; Pandey, D.K.; de Falco, V.; Upadhyay, A.; Kandimalla, R.; Chaudhary, A.; et al. Cancer chemotherapy and beyond: Current status, drug candidates, associated risks and progress in targeted therapeutics. Genes Dis. 2022, 10, 1367–1401. [Google Scholar] [CrossRef]
  10. Prossomariti, A.; Piazzi, G.; Alquatti, C.; Ricciardiello, L. Are Wnt/β-Catenin and PI3K/AKT/mTORC1 distinct pathways in colorectal cancer? Cell. Mol. Gastroenterol. Hepatol. 2020, 10, 491–506. [Google Scholar] [CrossRef]
  11. Wilken, R.; Veena, M.S.; Wang, M.B.; Srivatsan, E.S. Curcumin: A review of anti-cancer properties and therapeutic activity in head and neck squamous cell carcinoma. Mol. Cancer 2011, 10, 12. [Google Scholar]
  12. Hamaya, S.; Oura, K.; Morishita, A.; Masaki, T. Cisplatin in liver cancer therapy. Int. J. Mol. Sci. 2023, 24, 10858. [Google Scholar] [CrossRef] [PubMed]
  13. Chen, P.; Hu, M.D.; Deng, X.F.; Li, B. Genistein reinforces the inhibitory effect of Cisplatin on liver cancer recurrence and metastasis after curative hepatectomy. Asian Pac. J. Cancer Prev. 2013, 14, 759–764. [Google Scholar] [CrossRef]
  14. Sun, Y.; Zhou, Y.Q.; Liu, Y.K.; Zhang, H.Q.; Hou, G.G.; Meng, Q.G.; Hou, Y. Potential anti-neuroinflammatory NF-кB inhibitors based on 3,4-dihydronaphthalen-1(2H)-one derivatives. J. Enzym. Inhib. Med. Chem. 2020, 35, 1631–1640. [Google Scholar] [CrossRef] [PubMed]
  15. Sanoff, H.K.; Moon, D.H.; Moore, D.T.; Boles, J.; Bui, C.; Blackstock, W.; O’Neil, B.H.; Subramaniam, S.; McRee, A.J.; Carlson, C.; et al. Phase I/II trial of nano-camptothecin CRLX101 with capecitabine and radiotherapy as neoadjuvant treatment for locally advanced rectal cancer. Nanomedicine 2019, 18, 189–195. [Google Scholar] [CrossRef]
  16. Gajbhiye, K.R.; Salve, R.; Narwade, M.; Sheikh, A.; Kersharani, P.; Gajbhiye, V. Lipid polymer hybrid nanoparticles: A custom tailored next-generation approach for cancer therapeutics. Mol. Cancer 2023, 22, 160. [Google Scholar] [CrossRef]
  17. Parveen, S.; Gupta, P.; Kumar, S.; Banerjee, M. Lipid polymer hybrid nanoparticles as potent vehicles for drug delivery in nanoparticles. Med. Drug Discov. 2023, 20, 100165. [Google Scholar] [CrossRef]
  18. Ramasamy, T.; Haidar, Z.S.; Tran, T.H.; Choi, J.Y.; Jeong, J.H.; Shin, B.S.; Choi, H.G.; Yong, C.S.; Kim, J.O. Layer-by-layer assembly of liposomal nanoparticles with PEGylated polyelectrolytes enhances systemic delivery of multiple anticancer drugs. Acta Biomater. 2014, 10, 5116–5127. [Google Scholar] [CrossRef] [PubMed]
  19. Marin, E.; Tapeinos, C.; Sarasua, J.R.; Larranaga, A. Exploiting the layer-by-layer nanoarchitectonics for the fabrication of polymer capsules: A toolbox to provide multifunctional properties to target complex pathologies. Adv. Colloid Interface Sci. 2022, 304, 102680. [Google Scholar] [CrossRef]
  20. Urimi, D.; Hellsing, M.; Mahmoudi, N.; Söderberg, C.; Widenbring, R.; Gedda, L.; Edwards, K.; Loftsson, T.; Schipper, N. Structural characterization study of a lipid nanocapsule formulation intended for drug delivery applications using small-angle scattering techniques. Mol. Pharm. 2022, 19, 1068–1077. [Google Scholar] [CrossRef]
  21. Dabholkar, N.; Waghule, T.; Rapalli, V.K.; Gorantla, S.; Alexander, A.; Saha, R.N.; Singhvi, G. Lipid shell nanocapsules as smart generation lipid nanocarriers. J. Mol. Liq. 2021, 339, 117145. [Google Scholar] [CrossRef]
  22. Moura, R.P.; Pacheco, C.; Pêgo, A.P.; des Rieux, A.; Sarmento, B. Lipid nanocapsules to enhance drug bioavailability to the central nervous system. J. Control. Release 2020, 322, 390–400. [Google Scholar] [CrossRef] [PubMed]
  23. Gujrati, M.; Malamas, A.; Shin, T.; Jin, E.; Sun, Y.; Lu, Z.R. Multifunctional cationic lipid-based nanoparticles facilitate endosomal escape and reduction-triggered cytosolic siRNA release. Mol. Pharm. 2014, 11, 2734–2744. [Google Scholar] [CrossRef]
  24. Sun, D.; Sahu, B.; Gao, S.; Schur, R.M.; Vaidya, A.M.; Maeda, A.; Palczewski, K.; Lu, Z.R. Targeted multifunctional lipid eco plasmid DNA nanoparticles as efficient non-viral gene therapy for Leber’s congenital amaurosis. Mol. Ther. Nucleic Acids 2017, 7, 42–52. [Google Scholar] [CrossRef]
  25. Jorgensen, A.M.; Wibel, R.; Bernkop-Schnurch, A. Biodegradable cationic and ionizable cationic lipids: A roadmap for safer pharmaceutical excipients. Small 2023, 19, 2206968. [Google Scholar] [CrossRef]
  26. Jasiwal, M.; Dudhe, R.; Sharma, P.K. Nanoemulsion: An advanced mode of drug delivery system. 3 Biotech 2015, 5, 123–127. [Google Scholar] [CrossRef]
  27. Mushtaq, A.; Wani, S.M.; Malik, A.R.; Gull, A.; Ramniwas, S.; Nayik, G.A.; Ercisli, S.; Marc, R.A.; Ullah, R.; Bari, A. Recent insights into nanoemulsions: Their preparation, properties and applications. Food Chem. X 2023, 18, 100684. [Google Scholar] [CrossRef]
  28. de Groot, A.M.; Thanki, K.; Gangloff, M.; Falkenberg, E.; Zeng, X.; van Bijnen, D.C.J.; van Eden, W.; Franzyk, H.; Nielsen, H.M.; Broere, F.; et al. Immunogenicity testing of lipidoids in vitro and in silico: Modulating lipidoid-mediated TLR4 activation by nanoparticle design. Mol. Ther. Nucleic Acids 2018, 11, 159–169. [Google Scholar] [CrossRef]
  29. Khare, P.; Dave, K.M.; Kamte, Y.S.; Manoharan, M.A.; O’Donnell, L.A.; Manickam, D.S. Development of lipidoid nanoparticles for siRNA delivery to neural cells. AAPS J. 2021, 24, 8. [Google Scholar] [CrossRef] [PubMed]
  30. Zielińska, A.; Carreiró, F.; Oliveira, A.M.; Neves, A.; Pires, B.; Venkatesh, D.N.; Durazzo, A.; Lucarini, M.; Eder, P.; Silva, A.M.; et al. Polymeric nanoparticles: Production, characterization, toxicology and ecotoxicology. Molecules 2020, 25, 3731. [Google Scholar] [CrossRef]
  31. Yeh, Y.C.; Creran, B.; Rotello, V.M. Gold nanoparticles: Preparation, properties, and applications in bionanotechnology. Nanoscale 2012, 4, 1871–1880. [Google Scholar] [CrossRef]
  32. Arvizo, R.; Bhattacharya, R.; Mukherjee, P. Gold nanoparticles: Opportunities and challenges in nanomedicine. Expert Opin. Drug Deliv. 2010, 6, 753–763. [Google Scholar] [CrossRef]
  33. Tripathi, R.M.; Chung, S.J. Biogenic nanomaterials: Synthesis, characterization, growth mechanism, and biomedical applications. J. Microbiol. Methods 2019, 157, 65–80. [Google Scholar] [CrossRef]
  34. Ferdous, Z.; Nemmar, A. Health impact of silver nanoparticles: A review of the biodistribution and toxicity following various routes of exposure. Int. J. Mol. Sci. 2020, 7, 2375. [Google Scholar] [CrossRef] [PubMed]
  35. Jiang, J.; Pi, J.; Cai, J. The advancing of zinc oxide nanoparticles for biomedical applications. Bioinorg. Chem. Appl. 2018, 5, 1062562. [Google Scholar] [CrossRef] [PubMed]
  36. Rasmussen, J.W.; Martinez, E.; Louka, P.; Wingett, D.G. Zinc oxide nanoparticles for selective destruction of tumor cells and potential for drug delivery applications. Expert Opin. Drug Deliv. 2010, 7, 1063–1077. [Google Scholar] [CrossRef] [PubMed]
  37. Ali, A.; Zafar, H.; Zia, M.; Haq, I.U.; Phull, A.R.; Ali, J.S.; Hussain, A. Synthesis, characterization, applications, and challenges of iron oxide nanoparticles. Nanotechnol. Sci. Appl. 2016, 9, 49–67. [Google Scholar] [CrossRef]
  38. Malhotra, N.; Lee, J.S.; Liman, R.A.D.; Ruallo, J.M.S.; Villaflores, O.B.; Ger, T.R.; Hsiao, C.D. Potential toxicity of iron oxide magnetic nanoparticles: A review. Molecules 2020, 25, 3159. [Google Scholar] [CrossRef] [PubMed]
  39. Dourado, D.; Miranda, J.A.; de Oliveira, M.C.; Freire, D.T.; Xavier-Júnior, F.H.; Paredes-Gamero, E.J.; Alencar, É.D.N. Recent trends in curcumin-containing inorganic-based nanoparticles intended for in vivo cancer therapy. Pharmaceutics 2024, 16, 177. [Google Scholar] [CrossRef] [PubMed]
  40. Zhang, C.; Zhao, J.; Wang, W.; Geng, H.; Wang, Y.; Gao, B. Current advances in the application of nanomedicine in bladder cancer. Biomed. Pharmcother. 2023, 157, 14062. [Google Scholar] [CrossRef]
  41. Poshna, C.; Mailapalli, D.M. Modeling the particle size of nanomaterials synthesized in a planetary ball mill. OpenNano 2023, 14, 100191. [Google Scholar] [CrossRef]
  42. Dvornik, M.; Mikhailenko, E. The influence of the rotation frequency of a planetary ball mill on the limiting value of the specific surface area of the WC and Co nanopowders. Adv. Powder Technol. 2020, 31, 3937–3946. [Google Scholar] [CrossRef]
  43. Sen, S.; Xavier, J.; Kumar, N.; Ahmad, M.Z.; Ranjan, O.P. Exosomes as natural nanocarrier-based drug delivery system: Recent insights and future perspectives. 3 Biotech 2023, 3, 101. [Google Scholar] [CrossRef] [PubMed]
  44. Ulijn, R.V.; Bibi, N.; Jayawarna, V.; Thornton, P.D.; Todd, S.J.; Mart, R.J.; Smith, A.M.; Gough, J.E. Bioresponsive hydrogels. Mater. Today 2007, 10, 40–48. [Google Scholar] [CrossRef]
  45. Wilson, A.N.; Guiseppi-Elie, A. Bioresponsive hydrogels. Adv. Health Mater. 2013, 4, 520–532. [Google Scholar] [CrossRef] [PubMed]
  46. Jiang, J.; Thayumanavan, S. Synthesis and characterization of amine-functionalized polystyrene nanoparticles. Macromolecules 2005, 38, 5886–5891. [Google Scholar] [CrossRef]
  47. Machado, A.J.T.; Mataribu, B.; Serrão, C.; da Silva Silvestre, L.; Farias, D.F.; Bergami, E.; Corsi, I.; Marques-Santos, L.F. Single and combined toxicity of amino-functionalized polystyrene nanoparticles with potassium dichromate and copper sulfate on brine shrimp Artemia franciscana larvae. Environ. Sci. Pollut. Res. Int. 2021, 28, 45317–45334. [Google Scholar] [CrossRef]
  48. Hata, A.; Chen, Y.G. TGF-β Signaling from Receptors to Smads. Cold Spring Harb. Perspect. Biol. 2016, 8, a022061. [Google Scholar] [CrossRef]
  49. Baba, A.B.; Rah, B.; Bhat, G.R.; Mushtaq, I.; Parveen, S.; Hassan, R.; Zargar, M.H.; Afroze, D. Transforming growth factor-beta (TGF-β) signaling in cancer—A betrayal within. Front. Pharmacol. 2022, 13, 791272. [Google Scholar] [CrossRef]
  50. Parvani, J.G.; Gujrati, M.D.; Mack, M.A.; Schiemann, W.P.; Lu, Z.R. Silencing β3 integrin by targeted ECO/siRNA nanoparticles inhibits EMT and metastasis of triple-negative breast cancer. Cancer Res. 2015, 75, 2316–2325. [Google Scholar] [CrossRef]
  51. Liu, Y.; Xie, X.; Hou, X.; Shen, J.; Shi, J.; Chen, H.; He, Y.; Wang, Z.; Feng, N. Functional oral nanoparticles for delivering silibinin and cryptotanshinone against breast cancer lung metastasis. J. Nanobiotechnol. 2020, 18, 83. [Google Scholar] [CrossRef]
  52. Guo, D.D.; Li, Q.N.; Li, C.M.; Bi, H.S. Zinc oxide nanoparticles inhibit murine photoreceptor-derived cell proliferation and migration via reducing TGF-β and MMP-9 expression in vitro. Cell Prolif. 2015, 48, 198–208. [Google Scholar] [CrossRef] [PubMed]
  53. Casey, L.M.; Pearson, R.M.; Hughes, K.R.; Liu, J.; Rose, J.A.; North, M.G.; Wang, L.Z.; Lei, M.; Miller, S.D.; Shea, L.D. Conjugation of transforming growth factor beta to antigen-loaded poly(lactide-co-glycolide) nanoparticles enhances efficiency of antigen-specific tolerance. Bioconjugate Chem. 2018, 29, 813–823. [Google Scholar] [CrossRef] [PubMed]
  54. Morry, J.; Ngamcherdtrakul, W.; Gu, S.; Goodyear, S.M.; Castro, D.J.; Reda, M.M.; Sangvanich, T.; Yantasee, W. Dermal delivery of HSP47 siRNA with NOX4-modulating mesoporous silica-based nanoparticles for treating fibrosis. Biomaterials 2015, 66, 41–52. [Google Scholar] [CrossRef]
  55. Santarpia, L.; Lippman, S.M.; El-Naggar, A.K. Targeting the MAPK-RAS-RAF signaling pathway in cancer therapy. Expert Opin. Ther. Targets 2012, 16, 103–119. [Google Scholar] [CrossRef]
  56. Hu, J.-H.; Yang, J.-P.; Liu, L.; Li, C.-F.; Wang, L.-N.; Ji, F.-H.; Cheng, H. Involvement of CX3CR1 in bone cancer pain through the activation of microglia p38 MAPK pathway in the spinal cord. Brain Res. 2012, 1465, 1–9. [Google Scholar] [CrossRef] [PubMed]
  57. Yang, Y.; Kim, S.C.; Yu, T.; Yi, Y.-S.; Rhee, M.H.; Sung, G.-H.; Yoo, B.C.; Cho, J.Y. Functional roles of p38 mitogen-activated protein kinase in macrophage-mediated inflammatory responses. Mediat. Inflamm. 2014, 2014, 352371. [Google Scholar] [CrossRef]
  58. Pal, I.; Mandal, M. PI3K and Akt as molecular targets for cancer therapy: Current clinical outcomes. Acta Pharmacol. Sin. 2012, 33, 1441–1458. [Google Scholar] [CrossRef]
  59. Zhang, Y.; Wang, X. Targeting the Wnt/β-catenin signaling pathway in cancer. J. Hematol. Oncol. 2020, 13, 165. [Google Scholar] [CrossRef]
  60. Tran, M.A.; Gowda, R.; Sharma, A.; Park, E.J.; Adair, J.; Kester, M.; Smith, N.B.; Robertson, G.P. Targeting V600EB-Raf and Akt3 using nanoliposomal-small interfering RNA inhibits cutaneous melanocytic lesion development. Cancer Res. 2008, 68918, 7638–7649. [Google Scholar] [CrossRef]
  61. Clavreul, A.; Roger, E.; Pourbaghi-Masouleh, M.; Lemaire, L.; Tétaud, C.; Menei, P. Development and characterization of sorafenib-loaded lipid nanocapsules for the treatment of glioblastoma. Drug Deliv. 2018, 25, 1756–1765. [Google Scholar] [CrossRef] [PubMed]
  62. Lunova, M.; Prokhorov, A.; Jirsa, M.; Hof, M.; Olżyńska, A.; Jurkiewicz, P.; Kubinova, S.; Kubinová, S.; Lunov, O.; Dejneka, A. Nanoparticle core stability and surface functionalization drive the mTOR signaling pathway in hepatocellular cell lines. Sci. Rep. 2017, 7, 16049. [Google Scholar] [CrossRef]
  63. Uzhytchak, M.; Smolková, B.; Lunova, M.; Jirsa, M.; Frtús, A.; Kubinová, S.; Dejneka, A.; Lunov, O. Iron oxide nanoparticle-induced autophagic flux is regulated by interplay between p53-mTOR axis and Bcl-2 signaling in hepatic cells. Cells 2020, 9, 1015. [Google Scholar] [CrossRef] [PubMed]
  64. Unal, O.; Akkoc, Y.; Kocak, M.; Nalbat, E.; Dogan-Ekici, A.I.; Acar, H.Y.; Gozuacik, D. Treatment of breast cancer with autophagy inhibitory microRNAs carried by AGO2-conjugated nanoparticles. J. Nanobiotech. 2020, 18, 65. [Google Scholar] [CrossRef]
  65. Huijts, C.M.; Santegoets, S.J.; de Jong, T.D.; Verheul, H.M.; de Gruijl, T.D.; van der Vilet, H.J. Immunological effects of everolimus in patients with metastatic renal cell cancer. Int. J. Immunopathol. Pharmacol. 2017, 30, 341–342. [Google Scholar] [CrossRef]
  66. Kong, N.; Tao, W.; Ling, X.; Wang, J.; Xiao, Y.; Shi, S.; Ji, X.; Shajii, A.; Gan, S.T.; Kim, N.Y.; et al. Synthetic mRNA nanoparticle-mediated restoration of p53 tumor suppressor sensitizes p53-deficient cancers to mTOR inhibition. Sci. Transl. Med. 2019, 11, eaaw1565. [Google Scholar] [CrossRef] [PubMed]
  67. Kulkarni, A.A.; Roy, B.; Rao, P.S.; Wyant, G.A.; Mahmoud, A.; Ramachandran, M.; Sengupta, P.; Goldman, A.; Kotamraju, V.R.; Basu, S.; et al. Sengupta. Supramolecular nanoparticles that target phosphoinositide-3-kinase overcome insulin resistance and exert pronounced antitumor efficacy. Cancer Res. 2013, 73, 6987–6997. [Google Scholar] [CrossRef] [PubMed]
  68. Au, K.M.; Wang, A.Z.; Park, S.I. Pretargeted delivery of PI3K/mTOR small-molecule inhibitor-loaded nanoparticles for treatment of non-Hodgkin’s lymphoma. Sci. Adv. 2020, 6, eaaz9798. [Google Scholar] [CrossRef]
  69. Wu, Y.Y.; Wu, H.C.; Wu, J.E.; Huang, K.Y.; Yang, S.C.; Chen, S.X.; Tsao, C.J.; Hsu, K.F.; Chen, Y.L.; Hong, T.M. The dual PI3K/mTOR inhibitor BEZ235 restricts the growth of lung cancer tumors regardless of EGFR status, as a potent accompanist in combined therapeutic regimens. J. Exp. Clin. Cancer Res. 2019, 38, 282. [Google Scholar] [CrossRef]
  70. Dreaden, E.C.; Kong, Y.W.; Morton, S.W.; Correa, S.; Choi, K.Y.; Shopsowitz, K.E.; Renggli, K.; Drapkin, R.; Yaffe, M.B.; Hammond, P.T. Tumor-targeted synergistic blockade of MAPK and PI3K from a layer-by-layer nanoparticle. Clin. Cancer Res. 2015, 21, 4410–4419. [Google Scholar] [CrossRef]
  71. Lu, L.; Li, K.; Mao, Y.H.; Qu, H.; Yao, B.; Zhong, W.W.; Ma, B.; Wang, Z.Y. Gold-chrysophanol nanoparticles suppress human prostate cancer progression through inactivating AKT expression and inducing apoptosis and ROS generation in vitro and in vivo. Int. J. Oncol. 2017, 51, 1089–1103. [Google Scholar] [CrossRef]
  72. Yuan, J.; Dong, X.; Yap, J.; Hu, J. The MAPK and AMPK signalings: Interplay and implication in targeted cancer therapy. J. Hematol. Oncol. 2020, 13, 113. [Google Scholar] [CrossRef]
  73. Lucero-Acuña, A.; Jeffer, J.J.; Abril, E.R.; Nagle, R.B.; Guzman, R.; Pagel, M.D.; Meuillet, E.M. Nanoparticle delivery of an AKT/PDK1 inhibitor improves the therapeutic effect in pancreatic cancer. Int. J. Nanomed. 2014, 9, 5653–5665. [Google Scholar] [CrossRef]
  74. Shen, J.; Cai, W.; Ma, Y.; Xu, R.; Huo, Z.; Song, L.; Qiu, X.; Zhang, Y.; Li, A.; Cao, W.; et al. hGC33-modified and Sorafenib-loaded nanoparticles have a synergistic anti-hepatoma effect by inhibiting Wnt signaling pathway. Nanoscale Res. Lett. 2020, 15, 220. [Google Scholar] [CrossRef]
  75. Morales, J.; Li, L.; Fattah, F.J.; Dong, Y.; Bey, E.A.; Patel, M.; Gao, J.; Boothman, D.A. Review of poly (ADP-ribose) polymerase (PARP) mechanisms of action and rationale for targeting in cancer and other diseases. Crit. Rev. Eukaryot. Gene Expr. 2014, 24, 15–28. [Google Scholar] [CrossRef]
  76. Wang, Y.; Luo, W.; Wang, Y. PARP-1 and its associated nucleases in DNA damage response. DNA Repair 2019, 81, 102651. [Google Scholar] [CrossRef]
  77. Götting, I.; Jendrossek, V.; Matschke, J. A new twist in protein kinase B/Akt signaling: Role of altered cancer cell metabolism in AKT-mediated therapy resistance. Int. J. Mol. Sci. 2020, 21, 8563. [Google Scholar] [CrossRef]
  78. Zhang, D.; Baldwin, P.; Leal, A.S.; Carapellucci, S.; Sridhar, S.; Liby, K.T. A nano-liposome formulation of the PARP inhibitor Talazoparib enhances treatment efficacy and modulates immune cell populations in mammary tumors of BRCA-deficient mice. Theranostics 2019, 9, 6224–6238. [Google Scholar] [CrossRef]
  79. van de Ven, A.L.; Tangutoori, S.; Baldwin, P.; Qiao, J.; Gharagouzloo, C.; Seitzer, N.; Clohessy, J.G.; Makrigiorgos, G.M.; Cormack, R.; Pandolfi, P.P.; et al. Nanoformulation of Olaparib amplifies PARP inhibition and sensitizes PTEN/TP53-deficient prostate cancer to radiation. Mol. Cancer Ther. 2017, 16, 1279–1289. [Google Scholar] [CrossRef]
  80. Gonzales, J.; Kossatz, S.; Roberts, S.; Pirovano, G.; Brand, C.; Pérez-Medina, C.; Donabedian, P.; de la Cruz, M.J.; Mulder, W.J.M.; Reiner, T. Nanoemulsion-based delivery of fluorescent PARP inhibitors in mouse models of small cell lung cancer. Bioconjugate Chem. 2018, 29, 3776–3782. [Google Scholar] [CrossRef]
  81. Haynes, B.; Gajan, A.; Nangia-Makker, P.; Shekhar, M.P. RAD6B is a major mediator of triple negative breast cancer cisplatin resistance: Regulation of translesion synthesis/Fanconi anemia crosstalk and BRCA1 independence. Biochim. Biophys. Acta Mol. Basis Dis. 2021, 1866, 165561. [Google Scholar] [CrossRef]
  82. Haynes, B.; Zhang, Y.; Liu, F.; Li, J.; Petit, S.; Kothayer, H.; Bao, X.; Westwell, A.D.; Mao, G.; Shekar, M.P.V. Gold nanoparticle conjugated Rad6 inhibitor induces cell death in triple-negative breast cancer cells by inducing mitochondrial dysfunction and PARP-1 hyperactivation: Synthesis and characterization. Nanomedicine 2016, 12, 745–757. [Google Scholar] [CrossRef]
  83. Mensah, L.B.; Morton, S.W.; Li, J.; Xiao, H.; Quadir, M.A.; Elias, K.M.; Penn, E.; Richson, A.K.; Ghoroghchian, P.P.; Liu, J.; et al. Layer-by-layer nanoparticles for novel delivery of cisplatin and PARP inhibitors for platinum-based drug resistance therapy in ovarian cancer. Bioeng. Transl. Med. 2019, 4, e10131. [Google Scholar] [CrossRef]
  84. Goldberg, M.S.; Xing, D.; Ren, Y.; Orsulic, S.; Bhatia, S.N.; Sharp, P.A. Nanoparticle-mediated delivery of siRNA targeting Parp1 extends survival of mice bearing tumors derived from Brca1-deficient ovarian cancer cells. Proc. Natl. Acad. Sci. USA 2011, 108, 745–750. [Google Scholar] [CrossRef]
  85. Magalhães, J.A.; Arruda, D.C.; Baptista, M.S.; Tada, D.B. Co-encapsulation of methylene blue and PARP-Inhibitor into poly(lactic-co-glycolic acid) nanoparticles for enhanced PDT of cancer. Nanomaterials 2021, 11, 1514. [Google Scholar] [CrossRef]
  86. Jabir, M.; Sahib, U.I.; Taqi, Z.; Tasha, A.; Sulaiman, G.; Albukhaty, S.; Al-Shammari, A.; Alwahibi, M.; Soliman, D.; Dewir, Y.H.; et al. Linalool-loaded glutathione-modified gold nanoparticles conjugated with CALNN peptide as apoptosis inducer and NF-κB translocation inhibitor in SKOV-3 cell line. Int. J. Nanomed. 2020, 15, 9025–9047. [Google Scholar] [CrossRef]
  87. Yousefi, H.; Bahramy, A.; Zafari, N.; Delavar, M.R.; Nguyen, K.; Haghi, A.; Kandelouei, T.; Vittori, C.; Jazireian, P.; Maleki, S.; et al. Notch signaling pathway: A comprehensive prognostic and gene expression profile analysis in breast cancer. BMC Cancer 2022, 22, 1282. [Google Scholar] [CrossRef]
  88. Wang, Z.; Li, Y.; Kong, D.; Sarkar, F.H. The role of notch signaling pathway in epithelial-mesenchymal transition (EMT) during development and tumor aggressiveness. Curr. Drug Targets 2010, 11, 745–751. [Google Scholar] [CrossRef]
  89. Jia, Y.; Wang, Y.; Xie, J. The Hedgehog pathway: Role in cell differentiation, polarity and proliferation. Arch. Toxicol. 2015, 89, 179–191. [Google Scholar] [CrossRef]
  90. Mamaeva, V.; Niemi, R.; Beck, M.; Özliseli, E.; Desai, D.; Landor, S.; Gronroos, T.; Kronqvist, P.; Pettersen, I.K.N.; McCormack, E.; et al. Inhibiting notch activity in breast cancer stem cells by glucose functionalized nanoparticles carrying γ-secretase inhibitors. Mol. Ther. 2016, 24, 926–936. [Google Scholar] [CrossRef]
  91. Boinpelly, V.C.; Verma, R.K.; Srivastav, S.; Srivastava, R.K.; Shankar, S. α-Mangostin-encapsulated PLGA nanoparticles inhibit colorectal cancer growth by inhibiting Notch pathway. J. Cell. Mol. Med. 2020, 24, 11343–11354. [Google Scholar] [CrossRef]
  92. Singh, S.K.; Gordetsky, J.B.; Bae, S.; Acosta, E.P.; Lillard, J.W., Jr.; Singh, R. Selective targeting of the hedgehog signaling pathway by pbm nanoparticles in docetaxel-resistant prostate cancer. Cells 2020, 9, 1976. [Google Scholar] [CrossRef]
  93. Chenna, V.; Hu, C.; Pramanik, D.; Aftab, B.T.; Karikari, C.; Campbell, N.R.; Hong, S.-M.; Zhao, M.; Rudek, M.A.; Khan, S.R.; et al. A polymeric nanoparticle encapsulated small-molecule inhibitor of Hedgehog signaling (NanoHHI) bypasses secondary mutational resistance to smoothened antagonists. Mol. Cancer Ther. 2012, 11, 165–173. [Google Scholar] [CrossRef]
  94. Kim, J.; Dey, A.; Malhotra, A.; Liu, J.; Ahn, S.I.; Sei, Y.J.; Kenney, A.M.; Macdonald, T.J.; Kim, Y. Engineered biomimetic nanoparticle for dual targeting of the cancer stem-like cell population in sonic hedgehog medulloblastoma. Proc. Natl. Acad. Sci. USA 2020, 117, 24205–24212. [Google Scholar] [CrossRef]
  95. Bell, J.B.; Rink, J.S.; Eckerdt, F.; Clymer, J.; Goldman, S.; Thaxton, C.S.; Platanias, L.C. HDL nanoparticles targeting sonic hedgehog subtype medulloblastoma. Sci. Rep. 2018, 8, 1211. [Google Scholar] [CrossRef]
  96. Melamed, J.R.; Ioele, S.A.; Hannum, A.J.; Ullman, V.M.; Day, E.S. Polyethylenimine-spherical nucleic acid nanoparticles against gli1 reduce the chemoresistance and stemness of glioblastoma cells. Mol. Pharm. 2018, 15, 5135–5145. [Google Scholar] [CrossRef]
  97. Xu, Y.; Chenna, V.; Hu, C.; Sun, H.X.; Khan, M.; Bai, H.; Yang, X.R.; Zhu, Q.F.; Sun, Y.F.; Maitra, A.; et al. Polymeric nanoparticle-encapsulated hedgehog pathway inhibitor HPI-1 (NanoHHI) inhibits systemic metastases in an orthotopic model of human hepatocellular carcinoma. Clin. Cancer Res. 2012, 18, 1291–1302. [Google Scholar] [CrossRef]
  98. Piktel, E.; Ościłowska, I.; Suprewicz, Ł.; Depciuch, J.; Marcińczyk, N.; Chabielska, E.; Wolak, P.; Wollny, T.; Janion, M.; Parlinska-Wotjan, M.; et al. ROS-mediated apoptosis and autophagy in ovarian cancer cells treated with peanut-shaped gold nanoparticles. Int. J. Nanomed. 2021, 16, 1993–2011. [Google Scholar] [CrossRef]
  99. Yin, H.; Xiong, G.; Guo, S.; Xu, C.; Xu, R.; Guo, P.; Shu, D. Delivery of anti-miRNA for triple-negative breast cancer therapy using RNA nanoparticles targeting stem cell marker CD133. Mol. Ther. 2019, 27, 1252–1261. [Google Scholar] [CrossRef]
  100. Wan, X.; Liu, C.; Lin, Y.; Fu, J.; Lu, G.; Lu, Z. pH sensitive peptide functionalized nanoparticles for co-delivery of erlotinib and DAPT to restrict the progress of triple-negative breast cancer. Drug Deliv. 2019, 26, 470–480. [Google Scholar] [CrossRef]
  101. Ghaemi, B.; Hajipour, M.J. Tumor acidic environment directs nanoparticle impacts on cancer cells. J. Colloid Interface Sci. 2023, 634, 684–692. [Google Scholar] [CrossRef] [PubMed]
  102. Reda, M.; Ngamcherdtrakul, W.; Gu, S.; Bejan, D.S.; Siriwon, N.; Gray, J.W.; Yantasee, W. PLK1 and EGFR targeted nanoparticle as a radiation sensitizer for non-small cell lung cancer. Cancer Lett. 2019, 467, 9–18. [Google Scholar] [CrossRef]
  103. Xu, H.; Sun, Y.; Zeng, L.; Li, Y.; Hu, S.; He, S.; Chen, H.; Zou, Q.; Luo, B. Inhibition of cytosolic phospholipase A2 alpha increases chemosensitivity in cervical carcinoma through suppressing β-catenin signaling. Cancer Biol. Ther. 2019, 20, 912–921. [Google Scholar] [CrossRef]
  104. Tunset, H.M.; Feuerherm, A.J.; Selvik, L.M.; Johansen, B.; Moestue, S.A. Cytosolic phospholipase A2 alpha regulates TLR signaling and migration in metastatic 4T1 cells. Int. J. Mol. Sci. 2019, 20, 4800. [Google Scholar] [CrossRef]
  105. Gowda, R.; Dinavahi, S.S.; Iyer, S.; Banerjee, S.; Neves, R.I.; Pameijer, C.R.; Robertson, G.P. Nanoliposomal delivery of cytosolic phospholipase A(2) inhibitor arachidonyl trimethyl ketone for melanoma treatment. Nanomedicine 2018, 14, 863–873. [Google Scholar] [CrossRef]
  106. Tran, P.H.L.; Wang, T.; Yin, W.; Tran, T.T.D.; Nguyen, T.N.G.; Lee, B.J.; Duan, W. Aspirin-loaded nanoexosomes as cancer therapeutics. Int. J. Pharm. 2019, 572, 118786. [Google Scholar] [CrossRef]
  107. Sanaei, M.J.; Pourbagheri-Sigaroodi, A.; Kaveh, V.; Abolghasemi, H.; Ghaffari, S.H.; Momeny, M.; Bashash, D. Recent advances in immune checkpoint therapy in non-small cell lung cancer and opportunities for nanoparticle-based therapy. Eur. J. Pharmacol. 2021, 909, 174404. [Google Scholar] [CrossRef]
  108. Lao, Y.; Shen, D.; Zhang, W.; He, R.; Jiang, M. Immune checkpoint inhibitors in cancer therapy-how to overcome drug resistance? Cancers 2022, 14, 3575. [Google Scholar] [CrossRef]
  109. Zhao, Y.; Detering, L.; Sultan, D.; Cooper, M.L.; You, M.; Cho, S.; Meier, S.L.; Luehmann, H.; Sun, G.; Rettig, M.; et al. Gold nanoclusters doped with (64)Cu for CXCR4 positron emission tomography imaging of breast cancer and metastasis. ACS Nano 2016, 10, 5959–5970. [Google Scholar] [CrossRef]
  110. Sabit, H.; Abdel-Hakem, M.; Shoala, T.; Abdel-Ghany, S.; Abdel-Latif, M.M.; Almulhim, J.; Mansy, M. Nanocarriers: A reliable tool for the delivery of anticancer drugs. Pharmaceutics 2022, 14, 1566. [Google Scholar] [CrossRef]
  111. Thuru, X.; Magnez, R.; El-Bouazzati, H.; Vergoten, G.; Quesnel, B.; Bailly, C. Drug repurposing to enhance antitumor response to PD-1/PD-L1 immune checkpoint inhibitors. Cancers 2022, 14, 3368. [Google Scholar] [CrossRef] [PubMed]
  112. Cunningham, N.; Lapointe, R.; Lerouge, S. Biomaterials for enhanced immunotherapy. APL Bioeng. 2022, 6, 041502. [Google Scholar] [CrossRef]
  113. Zhao, J.; Ye, H.; Lu, Q.; Wang, K.; Chen, X.; Song, J.; Wang, H.; Lu, Y.; Cheng, M.; He, Z.; et al. Inhibition of post-surgery tumour recurrence via a sprayable chemo-immunotherapy gel releasing PD-L1 antibody and platelet-derived small EVs. J. Nanobiotechnol. 2022, 20, 62. [Google Scholar] [CrossRef] [PubMed]
  114. Guan, X.; Sun, L.; Shen, Y.; Jin, F.; Bo, X.; Zhu, C.; Han, X.; Li, X.; Chen, Y.; Xu, H.; et al. Nanoparticle-enhanced radiotherapy synergizes with PD-L1 blockade to limit post-surgical cancer recurrence and metastasis. Nat. Commun. 2022, 13, 2834. [Google Scholar] [CrossRef]
  115. Wicki, A.; Witzigmann, D.; Balasubramanian, V.; Huwyler, J. Nanomedicine in cancer therapy: Challenges, opportunities, and clinical applications. J. Control. Release 2015, 200, 138–157. [Google Scholar] [CrossRef]
  116. Rodríguez, F.; Caruana, P.; la Fuente, N.D.; Español, P.; Gámez, M.; Balart, J.; Llurba, E.; Rovira, R.; Ruiz, R.; Martín-Lorente, C.; et al. Nano-based approved pharmaceuticals for cancer treatment: Present and future challenges. Biomolecules 2022, 12, 784. [Google Scholar] [CrossRef]
  117. US NCI. Cancer Nano-Therapies in the Clinic and Clinical Trials. Available online: https://www.cancer.gov/nano/cancer-nanotechnology/current-treatments (accessed on 19 August 2024).
  118. Grivennikov, S.I.; Greten, F.R.; Karin, M. Immunity, inflammation, and cancer. Cell 2010, 140, 883–899. [Google Scholar] [CrossRef]
  119. You, P.; Fu, S.; Yu, K.; Xia, Y.; Wu, H.; Yang, Y.; Ma, C.; Liu, D.; Chen, X.; Wang, J.; et al. Scutellarin suppresses neuroinflammation via the inhibition of the AKT/NF-κB and p38/JNK pathway in LPS-induced BV-2 microglial cells. Naunyn Schmiedebergs Arch. Pharmacol. 2018, 391, 743–751. [Google Scholar] [CrossRef] [PubMed]
  120. Velagapudi, R.; Kumar, A.; Bhatia, H.S.; El-Bakoush, A.; Lepiarz, I.; Fiebich, B.L.; Olajide, O.A. Inhibition of neuroinflammation by thymoquinone requires activation of Nrf2/ARE signalling. Int. Immunopharmacol. 2017, 48, 17–29. [Google Scholar] [CrossRef]
  121. Hankittichai, P.; Lou, H.J.; Wikan, N.; Smith, D.R.; Potikanond, S.; Nimlamool, W. Oxyresveratrol inhibits IL-1β-induced inflammation via suppressing AKT and ERK1/2 activation in human microglia, HMC3. Int. J. Mol. Sci. 2020, 21, 6054. [Google Scholar] [CrossRef]
  122. Subedi, L.; Yumnam, S. Terpenoids from Abies holophylla attenuate LPS-Induced neuroinflammation in microglial cells by suppressing the JNK-related signaling pathway. Int. J. Mol. Sci. 2021, 22, 965. [Google Scholar] [CrossRef] [PubMed]
  123. Basak, U.; Sarkar, T.; Mukherjee, S.; Chakraborty, S.; Dutta, A.; Dutta, S.; Nayak, D.; Kaushik, S.; Das, T.; Sa, G. Tumor-associated macrophages: An effective player of the tumor microenvironment. Front. Immunol. 2023, 14, 1295257. [Google Scholar] [CrossRef]
  124. Gao, Y.; Zhuang, Z.; Lu, Y.; Tao, T.; Zhou, Y.; Liu, G.; Wang, H.; Zhang, D.; Wu, L.; Dai, H.; et al. Curcumin mitigates neuro-inflammation by modulating microglia polarization through inhibiting TLR4 axis signaling pathway following experimental subarachnoid hemorrhage. Front. Neurosci. 2019, 13, 1223. [Google Scholar] [CrossRef] [PubMed]
  125. Giacoppo, S.; Rajan, T.S.; Nicola, G.R.D.; Iori, R.; Bramanti, P.; Mazzon, E. Moringin activates Wnt canonical pathway by inhibiting GSK3β in a mouse model of experimental autoimmune encephalomyelitis. Drug Des. Dev. Ther. 2016, 10, 3291–3304. [Google Scholar] [CrossRef]
  126. Jo, S.H.; Kim, M.E.; Cho, J.H.; Lee, Y.; Lee, J.; Park, Y.D.; Lee, J.S. Hesperetin inhibits neuroinflammation on microglia by suppressing inflammatory cytokines and MAPK pathways. Arch. Pharm. Res. 2019, 42, 695–703. [Google Scholar] [CrossRef] [PubMed]
  127. Lee, T.H.; Chen, J.L.; Liu, P.S.; Tsai, M.M.; Wang, S.J.; Hsieh, H.L. Rottlerin, a natural polyphenol compound, inhibits upregulation of matrix metalloproteinase-9 and brain astrocytic migration by reducing PKC-δ-dependent ROS signal. J. Neuroinflamm. 2020, 17, 177. [Google Scholar] [CrossRef]
  128. Hu, X.J.; Xie, M.Y.; Kluxen, F.M.; Diel, P. Genistein modulates the anti-tumor activity of cisplatin in MCF-7 breast and HT-29 colon cancer cells. Arch. Toxicol. 2014, 88, 625–635. [Google Scholar] [CrossRef]
  129. Khoobchandani, M.; Katti, K.K.; Karikachery, A.R.; Thipe, V.C.; Srisrimal, D.; Mohandoss, D.D.K.; Darshakumar, R.D.; Joshi, C.M.; Katti, K.V. New approaches in breast cancer therapy through green nanotechnology and nano-ayurvedic medicine—Pre-clinical and pilot human clinical investigations. Int. J. Nanomed. 2020, 15, 181–197. [Google Scholar] [CrossRef]
  130. Clark, A.J.; Wiley, D.T.; Zuckerman, J.E.; Webster, P.; Chao, J.; Lin, J.; Yen, Y.; Davis, M.E. CRLX101 nanoparticles localize in human tumors and not in adjacent, nonneoplastic tissue after intravenous dosing. Proc. Natl. Acad. Sci. USA 2016, 113, 3850–3854. [Google Scholar] [CrossRef]
  131. Piha-Paul, S.A.; Thein, K.Z.; Souza, P.D.; Kefford, R.; Gangadhar, T.; Smith, C.; Schuster, S.; Zamboni, W.C.; Dees, C.E.; Markman, B. First-in-human, phase I/IIa study of CRLX301, a nanoparticle drug conjugate containing docetaxel, in patients with advanced or metastatic solid malignancies. Investig. New Drugs 2021, 39, 1047–1056. [Google Scholar] [CrossRef]
  132. Lu, J.; Gu, A.; Wang, W.; Huang, A.; Han, B.; Zhong, H. Polymeric micellar paclitaxel (Pm-Pac) prolonged overall survival for NSCLC patients without pleural metastasis. Int. J. Pharm. 2022, 623, 121961. [Google Scholar] [CrossRef] [PubMed]
  133. Zhu, Z.; Qian, Z.; Yan, Z.; Zhao, C.; Wang, H.; Ying, G. A phase I pharmacokinetic study of ursolic acid nanoliposomes in healthy volunteers and patients with advanced solid tumors. Int. J. Nanomed. 2013, 8, 129–136. [Google Scholar] [CrossRef]
  134. Sarkar, S.; Horn, G.; Moulton, K.; Oza, A.; Byler, S.; Kokolus, S.; Longacre, M. Cancer development, progression, and therapy: An epigenetic overview. Int. J. Mol. Sci. 2013, 14, 21087–21113. [Google Scholar] [CrossRef]
  135. Thompson, H.J.; Lutsiv, T. Natural products in precision oncology: Plant-based small molecule inhibitors of protein kinases for cancer chemoprevention. Nutrients 2023, 15, 1192. [Google Scholar] [CrossRef] [PubMed]
  136. Gurunathan, S.; Kang, M.H.; Qasim, M.; Kim, J.H. Nanoparticle-mediated combination therapy: Two-in-one approach for cancer. Int. J. Mol. Sci. 2018, 19, 3264. [Google Scholar] [CrossRef]
  137. Mitchell, M.J.; Billingsley, M.M.; Haley, R.M.; Wechsler, M.E.; Peppas, N.A.; Langer, R. Engineering precision nanoparticles for drug delivery. Nat. Rev. Drug Discov. 2021, 20, 101–124. [Google Scholar] [CrossRef]
  138. Azie, O.; Greenberg, Z.F.; Batich, C.D.; Dobson, J.P. Carbodiimide conjugation of latent transforming growth factor β1 to superparamagnetic iron oxide nanoparticles for remote activation. Int. J. Mol. Sci. 2019, 20, 3190. [Google Scholar] [CrossRef]
  139. Qie, Y.; Yuan, H.; von Romeling, C.A.; Chen, Y.; Liu, X.; Shih, K.D.; Knight, J.A.; Tun, H.W.; Wharen, R.E.; Jiang, W.; et al. Surface modification of nanoparticles enables selective evasion of phagocytic clearance by distinct macrophage phenotypes. Sci. Rep. 2016, 6, 26269. [Google Scholar] [CrossRef]
  140. Shi, Y.; van der Meel, R.; Chen, X.; Lammers, T. The EPR effect and beyond: Strategies to improve tumor targeting and cancer nanomedicine treatment efficacy. Theranostics 2020, 10, 7921–7924. [Google Scholar] [CrossRef]
Figure 1. Drug carriers used in advanced nanotechnology and their strengths and limitations [16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47]. NP, nanoparticle.
Figure 1. Drug carriers used in advanced nanotechnology and their strengths and limitations [16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47]. NP, nanoparticle.
Pharmaceutics 16 01169 g001
Table 1. Anticancer effect of TGF-β signaling-based nanotherapies.
Table 1. Anticancer effect of TGF-β signaling-based nanotherapies.
Nanomedicine NameDrug in NanomedicineDelivery SystemTarget CancerExperimental ModelEffect of Nanomedicine on CancerRef.
ECO/siRNA NPsβ3 Integrin siRNALipid ECO-based NPsTNBCMDA-MB-231 cell line and NME cell lineSilenced the expression of Integrin β3; lessened TGF-β mediated epithelial-mesenchymal transition and metastasis[50]
Poly-N-(2-hydroxypropyl) methacrylamide-coated W-LPNs (S/C-pW-LPNs)Silibinin and cryptotanshinonePoly-N-(2-hydroxypropyl) methylacrylamide-coated wheat germ agglutinin-modified lipid-polymer hybrid NPsBreast cancer4T1 breast cancer cells; 4T1 tumor-bearing nude mouse modelIncreased 4T1 cell toxicity; inhibited cell invasion and migration; reduced tumor progression and metastasis to the lungs[51]
ZP6Zinc oxideZp6 Capped with aminopolysiloxaneRetinal degenerative diseasesMurine photoreceptor-derived 661W cell lineFormation of apoptotic bodies; disruption of cell cycle; disruption of intracellular calcium homeostasis and increase in oxidative stress; reduction in the expression of TGF-β and matrix metalloprotease 9[52]
PLG(Ag) NPsTGF-β and OVA peptide PLGA NPsMultiple sclerosis and autoimmune encephalomyelitisMouse model for multiple sclerosis and autoimmune encephalomyelitisReduced inflammation in bone marrow-derived dendritic cells; induced regulatory T cells; reduced disease severity[53]
MSNP-PEI-PEGSiHSP47Polyethylenimine and polyethylene glycol coating on mesoporous silica NPFibrotic disease (scleroderma)TGF-β stimulated fibroblasts; bleomycin-induced scleroderma mouse modelReduced HSP47 protein expression; reduced NADPH oxidase 4 levels; reduced pro-fibrotic markers[54]
HSP47, heat shock protein 47; NP, nanoparticle; PLGA, poly(lactic-co-glycolic acid); TGF-β, transforming growth factor-beta; TNBC, triple-negative breast cancer.
Table 3. PARP signaling-based nanotherapies.
Table 3. PARP signaling-based nanotherapies.
Nanomedicine NameDrug in NanomedicineDelivery SystemTarget CancerExperimental ModelEffect of Nanomedicine on CancerRef.
NanoTalazoparibTalazoparibBilayer nano-liposomeBRCA-mutated metastatic breast cancerBRCA-deficient miceInduced DNA damage, cell cycle arrest, and inhibition of cell proliferation in tumors; modulated immune cell populations; decreased myeloid-derived suppressor cells in tumors and spleen[78]
NanoOlaparibOlaparibLipid-based injectable nanoformulationAdvanced prostate cancerPTEN/p53-deficient mouse with prostate cancerMade tumors more radiation-sensitive; caused significant tumor growth inhibition[79]
Nanoemulsion encapsulated PARPi-FLPARPi-FL (fluorescently labeled sensor for Olaparib)NanoemulsionSmall cell lung cancerSubcutaneous xenografts of small cell lung cancerIncreased blood half-life; improved delineation of small cell lung cancer xenografts[80]
SMI#9-GNPSMI#9gNPsTNBCCell culture models of TNBCInduced cytotoxicity in mesenchymal TNBC cells; enhanced cisplatin sensitivity when combined with cisplatin; selectively induced cell death through mitochondrial dysfunction and PARP1 stabilization/hyperactivation[82]
Liposomal NPsCisplatin and PARP inhibitorsLiposomal NPs with a terminal hyaluronic acid layerOvarian cancerLuciferase and CD44-expressing orthotopic OVCAR8 xenograft nude miceModerated systemic toxicity; reduced tumor metastasis; extended survival[83]
LipidoidssiRNA targeting PARP1 (siParp1)Lipidoids for delivering siRNAOvarian cancerMouse models of ovarian cancerInhibited cell growth, induced apoptosis in BRCA1-deficient cells, extended survival in mice with ovarian cancer cells[84]
PLGA NPs co-encapsulating methylene blueVeliparibPLGA NPsMelanomaIn vitro assays using B16F10-Nex2 cellsDecreased cell viability[85]
gNP-CALNNLinaloolgNPs capped with glutathione and conjugated with a CALNN peptideOvarian cancerIn vitro assays using SKOV-3 ovarian cancer cellsInduced apoptosis of ovarian cancer cells via activating caspase-8 and apoptosis-associated proteins[86]
gNP, gold NP; NP, nanoparticle; PARP, poly(ADP-ribose) polymerase; PLGA, poly(lactic-co-glycolic acid); TNBC, triple-negative breast cancer.
Table 6. Approved nanomedicines currently available for anticancer therapy [115,116,117].
Table 6. Approved nanomedicines currently available for anticancer therapy [115,116,117].
Institute (Approval Year)ProductCompanyDrug in NanomedicineDelivery SystemTarget Cancer
FDA (1994, 2006)OncasparEnzon-Sigma-tauPegaspargase/L-asparaginasePolymer conjugateAcute lymphoblastic leukemia
FDA (1996)DaunoXomeGilead SciencesDaunorubicinLiposomeKaposi’s sarcoma
FDA (1999)DepoCytPacira Pharmaceuticals CytarabineLiposomeNeoplastic meningitis
FDA (2005)AbraxaneAbraxis/CelgenePaclitaxelNP-bound albuminBreast and pancreatic cancer, NSCLC
FDA (2012)MarqiboTalon Therapeutics/Spectrum PharmaceuticalsVincristineLiposomeAcute lymphoblastic leukemia
FDA (2015)OnivydeMerrimack PharmaIrinotecanLiposomePancreatic cancer, colorectal cancer
FDA (1995, 1999, 2007), EMA (1996, 2000), Taiwan (1998)Doxil, Caelyx, Myocet, and Lipo-DoxJohnson and Johnson, Schering-Plough, Teva UK, and TTY BiopharmDoxorubicinLiposomeMetastatic breast cancer, ovarian cancer, Kaposi’s sarcoma, multiple myeloma
FDA (2017) EMA (2018)VyxeosCelator/Jazz PharmaDaunorubicin/CytarabineLiposomeAcute myeloid leukemia
EMA (2009)MepactTakeda PharmaceuticalsMifamurtide MTP-PELiposomeOsteosarcoma
EMA (2010, 2013)NanoThermMagForce Nanotechnologies AGThermal ablation using a magnetic fieldIron oxide nanoparticlesGlioblastoma, prostate, and pancreatic cancer
EMA (2019)Hensify (NBTXR3)NanobiotixNo drug with radiotherapyHafnium oxide nanoparticleLocally advanced soft tissue sarcoma (STS)
EMA (2019)PazenirRatiopharm GmbHPaclitaxelNP-bound albuminMetastatic breast cancer, metastatic adenocarcinoma of the pancreas, NSCLC
Republic of Korea (2007)Genexol-PMSamyang BiopharmaceuticalsPaclitaxelPEG-PLA polymeric micelleBreast, lung, and ovarian cancer
EMA, European Medicines Agency, FDA, US Food and Drug Administration, NP, nanoparticle; NSCLC, non-small cell lung cancer; PEG, polyethylene glycol; PLA, phospholipase A.
Table 7. Phytochemicals used in anticancer therapy.
Table 7. Phytochemicals used in anticancer therapy.
Source of PhytochemicalChemical StructureExperimental ModelAction Mechanism of PhytochemicalRef.
Pharmaceutics 16 01169 i001
Erigeron breviscapus
Pharmaceutics 16 01169 i002
Scutellarin
Lipopolysaccharide-induced BV-2 microglial cells
  • Inhibit the production of proinflammatory mediators by inhibiting MAPK and I-kappa B kinase (IKK)-dependent NFκB signaling pathway
[119]
Pharmaceutics 16 01169 i003
Black cumin seed of Nigella sativa
Pharmaceutics 16 01169 i004
Thymoquinone
Lipopolysaccharide-induced BV-2 microglial cells
  • Inhibit NFκB-dependent neuroinflammation in BV2 microglia via activating the antioxidant response element (ARE)/nuclear erythroid 2 related factor 2 (Nrf2) antioxidant pathway
[120]
Pharmaceutics 16 01169 i005
Artocarpus lakoocha
Pharmaceutics 16 01169 i006
Oxyresveratrol
Human microglial cells
  • Exerts anti-inflammatory roles in IL-1β-induced human microglial clone 3 cells by inhibiting extracellular signal-regulated kinases (ERKs) on MAPK signaling cascades and the AKT serine/threonine kinase on PI3K signaling cascades
[121]
Pharmaceutics 16 01169 i007
Abies holophylla
Pharmaceutics 16 01169 i008
Terpenoids
Lipopolysaccharide-activated BV2 murine microglial cells
  • Exert neuroprotective and anti-inflammatory effects by decreasing production of nitrite and increasing the production of nerve growth factor through the inhibition of JNK phosphorylation, thereby inhibiting the secretion of proinflammatory cytokines such as IL-1β, IL-6, TNF, and prostaglandin E2, and effectively decreasing neuroinflammation
[122]
Pharmaceutics 16 01169 i009
Curcuma longa
Pharmaceutics 16 01169 i010
Curcumin
Head and neck squamous carcinoma cells, TLR4(−/−) or wild type of subarachnoid hemorrhage-induced mice model
  • Possesses antioxidant, anticancer, and anti-inflammatory effects
  • Decrease neuroinflammation post-subarachnoid hemorrhage by inhibiting toll-like receptor/NFκB signaling pathway and sequentially a shift of microglia M1 phenotype to M2, which promotes tumor survival
[11,123,124]
Pharmaceutics 16 01169 i011
Moringa oleifera seed
Pharmaceutics 16 01169 i012
Moringin
Autoimmune encephalomyelitis mice model
  • Normalize the Wnt/β-catenin signaling pathway
  • Upregulate β-catenin and inhibit glycogen synthase kinase-3, which leads to the regulation of FoxP3 and CD4 expression in T cell activation, inhibition of COX-2, IL-6, and IL-1β, decreased apoptosis, and increased expression of antioxidant Nrf2
[125]
Pharmaceutics 16 01169 i013
Citrus fruits
Pharmaceutics 16 01169 i014
Hesperetin
Lipopolysaccharide-stimulated BV-2 microglial cells
  • Inhibit nitric oxide, decrease expression of IL-1β, IL-6, and MAPK, downregulate ERK1/2 phosphorylation, suppress astrocyte and microglial cell activation, and ultimately decrease neuroinflammation
[126]
Pharmaceutics 16 01169 i015
Mallotus philippinensis
Pharmaceutics 16 01169 i016
Rottlerin
Phorbol 12-myristate 13-acetate (PMA)-induced rat brain astrocytes
  • Inhibit metastasis-related matrix metalloproteases by inhibiting PKC-mediated ROS, inactivating ERK1/2, and suppressing the AP-1/c-Fos signaling pathway, which suppresses astrocyte migration in phorbol-12-myristate-13-acetate-induced rats
[127]
Pharmaceutics 16 01169 i017
Soy
Pharmaceutics 16 01169 i018
Genistein
Hepatectomy model of nude mice bearing human hepatocellular carcinoma xenografts, colon HT-29, and breast MCF-7 cancer cells
  • Boost the inhibitory role of cisplatin
  • Protect against tumor metastasis and recurrence following curative hepatectomy
  • Lower the dose requirement of cisplatin as well as improving anticancer activity in many malignancies, including prostate, lung, breast, and pancreatic cancers
[12,13,128]
Table 8. Combined application of nanotechnology with phytochemicals in clinical trials for anticancer therapy.
Table 8. Combined application of nanotechnology with phytochemicals in clinical trials for anticancer therapy.
Nanomedicine NamePhytochemical in NanomedicineDelivery SystemTarget CancerTarget PopulationEffect of Nanomedicine on Anticancer TherapyTreatment StageRef.
NSBMangiferringNPsBreast cancerFemale patients with stage IIIA or IIIB of breast carcinomaPatients who received nanomedicine alongside the standard care had a 100% clinical benefit rate when compared to those who only received the standard care; only one patient showed severe adverse effectsPilot preclinical trial[129]
CRLX101CamptothecinCyclodextrin-containing polymer NPsGastric, gastroesophageal or esophageal cancerPatients with gastroesophageal, esophageal, or gastric cancer who are on at least one line of systemic therapyDownregulation of tumor indicators such as topoisomerase I and carbonic anhydrase IX Phase II clinical trial[130]
CRLX101CamptothecinCyclodextrin-containing polymer NPsRectal cancerAdult patients with T3–4N0 or T1–4N+ of rectal cancerAsymptomatic lymphopenia was recorded with a high dose of the drug; downstaging occurred in 69% of patients; pathologic complete response was achieved in 19% of patients overall and 33% of patients at the weekly maximum tolerated dosePhase Ib/II clinical trial[15]
CRLX301DocetaxelCyclodextrin-containing polymersAdvanced or metastatic prostate and breast adenocarcinomaPatients with prostate or breast adenocarcinomaFound 19.4% of clinical benefit rate; presented some pharmacokinetic advantages over docetaxelPhase I/IIa clinical trial[131]
Pm-PacPaclitaxelPolymeric micellar NPsNSCLCPatients with advanced NSCLC without pleural metastasisIncreased progression-free survival and overall survival of patientsPhase III clinical trial[132]
UANLUrsolic acidNanoliposomesAdvanced solid tumors including non-Hodgkin’s lymphoma, Hodgkin’s lymphoma, Hepatoma, and gastric cancerHealthy volunteers and patients with advanced solid tumorsThey tested only pharmacokinetic parameters and safety; no accumulation with repeated doses of UANL; no adverse event in patients who received 37 mg/m2 of UANL; no adverse effect after the provision of the larger dosesPhase I clinical trial[133]
gNP, gold NP; NP, nanoparticle; NSCLC, non-small cell lung cancer; UANL, ursolic acid-loaded nanoliposomes.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kim, J.H.; Dareowolabi, B.O.; Thiruvengadam, R.; Moon, E.-Y. Application of Nanotechnology and Phytochemicals in Anticancer Therapy. Pharmaceutics 2024, 16, 1169. https://doi.org/10.3390/pharmaceutics16091169

AMA Style

Kim JH, Dareowolabi BO, Thiruvengadam R, Moon E-Y. Application of Nanotechnology and Phytochemicals in Anticancer Therapy. Pharmaceutics. 2024; 16(9):1169. https://doi.org/10.3390/pharmaceutics16091169

Chicago/Turabian Style

Kim, Jin Hee, Boluwatife Olamide Dareowolabi, Rekha Thiruvengadam, and Eun-Yi Moon. 2024. "Application of Nanotechnology and Phytochemicals in Anticancer Therapy" Pharmaceutics 16, no. 9: 1169. https://doi.org/10.3390/pharmaceutics16091169

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

Article Metrics

Back to TopTop