Next Article in Journal
Development and Optimization of Nigella sativa Nanoemulsion Loaded with Pioglitazone for Hypoglycemic Effect
Next Article in Special Issue
Muscle-like Scaffolds for Biomechanical Stimulation in a Custom-Built Bioreactor
Previous Article in Journal
The Composites of Polyamide 12 and Metal Oxides with High Antimicrobial Activity
Previous Article in Special Issue
Chitosan Hydrogels Based on the Diels–Alder Click Reaction: Rheological and Kinetic Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Polymer-Based Hybrid Nanoarchitectures for Cancer Therapy Applications

1
Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan 173229, India
2
Department of Chemistry, Changwon National University, Changwon 51140, Korea
3
Laboratory of Biological Active Macromolecular Systems, Institute of Bioorganic Chemistry, Uzbekistan Academy of Sciences, Tashkent 100125, Uzbekistan
4
Department of Pharmaceutical Chemistry, Tashkent Pharmaceutical Institute, Tashkent 100015, Uzbekistan
5
State Center for Expertise and Standardization of Medicines, Medical Devices, and Medical Equipment, State Unitary Enterprise, Tashkent 100002, Uzbekistan
6
Department of Chemistry, School of Physical and Mineral Sciences, University of Limpopo, Polokwane 0727, South Africa
7
Department of Physics, Faculty of Science, University of Zabol, Zabol 538-98615, Iran
8
Department of Chemistry, College of Natural Sciences, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Korea
*
Authors to whom correspondence should be addressed.
Polymers 2022, 14(15), 3027; https://doi.org/10.3390/polym14153027
Submission received: 14 June 2022 / Revised: 17 July 2022 / Accepted: 19 July 2022 / Published: 26 July 2022
(This article belongs to the Special Issue Polymers for Biomedical Engineering and Applications)

Abstract

:
Globally, cancer is affecting societies and is becoming an important cause of death. Chemotherapy can be highly effective, but it is associated with certain problems, such as undesired targeting and multidrug resistance. The other advanced therapies, such as gene therapy and peptide therapy, do not prove to be effective without a proper delivery medium. Polymer-based hybrid nanoarchitectures have enormous potential in drug delivery. The polymers used in these nanohybrids (NHs) provide them with their distinct properties and also enable the controlled release of the drugs. This review features the recent use of polymers in the preparation of different nanohybrids for cancer therapy published since 2015 in some reputed journals. The polymeric nanohybrids provide an advantage in drug delivery with the controlled and targeted delivery of a payload and the irradiation of cancer by chemotherapeutical and photodynamic therapy.

Graphical Abstract

1. Introduction

Cancer is an affliction on societies all around the world and cancer-related deaths have been continuously rising. According to GLOBOCAN, the year 2012 observed 14.1 million cases of cancer and 8.2 million cancer-related deaths, which increased to 18.1 million cases and 9.6 million deaths in 2018. As per the report published in 2021, about 19.3 million cases and 10 million cancer-related deaths occurred in the year 2020 [1,2,3]. Cancer has three major treatments: surgery, radiotherapy, and chemotherapy. Surgery is the oldest method for cancer treatment and was used even before the use of antiseptics and anesthetics; radiotherapy, though it is a modern therapeutic method, is highly localized, so chemotherapy is considered the most successful solution for cancer [4,5,6]. However, multidrug resistance (caused by resistance to drugs by cancer cells) and undesired cytotoxic actions on healthy cells are some of the problems faced in chemotherapy. More problems, such as the lack of selectivity, insufficient drug concentration, and drug degradation, also limit the full exploitation of chemotherapeutic agents for cancer therapy. It can be solved to some extent by using a variety of formulations, such asliposomes, micelles, etc., but better and modified formulations are currently needed [7,8,9]. Other cancer treatments involve gene therapy and peptide therapy, where both peptides and genes target the cancer cells with minimal to no impact on the healthy cells [10,11]. Even after some promising cancer treatments, the lack of a proper delivery system limits their use [12,13].
Nanotechnology enables the manipulation of matter at the nanoscale, especially matter from the1 to 100 nm scale, and the 21st century has seen an emergence of nanotechnology [14]. Nanotechnology has led to the molecular age of medicine, and it is advantageous, as most of the subcellular structures are on the nanoscale, e.g.,the width of the cell membrane (6–10 nm), the dimensions of proteins (1–20 nm), and the width of DNA (approx. 2.5 nm). Nanotechnology has proved to be very effective in cancer drug delivery, as the nanocarriers protect the drug from degradation and help to supply the drug in therapeutic concentrationswhilealso avoiding drug delivery to normal cells [15,16]. As nanotechnology uses various nanoparticles (nanocarriers) as building blocks, the properties of these nanoparticles make it possible to exploit their use in drug delivery [17], and these nanoparticles can be used to make different nanoformulations, nanocapsules, and nanoemulsions [18]. The presence of multiple components in a nanoformulation leads to the formation of nanohybrids (NHs), and these NHs contain a variety of components in them, from nanoparticles to polymers to the targeting agent. Furthermore, these nanohybrids can overcome the drawbacks of conventional drug delivery and work synergistically to produce better activity [19,20,21]. They have also proved their effectiveness in gene therapy for cancer, and they can deliver a variety of genetic material, such asimmune activators, microRNA inhibitors, smallinterfering RNA, and plasmid DNA [22]. The nanohybrids, being very small particles, exhibit deep tumor penetration, and because they contain multiple components, they can be customized for specific cancers. They have also found their way into photodynamic therapy because they exhibit excellent photodynamic properties and can be triggered by external stimuli such asphotosensitization [23,24]. The interest in using polymers in nanoformulations is also growing, and its effectiveness is dependent on the properties of the polymer as well as its nanostructure [25]. Polymers provide the controllable release of the drug and adjustability in the release rate for hydrophobic and hydrophilic drugs [26]. We have presented the recent uses of polymers in the preparation of nanohybrids for cancer therapy in this review.

2. Polyethylene Glycol-Based Nanohybrids

PEG is a commonly used polymer and is considered a gold standard for polymer-based drug delivery, as it enhances the biocompatibility and hydrophilicity of inorganic nanomaterials [27,28]. The generalized structures of PEG-based nanohybrids are given in Figure 1. Zheng et al. prepared C3N4-based nanocomposites (PCCN) for photodynamic antitumor therapy aimed at the hypoxic tumor by light-driven water splitting. The nanocomposites were prepared by doping carbon dots on C3N4 (carbon nitride) (CCN) and were then modified by adding them into PpIX-PEG-RGD (amphipathic polymer) formed by mixing RGD (tumor-targeting motif), PEG, and protoporphyrin photosensitizer (PpIX). The oxygen levels and ROS levels increased in the in vitro studies with light irradiation in the hypoxic as well as in the normoxic tumors due to the water splitting properties of C3N4. It also performed well in the in vivo studies, although the drug accumulated in the cancer cells (4T1) in a large amount, and wasalso accumulated in the kidney, lung, spleen, heart, and liver in some amount [29]. Pandey et al. developed a new method for the preparation of Cu-Cy nanoparticles using copper chloride, cysteamine, and PEG-4000. The method was more economical, as both the stirring and heating time were reduced in it. On exposure to microwave and UV irradiation, these nanoparticles produced single-oxygen RO species. Alongside this, these particles were found to have X-ray luminescence and photoluminescence that has not been reported previously. The nanoparticles containing coumarin as a probe molecule produced remarkable anticancer activity viaROS production on the KYSE-30 cells. Although these nanoparticles produced a significant anticancer activity on the KYSE-30 cells, further studies are necessary to explore their full potential in anticancer therapy [30]. Nanohybrids were prepared by functionalizing an amphiphilic PEG polymer containing SBMA (2-(methacryloyloxy)ethyl]dimethyl-(3-sulfopropyl)ammonium hydroxide) on graphene oxide (GO) and then loaded with IR780 to improve breast cancer phototherapy. The PEG improved the colloidal stability of the GO and the IR780 improved the photothermal heating. The in vitro study displayed a 27% reduction in the cell viability of the breast cancer cells when exposed to the unloaded nanohybrids, but this increased to 80% when the nanohybrids loaded with the IR780 were used. Further in vivo studies could provide some clues to their effect on the living system and the possibility of their use in cancer therapy [31].
The surface-passivated photoresponsive carbon nanohybrid dots (CNDs) with PEG200 and doped with nitrogen were prepared. The DOX (doxorubicin)-loaded NPs(nanoparticals) were then tested for the photosensitive delivery of the drug in the MCF-7 cell line by NIR two-photon excitation (TPE). The prepared NPs showed a 78% death of the MCF-7 cells, more than the DOX when used alone. Though the CNDs produced very significant cytotoxicity in the MCF-7 cells, further exploration is required to know their effects and efficiency in the living system [32]. Zhu et al. synthesized polyhedral photosensitizer/oligomeric silsesquioxane(POSS)-crosslinked nanohybrids for enhancing the drug-loading capacity in photodynamic therapy. POSS was crosslinked to Ce6 (chlorin e6); PEG was modified on the surface of the NPs to form POSS-Ce6-PEG nanohybrids. Improved cellular uptake and anticancer activity under light irradiation were seen in the in vitro studies. Although PEG enhanced the aqueous dispersion and prolonged the circulation time, overall, a high loading capacity and a better chemical stability were observed in the NHs. The nanohybrids also produced good results in the in vivo studies with negligible side effects on the female balb/c mice [33]. Curcumin-loaded zinc and cobalt ferrite-functionalized nanohybrids coated with chitosan and PEG, respectively, were prepared and tested for their drug delivery potential on MCF-7 cells. Both PEG and chitosan were used as coating/capping agents. The prepared nanohybrids enhanced the anticancer, apoptotic, and drug delivery potential. A higher payload delivery and anticancer activity were observed in zinc ferrite nanohybrids coated with chitosan [34]. Graphene oxide-based nanohybrids decorated with amphiphilic prodrug (PP-SS-DOX) were prepared and tested for their drug delivery and efficiency in cancer therapy. The conjugation of DOX (doxorubicin) and mPEG-PLGA(PP) via a disulfide bond(-SS-) led to the formation of the prodrug PP-SS-DOX, and its addition on GO along with PEG-incorporated FA (targeting agent) formed the NHs. The final GO/PP-SS-DOX/PEG-FA nanohybrids produced the reduction-sensitive DOX release due to the resolution of –SS- in the tumor environment and the higher cytotoxicity than the nontargeting nanohybrids in B16, MCF-7, and A549 cells. PEG, however, provided aqueous stability, but an overall improved and targeted drug release was observed due to the incorporation of PEG-FA and PP-SS-. The in vivo studies supported the claims of the improved cytotoxicity, as the tumor volume was least in the GO/PP-SS-DOX/PEG-FA-administered mice in comparison to the GO/PP-SS-DOX and free DOX. Along with that, no significant systemic toxicity was observed, the NHs reduced the cardiotoxicity of the DOX by targeted delivery, and they were also found to be blood compatible (no significant hemolysis). Though these nanohybrids could be excellent targeted drug delivery carriers for anticancer therapy, further clinical studies are necessary to consider them as an option [35]. High-intensity focused ultrasound (HIFU) and thermosensitive cerasome (HTSCs) nanohybrids were prepared by assembling CFL (cerasome-forming lipid) with LTSLs (low-temperature-sensitive liposomes) that included MSPC (1- stearoyl-2-hydroxy-sn-glycero-3-phosphocholine), DSPE-PEG-2000 (1,2-distearoyl-sn-glycero-3- phosphorethanol amine-N-[methoxy(polyethyleneglycol)-2000]), and DPPC (1,2-dipalmitiyl-sn-glycero-3-phosphocholine). The prepared NHs showed good bioavailability, prolonged blood circulation, and were thermostable at 37 °C, but exhibited a thermosensitive burst release at 42 °C. The HTSCs were very biocompatible, as they produced little to no toxicity on the HUVEC, T cells, and BMDC cells. The hydrophilic and lipophilic DOX-loaded HTSCs produced significant MDA-MB-231 tumor inhibition in the tumor-bearing mice with theHIFU treatment, with minimal side effects. All these properties make HTSCs a suitable thermosensitive drug delivery carrier, but more exploration is needed to use them in cancer therapy [36].
Titanate nanotube (TiINts)-based nanohybrids were prepared by coating titanate nanotubes with APTES (a siloxane), and then were coupled with the PEG3000 polymer (heterobifunctionalized) and Au@DTDTPA NPs (dithiolated diethylenetriaminepentaacetic acid acid-modified gold nanoparticles). The NHs were grafted with DTX (docetaxel). The nanohybrids enhanced the efficiency of the radiotherapy on the tumor-bearing animals by two folds. The loaded NHs (TiONts-AuNPs-PEG3000-DTX) were not as cytotoxic as the drug DOX, but they produced a prolonged cytotoxic activity on the PC-3 cell line. In vivo studies showed a promising decrease in the growth of the tumor with radiotherapy and a long retention time in the tumor (at least 20 days) on the PC-3 xenograft [37]. Phyto-liposomes loaded with PEGylated DOX-STS (STS–stigmasterol) and modified with HA (hyaluronic acid) nanohybrids were produced and tested for their anticancer potential on MCF-7 and MDA-MB-231 cells. The NHs performed better against MDA-MB-231. The prepared NHs showed a pH-dependent drug release, a better internalization in CD44-overexpressed cells, and accumulated more in the CD44-overexpressed MDA-MB-231 xenograft and less in the MCF-7 cells. The NHs produced better activity than the DOX alone, which was ensured by the longer circulation time due to the presence of PEG. The addition of HA led to the sustained release and higher release in the acidic tumor environment [38]. DTX (docetaxel) was functionalized on the titanate nanotube to form TiONts-DTX. For this, the TiONts surface was first modified with 3-aminopropyl triethoxysilane and then with PEG; furthermore, PMPI (p-maleimidophenyl isocyanate)-modified DTX was attached to form TiONts-DTX. These NHs were then modified with the chelating agent DOTA ((1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid) to form TiONts-DTX-DOTA.The cytotoxicity studies against 22Rv1 (androgen-dependent prostate cancer) cells showed that the NHs (TiONts-DTX) produced significant cytotoxicity.The IC50 value of the free DTX (3.5 nM) was better than the NHs, but the NHs kept the drug in the tumor site for a longer time and produced a more effective treatment than the free DOX. The NHs without the drug were nontoxic, making them safer to use. The in vivo study also supported the cytotoxic effect of the NHs, with slow tumor growth in the administered mice and with no significant effects on the vital organs (lung, kidney, liver, tumor, and bladder) [39]. Du et al. prepared self-assembling PEGylated pure dual drug-based nanohybrids (DNH) containing OXA (oxaliplatin) and 1-MT (1-methyl-D-tryptophan). The prepared NHs were then camouflaged with NKCM (natural killer cell membrane) to produce NK-DNH. The prepared NK-DNH showed a pH and intracellular reduction-based drug release and a better cellular uptake than DNH in the 4T1 cells. These NHs (NK-DNHs) also produced better activity than the OXA and DNHs. The anticancer potential of the NHs (NK-DNHs) was also confirmed by the in vivo studies performedon the 4T1 tumor-bearing BALB/c mice model, where the NHs reduced the tumor volume more than OXA and DNH. Although the HNs showed accumulation in the liver and spleen, they exhibited good anticancer activity on the 4T1 cells. Further modifications and studies are necessary to consider these NHs for cancer therapy [40]. Zhao et al. prepared a hollow-structured polymer–silica nanohybrid (HPSN) for the dual delivery of palladium phthalocyanine (PdPc) for photothermal therapy (PTT) and paclitaxel (Pax) for chemotherapeutic (CHT) effect. The HPSNs were formed by the self-assembly of the silica precursor and pluronic F108 with the PEG layer on it; cyclohexane was used for the hollowing of the structure. The biodistribution study showed that the HPSNs were accumulated most in the liver and then in the tumor, but the off-targeted HPSNs could be rapidly cleared through the hepatobiliary pathway from the body. The in vitro cytotoxicity studies on the HeLa cells showed that the combined PTT/CHT therapy of the NHs (Pax/PdPc@HPSNs+laser) produced a maximum reduction in the cell viability and the least recurrence compared to the other PdPc@HPSN, Pax@HPSN, and PdPc@HPSN+laser. A significant reduction in the tumor volume was also observed in the S180 tumor-inoculated nude mice in the case of the NHs (Pax/PdPc@HPSNs+laser), with a reduced recurrence as compared to the other individual therapies [41].
MSN (mesoporous silica nanoparticle)-based nanohybrids MSN-hyd-MOP (MOP = morpholine-4-acetyl-polyethylene glycol-b-polylactide) containing a charge reversal polymer connected via a hydrazone linker were prepared. The MSNs were taken as a core and the pH-responsive polymer as a shell. These NHs were further loaded with DOX (doxorubicin) and tested for their anticancer activity. The prepared nanohybrids showed a charge reversal and a pH-dependent drug release. The NHs are safe to use and the in vitro studies on the HepG2 cells showed that the NHs loaded with DOX (MSN-hyd-MOP-DOX) were not as cytotoxic as the standard drug doxorubicin when incubated for 24 h. However, the cytotoxicity of the loaded NHs was increased when the incubation time was increased to 48 h because the acidic excretion from the HepG2 cells was not enough for the charge reversal. Further modification in the formulation and further studies are necessary to produce a better agent for cancer therapy [42]. DOX (doxorubicin)-loaded Cu-Se (copper–selenium) NPs functionalized with PEG (polyethylene glycol) were prepared to enhance the aqueous solubility of the DOX and improve the efficiency of the photothermal therapy. The final PEG@Cu-Se+DOX nanohybrids were then tested for their activity on prostate cancer (LNCaP and DU145) cell lines. The NHs enhanced the aqueous solubility of the DOX and were highly biocompatible (did not produce any significant hemolysis). Cu-Se+DOX NHs had better cytotoxicity on both DU145 and LNCaP cells than the PEG@Cu-Se+DOX NPs. Further enhancement in the proposed structure could improve its effectiveness, and more studies would describe the full potential of these NHs [43]. Light-responsive nanohybrids (pGO-CuS/ICG) by combining PEGylated GO (graphene oxide), CuS (copper sulfide), and ICG (indocyanine green) as the NIR phototherapeutic agents were prepared. The in vitro studies of the prepared NHs were done on MCF-7 cells. The NHs entered the cells via the passive transmembrane pathway and produced photodynamic and photothermal effects on the NIR irradiation between808 nm and 940 nm. A maximum reduction in the cell viability of the MCF-7 cells was observed when the NHs were induced to produce PTT (at NIR laser 940 nm, 4 W/cm2, for 5 min) followed by PDT (at NIR laser 808 nm, 2 W/cm2, for 5 min). The ICG itself heavily reduced the cell viability on PDT (NIR laser 808 nm, 2 W/cm2, 5 min) [44]. Ma et al.synthesized nanohybrids for photochemotherapy by using rGO (reduced graphene oxide) sheets with Auclusters grown on them. The 3-(3-phenylureido) propanoic acid (PPA)–PEG (PPEG) was further introduced to form rGO/Au/PPEG nanohybrids. The DOX (doxorubicin) was loaded onto these NHs to check their photochemotherapeutic effect. A higher drug release was seen in the acidic environment of the tumor. The NHs produced more toxicity on the NIR irradiation with the rising temperature in response to the increasing concentration of the NHs and the intensity of the NIR irradiation. A strong reduction in the cell viability was observed in the case of the loaded NHs at a higher concentration ofthe NIR irradiation. Although the NHs produced a good anticancer activity, further alterations and animal and clinical studies could produce a better therapeutic agent for cancer [45].

3. Carbohydrate Polymer-BasedNHs

3.1. Carboxymethyl Cellulose-Based Nanohybrids

CMC is a cellulose derivative and has good biodegradability, biocompatibility, low cytotoxicity, and cell adhesion properties. CMC materials are widely used in drug delivery due to their high stability, reliability, pH sensitivity, drug binding capacity, long half-life, and biocompatibility [46,47,48,49]. Mansur et al. prepared quantum dot–biopolymer–drug nanohybrids and performedin vitro studies for their anticancer drug delivery and glioblastoma-targeting capability. ZnS fluorescent-QDs, carboxymethyl cellulose (CMC), and doxorubicin (DOX) formed ZnS@CMC-DOX nanohybrids. The blank nanohybrids did not produce any cytotoxicity in the healthy as well as glioblastoma cells. Free DOX showed more accumulated release percentage in an acidic environment than in a neutral one, whereas the NHs produced a similar accumulated drug release percentage in both the acidic (pH5.5) as well as the neutral pH (pH7.2). Drug-loaded nanohybrids produced a significant decrease in cell viability on prolonged exposure, but free DOX produced better activity than the NHs in the same exposure time [50]. Leonel et al.prepared magnetic (iron oxide) Fe3O4, MION, and cobalt-doped (CoxFe3−xO4, Co-MION) nanoparticles functionalized with CMC (carboxymethyl cellulose) core–shell nanohybrids and tested them for anticancer activity on HEK 293T and U87 cells. The magnetic nanohybrids (MION-CMC) acted as nanoheaters and killed U87 (glioblastoma) cells by hyperthermia under the influence of an alternating magnetic field at higher concentrations. However, further studies could help to understand the effect of these NHs on the biological system [51]. Carboxymethylcellulose-coated nanohybrids CMC/5-FU@MOF-5 were prepared by encapsulating the drug 5-FU (fluorouracil) in zinc-based MOF-5 (metal-organic framework), and then pH-sensitive CMC (carboxymethyl cellulose) was used as a protective polymer. The drug showed a sustained release in the GIT conditions in the in vitro studies. Along with that, the NHs produced some amount of toxicity against the HeLa cell line. Though, further modification in the formulation could improve the cytotoxic activity of the NHs [52]. Carvalho IC and group in 2019 prepared Poly-L-arginine and L-cysteine-functionalized CMC (carboxymethyl cellulose) conjugates (CMCelPolyArg and CMCelCys) and then capped them onto fluorescent AgInS2 QDs (AIS-QDs). The final nanohybrids (AIS-CMCel_Cys and AIS-CMCel_PolyArg) showed no cytotoxic effects in the in vitro studies on U-87 MG and HEK 293T cells. Both the NHs were successfully internalized by the U-87 MG and HEK 293T cells; however, a higher internalization was exhibited in the U-87 MG. The NHs were very biocompatible, as they were nontoxic and were effectively internalized in both the cell lines, but their effectiveness in brain cancer remained unexplored [53]. However, Carvalho IC and group in 2021 prepared a similar type of nanohybrid structure with some modifications in the NHs consisting of KLA (mitochondria-targeting peptide)-conjugated CMC_Cys (cysteine-modifiedcarboxymethylcellulose) with fluorescent AgInS2 and AIS (semiconducting core) for targeting glioblastoma (U-87 MG) cells. The NHs with CMC_Cys-KLA produced 70 times better activity than the NHs with just CMC_Cys and were more cytotoxic to the U8–7 MG cells. These nanohybrids were more lethal than DOX and they also produced a protective effect on the healthy cells. Although these NHs were good at killing glioblastoma cells, more studies could determine their reliability for cancer treatment [54]. Mansur et al. synthesized CMC (carboxymethyl cellulose)-functionalized Cu-In-S/ZnS and Cu-In-S QDs (ZCIS@CMC) and (CIS@CMC). FA (folic acid) was further conjugated along with the DOX onto these NHs to form ZCIS@CMC-FA-DOX. The NHs were found to be fluorescent and were tested for their anticancer potential on TNBC (FRα+), MCF7 (FRα−), and HEK 293T cells. The unloaded NHs were nontoxic, making them highly biocompatible; however, the loaded NHs produced better activity than the DOX on TNBC (triple-negative breast cancer), whereas it was less active on HEK 293T and MCF-7 cells, representing the specific targeting of the NHs. More studies on the nanostructures could open new gateways for the modification and exploitation for cancer therapy [55]. DOX (doxorubicin)-conjugated bifunctional magnetopolymersomes (MION@CMC-DOX) were prepared, and they consisted of CMC (carboxymethylcellulose) and MION (iron oxide nanoparticles). The in vitro cytotoxic study was done on U87 and HEK 239T cells. For comparison, theloaded NHs (MION@CMC-DOX) produced cytotoxic activity, whereas the unloaded MION@CMC had a low toxicity. Although, in the presence of an alternating magnetic field, the hyperthermic effect killed the cancer cells, even in the presence of an alternating magnetic field the loaded NHs (MION@CMC-DOX) were more lethal than the MION@CMCs. Though the NHs successfully killed glioblastoma cells, more modification and studies could make them an excellent agent for cancer therapy [56]. The generalized structures of the CMC-based nanohybrids are given in Figure 2.

3.2. Chitosan-Based Nanohybrids (Carbohydrate-Based)

Chitosan is a polysaccharide that supports many living organisms, and it is used in drug delivery because of its properties, such asefflux pump inhibition, permeation enhancement, transfection, in situ gelations, mucoadhesion, and controlled drug release [57,58,59,60].The pH- and temperature-responsive nanohybrid carriers consisting of SWNT and DMAA-TMC (Dimethyl acrylamide–trimethyl chitosan) copolymer grafting on SWCNT were designed and tested for their drug release potentials using molecular dynamic simulation. The carriers loaded with paclitaxel and doxorubicin showed a pH-responsive drug release in the acidic pH in the simulation studies [61]. Jia et al. prepared FA-containing chitosan oligosaccharide-grafted disulfide-containing polyethyleneimine copolymer-based silica nanohybrids against multidrug-resistant breast cancer cells. The NHs were fabricated with P-shRNA and paclitaxel for their codelivery. The nanohybrids showed pH-responsive drug release and redox-sensitive PshRNA release, alongside preventing premature drug loss and gene degradation before entering the cancer cells. The FA helped in the selective internalization of the NHs in the cancer cells and the in vitro studieson the MCF-7/ADR cells showed effective delivery and gene silencing. The NHs inhibited MCF-7/ADR proliferation and paclitaxel resistance was completely reversed with a multidrug-resistant reversion index of 50.59. Further in vivo studies can solidify these results [62]. The 5-FU (5-fluorouracil)-loaded nanospheres with Zn-MOF (zinc-based metalorganic framework) coated with chitosan and graphene oxide (5-Fu@CS/Zn-MOF@GO) were prepared and tested for their drug delivery and cytotoxic potential on breast cancer cells. The NHs showed a 45% loading capacity and exhibited a sustained and pH-sensitive release of 5-FU with a higher drug release in the acidic pH (pH 5.5). The blank NHs showed a good biocompatibility and biodegradability, whereas the loaded NHs reduced the cell viability of the MDA-MB-231 cells to 41.2%. Though the NHs produced some amount of cytotoxicity, the free 5-FU produced better toxicity than the NHs. Therefore, further modification and studies can improve the effectiveness of these NHs [63]. Nanohybrids containing chitosan-based hydrogels conjugated with cysteine and zinc oxide embedded in it were prepared. The hybrids were then loaded with Naringenin (NRG) and showed a loading capacity of 86.09%. The NHs exhibited a pH-sensitive drug release with a high release in the acidic pH (pH 5), while the NGR release was due to polymer erosion and non-Fickian diffusion. The NHs were found to be very biocompatible in the L929 cell viability assay and were found to double the cytotoxicity of the NRG in (A431) human skin cancer cell lines [64].
Nanohybrids containing a graphene oxide-Ag/chitosan-based bio–nano composite hydrogel bead loaded with DOX (doxorubicin) (CH/GO-Ag/DOX) were synthesized. Electrostatic interactions, hydrogen bonding, and a high number of pores provided a high DOX loading percentage in the NHs, and a prolonged and controlled drug release was observed. The cell viability assay on the SW480 cells showed that the blank NHs (CH/GO-Ag) produced no significant cytotoxicity, whereas the loaded NHs (CH/GO-Ag/DOX) produced some amount of toxicity on the SW480 cells, but the NHs were not as cytotoxic as the free DOX [65]. Luo et al.synthesized semi-interpreting PNIPAM (poly(N-isopropylacrylamide))/CMCS (carboxymethyl chitosan)/multiwalled CNT (carbon nanotubes) nanohybrid hydrogels and loaded them with DOX (doxorubicin). The NHs showed a pH- and temperature-responsive drug release. The in vitro studies showed that the blank NPs were nontoxic, and the drug-loaded NPs had the same activity as that of the DOX. Further studies are necessary to exploit these NHs for cancer therapy [66]. CNT-based (carbon nanotube) magneto/pH-responsive hybrid nanogels were prepared by coating the MnFe2O4 with chitosan and mixing them with f-CNTs. These NHs were then loaded with doxorubicin to form Chi-Mn MnFe2O4/CNT-DOX. Chitosan was used as the polymer in the NHs and chitosan-containing NHs had more drug loading capacity. These NHs were then tested for their anticancer activity on U-87 cells. The drug release was higher in the acidic medium and was even higher in NHs coated with chitosan, expressing that the chitosan was important for the pH-sensitive drug release. The NHs produced significant cytotoxicity on the U-87 cells, but further modifications in their structure can improve their effectiveness and animal studies can help to understand their effect on the biological system [67]. Magnetic polymeric nanohybrids, Sr, Fe-Hap (iron- and strontium-co-doped), and Sr-Hap (strontium-doped) hydroxyapatite nanoparticles were formed for the delivery of DOX (doxorubicin) with a coating of a chitosan derivative functionalized with cyclodextrin and complexed with platinum. The NHs displayed superparamagnetic and paramagnetic properties and a high loading capacity for DOX, followed by a sustained released at the physiological pH. The NHs were tested for their cytotoxicity on bone, liver, cervical, and lung cancer cell lines; they showed a dose- and time-dependent cytotoxicity, and an efficient cytotoxicity was seen in the case of the MG-63 cell lines. The metastatic anticancer study was performed on MG63-inoculated mice. Enhanced activity was observed in the case of the DOX-loaded Sr, FeHAp. The NHs proved to be good nanocarriers for cancer therapy and further clinical studies can determine their possible use [68]. A generalized structure of chitosan-based nanohybrids is given in Figure 3.

3.3. Pullulan-Based NHs

Pullulan is a biopolymer, and it is highly water soluble, biodegradable, biocompatible, noncarcinogenic, and nontoxic. Folate-hydrophobic-quaternized pullulan (polymer)-functionalized gold nanohybrids loaded with camptothecin (CPT-GNHs@FHQ-PUL) were prepared, and the NHs showed a pH-sensitive drug release thatproduced 2.82 times more cytotoxicity than the camptothecin on the human lung cancer cell line (Chago-k1), while they were less toxic against the normal cells (Wi-38). The internalization was mediated by the folate receptor and the NHs produced apoptotic cell death after administration [69].

3.4. Β-Glucan-Based NHs

Β-glucan from mushrooms are food ingredients with multiple biological properties that are also biocompatible and have a stable structure. These properties can be exploited in drug delivery. Li et al.integrated AuNRs (plasmonic gold nanorods) and Glu ((PTR) Pleurotus tuber-regium sclerotial β-glucan) to produce AuNR-Glu (gold nanorods coated with β-glucan) nanohybrids and tested them for their potential use in photothermal therapy on cancer cells. The prepared NHs exhibited good colloidal and photothermal stability with a low cytotoxicity. However, the reduction in the cell viability of the MCF-7 cells was higher in the case of the NHs than the AuNRs whenthe irradiation of the NIR-II laser with a power of 0.75 W/cm2 was struck, as the nanohybrid exhibited a photothermal effect [70]. The NHs were also studied for SW480 and HT-29 cells, and a similar kind of result was observed, with the increased cancer-cell-killing ability of the NHs on NIR irradiation. The results were also supported by the in vivo studies, as no significant increase in the tumor volume was observed in the HT-29 xenografts bearing athymic nude miceadministered with the NHs + laser irradiation of 1064 nm (1.0 W/cm2 for 5 min) [71].

4. Poly(lactic-co-glycolic Acid)-Based Nanohybrids

PLGA is an attractive option in drug delivery due to its biodegradable and biocompatible nature [72]. A generalized structure of the PLGA-based nanohybrids is given in Figure 4. Folic acid-functionalized andSPION (superparamagnetic iron oxide nanoparticles)/doxorubicin-loaded polymeric (poly(lactic-co–glycolic acid)) gold porous shell nanohybrids DOXO/SPION-PLGA NPs (DXSP-PGNH) were prepared for multifunctional use (for photothermal therapy, NIR-triggered drug release, magnetic- and folic-acid-targeted drug delivery, and MR imaging). These NHs were then tested for their potential in cancer theranostics on the HeLa cell line. The polymeric nanohybrids exhibited NIR-based drug release and induced localized cell death. The nanohybrids exhibited improved MRI properties and the FA receptor-based NPs (DXSP-PGNH-FA) showed a specific targeting of the cancer cells. The nanohybrids could be guided to tumors by magnet-assisted targeting. The high likelihood of cell death also increased in the case of the loaded nanohybrids supplied with the NIR, and it was better than the free DOXO [73]. Ray et al. intercalated MTX (methoteraxate) with Mg-Al-LDH (layered double hydroxide) and PLGA (poly (lactic-co-glycolic acid))-coated nanohybrids and tested their potential against osteosarcoma (MG-63 cell line). The nanohybrids produced significant anticancer activity in the in vitro studies, although the activity of the NHs was less than that of the MTX, and with the increase in the incubation time, the activity of the NHs became better than the NHs. In comparison to bare MTX, the NHs showed a longer retention in the in vivo studies on the New Zealand White rabbit model [74]. Jain et al. prepared NHs containing PLGA nanoparticles functionalized with HA (hyaluronic acid) and loaded with NIC (niclosamide). The ((NIC-PLGA NP)HA) NHs were highly cytocompatible and produced better cytotoxicity on the MDA-MB-231 than on the L929 cells, thus determining its cancer-targeting capabilities. Further in vivo studies are necessary to understand and estimate the possibilities of their use in therapeutics [75].

5. Polypyrrole-Based Nanohybrids

Polypyrrole is a polymer that can incorporate drugs and enzymes and is biologically compatible [76]. Cai et al. prepared controlled release Fe-soc-MOF@Polypyrrole core–shell nanohybrids. Fe-soc-MOF was prepared by the LSS method (liquid–solid–solution) and then integrated with polypyrrole (PPy) to form the Fe-soc-MOF@PPy nanohybrids for photothermal therapy. Laser irradiation at 808 nm triggered the nanohybrids to exhibit a photothermal effect in the cancer cells. The NHs produced a concentration-dependent darkening of the MR images and an incubation time-dependent cellular uptake. The in vitro study on the L929 cells and the 4T1 cells showed that there was no significant cytotoxicity on both the cell lines, even after the incubation for 48, though a concentration-dependent reduction in the cell viability was seen under the influence of the NIR at 808 nm. The in vivo studies on tumor-bearing mice confirmed the induced hyperthermic effect under the NIR irradiation. The tumor volume under NIR remained at a minimum and there was no significant reduction in the body weight. The NHs showed accumulation in some amount in the spleen and liver and a minor accumulation in the heart, kidney, and lungs, along with a clearance rate of 39.4% per week. These NHs have good anticancer potential, but further modification in them could produce better NPs [77]. Pillararene nanovalves and polypyrrole@UiO-66 nanohybrid multifunctional supramolecular materials (PPy@UiO-66@WP6@PEI-Fa) for targeted chemophotothermal therapy were designed and were then loaded with 5FU. The NHs were functionalized with FA-incorporated PEI for selectivity. The pH/temperature-responsive action of the nanoparticles allowed the drug release at the targeted site for 4 days. Then, the nanoparticles exhibited excellent synergistic chemophotothermal therapy for cervical cancer (HeLa) in the in vitro studies, whereas no significant effect was observed on the LO2 cells. The loaded NHs were moreactive than the blank NHs. The relative tumor volume was minimum in the in vivo studies, with no significant loss in body weight even after 12 days. This study could be a roadmap to a new anticancer agent [78]. A generalized structure of polypyrrole-based nanohybrids is given in Figure 5.

6. Peptide-BasedNHs

6.1. Casein-Based Nanohybrids

Casein is a dietary protein and is a promising drug delivery carrier, as it is easily available and has properties such as pH-dependent swelling behavior. Purushothaman et al. synthesized biotin-conjugated protein–inorganic (magnetic casein–calcium ferrite)nanohybrids with cinnamaldehyde loaded in them. The NHs showed a pH-based drug release in the acidic pH of the tumor microenvironment, and the superparamagnetic properties ensured a magnetically modulated drug delivery and a fast drug release. The NHs showed more enhanced activity than cinnamaldehyde on both the L929 and A549 cell lines, with the activity being 18 times greater than the drug. Further studies can help explore the full potential of the NHs [79]. Hesperidin (extracted from citrus peel)-loaded and progesterone-conjugated Casein-CaFe2O4 magnetic nanohybrids for ovarian and breast cancer were prepared. The NHs showed a higher drug encapsulation (encapsulating 89.54% of the drug) and alsoexhibited a magnetically induced and pH-sensitive drug release (high release in the acidic pH of 1.2–5.4) in the acidic environment of the tumor. The drug is released by the Fickian diffusion mechanism. The cytotoxicity of the NHs was thirty times that of hesperidin on both breast (MDA-MB-231) and ovarian (SKOV-3) cancer cell lines. The NHs were highly biocompatible, as they produced little to no effect on the L929 cells, but further animal studies could determine their effectiveness in the living system [80]. A generalized structure of the casein-based nanohybrids is given in Figure 6.

6.2. Pectin-Based NHs

Pectin is a naturally occurring polymer (a polysaccharide) that has the potential to be used in drug delivery and can be exploited for its properties. Hussien et al.prepared pectin-conjugated magnetic graphene oxide nanohybrids for the delivery of paclitaxel (PTX). The prepared nanohybrids had a good drug loading capacity and a pH-sensitive drug release. The anticancer activity on the MCF-7 and L-929 cell lines suggested that the nanohybrids alone were noncytotoxic (neither on the L-929 nor on the MCF-7 cells), but the loaded NHs produced a significant amount of concentration-dependent cytotoxicity. Free PAX was more cytotoxic than the NHs, suggesting further improvements can be done to increase the toxicity of the NHs [81].

6.3. Albumin-Based NHs

GO-based nanohybrids containing bovine serum albumin grafted with folic acid (FA-BSA/GO) and loaded with DOX (doxorubicin) were prepared. The NHs produced a pH-sensitive and sustained release with a higher drug release in the acidic environment (pH 5.0) of the tumor. The NHs also showed a high drug loading of 437.43 µg of DOX per mg of FA-BSA/GO. A higher cellular uptake was observed with low hemolysis, making the NHs compatible for animal studies. The NHs produced a concentration and incubation time-dependent anticancer effect on the MCF-7 and A549 cells. The NHs containing FA produced better activity than the NHs lacking FA [82].

6.4. Gelatin-Based NHs

Gelatin is a polymer that is excellent for drug delivery, as it is cheaply available, easily modifiable, nontoxic, biodegradable, and biocompatible. Cu-MOF/MTX@GM nanohybrids containing Cu-MOF (copper-based metal–organic framework) loaded with MTX (methotrexate) and inserted in GM (gelatin microsphere biopolymer) were prepared. A better drug release was seen in the acidic condition of the tumor. The loaded NHs produced better activity than the MTX in the in vitro cytotoxicity assay on the MCF-7 cells, while the unloaded NHs were less toxic than the MTX [83].

7. Poly(ethyleneimine)-Based Nanohybrids

PEI (Poly(ethyleneimine)) is a cationic water-soluble polymer that assists endosomal release and can deliver the load to the nucleus. A generalized structure of poly(ethyleneimine)-based nanohybrids is given in Figure 7. Aliabadi et al.prepared pH-responsive polymer graphene oxide (GO) nanohybrids and used the anticancer drug 5-fluorouracil as a carrier. The planer GO was taken for a support to encapsulate the 5-fluorouracil with tannic acid and poly(ethyleneimine). The resulting nanohybrid showed a pH-responsive drug release with a higher drug release in the acidic medium due to the degradation of the NHs. The NHs also showed a controlled release in the acidic environment, though further studies could explain the anticancer effect of the NHs [84]. The nanohybrids of MFNPs(mixed ferrite nanoparticles) and a smart block copolymer were prepared by modifying citrate-stabilized MFNPs (CA-MFNPs) with dual-responsive pluronic F-127 and PEI-crosslinked copolymer. RITC (isothiocyanate) and FA (folic acid) were attached to the NPs for cancer targeting and DOX (doxorubicin) was entrapped in it. The final NHs (DOX-FA-Poly-MFNPs) had a high payload capacity and a pH-responsive drug release both in the acidic pH and at 37 °C. The NPs also showed a FAR (folic acid receptor)-specified endocytosis in HeLa (HFAR) cells with a significant cytotoxicity; however, the NPs were nontoxic without the DOX, making them biocompatible, though further studies could help in understanding the future perspective for these NHs [85]. Huang et al.synthesized magnetic ternary nanohybrids for gene delivery in cancer therapy by using superparamagnetic iron oxide NPs decorated with hyaluronic acid and biodegradable cationic material to produce TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) by receptor and magnetic force dual targeting. The in vitro studies showed an enhanced uptake of MTNs by human mesenchymal cells (hMSCs) via magnetic force/CD44 mediation. The nanohybrids displayed strong anticancer activity on the hMSCs by activating caspase-3 apoptosis signaling, where they were visible in the MIR and showed a reduced proliferation of U87MG (human glioma) in the xenograft cancer model while prolonging the survival of the animals in the in vivo studies [86].

8. Pluronic F127-Based Nanohybrids

Pluronic F127 is an amphiphilic biocompatible copolymer and can be used as a delivery carrier. Wu et al. prepared charge-switchable nanohybrids (HNPs-DA) comprising pluronic F127 polymer, a SC-g-PEI-DMMA (stearoyl-polyethylenimine-2,3-dimethylmalefic anhydride) shell loaded with MNC (magnetic nanocluster), and PTX (paclitaxel). The in vitro studies showed darker T2-MRI images when the HepG2 cells were treated with HNPs-DA at a pH of 6.5 in comparison to a pH of 7.4, indicating a pH-sensitive release. Moreover, a higher cytotoxicity in the acidic tumor environment (pH 6.5) was observed in the HepG2 cells, further confirming the pH-based release, though further studies can help to understand and improve the potential of these NHs [87]. Polymer dots with pH- cleavable colorimetric nanosensor-based fluorescent polymer dots (L-PD) were prepared for the reduction-triggered release of PTX (paclitaxel). The matrices were synthesized by grafting 2-(dimethylamino) ethyl methacrylate (DMA) and 2-hydroxyethyl methacrylate (HEMA) to Pluronic F-127 and were taken as a base to prepare the NHs. The high level of glutathione (GSH) in the tumor microenvironment initiated the reduction and the drug release at the targeted site with a 100% drug release in the tumor environment. The cell viability assay on the MDCK and MDA-MB-231 cells showed that the blank NHs (L-PD) were nontoxic, whereas the NHs loaded with PTX were toxic only toward the MDA-MB-231 cells and produced little to no toxicity on the MDCK cells, further confirming the selectivity of the NHs. [88]. A generalized structure of the pluronic F127-based nanohybrids is given in Figure 8.

9. Other Polymer-Based Nanohybrids

Raloxifene hydrochloride intercalated layered double hydroxide (LDH) embedded in a polymer matrix of PCL (Poly(ε-caprolactone) with an appropriate surface charge for sustained drug release was prepared. The strong interaction between the drug and LDH led to a sustained release of the drug. The polymeric nanohybrid vehicle improved the in vitro anticancer activity of the hydrophobic drug, with an improved cellular uptake. Sustained drug release was also confirmed by the in vivo studies on an albino rat, with fewer side effects when compared to the pure drug. Polymeric NHs produced excellent and long-term antitumor effects, and further studies could help to improve the NHs for cancer therapy [89]. He et al. proposed a new strategy for the assembly of therapeutic peptides into stable NHs (peptide–auric), which further self-assembles into tumor-responsive nanoclusters. Therefore, Bcl 9 inhibitor/β-catenin copolymerized gold ion nanohybrid clusters using poly-L-lysine(PLL) (pCluster) were prepared by this strategy. Therapeutic peptides show insufficient internalization in diseased cells. The pClusters impaired the Wnt/β-catenin pathway in the in vitro and in vivo studies, and thus inhibited the tumor growth with little to no accumulation in the vital organs. The pClusters also showed a synergistic effect with the PD1/PD-L1 checkpoint blockade immunotherapy [90]. Doxorubicin was loaded into a silicate nanodisk (Laponite) and assembled with pH-sensitive poly(N-vinylpyrrolidone), followed by another anticancer drug, mitoxantrone (MXT), loaded onto it, and hence formed LDPM nanohybrids for cancer cells. The nanohybrids showed high drug encapsulation and longer colloidal stability. The NHs sequentially delivered the drugs, and an increased drug release was observed in the acidic tumor environment, where more toxicity was produced with the increasing concentration of the NHs [91]. Deng et al. synthesized a CuS@Cu2S@Au hollow nanoshell and modified it with thermosensitive polymer p(OEOMA-co-MEMA) (poly(oligo(ethylene oxide) methacrylate-co-2-(2- methoxyethoxy) ethyl methacrylate)) and RGD targeting molecules, and then loaded it with doxorubicin (DOX) for photoswitchable targeting and for enhancing the photothermal efficiency. The final HCuS@Cu2S@Au-P-DOX-RGD nanohybrids produced better activity with 5 min of laser irradiation (0.8 W cm−2, 808 nm) on the U87MG cells. The results were also confirmed by the in vivo studies on the U87MG tumor-bearing mice, as the tumor size remained minimum when treated with the NHs + NIR irradiation, and there was no significant change in the body weight [92]. Doxorubicin-encapsulated β-cyclodextrin-{poly(lactide)-poly(2-(dimethylamino) ethyl methacrylate)-poly[oligo(2-ethyl-2-oxazoline)methacrylate]}21[β-CD-(PLA-PDMAEMA-PEtOxMA)21] unimolecular micelle gold nanoparticles (GNPs) were prepared. The micelles showed a 41–61% entrapment efficiency and a fast drug release in the acidic tumor environment. In vitro and in vivo CT imaging showed more contrast enhancement than Omnipaque. The blank micelles produced no significant toxicity in the HepG2 cells, whereas the DOX-loaded micelles exhibited a concentration-dependent reduction in the cell viability with a more cytotoxic effect at higher concentrations. The in vivo studies on the HepG2 tumor-bearing nod–scid mice model showed that, though the DOX-loaded micelles reduced the tumor volume to some extent, the tumor volume was minimum in the case of the freeDOX, and a slight increase in body weight was observed, suggesting that the micelles were nontoxic to normal cells [93]. The pH- and light-responsive mesoporous silica nanohybrids (MSNs) containing the polymer PDMAEMA (poly(dimethylaminoethyl methacrylates) coating on them were prepared for the controlled release of doxorubicin. The PDMAEMA were functionalized with perylene and then added to the MSNs. A pH-responsive (higher release in acidic pH) and light-responsive drug release was seen, which can be enhanced by giving the combined stimuli. The loaded NHs produced significant cytotoxicity and produced the best activity in response to the light stimuli, further solidifying light-sensitive drug release [94]. Hexadecylamine polymer (HA-C16) containing magnetic nanoparticle (MNPs) liposomes, abbreviated as HA-MNP-LPs, were prepared and used to test the triggered drug release of DTX (docetaxel) in the breast cancer cell line (MCF-7). The liposomes of the HA-MNP-LPs showed better cellular uptake than the MNP-LPs. DTX-containing nanohybrids (DTX/HA-MNP-LPs) produced an IC50 value of 0.69µg/mL under NIR irradiation (808 nm) due to the 20% higher drug release [95]. Nanohybrids containing gold nanorods coated with HA (hyaluronic acid)-coated PSS (poly (sodium-4-styrenesulfonate))/DOX (doxorubicin)/(PLL)poly-L-lysine hydrobromide were tested for their potential in combinatorial therapy.The presence of HA enhanced the internalization of the nanohybrids in CD44-overexpressed ovarian, breast, and lung cancer. The drug release could be modulated either by enzymatic degradation or NIR laser irradiation (808 nm). The combinatorial therapy in vitro studies showed synergistic cytotoxicity on the MDA-MB-231 and HeLa cells with a maximum cytotoxicity at a power of 3 W/cm2. [96]. Polyvinylpyrrolidone-NIPPAm-lysine-based graphene oxide nanohybrids were prepared and fabricated with FU (fluorouracil) to form FU-GO/NHs. These NHs were then tested for their drug delivery potential on the MCF-7 cell line. A higher drug release was observed at a temperature (40 °C) higher than the body temperature (37 °C), and this release further increased in the acidic condition (pH 5.5, temperature 40 °C). FGO/NHs exhibit better cytotoxicity than the FU because of the increased internalization of the drug. The NHs produced 1.31–5.37% hemolysis, making them biocompatible, and could be considered for in vivo studies. The preparation affected p53 and PARP but produced no effect on Bax and Bcl-2 [97].
Dual-drug loading PAMAM (poly(amidoamine))dendrimer/carbon dot nanohybrids were synthesized, and DOX was loaded onto hydrothermally prepared sodium citrate carbon dots. TheCDs/DOX was complexed with G5-RGD-TPGS, which was formed by dendrimer PAMAM generation 5 modified with RGD peptide and TPGS (D-α-tocopheryl polyethylene glycol 1000 succinate), a drug efflux inhibitor. The final nanohybrid increased the ROS levels, decreased the ATP levels by P-glycoprotein inhibition, and hence produced their cytotoxic effect in the in vitro studies on the A549 cells, which, alongside the presence of CDs, enabled the simultaneous fluorescent imaging of the cancer cells. The RGD made the NHs highly specific to the αvβ3 integrin-expressing cancer cells. These NHs could produce MDR reversal and anticancer properties, but further studies are necessary to solidify these claims [98]. Yan et al.prepared SNH (peptide–auric spheroidal nanohybrids) for intracellular PPI (protein–protein interaction) targeting. PMI (a 12-mer hydrophobic and enzyme-intolerant p53 activator), BBI (Wnt inhibitor), and DPA (a 12-mer hydrophobic and dextrorotary (proteolytic-resistive) p53 activator) were used as a peptide in the nanohybrids, and these nanohybrids were tested for their anticancer potential on SW480(p53 mutant), A375(p53 wild type), MCF7(p53 wild type), and HCT116(p53 wild type) cells for both PMI- and DPA-loaded NHs, and on A549(unactivated Wnt), HepG2(activated Wnt), Hep3B(activated Wnt), and HCT116(activated Wnt) for BBI. The NHs inhibited the cell lines in a dose-dependent manner. The in vivo biodistribution studies showed an initial accumulation of NHs in the tumor in large quantities and in some amount in the kidney, liver, spleen, and lungs, but with time, the accumulation of the NHs increased in the tumor and declined in the other organs. Alongside the NHs, the administered models exhibited the lowest growth in tumor volume. The study suggested that SNHs maintained therapeutic safety while modulating the PPIs in the cells [99]. Patel et al.prepared nanohybrids with a graphene oxide surface modified with a polyurethane chain using diamine moieties, and these nanohybrids were then embedded with dexamethasone. The NHs produced a sustained drug release. The effect of the NHs on the biological system was observed in vitro on MDA-MB-231 cells. The NHs were found to be fluorescent, and a cell viability assay proved the biocompatibility of the NHs to be used as a carrier. However, studies on animal models could further justify their compatibility with biological systems [100]. PGEA ethanolamine-functionalized poly(glycidyl methacrylate)-functionalized dextran quantum dot nanohybrids (DQ-PGEA) were prepared for improved gene delivery. The NHs were stable, biocompatible, and effectively delivered the gene (antioncogene p53) to the breast cancer in the mouse model, as confirmed by the in vitro studies on the HEK293 and 4T1 cells. Further studies suggested that the NHs were fluorescent and accumulated in the tumor cells in breast cancer tumor-bearing mice. The tumor volume was found to be minimum in the case of DQ-PGEA/p53, further supporting the effectiveness of the NHs, with no significant damage to the major organ [101]. Ran et al. prepared polydopamine@ZIF-8 (PDA@ZIF-8) nanohybrids formed by covering ZIF-8 nanocrystals in a self-polymerizable dopamine nanoshell. The prepared nanohybrids were tested for their zinc-releasing and anticancer potential via in vitro and in vivo studies. The nanoparticles produced better results than the ZIF-8 nanocrystals. The in vitro studies on HCT8 and HEK293 cells showed that the NHs produced better cytotoxicity than ZIB-8 in the HCT8 cell while having a similar level of anticancer activity on the HEK293 cells. The NHs also internalized more levels of zinc in both the cell lines, and it was further confirmed by the in vivo studies [102]. Mehnath et al.synthesized the charge reversal and pH-responsive gold-poly(bis(carboxyphenoxy)phosphazene) PCPP nanohybrids loaded with CPT (camptothecin) and tested their intracellular drug delivery potential on breast cancer cells. Rapid drug release was observed in the acidic pH of the cancer microenvironment and the NHs were found to be fluorescent. The nanohybrids produced better cytotoxicity than the CPT by increasing the ROS levels and apoptotic protein expression [103]. Yang et al.synthesized oHA-APBA (oligomeric hyaluronic acid–aminophenylboronic acid) nanohybrids loaded with DOX (doxorubicin) and tested their cytotoxic effect on the MCF-7 cell line. The NHs were synthesized by entangling oHA-APBA into laponite RDS (LR) nanodisks. A pH-dependent, sustainable drug release, remarkable cellular internalization, and increased cytotoxicity were observed in the in vitro studies. The NHs produced batter activity than the free DOX in the MCF-7 cells [104].
Thermo-sensitive nanohybrids with Poly-N vinylimidazole and 1-vinyl-2-(hydroxymethyl)imidazole19 (UPVH) as a polymer were prepared and loaded with the drug (S)-10-Hydroxycamptothecin (HCPT). The nanohybrids exhibited thermosensitive drug release, with the loaded nanohybrids having ahigher drug release at a higher temperature. The fluorescent imaging of the NHs in the mice model suggests that the NHs accumulated in more quantity in the tumor, with little accumulation in the liver, spleen, and kidney. The tumor volume was found to be minimum in the animals administered with the NHs (CD-MSN@UP38). There was no significant toxicity observed after the administration of the NHs [105].AXT (Axitinib)-loaded nanohybrids by using hybrid liposomal nanoparticles coated with polypeptide (P-LNP/AXT) were synthesized. The NHs exhibited a loading capacity of 95.2% and were easily internalized by the SH-SY5YP, BT-474, and SCC7 cells, but were not very well internalized in RAW264.7. The cytotoxicity was not as good as AXT. The nanohybrids increased the level of poly (ADP-ribose) polymerase, caspase-3, and hydroxyl-HIF-1α expression, and decreased PECAM1, CD31, and Ki-67. Further studies suggested that the AXT-loaded NHs were moreactive in the biological system, as the tumor volume was found to be minimum in the case of P-LNP/AXT and was less as compared to the ATX-administered SCC7-tumor-bearing BALB/c nude mice. Alongside this, there was no reduction in the body weight of the mice, suggesting that the NHs were nontoxic on the normal cells [106]. Nanohybrids containing iron oxide nanoparticles coated with gold or silver and poly (butyl methacrylate-coacrylamide-co-methacrylic acid) were prepared. These NHs were loaded with letrozole and were then tested for their cytotoxic activity on L929 and MDA-MB-231in combination or absence of laser radiation. The NHs with the gold in them produced the best activity under the influence of laser radiation [107]. Che et al.prepared CO2-responsive magneto-polymeric nanohybrids (Fe3O4@SiO2-PDMAEMA) by using Fe3O4 NPs as the core and layer of SiO2, followed by a layer of the polymer PDMAEMA poly(N,N dimethylaminoethyl methacrylate), which were formed on it like a core–shell–corona structure. The prepared NHs were tested for the in vitro drug release of DOX (doxorubicin). The NHs showed a drug release induced by CO2 and inhibited by N2 treatment. The plain NHs were biocompatible and produced no significant cytotoxicity on the A549 cells [108]. Other polymers, such as porous organic polymers, and the importance of postsynthetic modifications, have been discussed by Kim et al., and these materials could prove to be promising in bioapplications. Alongside Poly(α-L-glutamic acid) (PGA),these polypeptides can also be exploited for theirbiomedical use, which has been discussed by zhang et al., as they have superior properties and a unique structure [109,110]. Some generalized structures of the remaining polymer-based nanohybrids are given in Figure 9. A list of all the polymer-based nanohybrids and their activity is given in Table 1.

10. Limitations and Challenges

The nanopolymers are designed to be inert, and hence mostly do not produce any toxicity by themselves, but some nanoparticles, such as TiO2 nanoparticles, are oncogenic [111,112]. Understanding the properties of the nanopolymers is necessary to improve the therapeutic effectiveness, but their properties in endo-lysosomal vesicles are hard to predict. The nanoparticles can disrupt cell function in many ways, including the production of ROS, in mitochondrial disruption, and others [113,114]. The NHs, although containing many components in them, are customizable in many ways, but one major limitation that was observed in various published articles was the lack of studies. The full potential of the NHs was unexplored in many of the cases. The undesirable accumulation in vital organs, the toxicity toward normal cells, and hemolysis were the problems found in some of the studies. Moreover, these smart delivery systems have, overtime, a low stability with antibodies, immunogenic adverse effects, and one of the most important is the high cost of preparation.

11. Successes

There are many successes in the use of NHs; as they are highly customizable, they solve many problems. One of the main advantages of NHs is the selective targeting of the cancer cells, as NHs can deliver the payload on the bases of the stimuli such aspH and temperature. Along with that, the delivery of the payload can also be triggered by external stimuli such as light, NIR, etc. Improved photodynamic and photothermal effects have been observed in the NHs, and they have produced remarkable results in hypoxic and even resistant tumors. Many modified nanoparticles target 3D tumor spheroid and targeting agents such asiRGD make the targeting more efficient in the animal models;moreover, there are many other strategies to improve the drug penetration in the tumor mass. More changes in shape, size, and further surface modifications can improve the therapeutic effect of the NPs in the animal models [115,116,117,118]. The NHs also produce fluorescence; in short, the NHs can create multifunctional carriers that can produce a variety of responses at the same time and could lead to better cancer therapeutic agents.

12. Conclusions

A variety of polymers have been used recently to prepare nanohybrids for cancer therapy. PEG is one of the most commonly used polymers to prepare nanohybrids, followed by other polymers such asCMC, chitosan, PLGA, polypyrroles, poly(ethyleneimine), casein, pluronic F127, and more. These polymers provide the nanohybrids with their distinct properties, such as pH-sensitive release and controlled release. The addition of components such asFA makes these nanohybrids highly target-specific. The polymeric nanohybrids provide an excellent delivery medium for anticancer molecules, genes, and peptides. They also solve the problems of MDR and the undesired side effects, while being safe to use with no significant cytotoxicity by themselves. Some of the NHs have also proved very effective in photodynamic, photothermal, and combined chemo/photothermal therapy. The use of organic/inorganic nanohybrids could help to combat cancer, but further research and exploration at the clinical level are required.

13. Prospect and Challenges

The nanohybrid polymers can provide a major breakthrough in cancer therapy, as they include many components in them. This will enable nanohybrids to act in a variety of ways on cancer cells, in particularvia photothermal therapy or chemotherapeutically. Nanohybrids could be an answer to the challenges faced in cancer cell targeting. However, the potential of many polymeric nanohybrids still need to be explored at the in vivo level and through clinical trials so to exploit their use in cancer therapy.

Author Contributions

Conceptualization, A.K. and D.K.; writing—original draft preparation, A.K., S.A., M.S. and D.K.; visualization, A.T., S.A. and I.A.; writing—review and editing, A.T., S.A., I.A., S.P., E.M. and A.R.; supervision, D.K. and S.P. All authors have read and agreed to the published version of the manuscript.

Funding

M.S., S.A., and S.A are thankful to the Ministry of Innovative Development of the Republic of Uzbekistan for the research fund grant number PZ-2021022512 under the project Development of technologies for targeted drug delivery with a long duration of action and increased safety based on nanostructured carriers.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare that there is no conflict of interest regarding the publication of this article.

References

  1. 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 A Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Torre, L.A.; Bray, F.; Siegel, R.L.; Ferlay, J.; Lortet-Tieulent, J.; Jemal, A. Global Cancer Statistics, 2012. CA A Cancer J. Clin. 2015, 65, 87–108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Galmarini, D.; Galmarini, C.M.; Galmarini, F.C. Cancer Chemotherapy: A Critical Analysis of Its 60 Years of History. Crit. Rev. Oncol./Hematol. 2012, 84, 181–199. [Google Scholar] [CrossRef]
  5. Wyld, L.; Audisio, R.A.; Poston, G.J. The Evolution of Cancer Surgery and Future Perspectives. Nat. Rev. Clin. Oncol. 2015, 12, 115–124. [Google Scholar] [CrossRef]
  6. Schaue, D.; Mcbride, W.H. Opportunities and Challenges of Radiotherapy for Treating Cancer. Nat. Rev. Clin. Oncol. 2015, 12, 527–540. [Google Scholar] [CrossRef]
  7. Raguz, S.; Yagüe, E. Resistance to Chemotherapy: New Treatments and Novel Insights into an Old Problem. Br. J. Cancer 2008, 99, 387–391. [Google Scholar] [CrossRef] [Green Version]
  8. Padma, V.V. An Overview of Targeted Cancer Therapy. BioMedicine 2015, 5, 1–6. [Google Scholar] [CrossRef]
  9. Xu, G.; Mcleod, H.L. Strategies for Enzyme/Prodrug Cancer Therapy 1. Clin. Cancer Res. 2001, 7, 3314–3324. [Google Scholar]
  10. Wu, D.; Gao, Y.; Qi, Y.; Chen, L.; Ma, Y.; Li, Y. Peptide-Based Cancer Therapy: Opportunity and Challenge. Cancer Lett. 2014, 351, 13–22. [Google Scholar] [CrossRef]
  11. El-Aneed, A. Current Strategies in Cancer Gene Therapy. Eur. J. Pharmacol. 2004, 498, 1–8. [Google Scholar] [CrossRef] [PubMed]
  12. Liu, X.; Wu, F.; Ji, Y.; Yin, L. Recent Advances in Anti-Cancer Protein/Peptide Delivery. Bioconjugate Chem. 2019, 30, 305–324. [Google Scholar] [CrossRef] [PubMed]
  13. Mali, S. Delivery Systems for Gene Therapy. Indian J. Hum. Genet. 2013, 19, 3–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Hulla, J.E.; Sahu, S.C.; Hayes, A.W. Nanotechnology: History and Future. Hum. Exp. Toxicol. 2015, 34, 1318–1321. [Google Scholar] [CrossRef]
  15. Thrall, J.H. Nanotechnology and Medicine. Radiology 2004, 230, 315–318. [Google Scholar] [CrossRef]
  16. Aslan, B.; Ozpolat, B.; Sood, A.K.; Lopez-Berestein, G. Nanotechnology in Cancer Therapy. J. Drug Target. 2013, 21, 904–913. [Google Scholar] [CrossRef] [Green Version]
  17. Nagarajan, R. Nanoparticles: Building Blocks for Nanotechnology. In Nanoparticles: Synthesis, Stabilization, Passivation, and Functionalization; Nagarajan, R., Alan Hatton, T., Eds.; ACS Publications: Washington, DC, USA, 2008; Volume 996, pp. 2–14. [Google Scholar]
  18. UlHaq, I.; Ijaz, S. Use of Metallic Nanoparticles and Nanoformulations as Nanofungicides for Sustainable Disease Management in Plants. In Nanobiotechnology in Bioformulations; Prasad, R., Kumar, V., Kumar, M., Choudhary, D., Eds.; Springer: Cham, Switzerland, 2019; pp. 289–316. [Google Scholar]
  19. Oshiro, J.A.; Abuçafy, M.P.; Manaia, E.B.; da Silva, B.L.; Chiari-Andréo, B.G.; Chiavacci, L.A. Drug Delivery Systems Obtained from Silica Based Organic-Inorganic Hybrids. Polymers 2016, 8, 91. [Google Scholar] [CrossRef] [Green Version]
  20. Attri, A.; Thakur, D.; Kaur, T.; Sensale, S.; Peng, Z.; Kumar, D.; Singh, R.P. Nanoparticles Incorporating a Fluorescence Turn-on Reporter forReal-Time Drug Release Monitoring, a Chemoenhancer and aStealth Agent: Poseidon’s Trident against Cancer? Mol. Pharmaceutics 2021, 18, 124–147. [Google Scholar] [CrossRef]
  21. Manatunga, D.C.; Godakanda, V.U.; de Silva, R.M.; de Silva, K.M.N. Recent Developments in the Use of Organic–Inorganic Nanohybrids for Drug Delivery. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2020, 12, e1605. [Google Scholar] [CrossRef]
  22. Li, J.; Shen, M.; Shi, X. Poly(Amidoamine) Dendrimer-Gold Nanohybrids in Cancer Gene Therapy: A Concise Overview. ACS Appl. Bio Mater. 2020, 3, 5590–5605. [Google Scholar] [CrossRef]
  23. Wang, Y.; Wang, F.; Shen, Y.; He, Q.; Guo, S. Tumor-Specific Disintegratable Nanohybrids Containing Ultrasmall Inorganic Nanoparticles: From Design and Improved Properties to Cancer Applications. Mater. Horiz. 2018, 5, 184–205. [Google Scholar] [CrossRef]
  24. Liang, C.; Zhang, X.; Wang, Z.; Wang, W.; Yang, M.; Dong, X. Organic/Inorganic Nanohybrids Rejuvenate Photodynamic Cancer Therapy. J. Mater. Chem. B 2020, 8, 4748–4763. [Google Scholar] [CrossRef] [PubMed]
  25. Cirillo, G.; Peitzsch, C.; Vittorio, O.; Curcio, M.; Farfalla, A.; Voli, F.; Dubrovska, A.; Iemma, F.; Kavallaris, M.; Hampel, S. When Polymers Meet Carbon Nanostructures: Expanding Horizons in Cancer Therapy. Future Med. Chem. 2019, 11, 2205–2231. [Google Scholar] [CrossRef] [PubMed]
  26. Liechty, W.B.; Kryscio, D.R.; Slaughter, B.V.; Peppas, N.A. Polymers for Drug Delivery Systems. Annu. Rev. Chem. Biomol. Eng. 2010, 1, 149–173. [Google Scholar] [CrossRef] [Green Version]
  27. Vimal, S.K.; Cao, H.; Dubey, A.; Agrawal, L.; Pathak, P.; Zuo, H.; Kumar, D.; Bhattacharyya, S. In vivo and in silico investigations of the pegylated gold nanoparticle treatment of amyotrophic lateral sclerosis in mice. New J. Chem. 2022, 46, 12252–12264. [Google Scholar] [CrossRef]
  28. Knop, K.; Hoogenboom, R.; Fischer, D.; Schubert, U.S. Poly(Ethylene Glycol) in Drug Delivery: Pros and Cons as Well as Potential Alternatives. Angew. Chem. Int. Ed. 2010, 49, 6288–6308. [Google Scholar] [CrossRef]
  29. Zheng, D.W.; Li, B.; Li, C.X.; Fan, J.X.; Lei, Q.; Li, C.; Xu, Z.; Zhang, X.Z. Carbon-Dot-Decorated Carbon Nitride Nanoparticles for Enhanced Photodynamic Therapy against Hypoxic Tumor via Water Splitting. ACS Nano 2016, 10, 8715–8722. [Google Scholar] [CrossRef]
  30. Pandey, N.K.; Chudal, L.; Phan, J.; Lin, L.; Johnson, O.; Xing, M.; Liu, J.P.; Li, H.; Huang, X.; Shu, Y.; et al. A Facile Method for the Synthesis of Copper-Cysteamine Nanoparticles and Study of ROS Production for Cancer Treatment. J. Mater. Chem. B 2019, 7, 6630–6642. [Google Scholar] [CrossRef]
  31. Leitão, M.M.; Alves, C.G.; de Melo-Diogo, D.; Lima-Sousa, R.; Moreira, A.F.; Correia, I.J. Sulfobetaine Methacrylate-Functionalized Graphene Oxide-IR780 Nanohybrids Aimed at Improving Breast Cancer Phototherapy. RSC Advances 2020, 10, 38621–38630. [Google Scholar] [CrossRef]
  32. Ardekani, S.M.; Dehghani, A.; Hassan, M.; Kianinia, M.; Aharonovich, I.; Gomes, V.G. Two-Photon Excitation Triggers Combined Chemo-Photothermal Therapy via Doped Carbon Nanohybrid Dots for Effective Breast Cancer Treatment. Chem. Eng. J. 2017, 330, 651–662. [Google Scholar] [CrossRef]
  33. Zhu, Y.X.; Jia, H.R.; Chen, Z.; Wu, F.G. Photosensitizer (PS)/Polyhedral Oligomeric Silsesquioxane (POSS)-Crosslinked Nanohybrids for Enhanced Imaging-Guided Photodynamic Cancer Therapy. Nanoscale 2017, 9, 12874–12884. [Google Scholar] [CrossRef]
  34. Sawant, V.J.; Bamane, S.R.; Shejwal, R.V.; Patil, S.B. Comparison of Drug Delivery Potentials of Surface Functionalized Cobalt and Zinc Ferrite Nanohybrids for Curcumin in to MCF-7 Breast Cancer Cells. J. Magn. Magn. Mater. 2016, 417, 222–229. [Google Scholar] [CrossRef]
  35. Huang, C.; Wu, J.; Jiang, W.; Liu, R.; Li, Z.; Luan, Y. Amphiphilic Prodrug-Decorated Graphene Oxide as a Multi-Functional Drug Delivery System for Efficient Cancer Therapy. Mater. Sci. Eng. C 2018, 89, 15–24. [Google Scholar] [CrossRef] [PubMed]
  36. Liang, X.; Gao, J.; Jiang, L.; Luo, J.; Jing, L.; Li, X.; Jin, Y.; Dai, Z. Nanohybrid Liposomal Cerasomes with Good Physiological Stability and Rapid Temperature Responsiveness for High Intensity Focused Ultrasound Triggered Local Chemotherapy of Cancer. ACS Nano 2015, 9, 1280–1293. [Google Scholar] [CrossRef] [PubMed]
  37. Loiseau, A.; Boudon, J.; Oudot, A.; Moreau, M.; Boidot, R.; Chassagnon, R.; Saïd, N.M.; Roux, S.; Mirjolet, C.; Millot, N. Titanate Nanotubes Engineered with Gold Nanoparticles and Docetaxel to Enhance Radiotherapy on Xenografted Prostate Tumors. Cancers 2019, 11, 1962. [Google Scholar] [CrossRef] [Green Version]
  38. Gautam, M.; Thapa, R.K.; Gupta, B.; Soe, Z.C.; Ou, W.; Poudel, K.; Jin, S.G.; Choi, H.-G.; Yong, C.S.; Kim, J.O. Phytosterol-Loaded CD44 Receptor-Targeted PEGylated Nano-Hybrid Phyto-Liposomes for Synergistic Chemotherapy. Expert Opin. Drug Deliv. 2020, 17, 423–434. [Google Scholar] [CrossRef]
  39. Loiseau, A.; Boudon, J.; Mirjolet, C.; Créhange, G.; Millot, N. Taxane-Grafted Metal-Oxide Nanoparticles as a New Theranostic Tool against Cancer: The Promising Example of Docetaxel-Functionalized Titanate Nanotubes on Prostate Tumors. Adv. Healthc. Mater. 2017, 6, 1700245. [Google Scholar] [CrossRef]
  40. Du, W.; Chen, C.; Sun, P.; Zhang, S.; Zhang, J.; Zhang, X.; Liu, Y.; Zhang, R.; Yan, C.; Fan, C.; et al. Eliciting an Immune Hot Tumor Niche with Biomimetic Drug-Based Multi-Functional Nanohybrids Augments Immune Checkpoint Blockade-Based Breast Cancer Therapy. Nanoscale 2020, 12, 3317–3329. [Google Scholar] [CrossRef]
  41. Zhao, L.; Yuan, W.; Tham, H.P.; Chen, H.; Xing, P.; Xiang, H.; Yao, X.; Qiu, X.; Dai, Y.; Zhu, L.; et al. Fast-Clearable Nanocarriers Conducting Chemo/Photothermal Combination Therapy to Inhibit Recurrence of Malignant Tumors. Small 2017, 13, 1700963. [Google Scholar] [CrossRef]
  42. Yuan, X.; Peng, S.; Lin, W.; Wang, J.; Zhang, L. Multistage PH-Responsive Mesoporous Silica Nanohybrids with Charge Reversal and Intracellular Release for Efficient Anticancer Drug Delivery. J. Colloid Interface Sci. 2019, 555, 82–93. [Google Scholar] [CrossRef]
  43. Liu, Y.; Guo, Q.; Sun, H.; Guo, N.; Zhang, J. Improved Therapeutic Efficiency of Photothermal Treatment and Nursing Care in Prostate Cancer by DOX Loaded PEG Coated Cu@Se Nano-Hybrid Vesicle. Process Biochem. 2020, 92, 78–84. [Google Scholar] [CrossRef]
  44. Wu, C.; Zhu, A.; Li, D.; Wang, L.; Yang, H.; Zeng, H.; Liu, Y. Photosensitizer-Assembled PEGylated Graphene-Copper Sulfide Nanohybrids as a Synergistic near-Infrared Phototherapeutic Agent. Expert Opin. Drug Deliv. 2016, 13, 155–165. [Google Scholar] [CrossRef]
  45. Ma, W.; Hu, Y.; Yang, H.; Zhang, Y.; Ding, J.; Chen, L. Au-Aided Reduced Graphene Oxide-Based Nanohybrids for Photo-Chemotherapy. Mater. Sci. Eng. C 2019, 95, 256–263. [Google Scholar] [CrossRef] [PubMed]
  46. Liu, Y.; Liu, K.; Li, C.; Wang, L.; Liu, J.; He, J.; Lei, J.; Liu, X. Self-Assembled Nanoparticles Based on a Carboxymethylcellulose–Ursolic Acid Conjugate for Anticancer Combination Therapy. RSC Adv. 2017, 7, 36256–36268. [Google Scholar] [CrossRef] [Green Version]
  47. Kharkwal, H.; Janaswamy, S. Natural Polymers for Drug Delivery; CABI: Wallingford, UK, 2016. [Google Scholar]
  48. Sorokin, A.V.; Kuznetsov, V.A.; Lavlinskaya, M.S. Synthesis of Graft Copolymers of Carboxymethyl Cellulose and N,N-Dimethylaminoethyl Methacrylate and Their Study as Paclitaxel Carriers. Polym. Bull. 2021, 78, 2975–2992. [Google Scholar] [CrossRef]
  49. Rahman, M.S.; Hasan, M.S.; Nitai, A.S.; Nam, S.; Karmakar, A.K.; Ahsan, M.S.; Shiddiky, M.J.A.; Ahmed, M.B. Recent Developments of Carboxymethyl Cellulose. Polymers 2021, 13, 1345. [Google Scholar] [CrossRef] [PubMed]
  50. Mansur, A.A.P.; Caires, A.J.; Carvalho, S.M.; Capanema, N.S.V.; Carvalho, I.C.; Mansur, H.S. Dual-Functional Supramolecular Nanohybrids of Quantum Dot/Biopolymer/Chemotherapeutic Drug for Bioimaging and Killing Brain Cancer Cells in Vitro. Colloids Surf. B Biointerfaces 2019, 184, 110507. [Google Scholar] [CrossRef]
  51. Leonel, A.G.; Mansur, H.S.; Mansur, A.A.P.; Caires, A.; Carvalho, S.M.; Krambrock, K.; Outon, L.E.F.; Ardisson, J.D. Synthesis and Characterization of Iron Oxide Nanoparticles/Carboxymethyl Cellulose Core-Shell Nanohybrids for Killing Cancer Cells in Vitro. Int. J. Biol. Macromol. 2019, 132, 677–691. [Google Scholar] [CrossRef]
  52. Javanbakht, S.; Hemmati, A.; Namazi, H.; Heydari, A. Carboxymethylcellulose-Coated 5-Fluorouracil@MOF-5 Nano-Hybrid as a Bio-Nanocomposite Carrier for the Anticancer Oral Delivery. Int. J. Biol. Macromol. 2020, 155, 876–882. [Google Scholar] [CrossRef]
  53. Carvalho, I.C.; Mansur, A.A.P.; Carvalho, S.M.; Florentino, R.M.; Mansur, H.S. L-Cysteine and Poly-L-Arginine Grafted Carboxymethyl Cellulose/Ag-In-S Quantum Dot Fluorescent Nanohybrids for in Vitro Bioimaging of Brain Cancer Cells. Int. J. Biol. Macromol. 2019, 133, 739–753. [Google Scholar] [CrossRef]
  54. Carvalho, I.C.; Mansur, A.A.P.; Carvalho, S.M.; Mansur, H.S. Nanotheranostics through Mitochondria-Targeted Delivery with Fluorescent Peptidomimetic Nanohybrids for Apoptosis Induction of Brain Cancer Cells. Nanotheranostics 2021, 5, 213–239. [Google Scholar] [CrossRef] [PubMed]
  55. Mansur, A.A.P.; Amaral-Júnior, J.C.; Carvalho, S.M.; Carvalho, I.C.; Mansur, H.S. Cu-In-S/ZnS@carboxymethylcellulose Supramolecular Structures: Fluorescent Nanoarchitectures for Targeted-Theranostics of Cancer Cells. Carbohydr. Polym. 2020, 247, 116703. [Google Scholar] [CrossRef] [PubMed]
  56. Carvalho, S.M.; Leonel, A.G.; Mansur, A.A.P.; Carvalho, I.C.; Krambrock, K.; Mansur, H.S. Bifunctional Magnetopolymersomes of Iron Oxide Nanoparticles and Carboxymethylcellulose Conjugated with Doxorubicin for Hyperthermo-Chemotherapy of Brain Cancer Cells. Biomater. Sci. 2019, 7, 2102–2122. [Google Scholar] [CrossRef] [PubMed]
  57. Bernkop-Schnürch, A.; Dünnhaupt, S. Chitosan-Based Drug Delivery Systems. Eur. J. Pharm. Biopharm. 2012, 81, 463–469. [Google Scholar] [CrossRef]
  58. Gooday, G.W. Biochemistry of Cell Walls and Membranes in Fungi; Kuhn, P.J., Trinci, A.P.J., Jung, M.J., Goosey, M.W., Copping, L.G., Eds.; Springer: Berlin/Heidelberg, Germany, 1990; ISBN 978-3-642-74217-0. [Google Scholar]
  59. Vincent, J.F.V. Biology of Fibrous Composites: Development Beyond the Cell Membrane. Science (1979) 1994, 265, 126–128. [Google Scholar]
  60. Kumar, M.N.V.R.; Muzzarelli, R.A.A.; Muzzarelli, C.; Sashiwa, H.; Domb, A.J. Chitosan Chemistry and Pharmaceutical Perspectives. Chem. Rev. 2004, 104, 6017–6084. [Google Scholar] [CrossRef]
  61. Dahri, M.; Akbarialiabad, H.; Jahromi, A.M.; Maleki, R. Loading and Release of Cancer Chemotherapy Drugs Utilizing Simultaneous Temperature and PH-Responsive Nanohybrid. BMC Pharmacol. Toxicol. 2021, 22, 1–10. [Google Scholar] [CrossRef]
  62. Jia, L.; Li, Z.; Zheng, D.; Li, Z.; Zhao, Z. A Targeted and Redox/PH-Responsive Chitosan Oligosaccharide Derivatives Based Nanohybrids for Overcoming Multidrug Resistance of Breast Cancer Cells. Carbohydr. Polym. 2021, 251, 117008. [Google Scholar] [CrossRef]
  63. Pooresmaeil, M.; Asl, E.A.; Namazi, H. A New PH-Sensitive CS/Zn-MOF@GO Ternary Hybrid Compound as a Biofriendly and Implantable Platform for Prolonged 5-Fluorouracil Delivery to Human Breast Cancer Cells. J. Alloy. Compd. 2021, 885, 160992. [Google Scholar] [CrossRef]
  64. George, D.; Maheswari, P.U.; Begum, K.M.M.S. Cysteine Conjugated Chitosan Based Green Nanohybrid Hydrogel Embedded with Zinc Oxide Nanoparticles towards Enhanced Therapeutic Potential of Naringenin. React. Funct. Polym. 2020, 148, 104480. [Google Scholar] [CrossRef]
  65. Rasoulzadehzali, M.; Namazi, H. Facile Preparation of Antibacterial Chitosan/Graphene Oxide-Ag Bio-Nanocomposite Hydrogel Beads for Controlled Release of Doxorubicin. Int. J. Biol. Macromol. 2018, 116, 54–63. [Google Scholar] [CrossRef] [PubMed]
  66. Luo, Y.-L.; Zhang, X.-Y.; Fu, J.-Y.; Xu, F.; Chen, Y.-S. Novel Temperature and PH Dual Sensitive PNIPAM/CMCS/MWCNTs Semi-IPN Nanohybrid Hydrogels: Synthesis, Characterization and DOX Drug Release. Int. J. Polym. Mater. Polym. Biomater. 2017, 66, 398–409. [Google Scholar] [CrossRef]
  67. Seyfoori, A.; Sarfarazijami, S.; Seyyed Ebrahimi, S.A. PH-Responsive Carbon Nanotube-Based Hybrid Nanogels as the Smart Anticancer Drug Carrier. Artif. Cells Nanomed. Biotechnol. 2019, 47, 1437–1443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Ramasamy, S.; Dhamecha, D.; Kaliyamoorthi, K.; Pillai, A.S.; Alexander, A.; Dhanaraj, P.; Menon, J.U.; Enoch, I.V.M.V. Magnetic Hydroxyapatite Nanomaterial–Cyclodextrin Tethered Polymer Hybrids as Anticancer Drug Carriers. Mater. Adv. 2021, 2, 3315–3327. [Google Scholar] [CrossRef]
  69. Laksee, S.; Supachettapun, C.; Muangsin, N.; Lertsarawut, P.; Rattanawongwiboon, T.; Sricharoen, P.; Limchoowong, N.; Chutimasakul, T.; Kwamman, T.; Hemvichian, K. Targeted Gold Nanohybrids Functionalized with Folate-Hydrophobic-Quaternized Pullulan Delivering Camptothecin for Enhancing Hydrophobic Anticancer Drug Efficacy. Polymers 2021, 13, 2670. [Google Scholar] [CrossRef]
  70. Li, X.; Zhou, J.; Liu, C.; Xiong, Q.; Duan, H.; Cheung, P.C.K. Stable and Biocompatible Mushroom β-Glucan Modified Gold Nanorods for Cancer Photothermal Therapy. J. Agric. Food Chem. 2017, 65, 9529–9536. [Google Scholar] [CrossRef]
  71. Li, X.; Zhou, J.; Dong, X.; Cheng, W.Y.; Duan, H.; Cheung, P.C.K. In Vitro and in Vivo Photothermal Cancer Therapeutic Effects of Gold Nanorods Modified with Mushroom β-Glucan. J. Agric. Food Chem. 2018, 66, 4091–4098. [Google Scholar] [CrossRef]
  72. Xu, Y.; Kim, C.-S.; Saylor, D.M.; Koo, D. Polymer Degradation and Drug Delivery in PLGA-Based Drug-Polymer Applications: A Review of Experiments and Theories. J. Biomed. Mater. Res. Part B Appl. Biomater. 2017, 105, 1692–1716. [Google Scholar] [CrossRef]
  73. Topete, A.; Alatorre-Meda, M.; Villar-Alvarez, E.M.; Carregal-Romero, S.; Barbosa, S.; Parak, W.J.; Taboada, P.; Mosquera, V. Polymeric-Gold Nanohybrids for Combined Imaging and Cancer Therapy. Adv. Healthc. Mater. 2014, 3, 1309–1325. [Google Scholar] [CrossRef]
  74. Ray, S.; Mishra, A.; Mandal, T.K.; Sa, B.; Chakraborty, J. Optimization of the Process Parameters for the Fabrication of a Polymer Coated Layered Double Hydroxide-Methotrexate Nanohybrid for the Possible Treatment of Osteosarcoma. RSC Adv. 2015, 5, 102574–102592. [Google Scholar] [CrossRef]
  75. Jain, N.K.; Prabhuraj, R.S.; Bavya, M.C.; Prasad, R.; Bandyopadhyaya, R.; Naidu, V.G.M.; Srivastava, R. Niclosamide Encapsulated Polymeric Nanocarriers for Targeted Cancer Therapy. RSC Adv. 2019, 9, 26572–26581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Geetha, S.; Rao, C.R.K.; Vijayan, M.; Trivedi, D.C. Biosensing and Drug Delivery by Polypyrrole. Anal. Chim. Acta 2006, 568, 119–125. [Google Scholar] [CrossRef] [PubMed]
  77. Cai, X.; Deng, X.; Xie, Z.; Shi, Y.; Pang, M.; Lin, J. Controllable Synthesis of Highly Monodispersed Nanoscale Fe-Soc-MOF and the Construction of Fe-Soc-MOF@polypyrrole Core-Shell Nanohybrids for Cancer Therapy. Chem. Eng. J. 2019, 358, 369–378. [Google Scholar] [CrossRef]
  78. Wu, M.X.; Yan, H.J.; Gao, J.; Cheng, Y.; Yang, J.; Wu, J.R.; Gong, B.J.; Zhang, H.Y.; Yang, Y.W. Multifunctional Supramolecular Materials Constructed from Polypyrrole@UiO-66 Nanohybrids and Pillararene Nanovalves for Targeted Chemophotothermal Therapy. ACS Appl. Mater. Interfaces 2018, 10, 34655–34663. [Google Scholar] [CrossRef]
  79. Purushothaman, B.K.; Maheswari, P.U.; Sheriffa Begum, K.M. PH and Magnetic Field Responsive Protein-Inorganic Nanohybrid Conjugated with Biotin: A Biocompatible Carrier System Targeting Lung Cancer Cells. J. Appl. Polym. Sci. 2021, 138, 49949. [Google Scholar] [CrossRef]
  80. Purushothaman, B.K.; Maheswari, P.U.; Begum, K.M.M.S. Magnetic Casein-CaFe2O4 Nanohybrid Carrier Conjugated with Progesterone for Enhanced Cytotoxicity of Citrus Peel Derived Hesperidin Drug towards Breast and Ovarian Cancer. Int. J. Biol. Macromol. 2020, 151, 293–304. [Google Scholar] [CrossRef]
  81. Hussien, N.A.; Işıklan, N.; Türk, M. Pectin-Conjugated Magnetic Graphene Oxide Nanohybrid as a Novel Drug Carrier for Paclitaxel Delivery. Artif. Cells Nanomed. Biotechnol. 2018, 46, 264–273. [Google Scholar] [CrossRef]
  82. Ma, N.; Liu, J.; He, W.; Li, Z.; Luan, Y.; Song, Y.; Garg, S. Folic Acid-Grafted Bovine Serum Albumin Decorated Graphene Oxide: An Efficient Drug Carrier for Targeted Cancer Therapy. J. Colloid Interface Sci. 2017, 490, 598–607. [Google Scholar] [CrossRef]
  83. Nezhad-Mokhtari, P.; Arsalani, N.; Javanbakht, S.; Shaabani, A. Development of Gelatin Microsphere Encapsulated Cu-Based Metal-Organic Framework Nanohybrid for the Methotrexate Delivery. J. Drug Deliv. Sci. Technol. 2019, 50, 174–180. [Google Scholar] [CrossRef]
  84. Aliabadi, M.; Yunessnialehi, A.; Shagholani, H.; Gerayeli, A. Planar Polymer-Graphene Oxide Nanohybrid as a 5-Fluorouacil Carrier in PH-Responsive Controlled Release. J. Drug Deliv. Sci. Technol. 2018, 43, 103–106. [Google Scholar] [CrossRef]
  85. Bhattacharya, D.; Behera, B.; Sahu, S.K.; Ananthakrishnan, R.; Maiti, T.K.; Pramanik, P. Design of Dual Stimuli Responsive Polymer Modified Magnetic Nanoparticles for Targeted Anti-Cancer Drug Delivery and Enhanced MR Imaging. New J. Chem. 2016, 40, 545–557. [Google Scholar] [CrossRef]
  86. Huang, R.Y.; Lin, Y.H.; Lin, S.Y.; Li, Y.N.; Chiang, C.S.; Chang, C.W. Magnetic Ternary Nanohybrids for Nonviral Gene Delivery of Stem Cells and Applications on Cancer Therapy. Theranostics 2019, 9, 2411–2423. [Google Scholar] [CrossRef] [PubMed]
  87. Wu, L.; Wu, M.; Lin, X.; Zhang, X.; Liu, X.; Liu, J. Magnetite Nanocluster and Paclitaxel-Loaded Charge-Switchable Nanohybrids for MR Imaging and Chemotherapy. J. Mater. Chem. B 2017, 5, 849–857. [Google Scholar] [CrossRef] [PubMed]
  88. Kim, S.G.; Ryplida, B.; Phuong, P.T.M.; Won, H.J.; Lee, G.; Bhang, S.H.; Park, S.Y. Reduction-Triggered Paclitaxel Release Nano-Hybrid System Based on Core-Crosslinked Polymer Dots with a Ph-Responsive Shell-Cleavable Colorimetric Biosensor. Int. J. Mol. Sci. 2019, 20, 5368. [Google Scholar] [CrossRef] [Green Version]
  89. Senapati, S.; Shukla, R.; Tripathi, Y.B.; Mahanta, A.K.; Rana, D.; Maiti, P. Engineered Cellular Uptake and Controlled Drug Delivery Using Two Dimensional Nanoparticle and Polymer for Cancer Treatment. Mol. Pharm. 2018, 15, 679–694. [Google Scholar] [CrossRef]
  90. He, W.; Wang, S.; Yan, J.; Qu, Y.; Jin, L.; Sui, F.; Li, Y.; You, W.; Yang, G.; Yang, Q.; et al. Self-Assembly of Therapeutic Peptide into Stimuli-Responsive Clustered Nanohybrids for Cancer-Targeted Therapy. Adv. Funct. Mater. 2019, 29, 1807736. [Google Scholar] [CrossRef]
  91. Zhou, B.; Wu, B.; Wang, J.; Qian, Q.; Wang, J.; Xu, H.; Yang, S.; Feng, P.; Chen, W.; Li, Y.; et al. Drug-Mediation Formation of Nanohybrids for Sequential Therapeutic Delivery in Cancer Cells. Colloids Surf. B Biointerfaces 2018, 163, 284–290. [Google Scholar] [CrossRef]
  92. Deng, X.; Li, K.; Cai, X.; Liu, B.; Wei, Y.; Deng, K.; Xie, Z.; Wu, Z.; Ma, P.; Hou, Z.; et al. A Hollow-Structured CuS@Cu2S@Au Nanohybrid: Synergistically Enhanced Photothermal Efficiency and Photoswitchable Targeting Effect for Cancer Theranostics. Adv. Mater. 2017, 29, 1701266. [Google Scholar] [CrossRef]
  93. Lin, W.; Yao, N.; Qian, L.; Zhang, X.; Chen, Q.; Wang, J.; Zhang, L. PH-Responsive Unimolecular Micelle-Gold Nanoparticles-Drug Nanohybrid System for Cancer Theranostics. Acta Biomater. 2017, 58, 455–465. [Google Scholar] [CrossRef]
  94. Wang, G.; Dong, J.; Yuan, T.; Zhang, J.; Wang, L.; Wang, H. Visible Light and PH Responsive Polymer-Coated Mesoporous Silica Nanohybrids for Controlled Release. Macromol. Biosci. 2016, 16, 990–994. [Google Scholar] [CrossRef]
  95. Nguyen, V.D.; Zheng, S.; Han, J.; Le, V.H.; Park, J.O.; Park, S. Nanohybrid Magnetic Liposome Functionalized with Hyaluronic Acid for Enhanced Cellular Uptake and Near-Infrared-Triggered Drug Release. Colloids Surf. B Biointerfaces 2017, 154, 104–114. [Google Scholar] [CrossRef] [PubMed]
  96. Mosquera, V.; Villar-Alvarez, E.; Cambón, A.; Pardo, A.; Mosquera, V.X.; Bouzas-Mosquera, A.; Topete, A.; Barbosa, S.; Taboada, P. Gold Nanorod-Based Nanohybrids for Combinatorial Therapeutics. ACS Omega 2018, 3, 12633–12647. [Google Scholar] [CrossRef]
  97. Ashjaran, M.; Babazadeh, M.; Akbarzadeh, A.; Davaran, S.; Salehi, R. Stimuli-Responsive Polyvinylpyrrolidone-NIPPAm-Lysine Graphene Oxide Nano-Hybrid as an Anticancer Drug Delivery on MCF7 Cell Line. Artif. Cells Nanomed. Biotechnol. 2019, 47, 443–454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Li, D.; Fan, Y.; Shen, M.; Bányai, I.; Shi, X. Design of Dual Drug-Loaded Dendrimer/Carbon Dot Nanohybrids for Fluorescence Imaging and Enhanced Chemotherapy of Cancer Cells. J. Mater. Chem. B 2019, 7, 277–285. [Google Scholar] [CrossRef]
  99. Yan, J.; Ji, F.; Yan, S.; You, W.; Ma, F.; Li, F.; Huang, Y.; Liu, W.; He, W. A General-Purpose Nanohybrid Fabricated by Polymeric Au(i)-Peptide Precursor to Wake the Function of Peptide Therapeutics. Theranostics 2020, 10, 8513–8527. [Google Scholar] [CrossRef]
  100. Patel, D.K.; Gupta, V.; Dwivedi, A.; Pandey, S.K.; Aswal, V.K.; Rana, D.; Maiti, P. Superior Biomaterials Using Diamine Modified Graphene Grafted Polyurethane. Polymer (Guildf) 2016, 106, 109–119. [Google Scholar] [CrossRef]
  101. Liu, Y.; Zhao, N.; Xu, F.J. PH-Responsive Degradable Dextran-Quantum Dot Nanohybrids for Enhanced Gene Delivery. ACS Appl. Mater. Interfaces 2019, 11, 34707–34716. [Google Scholar] [CrossRef]
  102. Ran, J.; Wang, C.; Zhang, J.; Wang, W.; Xiao, L.; Jia, S.; Wang, Z.; Wu, W.; Xiao, J.; Wu, X. New Insight into Polydopamine@ZIF-8 Nanohybrids: A Zinc-Releasing Container for Potential Anticancer Activity. Polymers 2018, 10, 476. [Google Scholar] [CrossRef] [Green Version]
  103. Mehnath, S.; Mukherjee, A.; Mariappan, R.; Vijayaanand, M.A.; Jeyaraj, M. Polyorganophosphazene Stabilized Gold Nanoparticles for Intracellular Drug Delivery in Breast Carcinoma Cells. Process Biochem. 2018, 72, 152–161. [Google Scholar] [CrossRef]
  104. Yang, Y.; Li, J.; Chen, F.; Qiao, S.; Li, Y.; Pan, W. Synthesis, Formulation, and Characterization of Doxorubicin-Loaded Laponite/Oligomeric Hyaluronic Acid-Aminophenylboronic Acid Nanohybrids and Cytological Evaluation against MCF-7 Breast Cancer Cells. AAPS PharmSciTech 2020, 21. [Google Scholar] [CrossRef]
  105. Dong, F.; Zheng, T.; Zhu, R.; Wang, S.; Tian, Y. An Engineered Thermo-Sensitive Nanohybrid Particle for Accurate Temperature Sensing at the Single-Cell Level and Biologically Controlled Thermal Therapy. J. Mater. Chem. B 2016, 4, 7681–7688. [Google Scholar] [CrossRef] [PubMed]
  106. Choi, J.Y.; Ramasamy, T.; Tran, T.H.; Ku, S.K.; Shin, B.S.; Choi, H.-G.; Yong, C.S.; Kim, J.O. Systemic Delivery of Axitinib with Nanohybrid Liposomal Nanoparticles Inhibits Hypoxic Tumor Growth. J. Mater. Chem. B 2015, 3, 408–416. [Google Scholar] [CrossRef] [PubMed]
  107. Hadilou, N.; Khoshgenab, A.N.; Amoli-Diva, M.; Sadighi-Bonabi, R. Remote Trice Light, Temperature, and PH-Actuation of Switchable Magneto-Plasmonic Nanocarriers for Combinational Photothermal and Controlled/Targeted Chemotherapies. J. Pharm. Sci. 2018, 107, 3123–3133. [Google Scholar] [CrossRef]
  108. Che, H.; Huo, M.; Peng, L.; Ye, Q.; Guo, J.; Wang, K.; Wei, Y.; Yuan, J. CO2-Switchable Drug Release from Magneto-Polymeric Nanohybrids. Polym. Chem. 2015, 6, 2319–2326. [Google Scholar] [CrossRef]
  109. Zhang, Y.; Song, W.; Lu, Y.; Xu, Y.; Wang, C.; Yu, D.-G.; Kim, I. Recent Advances in Poly(α-L-Glutamic Acid)-Based Nanomaterials for Drug Delivery. Biomolecules 2022, 12, 636. [Google Scholar] [CrossRef] [PubMed]
  110. Kim, J.H.; Kang, D.W.; Yun, H.; Kang, M.; Singh, N.; Kim, J.S.; Hong, C.S. Post-Synthetic Modifications in Porous Organic Polymers for Biomedical and Related Applications. Chem. Soc. Rev. 2022, 51, 43–56. [Google Scholar] [CrossRef]
  111. Onuma, K.; Sato, Y.; Ogawara, S.; Shirasawa, N.; Kobayashi, M.; Yoshitake, J.; Yoshimura, T.; Iigo, M.; Fujii, J.; Okada, F. Nano-Scaled Particles of Titanium Dioxide Convert Benign Mouse Fibrosarcoma Cells into Aggressive Tumor Cells. Am. J. Pathol. 2009, 175, 2171–2183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Albanese, A.; Tang, P.S.; Chan, W.C.W. The Effect of Nanoparticle Size, Shape, and Surface Chemistry on Biological Systems. Annu. Rev. Biomed. Eng. 2012, 14, 1–16. [Google Scholar] [CrossRef] [Green Version]
  113. AshaRani, P.V.; Low Kah Mun, G.; Hande, M.P.; Valiyaveettil, S. Cytotoxicity and Genotoxicity of Silver Nanoparticles in Human Cells. ACS Nano 2009, 3, 279–290. [Google Scholar] [CrossRef]
  114. Berneburg, M.; Kamenisch, Y.; Krutmann, J.; Röcken, M. “To Repair or Not to Repair ? No Longer a Question”: Repair of Mitochondrial DNA Shielding against Age and Cancer. Exp. Dermatol. 2006, 15, 1005–1015. [Google Scholar] [CrossRef]
  115. Priwitaningrum, D.L.; Blondé, J.-B.G.; Sridhar, A.; van Baarlen, J.; Hennink, W.E.; Storm, G.; le Gac, S.; Prakash, J. Tumor Stroma-Containing 3D Spheroid Arrays: A Tool to Study Nanoparticle Penetration. J. Control. Release 2016, 244, 257–268. [Google Scholar] [CrossRef] [PubMed]
  116. Sha, H.; Zou, Z.; Xin, K.; Bian, X.; Cai, X.; Lu, W.; Chen, J.; Chen, G.; Huang, L.; Blair, A.M.; et al. Tumor-Penetrating Peptide Fused EGFR Single-Domain Antibody Enhances Cancer Drug Penetration into 3D Multicellular Spheroids and Facilitates Effective Gastric Cancer Therapy. J. Control. Release 2015, 200, 188–200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Minchinton, A.I.; Tannock, I.F. Drug Penetration in Solid Tumours. Nat. Rev. Cancer 2006, 6, 583–592. [Google Scholar] [CrossRef] [PubMed]
  118. Xu, C.; Haque, F.; Jasinski, D.L.; Binzel, D.W.; Shu, D.; Guo, P. Favorable Biodistribution, Specific Targeting and Conditional Endosomal Escape of RNA Nanoparticles in Cancer Therapy. Cancer Lett. 2018, 414, 57–70. [Google Scholar] [CrossRef] [PubMed]
Figure 1. A generalized structure ofPEG-based nanohybrids.
Figure 1. A generalized structure ofPEG-based nanohybrids.
Polymers 14 03027 g001
Figure 2. A generalized structure of CMC-containing nanohybrids.
Figure 2. A generalized structure of CMC-containing nanohybrids.
Polymers 14 03027 g002
Figure 3. A generalized structure of chitosan-based nanohybrids.
Figure 3. A generalized structure of chitosan-based nanohybrids.
Polymers 14 03027 g003
Figure 4. A generalized structure of PLGA-based nanohybrids.
Figure 4. A generalized structure of PLGA-based nanohybrids.
Polymers 14 03027 g004
Figure 5. A generalized structure of polypyrrole-based nanohybrids.
Figure 5. A generalized structure of polypyrrole-based nanohybrids.
Polymers 14 03027 g005
Figure 6. A generalized structure of casein-based nanohybrids.
Figure 6. A generalized structure of casein-based nanohybrids.
Polymers 14 03027 g006
Figure 7. A generalized structure of Poly(ethyleneimine)-based nanohybrids.
Figure 7. A generalized structure of Poly(ethyleneimine)-based nanohybrids.
Polymers 14 03027 g007
Figure 8. A generalized structure of Pluronic F127-based nanohybrids.
Figure 8. A generalized structure of Pluronic F127-based nanohybrids.
Polymers 14 03027 g008
Figure 9. A generalized structure of other polymer-based nanohybrids.
Figure 9. A generalized structure of other polymer-based nanohybrids.
Polymers 14 03027 g009
Table 1. List of polymeric nanohybrids synthesized in recent years and their role.
Table 1. List of polymeric nanohybrids synthesized in recent years and their role.
NanohybridsPolymerActive AgentsCell LinePerformanceReferenceRole of Polymer
PCCNpolyethylene glycol (PEG)-4T1Photodynamic therapy by water splitting[29]acted as binder
Cu-Cy nanoparticles-KYSE-30MW-induced radical therapy by ROS generation[30]-
IR780-SPP-GO-MCF-7Photodynamic therapy[31]improved colloidal stability
CND-P@DOXdoxorubicin (DOX)MCF-7Drug delivery and photothermal therapy[32]surface passivation
POSS-Ce6-PEG-HeLaPhotodynamic cancer therapy[33]prolonged circulation time and improved aqueous dispersion
PEG-coated curcumin-loaded cobalt ferritecurcuminMCF-7Drug delivery[34]coating agent
GO/PP-SS-DOX/PEG-FAdoxorubicin (DOX)FR-positive MCF-7, B16, FR-negative A549Reduction-sensitive drug delivery[35]improved aqueous stability
HTSCsdoxorubicin (DOX)MDA-MB-231Temperature-responsive ultrasound-triggered local chemotherapy [36]kept pores open, pore stabilization and facilitated rapid drug release
TiONts-AuNPs-PEG3000-DTXdocetaxel (DTX)PC-3Drug delivery and radiotherapy[37]improved suspension stability
HA-DOX-STS-lipodoxorubicin (DOX)MCF-7 and MDA-MB-231CD44 receptor-targeted synergistic chemotherapy[38]ensured long circulation time
TiONts–DTXdocetaxel (DTX)22Rv1Drug delivery[39]enhanced individualization, enhanced stability
Drug-based nanohybrids (NK-DNH)oxaliplatin (OXA), 1-Methyl-D-tryptophan (1-MT)4T1Blockade-based breast cancer therapy[40]increased passive
targeting, decreased nonspecial
accumulation, facilitated self-assembly
HPSNpaclitaxelHeLaPhotothermal and chemotherapy[41]biodistribution, enhanced cellular uptake, and prevented the trapping of the nanoparticles in RES
MSN-hyd-MOPdoxorubicinHepG2Drug delivery[42]enhanced cellular internalization
PEG@Cu-Se+DOXdoxorubicin (DOX)LNCaP and DU145Drug delivery and photothermal therapy[43]enhanced the aqueous solubility of DOX
pGO-CuS/ICG-MCF-7Photodynamic therapy[44]-
rGO/Au/PPEGdoxorubicin (DOX) Photochemotherapy[45]improved biocompatibility
ZnS@CMC-DOXCarbohydrate-based nanohybridsCarboxymethyl cellulose (CMC)doxorubicin (DOX)U-87 MGDrug delivery[50]water-soluble capping ligand and biofunctional layer
MION-CMC and Co-MION-CMC-HEK 293T, U87 Magnetic hyperthermia therapy[51]stabilized ligand and functional biocompatible organic coating
CMC/5-FU@MOF-55- fluorouracil (5-FU)HeLaAnticancer oral delivery[52]protected 5-FU in digestive system and pH-sensitive release and to carry
CMCelPolyArg and QD nanoconjugates-HEK 293T, U-87 MGBioimaging and brain cancer cell targeting[53]stabilizing agent and capping ligand
AIS@CMC_Cys-based NHsdoxorubicin (DOX)U-87 MGMitochondria-targeted delivery[54]-
ZCIS@CMC-FA-DOXdoxorubicin (DOX)TNBC (FRα+), MCF7 (FRα-), HEK 293T, andTargeted drug delivery[55]nucleation, growth, and stabilization of nanocolloidal dispersions
MION@CMC-DOXdoxorubicin (DOX)U87Drug delivery[56]nanoparticlestabilization, biocompatibility, and water-soluble biopolymer ligand
SWCNT-based nanohybrids Chitosanpaclitaxel and doxorubicin-Drug delivery, computational studies[61]improved the biocompatibility and biodegradability; chitosan had a significant function in the DOX release mechanism and PAX uptake
FITC-PEG-CS-PEI/SNP-shRNA and paclitaxelMCF-7/ADR cellsDrug delivery[62]good water solubility and
excellent biocompatibility, as the polymer backbone to graft LMW
PEI using degradable disulfide linkages to construct copolymers with
suitable charge density and molecular weights
5-Fu@CS/Zn-MOF@GO5-FU (5-fluorouracil)MDA-MB 231Sustained and pH-sensitive drug release[63]monodispersion
CYS-CHGZ-NRGnaringenin (NRG)A431Drug delivery[64]chitosan stabilized the hydrogel and enabled a sustained release of NRG drug
CH/GO-Ag nanocomposite hydrogelDOX (doxorubicin)SW480 Drug delivery[65]sustained and controlled-release drug delivery
PNIPAM/CMCS/MWCNTs semi-IPN nanohybrid hydrogelsDOX (doxorubicin)L929Temperature-responsive and pH-responsive drug delivery[66]good biodegradability and
biocompatibility, pH-responsivity; carboxymethylated chitosan was used
Chi-MnFe2O4/CNTDOX (doxorubicin)U-87pH-sensitive drug release [67]greater pH-responsiveness and subsequently higher drug release
Chi-CD-Pt-fol-coated Sr–Fe, Sr-HAp, and Sr, Fe-HAp NPsDOX (doxorubicin)MG-63Drug delivery [68]complexed Pt with the polymer and tethered with folate
CPT-GNHs@FHQ-PULpullulancamptothecinChago-k1, KATO-III, HepG2Drug delivery[69]nontoxic, noncarcinogenic, biocompatible, biodegradable, and highly soluble
AuNR-Gluβ-glucan-MCF-7, HT-29, SW480 Photothermal therapy[70,71]low cytotoxicity and effective photothermal effect
DOXO/SPION-PLGA NPspoly (lactic-co-glycolic acid) (PLGA)doxorubicin (DOX)HeLaNIR-triggered and FA-receptor-targeted drug release[73]stabilization
PLGA-MTX and PLGA-LDH-MTX methoteraxate (MTX)MG-63Improved efficacy of drug[74]anionic, hydrophobic polymer: improved the overall therapeutic effect of MTX
NIC-PLGA NPNIC (niclosamide)MDA-MB-231, L929Drug delivery[75]good biocompatibility, degradability,
and an ample nanocarrier
Fe-soc-MOF@PolypyrrolePolypyrrole -L929, 4T1Photothermal therapy[77]biocompatible polymer with
strong absorption in the NIR region
Polypyrrole@UiO-66 nanohybrids5-FU (5-fluorouracil)HeLaChemophotothermal therapy[78]biocompatibility, high conductivity, and excellent
photothermal conversion efficiency
Casein-CFNP-I-BTProtein based NHsCaseincinnamaldehydeL929, A549pH- and magnetic-responsive drug delivery[79]drug carrier
CaseiIaFe2O4nanohybrid carrierhesperidinMDA-MB-231, SKOV-3Drug delivery[80]drug delivery
PEC-GO-Fe3O4-PACpectinPTX (paclitaxel)L-929, MCF-7Drug delivery[81]stabilizing agent
FA-BSA/GO/DOX albuminDOX (doxorubicin)MCF-7Targeted drug delivery[82]stabilizer and targeting
Cu-MOF/MTX@GMgelatin microsphere biopolymerMTX (methotrexate)MCF-7Drug delivery[83]pH-sensitive
TA-PEI-GO nanohybridPoly(ethyleneimine) baed nanohybrids 5-FU (5-fluorouracil)-pH-sensitive drug delivery[84]a water-soluble cationic polymer
DOX-FA-Poly-MFNPsDOX (doxorubicin)HeLa, HaCaTThermo/pH-sensitive drug release[85]dual-responsive triggering, i.e., pH and temperature
MTNTRAILU87MGGene delivery[86]-
Magnetite nanocluster charge-switchable nanohybridsPluronic F127 PTX (paclitaxel)HepG2Chemotherapy[87]charge-conversion ability via the co-hybridization; can also be used as the delivery carriers of
SPIONs aggregates
L-PDPTX (paclitaxel)MDA-MB-231Reduction-triggered drug release[88]formed the matrix for drug loading
LDH nanohybrids Other polymers(Poly(ε-caprolactone)raloxifene hydrochlorideHeLaDrug delivery[89]improved the therapeutic
efficacy of the hydrophobic anticancer drugs by enhancing bioavailability
pClusterpoly-L-lysine(PLL)β-ceteninA549, B16F10, HCT116, Hep3BTargeted therapy[90]triggered the self-assembly of the pParticle into the massive pCluster
LDPM nanohybridspoly(N-vinylpyrrolidone)DOX (doxorubicin), mitoxantrone (MXT)KB cellsDrug delivery[91]to improve
their colloidal stability and for pH sensitivity
HCuS@Cu2S@Au-P-RGD-DOXpoly(oligo(ethylene oxide)DOX (doxorubicin)U87MGPhotothermal therapy and photoswitchable drug delivery[92]thermosensitive polymer
β-CD-(PLA-PDMAEMA-PEtOxMA)21/Au/DOX{poly(lactide)-poly(2-(dimethylamino) ethyl methacrylate)-poly[oligo(2-ethyl-2-oxazoline)methacrylate]}DOX (doxorubicin)HepG2Theranostics[93]-
Polymer-coated MSNspoly(dimethylaminoethylmethacrylates)DOX (doxorubicin)MCF-7Controlled release[94]sensitive to visible light and pH
HA-MNP-LPshexadecylamine polymerDTX (docetaxel)MCF-7NIR-triggered drug delivery[95]NIR-stimulated drug release
(PSS/DOXO/PLL)2/HA-coated GNRspoly (sodium-4-styrenesulfonateDOX (doxorubicin)HeLa, MDA-MB-231Combinatorial therapeutics[96]to mask the toxic hexadecyltrimethyl ammonium bromide (CTAB) layer
FU-GO/NHspolyvinylpyrrolidoneFU (fluorouracil)MCF-7Temperature-sensitive drug delivery[97]-
Dendrimer/carbon dot nanohybrids Polyethylene glycol 1000 vitamin E succinate (TPGS)DOX (doxorubicin) MDR reversal[98]Pg-P inhibitor
Peptide-Au SNHpeptide-auricMDM2 antagonistSW480, MCF-7, A375, HCT116, Hep3B, HepG2, A549Peptide therapy[99]colloidal stability
PU nanohybrids polyurethanedexamethasone MDA-MB-231Drug delivery[100]GO coating material
DQ-PGEApoly(glycidyl methacrylate)antioncogene p534T1, HEK293 Gene delivery[101]pH-responsive drug release
polydopamine@ZIF-8polydopaminemelphalanMCF-7Zinc and drug delivery[102]negligible cytotoxicity and good biocompatibility, and were
stable in vivo for several weeks
CPT@PCPP-AuNPs poly(bis(carboxyphenoxy)phosphazene)CPT (camptothecin)MDA-MB-231Drug delivery[103]to avoid loss of drug at normal pH
DOX@LR/oHA-APBA nanohybridsoligomeric hyaluronic acid-aminophenylboronic acidDOX (doxorubicin)MCF-7Drug delivery[104]biocompatibility, and
controlled release
CD-MSN@UPPoly-Nvinylimidazole and 1-vinyl-2-(hydroxymethyl)imidazole19(S)-10-Hydroxycamptothecin-Temperature-sensitive drug delivery[105]-
P-LNP/AXTpolypeptideAXT (axitinib)SH-SY5YP, BT-474, SCC7Drug delivery[106]-
NP-PBAMspoly (butyl methacrylate-coacrylamide-co-methacrylic acid)letrozoleL929, MDA-MB-231pH-, light-, temperature-, and magnetically switchable drug delivery and photothermal therapy [107]drug carrier
Fe3O4@ SiO2–PDMAEMA nanoparticlespoly(N,N dimethylaminoethyl methacrylate)DOX (doxorubicin)A549CO2-switchable drug[108]reversibly binding with CO2 in water, exhibiting hydrophilic/hydrophobic chain conformation
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kumar, A.; Sharipov, M.; Turaev, A.; Azizov, S.; Azizov, I.; Makhado, E.; Rahdar, A.; Kumar, D.; Pandey, S. Polymer-Based Hybrid Nanoarchitectures for Cancer Therapy Applications. Polymers 2022, 14, 3027. https://doi.org/10.3390/polym14153027

AMA Style

Kumar A, Sharipov M, Turaev A, Azizov S, Azizov I, Makhado E, Rahdar A, Kumar D, Pandey S. Polymer-Based Hybrid Nanoarchitectures for Cancer Therapy Applications. Polymers. 2022; 14(15):3027. https://doi.org/10.3390/polym14153027

Chicago/Turabian Style

Kumar, Arun, Mirkomil Sharipov, Abbaskhan Turaev, Shavkatjon Azizov, Ismatdjan Azizov, Edwin Makhado, Abbas Rahdar, Deepak Kumar, and Sadanand Pandey. 2022. "Polymer-Based Hybrid Nanoarchitectures for Cancer Therapy Applications" Polymers 14, no. 15: 3027. https://doi.org/10.3390/polym14153027

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