Next Article in Journal
PLA/PBS Biocomposites for 3D FDM Manufacturing: Effect of Hemp Shive Content and Process Parameters on Printing Quality and Performances
Previous Article in Journal
Development of Self-Healing Polyurethane and Applications in Flexible Electronic Devices: A Review
Previous Article in Special Issue
Quantification of Hydrogen Peroxide in PVP and PVPVA Using 1H qNMR Spectroscopy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Research Progress on Polymer-Based Nanocarriers for Tumor-Targeted Delivery of Survivin siRNA

by
Luya Ren
1,2,†,
Shaoxia Wang
2,†,
Bin-Chun Li
1,* and
Guo-Bin Ding
1,2,*
1
Institute of Biotechnology, Shanxi University, Taiyuan 030006, China
2
Institutes of Biomedical Sciences, Inner Mongolia University, Hohhot 010070, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Polymers 2025, 17(17), 2279; https://doi.org/10.3390/polym17172279
Submission received: 18 July 2025 / Revised: 17 August 2025 / Accepted: 20 August 2025 / Published: 23 August 2025
(This article belongs to the Special Issue Polymers for Drug/Gene Delivery and Controlled Release)

Abstract

Survivin, a pivotal member of the inhibitor of apoptosis proteins (IAP) family, plays critical roles in cell cycle regulation and division. Survivin is overexpressed in most malignancies, making it an attractive therapeutic target. Due to its high specificity and potency, siRNA-based RNA interference (RNAi) has emerged as a powerful therapeutic strategy for effectively downregulating disease-related genes such as survivin in cancer therapy. However, naked siRNA suffers from rapid enzymatic degradation, poor cellular uptake, and off-target effects, severely limiting its therapeutic efficacy in vivo. Development of polymer-based nanocarriers for tumor-targeted delivery of survivin siRNA (siSurvivin) holds great potential to address these challenges. In this review, we first described the structure and function of survivin and summarized the survivin-targeted therapeutic strategy. Then, the siRNA delivery systems, particularly the polymeric nanocarriers, were introduced. Furthermore, a plethora of polymer-based nanocarriers for tumor-targeted siSurvivin delivery, including synthetic polymers (branched polymers, dendritic polymers, polymeric micelles), natural polymers (polysaccharides, proteins, and others), lipid-polymer hybrid nanoparticles, and polymer composite nanoparticles, were elaborated. Promising results underscore the potential of polymer-based nanocarriers for survivin siRNA delivery to enhance cancer therapy, providing a roadmap for future clinical translation.

1. Introduction

Cancer, as the second most common fatal disease in the world, has become a major public health problem that seriously threatens human health, with its incidence and mortality increasing year by year [1]. Its main feature is the uncontrolled growth of abnormal cells, which are able to break through the normal growth boundaries, invade the neighboring tissues, and even spread to other tissues of the human body through blood circulation and the lymphatic system [2]. Cancer, also known as a malignant tumor, is invasive and metastatic, and it is extremely destructive to the organism. The formation of cancer is usually associated with a variety of factors, including gene mutation, environmental factors, hereditary factors, and lifestyles [3]. These factors may trigger the activation of oncogenes or the inactivation of tumor suppressor genes, leading to abnormal cell proliferation. Traditional cancer treatments include chemotherapy, radiotherapy, and surgery. Although they can control cancer progression to a certain extent, the overall treatment effect is far from satisfactory due to their severe side effects [4].
In recent years, with the rapid development of nanotechnology, the application of nanoparticles in cancer therapy has gradually attracted extensive attention. Nanoparticles can leverage the enhanced permeability and retention (EPR) effect, then achieve passive targeted accumulation through the highly permeable blood vessels and damaged lymphatic drainage system of tumor tissues. To further enhance the accumulation of nanoparticles in tumors, antibodies, peptides, and other targeted ligands can be modified onto the surface of nanoparticles, enabling them to precisely recognize and bind to receptors on the surface of tumor cells [5]. Additionally, some studies have found that certain nanoparticles, even without modification or drug loading, can enhance anticancer effects by altering cell membrane permeability when used in combination with anticancer drugs [6]. This opens new possibilities for the application of nanotechnology in cancer therapy and provides innovative approaches for the delivery of nucleic acid drugs.
Small interfering RNA (siRNA), as a gene silencing tool, can specifically degrade target mRNA and inhibit the expression of specific genes [7]. Survivin, as a key member of the apoptosis inhibitory protein family, plays an important role in biological processes such as cell cycle regulation, cell division, and apoptosis inhibition, and exhibits overexpression in a variety of cancer cells [8]. Therefore, by designing siRNAs targeting the survivin gene, specific degradation of survivin mRNA can be achieved, and then the expression of survivin gene can be downregulated, thereby effectively inhibiting the proliferation of tumor cells. Based on this, targeted delivery of siRNA to silence the survivin gene in tumor cells has become a highly promising cancer treatment strategy.
Despite the great advantages of siRNA in cancer therapy, it still faces many challenges. First, siRNA has poor stability under physiological conditions and is easily degraded by nucleases, thus having a short half-life in blood [9]. Second, while siRNA exhibits high specificity, incomplete complementary pairing may occur within cells, leading to non-specific degradation and unpredictable changes in gene expression [10]. Additionally, siRNAs are difficult to enter into cells by themselves due to their anion charge [11]. These factors limit the efficacy of siRNA in cancer therapy. Therefore, developing carriers that can efficiently and safely deliver siRNA to target cells while enhancing its stability is crucial for fully realizing the therapeutic potential of siRNA. Polymeric nanocarriers are ideal delivery systems due to their biodegradability and biocompatibility [12]. They can form stable complexes with siRNA through electrostatic interactions or covalent bonding to protect siRNA from degradation [13]. However, some drawbacks of polymeric nanocarriers hindered their practical applications, such as low drug loading efficiency, high toxicity, uncontrollable drug release, and insufficient targeting. To address these issues, researchers have multifunctionally modified polymeric nanocarriers. For example, PEGylation of highly cytotoxic polyethyleneimine (PEI) significantly reduces its toxicity [14]; lipid modification not only reduces cytotoxicity but also further enhances transfection efficiency [15]. Additionally, by leveraging the unique physicochemical properties of the tumor microenvironment (TME), such as hypoxia, acidic pH, and elevated glutathione (GSH) levels, stimulus-responsive drug delivery systems can be designed to achieve targeted drug delivery and release [16]. Furthermore, combining lipids, inorganic materials, or metals with polymers to form composite delivery systems effectively overcomes the inherent limitations of polymers [17]. These innovative strategies not only optimize the drug-loading capacity and biocompatibility of nanoparticles but also significantly enhance siRNA delivery efficiency and therapeutic efficacy, providing promising solutions to overcome the limitations of traditional delivery systems.
In summary, an in-depth study of the mechanism of action of survivin, combined with advanced polymer nanocarrier-based delivery technology, will provide an important theoretical basis for the development of survivin-targeted siRNA delivery systems. This review stands out by systematically comparing various polymer nanoplatforms and their tumor-targeting strategies, offering unique perspectives on optimizing siRNA delivery efficiency. The aim is to provide an authoritative account and new insight into the research progress in recent years on the use of different polymer-based nano-platforms for tumor-targeted survivin siRNA delivery (Figure 1), thereby bridging the gap between preclinical studies and clinical applications in nanomedicine.

2. Introduction of Survivin

2.1. The IAP Family

The inhibitor of apoptosis proteins (IAPs) are a family of proteins and can directly bind to and inhibit caspases, effectively suppressing cell death by regulating cell division, cell cycle progression, and signal transduction. Meanwhile, IAPs are overexpressed in a variety of human cancers and preferentially expressed in malignant cells, so they are considered to be highly promising therapeutic targets, providing important directions and ideas for the design of cancer treatment strategies [18]. Currently, there are eight members of the IAP family, including NIAP, XIAP, cIAP1, cIAP2, ML-IAP, survivin, apollon, and ILP-2. Among them, XIAP, cIAP1, cIAP2, and survivin play important roles in tumor development, progression, and drug resistance due to their structural and functional uniqueness. Therefore, they have received extensive attention in recent years [19]. Survivin has emerged as a potential therapeutic target due to its key functions in cell cycle regulation, anti-apoptosis, promotion of angiogenesis, and metabolic reprogramming [20].

2.2. Survivin Structure

Survivin (BIRC5) is the smallest member of the Inhibitors of Apoptosis Proteins family, originally discovered by Ambrosini et al. in 1997, which is an evolutionarily conserved eukaryotic protein and exists as a stable homodimer [21]. Survivin consists of 142 amino acids and has a molecular weight of approximately 16.5 kDa. Compared to the structures of other members of the IAP family, survivin is relatively simple, containing only a BIR (Baculovirus IAP repeat) structural domain and an extended alpha helix. In addition, the first 10 amino acids at the N-terminus are proline-rich, representing the Mitochondrial Targeting Sequence (MTS) of survivin’s input to the mitochondria, and the α-helix region at the C-terminus has a Nuclear Export Signal (NES), which allows it to shuttle between the nucleus and the cytoplasm [21,22].

2.3. Survivin Function

Survivin is widely distributed in the cell, mainly in the cytoplasm, and also prominently present in the nucleus and mitochondria [23]. Survivin exerts different biological functions in different organelles (Figure 2).
In the cytoplasm and mitochondria, survivin mainly exhibits apoptosis inhibition [22]. Unlike other members of the IAPs family, survivin does not directly bind to caspases but enhances the inhibitory effect on caspases by interacting with other IAPs family members (e.g., XIAP) to achieve anti-apoptotic effects. First, survivin is able to utilize the BIR structural domain to bind to another member of the X-linked inhibitor of apoptosis protein (XIAP) to form the survivin–XIAP complex, which enhances the inhibitory effect of XIAP on caspases [24]. XIAP, as a known inhibitor of caspases, can directly bind to caspase-3, caspase-7, and caspase-9 and thus inhibit their activities. By binding to XIAP, survivin enhances the anti-apoptotic function of XIAP, thereby indirectly inhibiting caspase activity [25]. In addition, the BIR structural domain of survivin can also bind to SMAC (Second Mitochondria-Derived Activator of Caspases), which exerts an anti-apoptotic effect [26]. SMAC/DIABLO is a mitochondria-derived pro-apoptotic protein, which is found mainly in mitochondria. And in normal cells, SMAC is localized in the mitochondrial membrane gap. When the cell is stimulated by apoptosis, mitochondria will release SMAC protein into the cytoplasm to bind with IAPs to abolish the inhibitory effect of IAPs on caspases, which promotes the activation of caspases and ultimately leads to apoptosis [27]. The binding of survivin to SMAC prevents SMAC from functioning properly, thus inhibiting the process of apoptosis and indirectly protecting IAPs from SMAC inhibition, maintaining the inhibitory effect of IAPs on caspases. Survivin also binds to HBXIP (HBx-Interacting Protein, Hepatitis B X Interacting Protein, a cofactor of survivin) and jointly inhibits apoptosis. The BIR structural domain of survivin interacts with HBXIP to form the HBXIP–survivin complex, which is capable of binding to pro-caspase-9, preventing the recruitment of pro-caspase-9 to apoptosis protease activating factor 1 (Apaf-1) [28]. Apaf-1 is a key regulatory protein in apoptosis, which activates caspase-9 and initiates the apoptotic cascade [29]. By blocking the binding of pro-caspase-9 to Apaf-1, the HBXIP–survivin complex inhibited the activation of caspase-9, thereby suppressing apoptosis. At the same time, survivin can also effectively prevent the release of cytochrome C from mitochondria to cytoplasm via regulating the Bax/Bcl-2 pathway, preventing the formation of the Cyt c/Apaf-1/Caspase-9 complex, and blocking the activation pathway of caspase-3, ultimately inhibiting the apoptosis [30]. In addition to effectively inhibiting caspase-dependent apoptosis, survivin also protects cells from apoptosis by preventing the release of AIF (Apoptosis-Inducing Factor) from mitochondria [31]. AIF, a protein located inside the mitochondria, plays an important role in regulating cell apoptosis. During the process of apoptosis, AIF is released from mitochondria and transferred to the nucleus, which in turn triggers chromatin condensation and DNA fragmentation and ultimately leads to apoptosis [32]. In addition, it has been reported that survivin binds to AIP [33] (aryl hydrocarbon receptor-interacting protein) and Hsp90 [22] (heat shock protein 90) and achieves anti-apoptotic effects via enhancing the stability of survivin.
In the nucleus, survivin is a key member of the chromosomal passenger complex (CPC) and plays an important role in cell cycle regulation, especially during mitosis. It is expressed in all phases of the cell cycle but reaches a peak in the G2/M phase and decreases significantly in the G1 phase [34]. CPC is a protein complex that plays a key regulatory role in mitosis and consists of four core components: aurora B kinase, INCENP, borealin, and survivin. Among them, aurora B kinase ensures the correct attachment of chromosomes to microtubules by phosphorylating kinetochore region proteins and activating the spindle assembly checkpoint (SAC). INCENP, survivin, and borealin are involved in the correct localization of the centromere [35]. Survivin interacts with aurora B kinase, regulates kinase activity, and assists it in correcting misconnections between chromosomes and spindle microtubules, and participates in the processes of chromosome separation, spindle assembly, and cytoplasmic division. This makes survivin a key regulator of cell mitosis and cell survival, assisting in the correct separation of chromosomes and ensuring that mitosis proceeds smoothly [36].

2.4. Survivin-Targeted Therapeutic Strategies

Survivin is typically expressed in developing and proliferating cells and is hardly expressed in normal cells. It is often overexpressed in most cancers, such as breast, liver, ovarian, lung, and prostate cancers, and is closely associated with the development, progression, and drug resistance of various cancers. Thus, survivin has received extensive attention as a potential target for tumor therapy [37]. Up to now, there are five surviving targeted cancer therapeutic strategies [38]. (a) Inhibitors that disrupt the interactions of survivin with its chaperone proteins. Examples include Shepherdin [39], AICAR [40] (disrupts the interaction between Hsp90 and survivin), PZ-6-QN [26], UC-112 and its analogs [41] (disrupts the interaction between survivin and SMAC). (b) Inhibitors that disrupt homodimerization of survivin. Examples include Abbot 8 [42], LQZ-7, LQZ-7i [43] etc. They degrade survivin by acting at the dimerization interface of survivin, thereby inhibiting cancer cell survival and promoting apoptosis. (c) Inhibitors that inhibit the transcription of the survivin gene. Inhibitors of survivin gene transcription include YM155, FL118 [44], and WM-127 [45], etc. Among them, YM155 is the first inhibitor targeting the survivin promoter, which can significantly inhibit survivin expression at the protein and mRNA levels and exert anticancer effects through a variety of mechanisms [46], but it has poor results in clinical trials and is mainly limited by stability issues [47]. FL118 and WM-127 also work by inhibiting survivin gene transcription to reduce its expression. (d) Survivin or its peptides for immunotherapy. This class mainly includes Survivin-2B80-88, SurVaxM (SVN53-67/M57-KLH), peptide cocktail vaccine EMD640744, and other specific survivin-based peptides, etc., which are mainly used for vaccine development and combination therapy and especially show great potential in combination therapy. (e) Inhibitors that induce survivin mRNA degradation. Inhibitors for the degradation of survivin mRNA mainly include antisense oligonucleotides (ASOs), ribozymes, and small interfering RNAs (siRNAs) [38]. Among them, two antisense oligonucleotides, ISIS 23722/LY2181308 and SPC3042/EZN-3042, are under investigation in clinical trials, but the results are not satisfactory [48]. Ribozyme-based treatment progressed slowly, and no significant result was obtained [49]. In contrast, siRNAs have been widely employed due to their high specificity, ability to achieve precise gene silencing through complete complementary pairing with target mRNAs, and simple design and synthesis at low cost [50]. However, the poor stability and low transfection efficiency of naked siRNAs greatly limited their application in tumor therapy [51], so delivery vectors are needed to improve their stability and cellular uptake efficiency.

3. siRNA Delivery Systems

3.1. Overview of siRNA Delivery Systems

Researchers have developed a variety of viral and non-viral vectors for siRNA delivery over the past years, aiming to enhance their stability, improve transfection efficiency, and reduce off-target effects. Viral vectors were developed relatively early, and these vectors have efficient transfection ability, and some lentiviral vectors and adeno-associated viral vectors are also capable of achieving long-term and stable expression of siRNAs [52]. However, the immunogenicity of viral vectors, the potential risk of viral recombination, and the biosafety issues associated with insertion mutagenesis, together with their complex preparation process, high production cost, and limited cargo capacity, have restricted their widespread use in clinical applications [53].
Due to the above-mentioned limitations of viral vectors, non-viral vectors are gradually gaining attention as an alternative. Non-viral vectors mainly include lipid nanoparticles [54], polymer nanoparticles [12], inorganic nanoparticles [55], and biomimetic nanoparticles [56], etc., which have the advantages of low immunogenicity, low off-target effects and toxicity, easy large-scale production, applicability to a variety of delivery routes, and long-term stability. In recent years, with the rapid development of nanotechnology and biomaterials science, the application of non-viral vectors in siRNA delivery has made significant progress, providing new possibilities for the clinical translation of siRNA agents. Among them, polymer-based nanoscale delivery systems have become a type of highly promising tool for siRNA, delivery especially in clinical applications, due to their excellent biodegradability, biocompatibility, and adjustable physicochemical properties [57].

3.2. Polymer-Based Nanoscale Delivery Systems

Polymer nanoparticles (PNPs) are prepared from natural or synthetic polymeric materials by physical or chemical methods, with the size usually ranging from 1 to 1000 nm [58]. Natural polymers such as hyaluronic acid, cyclodextrins, and chitosan exhibit significant advantages in drug delivery systems due to their unique physicochemical properties; for example, hyaluronic acid has good moisturizing and targeting properties and is able to specifically bind to the CD44 receptor, which enables precise drug delivery to tumor cells [59]. Due to its cationic properties, chitosan, is able to interact with the negative charge on the cell surface to enhance the cellular uptake and adhesion of drugs [60]. The unique molecular structure of cyclodextrins can be used to encapsulate drug molecules and improve drug stability and solubility [61].
Compared to natural polymers, synthetic polymeric composites have the characteristic of easy modification. Polymers such as polyethyleneimine (PEI), polypropyleneimine (PPI), polylactic acid (PLA), and polyethylene glycol (PEG) can be chemically modified to endow different drug release profiles. In addition, polymeric nanoparticles can achieve responsive drug release based on environmental factors (e.g., pH, temperature, enzymes, etc.) to further enhance therapeutic efficacy. This versatility and modifiability make polymer nanoparticles popular in the field of drug delivery [62].
However, with the increasing demands on the performance of conventional delivery vehicles and the limitations of liposomes (Lipid Nanoparticles, LNPs) in terms of stability and targeting ability, researchers began to explore some innovative solutions [63]. They combined lipids with polymers to develop lipid-polymer nanoparticles (LPNs), a hybrid structure that not only retains the biocompatibility of lipids but also enhances their stability by leveraging the properties of polymers. Meanwhile, the targeting ability and delivery efficiency of LPNs can be further enhanced by surface modification [64]. For example, polysaccharide-modified lipid nanoparticles showed higher stability and targeting ability in drug delivery [65]. Meanwhile, researchers have also attempted to combine metals [66] or inorganic materials [67] with polymers to further optimize delivery efficiency and reduce cytotoxicity. These innovative strategies provide new ideas and directions for the development of drug delivery systems.
Thus, we will systematically describe the research progress of polymer-based nanocarriers for targeted delivery of survivin siRNA from four aspects: synthetic polymeric nanoparticles, natural polymeric nanoparticles, lipid polymer nanoparticles, and other polymeric nanoparticles. The advantages and disadvantages of the four polymer-based survivin siRNA delivery systems were summarized in Table 1.

4. Polymeric Nanocarriers for Targeted Survivin siRNA Delivery

4.1. Targeted Delivery of Survivin siRNA by Synthetic Polymeric Nanoparticles

4.1.1. Branched and Hyperbranched Polymers

Polyethyleneimine (PEI) is a highly cationic polymer, which is often regarded as the “gold standard” polymer in gene transfection due to its excellent transfection efficiency, gene complexation ability, and endosome escape activity [73]. However, high cytotoxicity limits its wide application [74]. To reduce cytotoxicity while maintaining efficient transfection, researchers have developed various modification strategies. In our previous study, polylactide (PLA) was integrated to polyethyleneimine via synthesizing PEI–PLA copolymer, PLA modification significantly improved the cytocompatibility of PEI and retained the high transfection efficiency [75] (Table 2). And pH low insertion peptide decoration further improved the biocompatibility of PEI–PLA copolymer, while pH-responsive and effective siRNA delivery was achieved [76].
Hamidreza et al. synthesized octanoic acid (OA)-, palmitic acid (PA)-, oleic acid (OA)-, and linoleic acid (LA)-modified PEIs by introducing lipids on low molecular weight PEIs (2 kDa). These lipid modifications significantly enhanced the cellular uptake efficiency and delivery capacity of PEIs, and reduced cytotoxicity. Among them, octanoic acid-modified PEI showed the best survivin silencing efficiency, which significantly reduced cell survival and induced apoptosis [77]. In 2018, Manoj et al. used linoleic acid-modified polyethyleneimine (PEI–LA) as the main carrier to deliver survivin siRNA and introduced polymers such as hyaluronic acid (HA), polyacrylic acid (PA), chondroitin sulfate (DS), and methylcellulose (MC) to optimize delivery efficiency and cellular uptake, which significantly improved cellular viability and cellular uptake when compared to PEI–LA. Compared with PEI–LA alone, their incorporation significantly improved the cellular uptake and release of siSurvivin, and, in particular,, the incorporation of HA and PA was able to effectively inhibit the growth of breast cancer cells with fewer side effects on non-malignant cells [78].
In addition to lipid modification, Hou et al. combined PEI with T7 peptide-modified AIE/Gd nanoparticles to form Sur@T7-AIE-Gd NPs, a nanocarrier with bifunctional imaging of magnetic resonance imaging (MRI) and aggregation-induced emission (AIE) imaging, which enabled precise delivery, real-time monitoring, and highly efficient treatment of hepatocellular carcinoma (HCC) (Figure 3A) [79]. In another pioneering study, Zhupanyn et al. for the first time combined natural extracellular vesicles (ECVs) derived from different cell lines with PEI to construct a complex for siRNA (siSurvivin) delivery. In a PC3 tumor xenograft nude mouse model, PEI/siSurvivin complexes modified with ECVs were injected into the tail vein and significantly inhibited tumor growth [80]. In addition, Cao et al. designed a pH-responsive polyethyleneimine betaine-functionalized single-walled carbon nanotube (SWCNT) complex for co-delivery of siSurvivin and DOX, and the results showed that the DOX-SPBB-siRNA complex significantly reduced the tumor volume in an A549 tumor xenograft nude mouse model (Figure 3B) [81]. In contrast, Jin et al. constructed a pH-responsive nanoparticle for co-delivery of paclitaxel (PTX) and siSurvivin by coating polyethyleneimine-poly (lactic acid) (PEI–PLA) with poly (ethylene glycol)-poly (aspartic acid) (PEG-PAsp) (Figure 3C). Due to the pH-responsive property of PEG-PAsp, the nanoparticles presented better drug release and cellular uptake at pH 5.5. In vitro experiments showed that the nanoparticles induced apoptosis in A549 cells (up to 80.81%) and blocked the cell cycle in the G2/M phase [82].
A high level of GSH in cancer cells is indispensable for reactive oxygen species (ROS) scavenging, which makes it a promising target for cancer therapy [90]. Wang et al. designed a multifunctional nanoscale system based on quantum dots (QDs)-modified hollow manganese dioxide (QH-MnO2) for the combined delivery of paclitaxel (PTX) and survivin siRNA to enhance chemotherapy efficacy. The delivery system first used SiO2 nanoparticles as templates, uniformly coating their surfaces with a layer of MnO2 via chemical vapor deposition to form SiO2@MnO2 composite structures. Subsequently, Na2CO3 solution was used to selectively dissolve the SiO2 templates. As the templates gradually dissolved, hollow MnO2 nanoparticles (H-MnO2) were ultimately obtained. Next, PTX is physically adsorbed onto the interior of H-MnO2, forming H-MnO2@PTX. Subsequently, InP/ZnS quantum dots are combined with H-MnO2@PTX via electrostatic interactions, forming QH-MnO2@PTX. To enable siRNA attachment, the surface of QH-MnO2@PTX is modified with polyethyleneimine (PEI) to impart a positive charge, enabling electrostatic interactions with siRNA to form QH-MnO2@PTX-siRNA. Following endocytosis, GSH-induced degradation of QH-MnO2 triggers the release of PTX and siRNA while restoring the fluorescence of InP/ZnS quantum dots, thereby enabling real-time tracking and precise localization of therapeutic drugs. This approach achieved a significant tumor inhibition rate of 95.3% in the MDA-MB-231 tumor-bearing nude mouse model [83].
Chen et al. designed a reduction-responsive nanocarrier (mPEG-g-γ-PGA/SSBPEI@siRNA) for effective siSurvivin delivery to A549 cells (Figure 3D). This nanocarrier consisted of a cationic carrier SSBPEI for siRNA complexation, an mPEG shell for nanocarrier stabilization, and γ-PGA for efficient cellular uptake [84]. The same group further designed dual-targeted and glutathione (GSH)-responsive nanoparticles (SSBPEI-DOX@siRNAs/iRGD-PEG-HA) for co-delivery of DOX and siRNA cocktails (survivin siRNA, Bcl-2 siRNA, and ABCG2 siRNA) to ovarian cancer stem cells. The system first connects BPEI-SH with DOX-SH through an oxidation reaction to form SSBPEI-DOX, followed by electrostatic interaction to complex with siRNAs, resulting in SSBPEI-DOX@siRNAs. To enhance the stability and targeting of the nanodelivery system, tumor-targeting peptide iRGD, hyaluronic acid (HA), and polyethylene glycol (PEG) were further incorporated, self-assembling via electrostatic interactions to form SSBPEI-DOX@siRNAs/iRGD-PEG-HA nanoparticles. The system targets the CD44 receptor on the membrane of CSCs via HA and the NRP-1 receptor via iRGD, enabling specific recognition and endocytosis of ovarian CSCs. Upon entering the cells, the disulfide bonds (-SS-) in SSBPEI-DOX can be specifically reduced due to the high glutathione content in the tumor microenvironment, thereby triggering the release of siRNAs and DOX. The experimental results showed that the nanoparticles significantly enhanced the antitumor effect of DOX compared with free DOX and also greatly suppressed the migration and invasion of A2780/DDP-derived CSCs [85].
These findings indicate that the cytotoxicity of PEI was significantly reduced and the anti-tumor effect was significantly enhanced by various modification strategies, providing ideas and methods for the application of PEI in tumor-targeted gene therapy.

4.1.2. Dendrimers

Dendrimers are a class of highly structurally ordered synthetic macromolecules, rich in modifiable functional groups at the periphery of the molecule, which can significantly enhance the solubility of hydrophobic drugs, thereby enhancing their bioavailability and stability [91]. Common dendrimers include polyamide-amine (PAMAM) [92] and polypropyleneimine (PPI) [93]. In addition, cationic dendritic polymers can protect nucleic acids from degradation by binding to them through electrostatic interaction.
In this regard, Jugel et al. developed a PPI polymer-based siSurvivin-targeted delivery system, which achieved efficient cellular uptake and gene silencing of siRNAs through a caveolin/lipid raft-mediated internalization mechanism, thereby effectively inhibiting tumor growth. In this system, the PPI was modified with maltose, which significantly reduced cytotoxicity and dramatically improved biocompatibility; moreover, a single-chain antibody fragment (scFv) was introduced into the system, which was able to specifically recognize prostate stem cell antigens (PSCA), thus precisely entering into the target cells. Meanwhile, by taking advantage of the extremely high affinity between streptavidin and biotin, biotinylated scFv is stably bound to biotinylated PPIs by streptavidin, forming an intact polyplex structure and directing it to bind specifically to PSCA-positive cells. The luciferase activity assay indicated that the polyplex showed significant siRNA delivery efficiency and gene silencing effect in 293TPSCA/ffLuc cells. In the PC3PSCA xenograft tumor mouse model, PSCA-targeted siSurv polyplex reduced the tumor volume by about 50% compared to the control group, showing significant tumor growth inhibition [86]. Salve et al. designed a hyperbranched bis-MPA-based H40-TEPA-PEG-MUC1@siSurvivin delivery system. In this study, hyperbranched bis-MPA polyester (H40) was conjugated to tetraethylenepentamine (TEPA) to obtain H40-TEPA, and then H40-TEPA and mucin-1 (MUC1) aptamer were covalently coupled to the two ends of heterobifunctional polyethylene glycol (PEG) linker. The interaction of the MUC1 aptamer with the MUC1 receptor on MCF-cells greatly enhanced the cellular uptake of aptamer-conjugated targeted dendrimer. The experimental results showed that this delivery system exhibited a significant gene silencing effect in MCF-7 cells, which could reduce the expression level of Survivin mRNA by 2.5-fold compared with non-targeted dendritic polymers (1.3-fold) and Lipofectamine-2000 (2-fold), demonstrating a superior gene silencing efficiency [87].

4.1.3. Polymeric Micelles

Polymeric micelles (PMs), as a class of highly efficient nanocarriers, have a unique “core–shell” structure that not only significantly improves the solubility, stability, and bioavailability of hydrophobic drugs [94], but also enables targeted delivery and stimulates responsive drug release through surface functionalization, thereby enhancing therapeutic efficacy and reducing systemic toxicity [95] (Table 2). Based on this property, Salzano et al. constructed a PM-based siRNA/PTX co-delivery system by conjugating siSurvivin to phospholipids (PL) via disulfide bonds and self-assembling with polyethylene glycol-phosphatidylethanolamine (PEG2000-PE) to form PM and by co-encapsulating paclitaxel (PTX). This system significantly inhibited cell viability (about 70% reduction in ovarian cancer cell line) and enhanced cell sensitivity to PTX in a variety of cancer cell lines, while effectively overcoming drug resistance by down-regulating survivin protein level and disrupting microtubule structure in drug-resistant cell line SKOV3-tr [88]. Subsequently, the same group further optimized the preparation process to increase the PTX encapsulation efficiency to 90% and verified its anti-tumor effect in the SKOV3-tr ovarian cancer drug-resistant nude mice model, where the survivin siRNA/PTX PM co-delivery system resulted in a 4-fold reduction in tumor volume compared to a single drug, as well as down-regulated the expression of survivin and promoted drug enrichment in tumor tissues [89].

4.2. Targeted Delivery of Survivin siRNA by Natural Polymeric Nanoparticles

4.2.1. Polysaccharide

Chitosan (CS), a natural cationic polysaccharide, is the second most abundant polymer in nature after cellulose [96], and its surface amino groups can bind negatively charged substances (e.g., nucleic acids, proteins) by electrostatic interaction [97], a property that gives it a significant advantage in siRNA delivery systems as the only positively charged polysaccharide [98]. However, drug release issues and low solubility of chitosan at pH > 6.5 and in organic solvents limit its chemical modification and siRNA encapsulation efficiency [99]. Therefore, improving the solubility and drug release properties of chitosan is particularly important for the development of efficient siRNA delivery systems (Table 3).
It has been reported that chemical modifications (e.g., phosphorylation, carboxymethylation, quaternization, etc.) can significantly improve the solubility of chitosan and endow it with new functions [115]. Recently, Sader et al. constructed a dual self-assembled nanoparticle based on dermatan sulfate (DS) and chitosan, in which chitosan was used to form amphiphilic chitosan graft copolymer (CS-g-PMMA) by grafting poly(methylmethacrylate) (PMMA) and subsequently formed a polyelectrolyte complex (PEC) with DS via electrostatic interactions for the delivery of survivin siRNA (siSurvivin) (Figure 4A). Compared with the control group, 4T1 cells treated with siSurvivin-loaded nanoparticles showed a significant decrease in cell viability, migration ability, and sphere size, confirming that this delivery system is effective in achieving gene silencing [100]. Li et al. introduced quaternary ammonium into the hydroxyl groups at the C-6 position of chitosan via click chemistry to obtain 6-N, N, N-trimethyltriazole chitosan (CS) with high gene transfection efficiency. Erlotinib (Er), a near-infrared dye (Cy7), and quaternary ammonium were conjugated onto the chitosan skeleton to form CE7Q (Figure 4D). CQ and CE7Q were mixed and used to condense Survivin shRNA-expressing plasmid (SV) to obtain CE7Q/CQ/S nanocomplexes. This system could recognize epidermal growth factor receptor (EGFR) and enter into EGFR-mutated non-small cell lung cancer (NSCLC) cells, and a stimuli-responsive release profile was achieved by near-infrared laser irradiation at pH 5.4. CE7Q/CQ/S markedly downregulated survivin expression in all three cell lines (A549, PC9, and H1975) and displayed superior antitumor efficacy in vitro and in vivo by integrating chemo/gene/photothermal triple therapies into one nanoplatform [101]. On the other hand, Sun et al. successfully synthesized poly (ethylene glycol)-modified chitosan (PEG-CS) by coupling PEG molecules to the hydroxyl group of chitosan, and PEG-CS was employed to condense survivin siRNA to obtain PEG-CS/siRNA nanoparticles. PEG-CS/siSurvivin showed a significant inhibitory effect on 4T1 cells, with the cell survival rate decreasing from 62% to 48% and the apoptosis rate elevated from 5.32% to 16.03% as compared to the naked siRNA group. Furthermore, in a 4T1 tumor-bearing mouse model, PEG-CS/siSurvivin inhibited tumor growth and lung metastasis effectively [102].
In another study, Yang et al. directly grafted TAT peptides to the primary amino groups of chitosan (CS) to synthesize TAT-g-CS copolymer, which was used to bind survivin-targeted siRNA. The cellular uptake efficiency of TAT-g-CS/siRNA nanoparticles was 1.3-fold higher than that of unmodified CS/siRNA nanoparticles. TAT-g-CS/siRNASur nanoparticles strongly inhibited the proliferation of 4T1-Luc tumor cells via triggering cell apoptosis and markedly suppressed the in vivo growth and metastasis of malignant breast tumors [103].
Construction of chitosan-based stimuli-responsive nanocarriers can significantly improve the drug release efficiency in tumor tissues, thus enhancing the therapeutic effect of siRNA delivery systems [116]. For example, Zhang et al. grafted p-mercaptobenzoic acid (MA) on N, N, N-trimethyl chitosan (TMC) to obtain MA-TMC (MT) polymer, which could self-assemble into MT nanoparticles (NPs) with a core–shell structure and redox-responsive property. Then, paclitaxel (PTX), survivin shRNA-expressing plasmid (iSur-pDNA), and recombinant human interleukin-2 (rhIL-2) were loaded into MT NPs to form MT/PTX/pDNA/rhIL-2 NPs. PTX was quickly released from MT/PTX/pDNA/rhIL-2 NPs under simulated intracellular reductive conditions (10 mM GSH), and the release rate reached up to 98% at 24 h, significantly higher than that of GSH-free conditions (31%). In addition, the tumor inhibition rate of MT/PTX/pDNA/rhIL-2 NPs reached 98.7% in the H22 tumor-bearing mice model, which was significantly higher than that of the treatment groups using the three drugs alone, indicating that this system could dramatically enhance the antitumor effect [104].
Due to the increased water solubility and non-toxicity as compared to chitosan, chitooligosaccharides (COSs, also known as chitosan oligomers or chitooligomers) received considerable attention for pharmacological and medical applications [117]. Phenylboronic acid (PBA)-modified COS (PBA-COS) was synthesized by Liu et al. and employed to deliver survivin-targeted siRNA (siSur) (Figure 4C). The prepared PBA20%-COS3000/siSur nanoparticles remarkably inhibited the cell proliferation and significantly inhibited the growth and distant metastasis of B16F10 melanoma cells in the melanoma mouse model [105].
David et al. developed a targeted stealth magnetic siRNA nanocarrier (TS-MSN) as a delivery system for delivery of survivin siRNA (Figure 4B) [106,107]. In this system, superparamagnetic iron oxide nanoparticles (SPIONs) was used as the core, while the shell was composed of polyethylene glycol (PEG), anti-HER2 single-chain antibody fragments (scFv), chitosan, poly-L-arginine (PLR), and survivin siRNA. TS-MSN showed an improved cellular internalization and enhanced the anticancer effect of DOX via downregulating survivin expression.
In addition to chitosan, amylose, as another natural polysaccharide polymer, has become an important carrier material in the field of drug delivery by virtue of its rich source of raw materials, good biocompatibility, and low toxicity [118]. Based on the unique properties of amylose, Zhang et al. developed CSP/TPE@siRNA-SP94 NPs for survivin siRNA delivery. Cationized amylose (CA), superparamagnetic iron oxide (SPIO) NPs, and tetraphenylethylene (TPE) self-assembled into nanospheres (CSP/TPE), followed by surface functionalization with S94 peptides and survivin siRNA loading. CSP/TPE@siRNA-SP94 NPs exhibited excellent in vitro and in vivo fluorescence and magnetic resonance imaging (MRI) properties and remarkably inhibited tumor growth in nude mice with Huh-7 tumors [108].
These innovative strategies not only effectively address the limitations of chitosan in drug delivery but also provide new research ideas and development directions for the application of chitosan and its derivatives in the biomedical field.

4.2.2. Protein

Among the natural polymer systems, protein-based polymers (e.g., silk protein, fish protein, collagen, etc.) reduce the toxicity and immunogenicity of drugs by virtue of their excellent biocompatibility and biodegradability. Meanwhile, their rich functional groups can be coupled with drugs to realize precise drug delivery, showing significant application advantages [119] (Table 3).
Norouzi et al. prepared silk fibroin nanoparticles (SFNPs) by extracting silk fibroin protein from silkworms, then modified the surface of SFNPs with PEI through electrostatic interaction to enhance the binding capacity of the carrier with siRNA. Subsequently, by optimizing the ratio of SFNPs to DOX, they successfully constructed a DOX/siRNA/PEI/SFNPs nanodelivery system. This nanodelivery system had a significant antitumor effect in a 4T1 tumor-bearing mice model, with a tumor growth inhibition rate of 84.19%, and did not cause significant weight loss or other systemic toxicity, providing a new strategy for breast cancer treatment [109].
As a cationic peptide rich in arginine residues, protamine can bind to negatively charged nucleic acids to form stable complexes through electrostatic interactions, thus effectively protecting them from nuclease degradation [120]. Currently, the clinical safety of protamine has been verified and has shown excellent biocompatibility [106]. Based on the properties of protamine, Xu et al. developed an aptamer-protamine-siRNA nanoparticle (APR), which consists of protamine, ErbB3 aptamer, and siSurvivin, for targeting and recognizing ErbB3-positive breast cancer cells. In this system, the ErbB3 aptamer can specifically recognize and bind to the ErbB3 receptor overexpressed on the surface of breast cancer cells, realizing the precise delivery of siRNA. Meanwhile, protamine not only enhances the stability of the complex but also improves the intracellular delivery efficiency of siRNA through the electrostatic binding of siSurvivin. The apoptosis rate of cells is significantly increased to 37.90% after APR treatment as compared to 4.15% of the control group. APR nanoparticles demonstrated a significant therapeutic effect in the MCF-7-bearing nude mice model [110]. In another study, Ma et al. further improved the specific enrichment of the nanocarriers in tumor tissues by loading both siSurvivin and Pt (IV) prodrugs using protamine/hyaluronic acid nanocarriers, Pt (IV) was integrated into the nanoparticles through chemical conjugation with the coating polymer polyglutamic acid (PGA). Surface modification of the nanocarriers by PGA effectively enhanced their stability and prolonged the circulation time in blood. In a nude mouse A549/DDP tumor model, this delivery system achieved 82.46% tumor growth inhibition [111].

4.2.3. Others

In addition to polysaccharides and protein-based natural polymers, other natural materials have been widely used in the design of delivery systems [121] (Table 3). For example, Liu et al. conjugated the carboxyl group of lysine (-COOH) with hydrazine (-NH-NH2) to form lysine-hydrazine, which was subsequently polymerized with HG-CA monomer under acidic conditions (pH 5.0) to form a biodynamic polymer (Lys-biodynamer) with dynamic covalent bonds. This polymer is pH-responsive, capable of releasing siSurvivin in acidic environments, has good biocompatibility and low cytotoxicity, and has dynamic covalent bonds such as imine and acyl-hydrazone bonds in its structure that can be reversibly broken and reorganized in specific environments. The adaptability of the polymer in different environments enables it to remain stable in complex biological environments while releasing the drug when needed to achieve controlled release [112]. Han et al. linked phenylalanine dipeptide (FF) with PEG–Arg–Ala (PEG–RA) sequence at the N-terminus to improve its hydrophilicity and serum stability. Arginine is frequently present in cell-penetrating peptides, which can enhance cellular uptake. Alanine serves as a spacer, which effectively avoids the steric hindrance [113].
Zhao et al. designed VE-Su-Sper/DSPE-PEG2000/siSurvivin nanocomplexes by taking advantage of the properties of spermine and vitamin E. Spermine is able to specifically interact with the major groove of the siRNA helix, while vitamin E enabled the complexes to interact with the structural domains of lipid rafts on the cellular membranes, which effectively avoided lysosomal trapping and improved the intracellular delivery efficiency of siRNA. This nanocomplex was able to effectively silence the Eg5 and Survivin genes and arrest the cell cycle at the G2/M phase. VE-Su-Sper/DSPE-PEG2000/siSurvivin nanocomplex significantly inhibited the tumor growth in HepG2-tumor-bearing mice, and the tumor volume and weight were about 4-fold smaller than other groups, and survivin expression was significantly reduced in tumor tissues [114].

4.3. Targeted Delivery of Survivin siRNA by Liposomal Polymer Nanoparticles

Lipid polymer hybrid nanoparticles (LPHNPs) are hybrid delivery systems with a polymeric core enclosed by a lipidic shell [122]. Integrating the structural properties of liposomes and polymers. The polymeric core is commonly composed of polylactic acid (PLA), polycaprolactone (PCL), pluronic F-68, chitosan, etc., while the lipid shell is made up of myristic acid, phosphatidylcholine (PC), cholesterol, 1,2-dipalmitoylsn-glycero-3-phosphocholine (DPPC), stearic acid, 1,2-distearoyl-sn-glycero-3-phosphoethanolamine (DSPE), soya phosphatidylcholine (SPC), and 1,2-dilauroyl-sn-glycero-3-phosphocholine (DLPC). LPHNPs combine the good structural integrity, storage stability, and controlled release quality of polymeric cores with the great biocompatibility and bioavailability of lipid layers [71].
CD44 is a non-kinase transmembrane glycoprotein, and its high expression is associated with enhanced migration and invasion of tumor cells and resistance to chemotherapy [123]. Overexpression of HER2, a tyrosine kinase transmembrane glycoprotein, can lead to abnormal proliferation and enhanced invasiveness of tumor cells in breast cancer patients, which accelerates the tumor progression and leads to poor prognosis [124]. Notably, recurrent breast cancers may show CD44 and HER2 co-expression or single expression patterns. Therefore, dual-targeting strategies against CD44 and HER2 can more comprehensively cover different subtypes of tumor cells and improve the precision and effectiveness of treatment. Chen et al. developed a HER2/CD44-targeted hydrogel nanobot (termed as ALPR) by embedding the cationic liposome DOTAP/DOPE-based nanocomplexes containing pro-apoptotic peptide and survivin siRNA into Herceptin/hyaluronic acid cross-linked nanohydrogels (Herceptin-HA) (Figure 5A). ALPR can efficiently deliver Herceptin, peptide, and survivin siRNA to SKBR-3, MDA-MB-231, and MCF-7 cells. Moreover, ALPR exhibited excellent antitumor effects on SKBR-3 and MDA-MB-231 heterogeneous xenograft models with a tumor growth inhibition higher than 93% [125].
Yang et al. developed an siRNA delivery system, cRGD-PSH-NP, based on a modified polyethyleneimine (PSH) and DSPE-PEG2000-cRGD (Figure 5D). cRGD-PSH-NP loaded with survivin siRNA (cRGD-PSH-NP/S) was made up of egg phosphatidylcholine, cationic PSH, PEGylated lipids, survivin siRNA, and a targeting ligand (cRGD peptide). cRGD-PSH-NP/S markedly downregulated survivin expression both in vitro and in vivo and exhibited potent tumor inhibition (74.71%) in HepG2-bearing nude mice [126]. Bi et al. designed a transferrin (Tf)-functionalized and liposome-based system (Tf-L-SN38/P/siRNA) for codelivery of SN38 prodrug (a topoisomerase inhibitor) and survivin siRNA (Figure 5C). Tf-L-SN38/P/siRNA was more effective than liposomes carrying a single component and induced potent tumor inhibition (76.8%) in HeLa cell xenograft tumor-bearing nude mice [127]. Gibson prepared switchable lipid nanoparticles (switchable LNP) using CSL, DSPC, cholesterol, and DSPE-PEG2000 by microfluidics and manual extrusion methods, and LNP was used for survivin siRNA complexation to obtain siLNP. They found that survivin downregulation by siLNP enhanced the cytotoxicity of carboplatin and melphalan to Y79 and primary retinoblastoma cells [128].
In addition to being modified with synthetic polymers, liposomes can also be decorated with polysaccharides such as chitosan [129]. Chen et al. constructed a stimuli-responsive polysaccharide-enveloped liposome by layer-by-layer deposition of chitosan (CS) and hyaluronic acid (HA) onto the liposome and then loading survivin-shRNA gene and hyaluronidase (HAase) sequentially (Figure 5B). The as-prepared HA/HAase/CS/liposome/shRNA (HCLR) In the HA/HAase/CS/liposome/survivin-shRNA (HCLR) system, HA promoted tumor targeting by virtue of its specific binding to the CD44 receptor on the surface of tumor cells, resulting in much higher tumor accumulation than unmodified liposomes, and prolonging the retention time of liposomes in the blood circulation due to its negative charge. CS undergoes protonation in the tumor microenvironment (pH 6.5) to promote cellular uptake, resulting in a cellular uptake higher than 85%, which was far greater than pH 7.4. Meanwhile, pH-triggered HAase release decreased the extracellular matrix density and improved the diffusion of nanocarriers into tumor tissues, accounting for the better tumor penetration ability of HCLR. HCLR exhibited superior in vivo tumor suppression capability via downregulation of survivin and triggering cell apoptosis, and no visible damage was observed in normal tissues [130]. Similarly, Eljack’s team designed lipid-based nanoparticles (LBNP) for the co-delivery of tyrosine kinase inhibitor (TKI) Lapatinib (LAPA) and survivin siRNA (siSurvivin). This nanocarrier is based on lipid nanocapsules (LNCs) coated with a cationic chitosan shell. Chitosan was grafted on the surface of LNCs via a transacylation reaction. The siSurvivin-LAPA_LBNP had favorable physicochemical properties, showed a high cellular uptake in human epidermal growth factor receptor 2 overexpressed (HER2+) breast cancer cell line SK-BR-3, and exerted a significant synergistic cytotoxic effect as compared to siCtrl.-LAPA_LBNP. The IC50 value of siSurvivin-LAPA_LBNP to SK-BR-3 cells was 76.8 nM, which is much lower than LAPA ditosylate (159.0 nM) and siCtrl.-LAPA_LBNP (99.7 nM) [131]. In addition, Xie et al. developed a one-pot modular assembly strategy by combining octreotide, a cell-penetrating peptide (CPP), and glutamic acid. The three modules were assembled on the surface of the siRNA/liposome complex to obtain a multifunctional integrated survivin siRNA delivery system (OCA-CC). OCA-CC demonstrated enhanced cytosolic delivery in three tumor cells (A549, MCF-7 and PANC-1) and displayed remarkable antitumor efficacy in vitro and in vivo via silencing survivin expression [132].
It can be realized that the design of lipid-polymer hybrids breaks through the limitations of the traditional delivery system and significantly improves the targeting and cellular uptake efficiency of the drug delivery systems, which provides a new strategy for the design of future drug delivery systems (Table 4).
Figure 5. (A) Schematic illustration of the preparation of ALPR by embedding LPR into Herceptin-HA. Reprinted with permission from [125]. Copyright 2023, Wiley-VCH Verlag; (B) Schematic of HCLR Nanocarrier Fabrication and the In Vivo Fate in Breast Tumor Targeting Gene Delivery. Reprinted with permission from [130]. Copyright 2020, American Chemical Society; (C) The liposome structure and effect mechanisms of Tf-L-SN38/P/siRNA. Reprinted with permission from [127]. Copyright 2018, Dove Medical Press; (D) The structural formulae of DSPE-PEG 2000-cRGD, PSH, and schematic diagram of the structure of cRGD-PSH-NP and its application for siRNA delivery to cancer cells. (a) Structural formulae of DSPE-PEG2000-cRGD. (b) Structural formulae of PSH. (c) Schematic diagram of the structure of cRGD-PSH-NP and its application for siRNA delivery to cancer cells. Reprinted with permission from [126]. Copyright 2021, Taylor and Francis.
Figure 5. (A) Schematic illustration of the preparation of ALPR by embedding LPR into Herceptin-HA. Reprinted with permission from [125]. Copyright 2023, Wiley-VCH Verlag; (B) Schematic of HCLR Nanocarrier Fabrication and the In Vivo Fate in Breast Tumor Targeting Gene Delivery. Reprinted with permission from [130]. Copyright 2020, American Chemical Society; (C) The liposome structure and effect mechanisms of Tf-L-SN38/P/siRNA. Reprinted with permission from [127]. Copyright 2018, Dove Medical Press; (D) The structural formulae of DSPE-PEG 2000-cRGD, PSH, and schematic diagram of the structure of cRGD-PSH-NP and its application for siRNA delivery to cancer cells. (a) Structural formulae of DSPE-PEG2000-cRGD. (b) Structural formulae of PSH. (c) Schematic diagram of the structure of cRGD-PSH-NP and its application for siRNA delivery to cancer cells. Reprinted with permission from [126]. Copyright 2021, Taylor and Francis.
Polymers 17 02279 g005

4.4. Targeted Delivery of Survivin siRNA by Organic/Inorganic Hybrid Nanomaterials

Nanohybrids integrate the biocompatibility of organic polymers/peptides and diverse functionalities of inorganic nanomaterials, and these nanohybrids have been widely employed in various biomedical applications, including drug delivery and cancer immunotherapy [17]. Calcium phosphate (CaP) nanoparticles have been developed for DNA and siRNA delivery due to the simplicity of preparation, intrinsic biocompatibility, and biodegradability [133]. For example, the CaP nanoparticle-based siSurvivin delivery system designed by Mitrach et al. achieved highly efficient loading of siRNA through electrostatic binding and acidic environment-triggered siRNA release. In addition, the introduction of a new terpolymer (o14PEGMA) also improved the dispersion stability of the nanoparticles, enhanced the cellular uptake efficiency, and significantly improved the delivery performance of the vector while reducing the potential cytotoxicity of the conventional CaP [134]. Kara et al. modified the CaP nanoparticles with arginine to simultaneously bind two siRNAs against survivin and cyclin B1. CaP-Arg-siRNAs significantly suppressed the expression of survivin and cyclin B1, resulting in a marked decrease in cell growth and induced apoptosis drastically [135]. Bi et al. synthesized RGDV-functionalized nanodiamond NDCONH(CH2)2NH-VDGR for survivin siRNA delivery. The NDCONH(CH2)2NH-VDGR/survivin-siRNA nanoparticle reduced survivin expression and displayed a potent inhibitory effect on tumor growth in vitro and in vivo [136].
Carbon nanotubes (CNTs), a nanoscale tubular structured material formed by carbon atoms arranged in a hexagonal pattern, have been widely used in biomedical applications due to their excellent thermal and optical properties. CNTs are able to efficiently convert near-infrared light into heat energy for photothermal therapy (PTT) and can be utilized to efficiently load different drugs in a covalent or non-covalent manner [137]. However, CNTs still face many challenges in biomedical applications, such as their inherent cytotoxicity and immunoreactivity as well as their dispersion in physiological environments, which limit their applications to some extent [138]. Therefore, to overcome these challenges and further enhance the potential of CNTs in biomedical applications, Zhao’s team introduced temperature-sensitive lipid peptide lipid (PL) and sucrose laurate (SL) to modify the single-walled carbon nanotubes (SCNTs) and multi-walled carbon nanotubes (MCNTs) for the delivery of survivin siRNA, and the modified CNTs demonstrated excellent biocompatibility and temperature-sensitive properties. Agarose gel electrophoresis showed that siRNA release rates from SCNT-PS/siRNA and MCNT-PS/siRNA were 80% and 67%, respectively, between 40 and 43 °C. The modified CNTs (SCNT-PS and MCNT-PS) did not show significant cytotoxicity to both HeLa cells and MCR-5 cells at high concentrations and had good cytocompatibility. Meanwhile, the combination of photothermal therapy (PTT) and gene therapy (GT) triggered substantial apoptosis in HeLa cells, with apoptosis rates of 63.8% and 60.2% for the SCNT-PS and the MCNT-PS groups, respectively. More importantly, SCNT-PS/siRNA and MCNT-PS/siRNA combined with PTT exhibited strong in vivo antitumor activity in nude mice with HeLa tumor xenografts, and the tumors disappeared completely in two mice in the SCNT-PS+P+G group [139].
Nanoscale metal–organic frameworks (MOFs) have shown high radio-enhancement effects by a unique radiotherapy-radiodynamic therapy mechanism. Ma et al. developed a hafnium-based cationic metal–organic layer (CMOL) for the delivery of siRNA cocktails targeting survivin, HIF-1α, and TGF-β to enhance the effect of cancer radiotherapy and to overcome the radioresistance of tumor cells. In this system, CMOL functions as an effective reactive oxygen species (ROS) generator, and the cationic framework serves as an siRNA cocktail carrier to synergistically downregulate survivin levels. It was shown that the cellular uptake of CMOL@ siRNA in 4T1 cells increased by almost 10-fold as compared to free siRNA. In combination with low-dose X-ray irradiation, CMOL@ siRNA exerted remarkable antitumor efficacy with 96.9% and 91.4% tumor growth inhibition in subcutaneous CT26 and 4T1 tumor models, respectively [140]. Cai et al. designed and prepared a cell-penetrating peptide (CPP)-functionalized metal–organic framework nanoplatform (termed PEG-CPP33@NPs) for co-delivery of oridonin (ORI) and survivin siRNA. The results showed that PEG-CPP33@NPs significantly increased the cellular uptake efficiency of siRNA in A549 cells and showed stronger tumor accumulation as compared with unmodified ZIF-90. In addition, PEG-CPP33@NPs induced a much higher apoptosis rate (60.73%) in A549 cells than unmodified siRNA@ZIF-90 (30.13%) and displayed the strongest in vivo antitumor effect with a tumor inhibition rate of 61.04% via co-delivery of ORI and survivin siRNA [141]. Additionally, Wan et al. achieved specific recognition and binding of the nanocarrier to the asialoglycoprotein receptor (ASGPR) on the surface of hepatocellular carcinoma (HCC) cells by modifying the surface of the metal–organic framework (MOF) with N-acetylgalactosamine (GalNAc), thereby enabling efficient delivery of survivin siRNA. Both in vitro and in vivo experimental results demonstrated that GalNAc-modified MOFs could specifically accumulate in HCC tumor tissues and were efficiently internalized by HCC cells. The protective effect of MOFs effectively enhanced the stability of siRNA, significantly downregulating survivin expression in HCC tumors, thereby inhibiting cell proliferation and inducing apoptosis to achieve tumor growth suppression [142].
All these studies have shown that the combination of inorganic materials/metal frameworks and polymers not only bridges the gap between the two but also significantly improves the overall performance of the nano-delivery system, which provides an effective strategy for overcoming radioresistance and enhancing the efficacy of radiotherapy (Table 5).

5. Conclusions

In summary, the survivin siRNA delivery system based on various polymers effectively overcomes the limitations of siRNA, such as poor stability, easy degradation, immunogenicity, and low delivery efficiency. By constructing various polymer-based nano-delivery systems, such as the synthetic polymer nano-delivery system with PEI as the core, the natural polymer nano-delivery system with chitosan as the core, and the composite delivery system combining lipids/inorganic materials/metals and polymers as introduced in this paper, not only did they improve the stability and delivery efficiency of siRNAs, but they also reduced the cytotoxicity of the delivery system, which made the survivin siRNA delivery system more effective. Survivin siRNA can exert its antitumor effect more effectively and inhibit the proliferation, invasion, and drug resistance of tumor cells, providing new strategies and ideas for cancer treatment. Meanwhile, the multi-drug combination therapy strategy shows a broad prospect in the field of tumor treatment. Combining the survivin siRNA-based gene therapy with various therapeutic approaches, such as chemotherapy, radiotherapy, immunotherapy, and multimodal imaging technology, it is expected to overcome the limitations of single therapy and yield new breakthroughs and hope for cancer treatment.

Author Contributions

Conceptualization, G.-B.D.; software, L.R., S.W., and B.-C.L.; validation, G.-B.D. and L.R.; formal analysis, L.R., S.W., G.-B.D., and B.-C.L.; resources, G.-B.D. and B.-C.L.; data curation, L.R. and S.W.; writing—original draft preparation, L.R., S.W., and G.-B.D.; writing—review and editing, G.-B.D. and B.-C.L.; visualization, L.R. and S.W.; supervision, G.-B.D. and B.-C.L.; project administration, G.-B.D. and B.-C.L.; funding acquisition, G.-B.D. All authors have read and agreed to the published version of the manuscript.

Funding

The authors would like to acknowledge the financial support from the National Natural Science Foundation of China (32460246 to G.-B.D.), the Central Guiding Local Science and Technology Development Fund (No. YDZJSX20231A004 to G.-B.D.), and the Natural Science Foundation Project of Inner Mongolia Autonomous Region (No. 2024MS03065 to G.-B.D.).

Institutional Review Board Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

AICAR5-Aminoimidazole-4-carboxamide ribonucleotide
AIEAggregation-induced emission
AIFApoptosis-inducing factor
ALPRHER2/CD44-targeted hydrogel nanobot
ASOsAntisense oligonucleotides
CACationized amylose
cIAP1Cellular inhibitor of apoptosis protein 1
cIAP2Cellular inhibitor of apoptosis protein 2
COSChitooligosaccharides
CPCChromosal passenger complex
CPPCell penetrating peptide
DDPCis-Diaminodichloroplatinum/Cisplatin
DIABLODirect inhibitor of apoptosis-binding proteins with low pI
DLPC1,2-dilauroyl-.sn-glycero-3-phosphocholine
DOXDoxorubicin
DPPC1,2-dipalmitoylsn-glycero-3-phosphocholine
DSPE1,2-distearoyl-sn-glycero-3-phosphoethanolamine
ECVsExtra cellular vesicles
HAHyaluronic acid
HCCHepatocellular carcinoma
IAP Inhibitor of apoptosis proteins
INCENPInner centromere protein
LALinolic acid
LNCsLipid nanocapsules
LNPsLiquid nanoparticles
LPHNPsLipid polymer hybrid nanoparticles
MAMercaptobenzoic acid
ML-IAPMelanoma inhibitor of apoptosis protein
MTSMitochondrial targeting sequence
NAIPNeuronal apoptosis inhibitory protein
NESNuclear export signal
OAOleic acid
ORIOridonin
PAPalmitic acid
PAMAMPolyamide-amine
PBAPhenylboronic acid
PCPhosphatidyl choline
PCLPolycaprolactone
PEG-CSPoly (ethylene glycol)-modified chitosan
PEG-PAspPoly(ethylene glycol)-poly (aspartic acid)
PEIPolyethyleneimine
PEI–PLAPolyehtyleneimine-poly(lactic acid)
PGAPolyglutamic acid
PMPolymeric micelles
PNPsPolymer nanoparticles
PSCAProstate stem cell antigens
PSHPEI–SS–HA
PTTPhotothermal therapy
PTXPachitaxel
RNAiRNA interference
ROSReactive oxygen species
SACSpindle assembly check point
SFNPsSilk fibroin nanoparticles
siRNASmall interfering RNA
SMACSecond mitochondria derived activator of caspases
SPCSoya phosphatidylcholine
SPIOSuperparamagnetic iron oxide
SurvivinBIRC5
TEPATetraethylenepentamine
TKITyrosine kinase inhibitor
TMCN, N, N-trimethyl chitosan
TPETetraphenylethylene
XIAPX-linked inhibitor of apoptosis protein

References

  1. Kiri, S.; Ryba, T. Cancer, metastasis, and the epigenome. Mol. Cancer 2024, 23, 154. [Google Scholar] [CrossRef] [PubMed]
  2. Brown, J.S.; Amend, S.R.; Austin, R.H.; Gatenby, R.A.; Hammarlund, E.U.; Pienta, K.J. Updating the Definition of Cancer. Mol. Cancer Res. 2023, 21, 1142–1147. [Google Scholar] [CrossRef]
  3. Judasz, E.; Lisiak, N.; Kopczyński, P.; Taube, M.; Rubiś, B. The Role of Telomerase in Breast Cancer’s Response to Therapy. Int. J. Mol. Sci. 2022, 23, 12844. [Google Scholar] [CrossRef]
  4. Granja, A.; Pinheiro, M.; Reis, S. Epigallocatechin Gallate Nanodelivery Systems for Cancer Therapy. Nutrients 2016, 8, 307. [Google Scholar] [CrossRef] [PubMed]
  5. Chen, Z.; Kankala, R.K.; Long, L.; Xie, S.; Chen, A.; Zou, L. Current understanding of passive and active targeting nanomedicines to enhance tumor accumulation. Coord. Chem. Rev. 2023, 481, 215051. [Google Scholar] [CrossRef]
  6. Genc, S.; Taghizadehghalehjoughi, A.; Yeni, Y.; Jafarizad, A.; Hacimuftuoglu, A.; Nikitovic, D.; Docea, A.O.; Mezhuev, Y.; Tsatsakis, A. Fe3O4 Nanoparticles in Combination with 5-FU Exert Antitumor Effects Superior to Those of the Active Drug in a Colon Cancer Cell Model. Pharmaceutics 2023, 15, 245. [Google Scholar] [CrossRef]
  7. Xu, J.; Chen, G.; Mo, C.; Sha, Y.; Luo, S.; Ou, M. Development and evaluation of siRNA-mediated gene silencing strategies for ADO2 therapy utilizing iPSCs model and DMPC-SPIONs delivery system. Stem Cell Res. Ther. 2025, 16, 66. [Google Scholar] [CrossRef]
  8. Jaiswal, P.K.; Goel, A.; Mittal, R.D. Survivin: A molecular biomarker in cancer. Ind. J. Med. Res. 2015, 141, 389–397. [Google Scholar]
  9. Dong, Y.; Siegwart, D.J.; Anderson, D.G. Strategies, design, and chemistry in siRNA delivery systems. Adv. Drug Deliv. Rev. 2019, 144, 133–147. [Google Scholar] [CrossRef]
  10. Gorabi, A.M.; Kiaie, N.; Aslani, S.; Jamialahmadi, T.; Johnston, T.P.; Sahebkar, A. Prospects for the potential of RNA interference in the treatment of autoimmune diseases: Small interfering RNAs in the spotlight. J. Autoimmun. 2020, 114, 102529. [Google Scholar] [CrossRef] [PubMed]
  11. Sajid, M.I.; Moazzam, M.; Kato, S.; Yeseom Cho, K.; Tiwari, R.K. Overcoming Barriers for siRNA Therapeutics: From Bench to Bedside. Pharmaceuticals 2020, 13, 294. [Google Scholar] [CrossRef]
  12. Sun, Y.; Li, B.; Cao, Q.; Liu, T.; Li, J. Targeting cancer stem cells with polymer nanoparticles for gastrointestinal cancer treatment. Stem Cell Res. Ther. 2022, 13, 489. [Google Scholar] [CrossRef]
  13. Kozielski, K.L.; Tzeng, S.Y.; Green, J.J. Bioengineered nanoparticles for siRNA delivery. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2013, 5, 449–468. [Google Scholar] [CrossRef] [PubMed]
  14. Velluto, D.; Thomas, S.N.; Simeoni, E.; Swartz, M.A.; Hubbell, J.A. PEG-b-PPS-b-PEI micelles and PEG-b-PPS/PEG-b-PPS-b-PEI mixed micelles as non-viral vectors for plasmid DNA: Tumor immunotoxicity in B16F10 melanoma. Biomaterials 2011, 32, 9839–9847. [Google Scholar] [CrossRef] [PubMed]
  15. Dong, S.; Zhou, X.; Yang, J. TAT modified and lipid—PEI hybrid nanoparticles for co-delivery of docetaxel and pDNA. Biomed. Pharmacother. 2016, 84, 954–961. [Google Scholar] [CrossRef]
  16. Sun, T.; Jiang, C. Stimuli-responsive drug delivery systems triggered by intracellular or subcellular microenvironments. Adv. Drug Deliv. Rev. 2023, 196, 114773. [Google Scholar] [CrossRef]
  17. Li, J.; Lu, W.; Yang, Y.; Xiang, R.; Ling, Y.; Yu, C.; Zhou, Y. Hybrid Nanomaterials for Cancer Immunotherapy. Adv. Sci. 2023, 10, e2204932. [Google Scholar] [CrossRef]
  18. Sabour, R.; Harras, M.F.; Mohamed Al Kamaly, O.; Altwaijry, N. Discovery of Novel 3-Cyanopyridines as Survivin Modulators and Apoptosis Inducers. Molecules 2020, 25, 4892. [Google Scholar] [CrossRef] [PubMed]
  19. Kumar, S.; Fairmichael, C.; Longley, D.B.; Turkington, R.C. The Multiple Roles of the IAP Super-family in cancer. Pharmacol. Ther. 2020, 214, 107610. [Google Scholar] [CrossRef]
  20. Martínez-Sifuentes, M.A.; Bassol-Mayagoitia, S.; Nava-Hernández, M.P.; Ruiz-Flores, P.; Ramos-Treviño, J.; Haro-Santa Cruz, J.; Hernández-Ibarra, J.A. Survivin in Breast Cancer: A Review. Genet. Test. Mol. Biomark. 2022, 26, 411–421. [Google Scholar] [CrossRef]
  21. Wheatley, S.P.; Altieri, D.C. Survivin at a glance. J. Cell Sci. 2019, 132, jcs223826. [Google Scholar] [CrossRef] [PubMed]
  22. Albadari, N.; Li, W. Survivin Small Molecules Inhibitors: Recent Advances and Challenges. Molecules 2023, 28, 1376. [Google Scholar] [CrossRef] [PubMed]
  23. Frassanito, M.A.; Saltarella, I.; Vinella, A.; Muzio, L.L.; Pannone, G.; Fumarulo, R.; Vacca, A.; Mariggiò, M.A. Survivin overexpression in head and neck squamous cell carcinomas as a new therapeutic target (Review). Oncol. Rep. 2019, 41, 2615–2624. [Google Scholar] [CrossRef]
  24. Hagenbuchner, J.; Oberacher, H.; Arnhard, K.; Kiechl-Kohlendorfer, U.; Ausserlechner, M.J. Modulation of Respiration and Mitochondrial Dynamics by SMAC-Mimetics for Combination Therapy in Chemoresistant Cancer. Theranostics 2019, 9, 4909–4922. [Google Scholar] [CrossRef]
  25. Cheung, C.H.A.; Chang, Y.C.; Lin, T.Y.; Cheng, S.M.; Leung, E. Anti-apoptotic proteins in the autophagic world: An update on functions of XIAP, Survivin, and BRUCE. J. Biomed. Sci. 2020, 27, 31. [Google Scholar] [CrossRef]
  26. Park, S.H.; Shin, I.; Park, S.H.; Kim, N.D.; Shin, I. An Inhibitor of the Interaction of Survivin with Smac in Mitochondria Promotes Apoptosis. Chem. Asian J. 2019, 14, 4035–4041. [Google Scholar] [CrossRef]
  27. Santhanam, M.; Kumar Pandey, S.; Shteinfer-Kuzmine, A.; Paul, A.; Abusiam, N.; Zalk, R.; Shoshan-Barmatz, V. Interaction of SMAC with a survivin-derived peptide alters essential cancer hallmarks: Tumor growth, inflammation, and immunosuppression. Mol. Ther. 2024, 32, 1934–1955. [Google Scholar] [CrossRef]
  28. Mahmoudian-Sani, M.R.; Alghasi, A.; Saeedi-Boroujeni, A.; Jalali, A.; Jamshidi, M.; Khodadadi, A. Survivin as a diagnostic and therapeutic marker for thyroid cancer. Pathol. Res. Pract. 2019, 215, 619–625. [Google Scholar] [CrossRef]
  29. Shakeri, R.; Kheirollahi, A.; Davoodi, J. Apaf-1: Regulation and function in cell death. Biochimie 2017, 135, 111–125. [Google Scholar] [CrossRef]
  30. Liu, Y.; Lear, T.; Iannone, O.; Shiva, S.; Corey, C.; Rajbhandari, S.; Jerome, J.; Chen, B.B.; Mallampalli, R.K. The Proapoptotic F-box Protein Fbxl7 Regulates Mitochondrial Function by Mediating the Ubiquitylation and Proteasomal Degradation of Survivin. J. Biol. Chem. 2015, 290, 11843–11852. [Google Scholar] [CrossRef]
  31. Kim, S.; Lee, M.; Kim, N.Y.; Kwon, Y.S.; Nam, G.S.; Lee, K.; Kwon, K.M.; Kim, D.K.; Hwang, I.H. Oxidative tryptamine dimers from Corynebacterium durum directly target survivin to induce AIF-mediated apoptosis in cancer cells. Biomed. Pharmacother. 2024, 173, 116335. [Google Scholar] [CrossRef]
  32. Novo, N.; Ferreira, P.; Medina, M. The apoptosis-inducing factor family: Moonlighting proteins in the crosstalk between mitochondria and nuclei. IUBMB Life 2021, 73, 568–581. [Google Scholar] [CrossRef] [PubMed]
  33. Kang, B.H.; Xia, F.; Pop, R.; Dohi, T.; Socolovsky, M.; Altieri, D.C. Developmental control of apoptosis by the immunophilin aryl hydrocarbon receptor-interacting protein (AIP) involves mitochondrial import of the survivin protein. J. Biol. Chem. 2011, 286, 16758–16767. [Google Scholar] [CrossRef]
  34. Siragusa, G.; Tomasello, L.; Giordano, C.; Pizzolanti, G. Survivin (BIRC5): Implications in cancer therapy. Life Sci. 2024, 350, 122788. [Google Scholar] [CrossRef]
  35. Chen, Y.H.; Chen, H.H.; Wang, W.J.; Chen, H.Y.; Huang, W.S.; Kao, C.H.; Lee, S.R.; Yeat, N.Y.; Yan, R.L.; Chan, S.J.; et al. TRABID inhibition activates cGAS/STING-mediated anti-tumor immunity through mitosis and autophagy dysregulation. Nat. Commun. 2023, 14, 3050. [Google Scholar] [CrossRef]
  36. Vader, G.; Kauw, J.J.; Medema, R.H.; Lens, S.M. Survivin mediates targeting of the chromosomal passenger complex to the centromere and midbody. EMBO Rep. 2006, 7, 85–92. [Google Scholar] [CrossRef]
  37. Lyu, H.; Huang, J.; He, Z.; Liu, B. Epigenetic mechanism of survivin dysregulation in human cancer. Sci. China Life Sci. 2018, 61, 808–814. [Google Scholar] [CrossRef]
  38. Li, F.; Aljahdali, I.; Ling, X. Cancer therapeutics using survivin BIRC5 as a target: What can we do after over two decades of study? J. Exp. Clin. Cancer Res. 2019, 38, 368. [Google Scholar] [CrossRef]
  39. Venkatesan, N.; Kanwar, J.R.; Deepa, P.R.; Navaneethakrishnan, S.; Joseph, C.; Krishnakumar, S. Targeting HSP90/Survivin using a cell permeable structure based peptido-mimetic shepherdin in retinoblastoma. Chem. Biol. Interact. 2016, 252, 141–149. [Google Scholar] [CrossRef]
  40. Daglioglu, C.; Okutucu, B. Therapeutic Effects of AICAR and DOX Conjugated Multifunctional Nanoparticles in Sensitization and Elimination of Cancer Cells via Survivin Targeting. Pharm. Res. 2017, 34, 175–184. [Google Scholar] [CrossRef]
  41. Wang, Q.; Arnst, K.E.; Xue, Y.; Lei, Z.N.; Ma, D.; Chen, Z.S.; Miller, D.D.; Li, W. Synthesis and biological evaluation of indole-based UC-112 analogs as potent and selective survivin inhibitors. Eur. J. Med. Chem. 2018, 149, 211–224. [Google Scholar] [CrossRef]
  42. Ibrahim, T.M.; Ernst, C.; Lange, A.; Hennig, S.; Boeckler, F.M. Small-Molecule Intervention At The Dimerization Interface Of Survivin By Novel Rigidized Scaffolds. Drug Des. Devel Ther. 2019, 13, 4247–4263. [Google Scholar] [CrossRef]
  43. Peery, R.; Kyei-Baffour, K.; Dong, Z.; Liu, J.; de Andrade Horn, P.; Dai, M.; Liu, J.Y.; Zhang, J.T. Synthesis and Identification of a Novel Lead Targeting Survivin Dimerization for Proteasome-Dependent Degradation. J. Med. Chem. 2020, 63, 7243–7251. [Google Scholar] [CrossRef]
  44. Ling, X.; Wu, W.; Aljahdali, I.A.M.; Liao, J.; Santha, S.; Fountzilas, C.; Boland, P.M.; Li, F. FL118, acting as a ‘molecular glue degrader’, binds to dephosphorylates and degrades the oncoprotein DDX5 (p68) to control c-Myc, survivin and mutant Kras against colorectal and pancreatic cancer with high efficacy. Clin. Transl. Med. 2022, 12, e881. [Google Scholar] [CrossRef]
  45. Yin, H.; Que, R.; Liu, C.; Ji, W.; Sun, B.; Lin, X.; Zhang, Q.; Zhao, X.; Peng, Z.; Zhang, X.; et al. Survivin-targeted drug screening platform identifies a matrine derivative WM-127 as a potential therapeutics against hepatocellular carcinoma. Cancer Lett. 2018, 425, 54–64. [Google Scholar] [CrossRef]
  46. Cheng, S.M.; Lin, T.Y.; Chang, Y.C.; Lin, I.W.; Leung, E.; Cheung, C.H.A. YM155 and BIRC5 downregulation induce genomic instability via autophagy-mediated ROS production and inhibition in DNA repair. Pharmacol. Res. 2021, 166, 105474. [Google Scholar] [CrossRef]
  47. Hong, M.; Ren, M.Q.; Silva, J.; Paul, A.; Wilson, W.D.; Schroeder, C.; Weinberger, P.; Janik, J.; Hao, Z. YM155 inhibits topoisomerase function. Anticancer. Drugs 2017, 28, 142–152. [Google Scholar] [CrossRef]
  48. Carrasco, R.A.; Stamm, N.B.; Marcusson, E.; Sandusky, G.; Iversen, P.; Patel, B.K. Antisense inhibition of survivin expression as a cancer therapeutic. Mol. Cancer Ther. 2011, 10, 221–232. [Google Scholar] [CrossRef]
  49. Zhang, H.; Vandesompele, J.; Braeckmans, K.; De Smedt, S.C.; Remaut, K. Nucleic acid degradation as barrier to gene delivery: A guide to understand and overcome nuclease activity. Chem. Soc. Rev. 2024, 53, 317–360. [Google Scholar] [CrossRef] [PubMed]
  50. Purewal, J.S.; Doshi, G.M. RNAi in psoriasis: A melodic exploration of miRNA, shRNA, and amiRNA with a spotlight on siRNA. Eur. J. Pharmacol. 2024, 985, 177083. [Google Scholar] [CrossRef] [PubMed]
  51. Chi, X.; Gatti, P.; Papoian, T. Safety of antisense oligonucleotide and siRNA-based therapeutics. Drug Discov. Today 2017, 22, 823–833. [Google Scholar] [CrossRef]
  52. Li, X.; Le, Y.; Zhang, Z.; Nian, X.; Liu, B.; Yang, X. Viral Vector-Based Gene Therapy. Int. J. Mol. Sci. 2023, 24, 7736. [Google Scholar] [CrossRef]
  53. Zhou, Z.; Liu, X.; Zhu, D.; Wang, Y.; Zhang, Z.; Zhou, X.; Qiu, N.; Chen, X.; Shen, Y. Nonviral cancer gene therapy: Delivery cascade and vector nanoproperty integration. Adv. Drug Deliv. Rev. 2017, 115, 115–154. [Google Scholar] [CrossRef]
  54. El Moukhtari, S.H.; Garbayo, E.; Amundarain, A.; Pascual-Gil, S.; Carrasco-León, A.; Prosper, F.; Agirre, X.; Blanco-Prieto, M.J. Lipid nanoparticles for siRNA delivery in cancer treatment. J. Control Release 2023, 361, 130–146. [Google Scholar] [CrossRef]
  55. Thirumurugan, S.; Ramanathan, S.; Muthiah, K.S.; Lin, Y.C.; Hsiao, M.; Dhawan, U.; Wang, A.N.; Liu, W.C.; Liu, X.; Liao, M.Y.; et al. Inorganic nanoparticles for photothermal treatment of cancer. J. Mater. Chem. B 2024, 12, 3569–3593. [Google Scholar] [CrossRef]
  56. Oroojalian, F.; Beygi, M.; Baradaran, B.; Mokhtarzadeh, A.; Shahbazi, M.A. Immune Cell Membrane-Coated Biomimetic Nanoparticles for Targeted Cancer Therapy. Small 2021, 17, e2006484. [Google Scholar] [CrossRef]
  57. Hu, Y.; Zhang, W.; Chu, X.; Wang, A.; He, Z.; Si, C.L.; Hu, W. Dendritic cell-targeting polymer nanoparticle-based immunotherapy for cancer: A review. Int. J. Pharm. 2023, 635, 122703. [Google Scholar] [CrossRef]
  58. Beach, M.A.; Nayanathara, U.; Gao, Y.; Zhang, C.; Xiong, Y.; Wang, Y.; Such, G.K. Polymeric Nanoparticles for Drug Delivery. Chem. Rev. 2024, 124, 5505–5616. [Google Scholar] [CrossRef]
  59. Ziranu, P.; Pretta, A.; Aimola, V.; Cau, F.; Mariani, S.; D’Agata, A.P.; Codipietro, C.; Rizzo, D.; Dell’Utri, V.; Sanna, G.; et al. CD44: A New Prognostic Marker in Colorectal Cancer? Cancers 2024, 16, 1569. [Google Scholar] [CrossRef]
  60. Aibani, N.; Rai, R.; Patel, P.; Cuddihy, G.; Wasan, E.K. Chitosan Nanoparticles at the Biological Interface: Implications for Drug Delivery. Pharmaceutics 2021, 13, 1686. [Google Scholar] [CrossRef]
  61. Mousazadeh, H.; Pilehvar-Soltanahmadi, Y.; Dadashpour, M.; Zarghami, N. Cyclodextrin based natural nanostructured carbohydrate polymers as effective non-viral siRNA delivery systems for cancer gene therapy. J. Control Release 2021, 330, 1046–1070. [Google Scholar] [CrossRef]
  62. Taghipour-Sabzevar, V.; Sharifi, T.; Moghaddam, M.M. Polymeric nanoparticles as carrier for targeted and controlled delivery of anticancer agents. Ther. Deliv. 2019, 10, 527–550. [Google Scholar] [CrossRef]
  63. Liu, Y.; Huang, Y.; He, G.; Guo, C.; Dong, J.; Wu, L. Development of mRNA Lipid Nanoparticles: Targeting and Therapeutic Aspects. Int. J. Mol. Sci. 2024, 25, 10166. [Google Scholar] [CrossRef]
  64. Mainini, F.; Eccles, M.R. Lipid and Polymer-Based Nanoparticle siRNA Delivery Systems for Cancer Therapy. Molecules 2020, 25, 2692. [Google Scholar] [CrossRef]
  65. Bunwatcharaphansakun, P.; Yodsanit, N.; Keaswejjareansuk, W.; Gonil, P.; Wongkhieo, S.; Jantimaporn, A.; Kengkittipat, W.; Kaewmalun, S.; Sawangrat, N.; Yata, T.; et al. Innovative Hybrid Nanocarriers of GnRH Peptide-Modified Chitosan-Coated Lipid Nanoparticles as a Targeted Chemotherapy for Reproductive-Related Cancers. J. Biomed. Mater. Res. A 2025, 113, e37843. [Google Scholar] [CrossRef]
  66. Xu, Y.; Reheman, A.; Feng, W. Recent research progress on metal ions and metal-based nanomaterials in tumor therapy. Front. Bioeng. Biotechnol. 2025, 13, 1550089. [Google Scholar] [CrossRef]
  67. Hou, H.; Liu, X.; Liu, J.; Wang, Y. Carbohydrate polymer-based nanoparticles with cell membrane camouflage for cancer therapy: A review. Int. J. Biol. Macromol. 2025, 289, 138620. [Google Scholar] [CrossRef]
  68. Eftekhari, A.; Kryschi, C.; Pamies, D.; Gulec, S.; Ahmadian, E.; Janas, D.; Davaran, S.; Khalilov, R. Natural and synthetic nanovectors for cancer therapy. Nanotheranostics 2023, 7, 236–257. [Google Scholar] [CrossRef]
  69. Wong, K.H.; Lu, A.; Chen, X.; Yang, Z. Natural Ingredient-Based Polymeric Nanoparticles for Cancer Treatment. Molecules 2020, 25, 3620. [Google Scholar] [CrossRef]
  70. Kuna, K.; Baddam, S.R.; Kalagara, S.; Akkiraju, P.C.; Tade, R.S.; Enaganti, S. Emerging natural polymer-based architectured nanotherapeutics for the treatment of cancer. Int. J. Biol. Macromol. 2024, 262 Pt 1, 129434. [Google Scholar] [CrossRef]
  71. Gajbhiye, K.R.; Salve, R.; Narwade, M.; Sheikh, A.; Kesharwani, P.; Gajbhiye, V. Lipid polymer hybrid nanoparticles: A custom-tailored next-generation approach for cancer therapeutics. Mol. Cancer 2023, 22, 160. [Google Scholar] [CrossRef]
  72. Yang, C.; Lin, Z.I.; Chen, J.A.; Xu, Z.; Gu, J.; Law, W.C.; Yang, J.H.C.; Chen, C.K. Organic/Inorganic Self-Assembled Hybrid Nano-Architectures for Cancer Therapy Applications. Macromol. Biosci. 2022, 22, e2100349. [Google Scholar] [CrossRef]
  73. Xue, L.; Yan, Y.; Kos, P.; Chen, X.; Siegwart, D.J. PEI fluorination reduces toxicity and promotes liver-targeted siRNA delivery. Drug Deliv. Transl. Res. 2021, 11, 255–260. [Google Scholar] [CrossRef]
  74. Warrier, N.M.; Agarwal, P.; Kumar, P. Emerging Importance of Survivin in Stem Cells and Cancer: The Development of New Cancer Therapeutics. Stem Cell Rev. Rep. 2020, 16, 828–852. [Google Scholar] [CrossRef]
  75. Ding, G.B.; Meng, X.; Yang, P.; Li, B.; Stauber, R.H.; Li, Z. Integration of Polylactide into Polyethylenimine Facilitates the Safe and Effective Intracellular siRNA Delivery. Polymers 2020, 12, 445. [Google Scholar] [CrossRef]
  76. Ding, G.-B.; Ma, X.; Meng, X.; Yang, P.; Stauber, R.H.; Li, Z. pH low insertion peptide (pHLIP)-decorated polymeric nanovehicle for efficient and pH-responsive siRNA translocation. Mater. Des. 2021, 212, 110197. [Google Scholar] [CrossRef]
  77. Montazeri Aliabadi, H.; Landry, B.; Mahdipoor, P.; Uludağ, H. Induction of apoptosis by survivin silencing through siRNA delivery in a human breast cancer cell line. Mol. Pharm. 2011, 8, 1821–1830. [Google Scholar] [CrossRef]
  78. Parmar, M.B.; K C, R.B.; Löbenberg, R.; Uludağ, H. Additive Polyplexes to Undertake siRNA Therapy against CDC20 and Survivin in Breast Cancer Cells. Biomacromolecules 2018, 19, 4193–4206. [Google Scholar] [CrossRef]
  79. Hou, Y.C.; Zhang, C.; Zhang, Z.J.; Xia, L.; Rao, K.Q.; Gu, L.H.; Wu, Y.C.; Lv, Z.C.; Wu, H.X.; Zuo, X.L.; et al. Aggregation-Induced Emission (AIE) and Magnetic Resonance Imaging Characteristics for Targeted and Image-Guided siRNA Therapy of Hepatocellular Carcinoma. Adv. Healthc. Mater. 2022, 11, e2200579. [Google Scholar] [CrossRef]
  80. Zhupanyn, P.; Ewe, A.; Büch, T.; Malek, A.; Rademacher, P.; Müller, C.; Reinert, A.; Jaimes, Y.; Aigner, A. Extracellular vesicle (ECV)-modified polyethylenimine (PEI) complexes for enhanced siRNA delivery in vitro and in vivo. J. Control Release 2020, 319, 63–76. [Google Scholar] [CrossRef]
  81. Cao, Y.; Huang, H.Y.; Chen, L.Q.; Du, H.H.; Cui, J.H.; Zhang, L.W.; Lee, B.J.; Cao, Q.R. Enhanced Lysosomal Escape of pH-Responsive Polyethylenimine-Betaine Functionalized Carbon Nanotube for the Codelivery of Survivin Small Interfering RNA and Doxorubicin. ACS Appl. Mater. Interfaces 2019, 11, 9763–9776. [Google Scholar] [CrossRef]
  82. Jin, M.; Jin, G.; Kang, L.; Chen, L.; Gao, Z.; Huang, W. Smart polymeric nanoparticles with pH-responsive and PEG-detachable properties for co-delivering paclitaxel and survivin siRNA to enhance antitumor outcomes. Int. J. Nanomed. 2018, 13, 2405–2426. [Google Scholar] [CrossRef]
  83. Niu, B.; Liao, K.; Zhou, Y.; Wen, T.; Quan, G.; Pan, X.; Wu, C. Application of glutathione depletion in cancer therapy: Enhanced ROS-based therapy, ferroptosis, and chemotherapy. Biomaterials 2021, 277, 121110. [Google Scholar] [CrossRef]
  84. Wang, Y.; Hao, Z.; Zhang, F.; Liu, Z.; Pang, H.; Guo, H.; Liu, J.; Zhang, H.; Zhang, R.; Li, X.; et al. Endogenous Stimuli-Responsive Hollow Manganese Dioxide/Semiconductor Nanostructures for siRNA-Enhanced Chemotherapy. ACS Appl. Nano Mater. 2025, 8, 8751–8760. [Google Scholar] [CrossRef]
  85. Chen, L.; Wang, S.; Liu, Q.; Zhang, Z.; Lin, S.; Zheng, Q.; Cheng, M.; Li, Y.; Cheng, C. Reduction sensitive nanocarriers mPEG-g-γ-PGA/SSBPEI@siRNA for effective targeted delivery of survivin siRNA against NSCLC. Colloids Surf. B Biointerfaces 2020, 193, 111105. [Google Scholar] [CrossRef] [PubMed]
  86. Chen, L.; Luo, J.; Zhang, J.; Wang, S.; Sun, Y.; Liu, Q.; Cheng, C. Dual Targeted Nanoparticles for the Codelivery of Doxorubicin and siRNA Cocktails to Overcome Ovarian Cancer Stem Cells. Int. J. Mol. Sci. 2023, 24, 11575. [Google Scholar] [CrossRef]
  87. Li, H.; Zha, S.; Li, H.; Liu, H.; Wong, K.L.; All, A.H. Polymeric Dendrimers as Nanocarrier Vectors for Neurotheranostics. Small 2022, 18, e2203629. [Google Scholar] [CrossRef] [PubMed]
  88. Wong, K.H.; Guo, Z.; Law, M.K.; Chen, M. Functionalized PAMAM constructed nanosystems for biomacromolecule delivery. Biomater. Sci. 2023, 11, 1589–1606. [Google Scholar] [CrossRef] [PubMed]
  89. Tietze, S.; Schau, I.; Michen, S.; Ennen, F.; Janke, A.; Schackert, G.; Aigner, A.; Appelhans, D.; Temme, A. A Poly(Propyleneimine) Dendrimer-Based Polyplex-System for Single-Chain Antibody-Mediated Targeted Delivery and Cellular Uptake of SiRNA. Small 2017, 13, 1700072. [Google Scholar] [CrossRef]
  90. Jugel, W.; Aigner, A.; Michen, S.; Hagstotz, A.; Ewe, A.; Appelhans, D.; Schackert, G.; Temme, A.; Tietze, S. Targeted RNAi of BIRC5/Survivin Using Antibody-Conjugated Poly(Propylene Imine)-Based Polyplexes Inhibits Growth of PSCA-Positive Tumors. Pharmaceutics 2021, 13, 676. [Google Scholar] [CrossRef]
  91. Salve, R.; Haldar, N.; Shaikh, A.; Samanta, R.; Sengar, D.; Patra, S.; Gajbhiye, V. MUC1 aptamer-tethered H40-TEPA-PEG nanoconjugates for targeted siRNA-delivery and gene silencing in breast cancer cells. Front. Bioeng. Biotechnol. 2024, 12, 1383495. [Google Scholar] [CrossRef]
  92. Zhou, Z.; Feng, Y.; Jiang, M.; Yao, Z.; Wang, J.; Pan, F.; Feng, R.; Zhao, C.; Ma, Y.; Zhou, J.; et al. Ionizable polymeric micelles (IPMs) for efficient siRNA delivery. Nat. Commun. 2025, 16, 360. [Google Scholar] [CrossRef] [PubMed]
  93. Hwang, D.; Ramsey, J.D.; Kabanov, A.V. Polymeric micelles for the delivery of poorly soluble drugs: From nanoformulation to clinical approval. Adv. Drug Deliv. Rev. 2020, 156, 80–118. [Google Scholar] [CrossRef]
  94. Salzano, G.; Riehle, R.; Navarro, G.; Perche, F.; De Rosa, G.; Torchilin, V.P. Polymeric micelles containing reversibly phospholipid-modified anti-survivin siRNA: A promising strategy to overcome drug resistance in cancer. Cancer Lett. 2014, 343, 224–231. [Google Scholar] [CrossRef] [PubMed]
  95. Salzano, G.; Navarro, G.; Trivedi, M.S.; De Rosa, G.; Torchilin, V.P. Multifunctional Polymeric Micelles Co-loaded with Anti-Survivin siRNA and Paclitaxel Overcome Drug Resistance in an Animal Model of Ovarian Cancer. Mol. Cancer Ther. 2015, 14, 1075–1084. [Google Scholar] [CrossRef]
  96. Zaiki, Y.; Iskandar, A.; Wong, T.W. Functionalized chitosan for cancer nano drug delivery. Biotechnol. Adv. 2023, 67, 108200. [Google Scholar] [CrossRef]
  97. Bian, X.; Yu, X.; Lu, S.; Jia, L.; Li, P.; Yin, J.; Tan, S. Chitosan-based nanoarchitectures for siRNA delivery in cancer therapy: A review of pre-clinical and clinical importance. Int. J. Biol. Macromol. 2025, 284 Pt 1, 137708. [Google Scholar] [CrossRef] [PubMed]
  98. Edo, G.I.; Yousif, E.; Al-Mashhadani, M.H. Chitosan: An overview of biological activities, derivatives, properties, and current advancements in biomedical applications. Carbohydr. Res. 2024, 542, 109199. [Google Scholar] [CrossRef]
  99. Dubey, S.K.; Bhatt, T.; Agrawal, M.; Saha, R.N.; Saraf, S.; Saraf, S.; Alexander, A. Application of chitosan modified nanocarriers in breast cancer. Int. J. Biol. Macromol. 2022, 194, 521–538. [Google Scholar] [CrossRef]
  100. Li, X.; Wang, Y.; Feng, C.; Chen, H.; Gao, Y. Chemical Modification of Chitosan for Developing Cancer Nanotheranostics. Biomacromolecules 2022, 23, 2197–2218. [Google Scholar] [CrossRef]
  101. Sader, D.; Zlotver, I.; Moya, S.; Calabrese, G.C.; Sosnik, A. Doubly self-assembled dermatan sulfate/chitosan nanoparticles for targeted siRNA delivery in cancer therapy. J. Colloid Interface Sci. 2025, 680 Pt B, 763–775. [Google Scholar] [CrossRef]
  102. Li, Z.; Zhu, L.; Liu, W.; Zheng, Y.; Li, X.; Ye, J.; Li, B.; Chen, H.; Gao, Y. Near-infrared/pH dual-responsive nanocomplexes for targeted imaging and chemo/gene/photothermal tri-therapies of non-small cell lung cancer. Acta Biomater. 2020, 107, 242–259. [Google Scholar] [CrossRef] [PubMed]
  103. Sun, P.; Huang, W.; Jin, M.; Wang, Q.; Fan, B.; Kang, L.; Gao, Z. Chitosan-based nanoparticles for survivin targeted siRNA delivery in breast tumor therapy and preventing its metastasis. Int. J. Nanomed. 2016, 11, 4931–4945. [Google Scholar] [CrossRef] [PubMed]
  104. Yang, F.; Huang, W.; Li, Y.; Liu, S.; Jin, M.; Wang, Y.; Jia, L.; Gao, Z. Anti-tumor effects in mice induced by survivin-targeted siRNA delivered through polysaccharide nanoparticles. Biomaterials 2013, 34, 5689–5699. [Google Scholar] [CrossRef] [PubMed]
  105. Fang, G.; Hao, P.; Qiao, R.; Liu, B.X.; Shi, X.; Wang, Z.; Sun, P. Stimuli-responsive chitosan based nanoparticles in cancer therapy and diagnosis: A review. Int. J. Biol. Macromol. 2024, 283 Pt 3, 137709. [Google Scholar] [CrossRef]
  106. Zhang, L.; Li, Q.; Chen, J.; Tang, C.; Yin, C. Enhanced antitumor efficacy of glutathione-responsive chitosan based nanoparticles through co-delivery of chemotherapeutics, genes, and immune agents. Carbohydr. Polym. 2021, 270, 118384. [Google Scholar] [CrossRef]
  107. Anil, S. Potential Medical Applications of Chitooligosaccharides. Polymers 2022, 14, 3558. [Google Scholar] [CrossRef]
  108. Liu, X.; Chen, L.; Zhang, Y.; Xin, X.; Qi, L.; Jin, M.; Guan, Y.; Gao, Z.; Huang, W. Enhancing anti-melanoma outcomes in mice using novel chitooligosaccharide nanoparticles loaded with therapeutic survivin-targeted siRNA. Eur. J. Pharm. Sci. 2021, 158, 105641. [Google Scholar] [CrossRef]
  109. Bruniaux, J.; Allard-Vannier, E.; Aubrey, N.; Lakhrif, Z.; Ben Djemaa, S.; Eljack, S.; Marchais, H.; Hervé-Aubert, K.; Chourpa, I.; David, S. Magnetic nanocarriers for the specific delivery of siRNA: Contribution of breast cancer cells active targeting for down-regulation efficiency. Int. J. Pharm. 2019, 569, 118572. [Google Scholar] [CrossRef]
  110. Eljack, S.; Allard-Vannier, E.; Misericordia, Y.; Hervé-Aubert, K.; Aubrey, N.; Chourpa, I.; Faggad, A.; David, S. Combination of Nanovectorized siRNA Directed against Survivin with Doxorubicin for Efficient Anti-Cancer Activity in HER2+ Breast Cancer Cells. Pharmaceutics 2022, 14, 2537. [Google Scholar] [CrossRef]
  111. Lyu, Y.; Yu, M.; Liu, Q.; Zhang, Q.; Liu, Z.; Tian, Y.; Li, D.; Changdao, M. Synthesis of silver nanoparticles using oxidized amylose and combination with curcumin for enhanced antibacterial activity. Carbohydr. Polym. 2020, 230, 115573. [Google Scholar] [CrossRef]
  112. Zhang, H.; Deng, L.; Liu, H.; Mai, S.; Cheng, Z.; Shi, G.; Zeng, H.; Wu, Z. Enhanced fluorescence/magnetic resonance dual imaging and gene therapy of liver cancer using cationized amylose nanoprobe. Mater. Today Bio 2022, 13, 100220. [Google Scholar] [CrossRef] [PubMed]
  113. Varanko, A.; Saha, S.; Chilkoti, A. Recent trends in protein and peptide-based biomaterials for advanced drug delivery. Adv. Drug Deliv. Rev. 2020, 156, 133–187. [Google Scholar] [CrossRef] [PubMed]
  114. Norouzi, P.; Motasadizadeh, H.; Atyabi, F.; Dinarvand, R.; Gholami, M.; Farokhi, M.; Shokrgozar, M.A.; Mottaghitalab, F. Combination Therapy of Breast Cancer by Codelivery of Doxorubicin and Survivin siRNA Using Polyethylenimine Modified Silk Fibroin Nanoparticles. ACS Biomater. Sci. Eng. 2021, 7, 1074–1087. [Google Scholar] [CrossRef]
  115. Yoo, H.; Mok, H. Evaluation of multimeric siRNA conjugates for efficient protamine-based delivery into breast cancer cells. Arch. Pharm. Res. 2015, 38, 129–136. [Google Scholar] [CrossRef]
  116. Xu, X.; Li, L.; Li, X.; Tao, D.; Zhang, P.; Gong, J. Aptamer-protamine-siRNA nanoparticles in targeted therapy of ErbB3 positive breast cancer cells. Int. J. Pharm. 2020, 590, 119963. [Google Scholar] [CrossRef]
  117. Ma, S.; Li, X.; Ran, M.; Ji, M.; Gou, J.; Yin, T.; He, H.; Wang, Y.; Zhang, Y.; Tang, X. Fabricating nanoparticles co-loaded with survivin siRNA and Pt(IV) prodrug for the treatment of platinum-resistant lung cancer. Int. J. Pharm. 2021, 601, 120577. [Google Scholar] [CrossRef]
  118. Fazal, T.; Murtaza, B.N.; Shah, M.; Iqbal, S.; Rehman, M.U.; Jaber, F.; Dera, A.A.; Awwad, N.S.; Ibrahium, H.A. Recent developments in natural biopolymer based drug delivery systems. RSC Adv. 2023, 13, 23087–23121. [Google Scholar] [CrossRef]
  119. Liu, Y.; Ashmawy, S.; Latta, L.; Weiss, A.V.; Kiefer, A.F.; Nasr, S.; Loretz, B.; Hirsch, A.K.H.; Lee, S.; Lehr, C.M. pH-Responsive Dynaplexes as Potent Apoptosis Inductors by Intracellular Delivery of Survivin siRNA. Biomacromolecules 2023, 24, 3742–3754. [Google Scholar] [CrossRef]
  120. Han, W.; Yuan, Y.; Li, H.; Fu, Z.; Wang, M.; Guan, S.; Wang, L. Design and anti-tumor activity of self-loaded nanocarriers of siRNA. Colloids Surf. B Biointerfaces 2019, 183, 110385. [Google Scholar] [CrossRef] [PubMed]
  121. Zhao, X.; Xu, Q.; Wang, Q.; Liang, X.; Wang, J.; Jin, H.; Man, Y.; Guo, D.; Gao, F.; Tang, X. Induced Self-Assembly of Vitamin E-Spermine/siRNA Nanocomplexes via Spermine/Helix Groove-Specific Interaction for Efficient siRNA Delivery and Antitumor Therapy. Adv. Healthc. Mater. 2024, 13, e2303186. [Google Scholar] [CrossRef]
  122. Mukherjee, A.; Waters, A.K.; Kalyan, P.; Achrol, A.S.; Kesari, S.; Yenugonda, V.M. Lipid-polymer hybrid nanoparticles as a next-generation drug delivery platform: State of the art, emerging technologies, and perspectives. Int. J. Nanomed. 2019, 14, 1937–1952. [Google Scholar] [CrossRef]
  123. Hassn Mesrati, M.; Syafruddin, S.E.; Mohtar, M.A.; Syahir, A. CD44: A Multifunctional Mediator of Cancer Progression. Biomolecules 2021, 11, 1850. [Google Scholar] [CrossRef]
  124. Marchiò, C.; Annaratone, L.; Marques, A.; Casorzo, L.; Berrino, E.; Sapino, A. Evolving concepts in HER2 evaluation in breast cancer: Heterogeneity, HER2-low carcinomas and beyond. Semin. Cancer Biol. 2021, 72, 123–135. [Google Scholar] [CrossRef] [PubMed]
  125. Chen, J.; Li, J.; Sun, X.; Lu, H.; Liu, K.; Li, Z.; Guan, J.; Song, H.; Wei, W.; Ge, Y.; et al. Precision Therapy of Recurrent Breast Cancer through Targeting Different Malignant Tumor Cells with a HER2/CD44-Targeted Hydrogel Nanobot. Small 2023, 19, e2301043. [Google Scholar] [CrossRef]
  126. Yang, S.; Wang, D.; Zhang, X.; Sun, Y.; Zheng, B. cRGD peptide-conjugated polyethylenimine-based lipid nanoparticle for intracellular delivery of siRNA in hepatocarcinoma therapy. Drug Deliv. 2021, 28, 995–1006. [Google Scholar] [CrossRef] [PubMed]
  127. Bi, Y.; Lee, R.J.; Wang, X.; Sun, Y.; Wang, M.; Li, L.; Li, C.; Xie, J.; Teng, L. Liposomal codelivery of an SN38 prodrug and a survivin siRNA for tumor therapy. Int. J. Nanomed. 2018, 13, 5811–5822. [Google Scholar] [CrossRef]
  128. Passos Gibson, V.; Derbali, R.M.; Phan, H.T.; Tahiri, H.; Allen, C.; Hardy, P.; Chain, J.L. Survivin silencing improved the cytotoxicity of carboplatin and melphalan in Y79 and primary retinoblastoma cells. Int. J. Pharm. 2020, 589, 119824. [Google Scholar] [CrossRef]
  129. Tan, C.; Zhang, Y.; Abbas, S.; Feng, B.; Zhang, X.; Xia, W.; Xia, S. Biopolymer-Lipid Bilayer Interaction Modulates the Physical Properties of Liposomes: Mechanism and Structure. J. Agric. Food Chem. 2015, 63, 7277–7285. [Google Scholar] [CrossRef] [PubMed]
  130. Chen, H.; Fan, X.; Zhao, Y.; Zhi, D.; Cui, S.; Zhang, E.; Lan, H.; Du, J.; Zhang, Z.; Zhang, S.; et al. Stimuli-Responsive Polysaccharide Enveloped Liposome for Targeting and Penetrating Delivery of survivin-shRNA into Breast Tumor. ACS Appl. Mater. Interfaces 2020, 12, 22074–22087. [Google Scholar] [CrossRef]
  131. Eljack, S.; David, S.; Chourpa, I.; Faggad, A.; Allard-Vannier, E. Formulation of Lipid-Based Nanoparticles for Simultaneous Delivery of Lapatinib and Anti-Survivin siRNA for HER2+ Breast Cancer Treatment. Pharmaceuticals 2022, 15, 1452. [Google Scholar] [CrossRef]
  132. Xie, D.; Du, J.; Bao, M.; Zhou, A.; Tian, C.; Xue, L.; Ju, C.; Shen, J.; Zhang, C. A one-pot modular assembly strategy for triple-play enhanced cytosolic siRNA delivery. Biomater. Sci. 2019, 7, 901–913. [Google Scholar] [CrossRef]
  133. Xu, X.; Li, Z.; Zhao, X.; Keen, L.; Kong, X. Calcium phosphate nanoparticles-based systems for siRNA delivery. Regen. Biomater. 2016, 3, 187–195. [Google Scholar] [CrossRef]
  134. Mitrach, F.; Schmid, M.; Toussaint, M.; Dukic-Stefanovic, S.; Deuther-Conrad, W.; Franke, H.; Ewe, A.; Aigner, A.; Wölk, C.; Brust, P.; et al. Amphiphilic Anionic Oligomer-Stabilized Calcium Phosphate Nanoparticles with Prospects in siRNA Delivery via Convection-Enhanced Delivery. Pharmaceutics 2022, 14, 326. [Google Scholar] [CrossRef]
  135. Kara, G.; Parlar, A.; Cakmak, M.C.; Cokol, M.; Denkbas, E.B.; Bakan, F. Silencing of survivin and cyclin B1 through siRNA-loaded arginine modified calcium phosphate nanoparticles for non-small-cell lung cancer therapy. Colloids Surf. B Biointerfaces 2020, 196, 111340. [Google Scholar] [CrossRef]
  136. Bi, Y.; Zhang, Y.; Cui, C.; Ren, L.; Jiang, X. Gene-silencing effects of anti-survivin siRNA delivered by RGDV-functionalized nanodiamond carrier in the breast carcinoma cell line MCF-7. Int. J. Nanomed. 2016, 11, 5771–5787. [Google Scholar] [CrossRef] [PubMed]
  137. Negri, V.; Pacheco-Torres, J.; Calle, D.; López-Larrubia, P. Carbon Nanotubes in Biomedicine. Top. Curr. Chem. 2020, 378, 15. [Google Scholar] [CrossRef] [PubMed]
  138. Hassan, H.; Diebold, S.S.; Smyth, L.A.; Walters, A.A.; Lombardi, G.; Al-Jamal, K.T. Application of carbon nanotubes in cancer vaccines: Achievements, challenges and chances. J. Control Release 2019, 297, 79–90. [Google Scholar] [CrossRef]
  139. Zhao, Y.; Zhao, T.; Cao, Y.; Sun, J.; Zhou, Q.; Chen, H.; Guo, S.; Wang, Y.; Zhen, Y.; Liang, X.J.; et al. Temperature-Sensitive Lipid-Coated Carbon Nanotubes for Synergistic Photothermal Therapy and Gene Therapy. ACS Nano 2021, 15, 6517–6529. [Google Scholar] [CrossRef] [PubMed]
  140. Ma, X.; Jiang, X.; Wang, Z.; Fan, Y.; Li, J.; Chow, C.; Wang, C.; Deng, C.; Lin, W. Cationic Metal-Organic Layer Delivers siRNAs to Overcome Radioresistance and Potentiate Cancer Radiotherapy. Angew. Chem. Int. Ed. Engl. 2025, 64, e202419409. [Google Scholar] [CrossRef]
  141. Cai, M.; Yao, Y.; Yin, D.; Zhu, R.; Fu, T.; Kong, J.; Wang, K.; Liu, J.; Yao, A.; Ruan, Y.; et al. Enhanced lysosomal escape of cell penetrating peptide-functionalized metal-organic frameworks for co-delivery of survivin siRNA and oridonin. J. Colloid Interface Sci. 2023, 646, 370–380. [Google Scholar] [CrossRef] [PubMed]
  142. Wan, X.; Ge, Y.; Zhu, W.; Zhang, J.; Pan, W.; Li, N.; Tang, B. GalNAc-functionalized metal-organic frameworks for targeted siRNA delivery: Enhancing survivin silencing in hepatocellular carcinoma. Biomater. Sci. 2025, 13, 2704–2712. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The classification of polymer-based nanocarriers for survivin siRNA delivery. There are mainly four kinds of polymeric nanocarriers for survivin siRNA delivery: synthetic polymers, natural polymers, lipid-polymer nanoparticles, and organic/inorganic hybrid nanomaterials.
Figure 1. The classification of polymer-based nanocarriers for survivin siRNA delivery. There are mainly four kinds of polymeric nanocarriers for survivin siRNA delivery: synthetic polymers, natural polymers, lipid-polymer nanoparticles, and organic/inorganic hybrid nanomaterials.
Polymers 17 02279 g001
Figure 2. Schematic diagram of the function and mechanism of action of survivin. Survivin mainly inhibits apoptosis in the cytoplasm and mitochondria and regulates the cell cycle in the nucleus.
Figure 2. Schematic diagram of the function and mechanism of action of survivin. Survivin mainly inhibits apoptosis in the cytoplasm and mitochondria and regulates the cell cycle in the nucleus.
Polymers 17 02279 g002
Figure 3. (A) The scheme of the fabrication and working principle of Sur@T7-AIE-Gd nanoparticles (NPs). Reprinted with permission from [79]. Copyright 2022, John Wiley and Sons; (B) Schematic diagram of DOX–SPBB–siRNA nanocarriers in A549 lung cancer cells. Reprinted with permission from [81]. Copyright 2019, American Chemical Society; (C) Schematical illustration of preparation of PTX/siRNA-loaded layer-by-layer nanoparticle delivery system and its intracellular therapeutic mechanism. Reprinted with permission from [82]. Copyright 2018, Dove Medical Press; (D) Schematic illustration of the targeted delivery of siRNAs to tumor cells by mPEG-g-γ-PGA/SSBPEI@siRNA nanoparticles. Reprinted with permission from [84]. Copyright 2020, Elsevier.
Figure 3. (A) The scheme of the fabrication and working principle of Sur@T7-AIE-Gd nanoparticles (NPs). Reprinted with permission from [79]. Copyright 2022, John Wiley and Sons; (B) Schematic diagram of DOX–SPBB–siRNA nanocarriers in A549 lung cancer cells. Reprinted with permission from [81]. Copyright 2019, American Chemical Society; (C) Schematical illustration of preparation of PTX/siRNA-loaded layer-by-layer nanoparticle delivery system and its intracellular therapeutic mechanism. Reprinted with permission from [82]. Copyright 2018, Dove Medical Press; (D) Schematic illustration of the targeted delivery of siRNAs to tumor cells by mPEG-g-γ-PGA/SSBPEI@siRNA nanoparticles. Reprinted with permission from [84]. Copyright 2020, Elsevier.
Polymers 17 02279 g003
Figure 4. (A) The conceptual strategy to nanoencapsulate siRNA within doubly self-assembled CS/DS polymeric nanoparticles under a microfluidics setup. Reprinted with permission from [100]. Copyright 2025, Academic Press. (B) Synthesis of siRNA nanocarriers and internalization process in HER2+ cells. Reprinted with permission from [106]. Copyright 2019, Elsevier. (C) Schematic illustration for synthesizing PBA-modified COS. Reprinted with permission from [105]. Copyright 2021, Elsevier. (D) Near-infrared/pH dual-responsive nanocomplexes for targeted imaging and chemo/gene/photothermal tri-therapies of non-small cell lung cancer. Reprinted with permission from [101]. Copyright 2020, Elsevier.
Figure 4. (A) The conceptual strategy to nanoencapsulate siRNA within doubly self-assembled CS/DS polymeric nanoparticles under a microfluidics setup. Reprinted with permission from [100]. Copyright 2025, Academic Press. (B) Synthesis of siRNA nanocarriers and internalization process in HER2+ cells. Reprinted with permission from [106]. Copyright 2019, Elsevier. (C) Schematic illustration for synthesizing PBA-modified COS. Reprinted with permission from [105]. Copyright 2021, Elsevier. (D) Near-infrared/pH dual-responsive nanocomplexes for targeted imaging and chemo/gene/photothermal tri-therapies of non-small cell lung cancer. Reprinted with permission from [101]. Copyright 2020, Elsevier.
Polymers 17 02279 g004
Table 1. Advantages and disadvantages of four nanoparticle delivery systems.
Table 1. Advantages and disadvantages of four nanoparticle delivery systems.
Delivery SystemAdvantagesDisadvantagesRef.
Synthetic polymersEase of functionalization, high stability, high drug loading capacity, and long circulation time enable sustained drug release.Certain toxicity, immunogenicity, and safety risks; the preparation process is complex.[68]
Natural polymers Wide range of sources, good biocompatibility and biodegradability, low toxicity, and non-immunogenicity.Off-target effect, stability, and longevity during storage and circulation in complex biological environments[69,70]
Lipid-polymer nanoparticlesStability, high loading of cargo, increased biocompatibility, rate-limiting controlled release, and prolonged drug half-life.High dose may cause immune reactions, preparation is complex and costly, and long-term stability still needs to be studied.[71]
Organic/inorganic hybrid nanomaterialsCombining the high stability and targeting properties of inorganic materials with the biocompatibility and biodegradability of organic materials, this approach enhances the intelligent response capability and structural stability of the carrier. Easy functionalization and low cytotoxicity.Complex design, high preparation costs, batch-to-batch consistency issues, scalability for large-scale production, and biosafety.[72]
Table 2. Targeted delivery of survivin siRNA by synthetic polymeric nanoparticles.
Table 2. Targeted delivery of survivin siRNA by synthetic polymeric nanoparticles.
Synthetic Polymer-Based Survivin siRNA Delivery Systems
NanoparticleSurvivin SequenceCancer TypesCell LinesOutcomeRef.
PEI-CA6.9/siRNA-Breast cancerMDA-MB-231 cellOctanoic acid-modified PEI showed the best survivin silencing efficiency, which significantly reduced cell survival and induced apoptosis.[77]
CDC20/surviving siRNA polymer polyplexes-Breast cancerMDA-MB-231 MDA-MB-436
MCF-7 cell
Polyethyleneimine modified with linoleic acid (PEI–LA) significantly increased cell viability and cellular uptake, improved the cellular uptake and release of siSurvivin, and effectively inhibited the growth of breast cancer cells.[78]
Sur@T7-AIF-GD NPS-Hepatocellular carcinomaLM3 cellT7 peptide enhances targeting, dual-functional imaging with Gd and AIE magnetic resonance imaging (MRI) and aggregation-induced emission (AIE) imaging, enabling precise delivery, real-time monitoring, and efficient treatment of hepatocellular carcinoma.[79]
ECV-modified PEI/siRNASense: 5′-AUUCACCAAGGG-UUAAUUCdTdT-3′Prostate and Osteosarcoma carcinoma
Ovarian and Colorectal cancer
SKOV3 PC3 HCT-116 Saos-2 cellIn the PC3 tumor xenograft nude mouse model, the ECV-modified PEI/siSurvivin complex significantly inhibited tumor growth.[80]
DOX-SPBB-siRNASense: 5′-GAGCAGUUUGAAGAAUUATT-3′
Antisense: 5′-UAAUUCUUC-AAACUGCUUCTT-3′
Lung cancerA549 cellDOX-SPBB-siRNA complex significantly reduced the tumor volume in an A549 tumor xenograft nude mouse model.[81]
PEI–PLA/PTX/siRNA-/PEG-PAspSense: 5′-GCAUUCGUCCGG-UUGCGCUTT-3′Lung cancercellPEG-PAsp has pH-responsive properties, enabling the PEG-PAsp-modified delivery system to induce apoptosis and arrest the cell cycle at the G2/M phase in A549 cells.[82]
QH-MnO2@PTX-siRNASense: 5′-CACCGCAUCUCU-ACAUUCATT-3′
Antisense: 5′-UGAAUGUAG-AGAGCGGUGTT-3′
Breast cancerMDA-MB-231 cellMnO2 nanoparticles exhibit superior responsiveness to high levels of GSH within cancer cells. The fluorescence recovery function of InP/ZnS quantum dots enables real-time monitoring of drug release in the tumor microenvironment.[83]
mPEG-g-γ-PGA/SSBPEI@siRNA-Lung cancerA549 cellPEI containing disulfide bonds breaks down in the high GSH environment of tumors to release drugs.[84]
SSBPEI-DOX@ siRNAs/iRGD-PEG-HA-Ovarian cancerA2780 cellThe nanoparticles significantly enhanced the antitumor effect of DOX compared with free DOX and also greatly suppressed the migration and invasion of A2780/DDP-derived CSCs.[85]
scFv-P-BAP-siSurv-polyplexSense: 5′-GAAUUAACCCUU-GGUGAAU(tt)-3′Prostate carcinomaPC3 cellMaltose-modified PPI significantly reduced cytotoxicity and improved biocompatibility. The single-chain antibody fragment (scFv) delivery system can specifically recognize prostate stem cell antigen (PSCA), thereby precisely entering target cells. The siRNA delivered by the delivery system significantly inhibited tumor growth.[86]
H40-TEPA-PEG-MUC1 aptamerSense: 5′-GAAAGAAUUUGA-GGAAACUdTdT -3′
Antisense: 5′-AGUUUCCUC-AAAUUCUUUCdTdT 3′
Breast cancerMCF-7 cellIt exhibits significant gene silencing effects in MCF-7 cells and demonstrates superior gene silencing efficiency compared to non-targeted dendritic polymers and Lipofectamine-2000.[87]
Survivin siRNA/PTX PMSense: 5′-GCAUUCGUCCGG-UUGCGCUdTdT-3′Breast cancerSKOV3-tr cell
MDA-MB 231 cell
Significantly inhibited cell viability and enhanced cell sensitivity to PTX in a variety of cancer cell lines, while effectively overcoming drug resistance by down-regulating survivin protein level and disrupting microtubule structure in drug-resistant cell line SKOV3-tr.[88]
siRNA/PTX PMSense: 5′-GCAUUCGUCCGG-UUGCGCUdTdT-3′Ovarian cancerSKOV3-tr cellPTX encapsulation efficiency to 90%, downregulated the expression of survivin, and promoted drug enrichment in tumor tissues.[89]
Table 3. Targeted delivery of survivin siRNA by natural polymeric nanoparticles.
Table 3. Targeted delivery of survivin siRNA by natural polymeric nanoparticles.
Natural Polymer-Based Survivin siRNA Delivery Systems
NanoparticleSurvivin SequenceCancer TypesCell LinesOutcomeRef.
SUR siRNA-CS-g-PMMA/DSSense: 5′-GAACAUCAUCAU-CCAGGACTT-3′Breast cancer4T1 cell4T1 cells treated with siSurvivin-loaded nanoparticles showed a significant decrease in cell viability, migration ability, and sphere size, confirming that this delivery system is effective in achieving gene silencing.[100]
CE7Q/CQ/S/Survivin shRNASurvivin-shRNA: 5′-GAATTA-ACCCTTGGTGAAT-3′Lung cancerA549 cellThis system could recognize epidermal growth factor receptor and enter into EGFR-mutated non-small cell lung cancer cells, and a stimuli-responsive release profile was achieved by near-infrared laser irradiation at pH 5.4 and displayed superior antitumor efficacy in vitro and in vivo.[101]
PEG-CS/siRNA-Breast cancer4T1 cellPEG-CS/siSurvivin showed a significant inhibitory effect on 4T1 cells, while the cell survival rate decreased and the apoptosis rate elevated as compared to the naked siRNA group, inhibiting tumor growth and lung metastasis effectively.[102]
TAT-g-CS/siSsurSense: 5′-GAACAUCAUCAU-CCAGGAC-3′Breast cancerMCF-7
4T1-Luc
Improved cellular uptake efficiency, inhibited the proliferation of 4T1-Luc tumor cells, and significantly inhibited the in vivo growth and metastasis of malignant breast tumors.[103]
MT/PTX/pDNA/rhIL-2 NPs-Hepatocellular carcinomaQGY-7703Formation of MT nanoparticles with redox properties. The release rate significantly improved. Tumor inhibition rate was significantly higher than the treatment group using the three drugs alone, enhancing the antitumor effect.[104]
PBA-COS/siRNASense: 5′-GCAUUCGUCCGG-UUGCGCUTT-3′
Antisense: 5′-AGCGCAACCGGACGAAUGCTT-3′
MelanomaB16F10 cellSignificantly inhibited melanoma cell proliferation, growth, and metastasis.[105]
TS-MSN/siRNASense: 5′-GUCUGGACCUCA-UGUUGUUdTdT-3′Breast cancerMDA-MB-231 cellImproved cellular internalization and enhanced gene silencing efficiency.[106]
TS-MSN siSurvivinSense: 5′-GUCUGGACCUCA-UGUUGUUdTdT-3′
Antisense: 5′-AACAACAUG-AGGUCCAGACdTdT-3′
Breast cancerSK-BR-3 cellTS-MSN improved cellular internalization and enhanced the anticancer effect of DOX by downregulating survivin expression.[107]
CSP/TPE@siRNA-SP94sense: 5′-CACCGCAUCUCU-ACAUUCATT-3′
Antisense: 5′-UGAAUGUAG-AGAUGCGGUGTT-3′
Hepatocellular carcinomaHuh-7 cellIt exhibits excellent fluorescence and magnetic resonance imaging performance both in vitro and in vivo and significantly inhibits tumor growth in a nude mouse model carrying Huh-7 tumors.[108]
DOX/siRNA/PEI-SFNPs-Breast cancer4T1 cellIt effectively inhibited tumor growth without causing significant weight loss or other systemic toxicity, providing a new strategy for breast cancer treatment.[109]
APRSense: 5′-GGACCACCGCAUCUCUACAdTdT-3′
Antisense: 3′-dTdTCCUGGU-GGCGUAGAGAUGU-5′
Breast cancerMCF-7 cellErbB3 aptamer can specifically recognize and bind to the ErbB3 receptor overexpressed on the surface of breast cancer cells, realizing the precise delivery of siRNA. Protamine not only enhances the stability of the complex but also improves the intracellular delivery efficiency of siRNA through the electrostatic binding of siSurvivin.[110]
NP-siRNA/Pt (IV)Sense: 5′-GAAUUAACCCUU-GGUGAUTT-3′
Antisense: 3′-AUUCACCAAG GGUUAAUUCTT-5′
Lung cancerA549 cellCo-loading siSurvivin and Pt(IV) prodrug enhanced the tumor tissue-specific accumulation of the nanocarrier. Surface modification of the nanocarrier with PGA effectively improved its stability and prolonged its circulation time in the blood.[111]
XL-DPs/siRNA-Lung cancerA549 cellThe polymer is pH-responsive and releases siSurvivin in acidic environments. It has good biocompatibility and low cytotoxicity.[112]
PEG–RAFF-siRNASense: 5′-GAGACAGAAUAG-AGUGAUATT-3′
Antisense: 5′-UAUCACUCU-AUUCUGUCUCTT-3′
Breast cancerMCF-7 cellArginine is frequently present in cell-penetrating peptides, which can enhance cellular uptake. Alanine serves as a spacer, which effectively avoids the steric hindrance.[113]
VE-Sper/DSPE-PEG2000/siRNASense: 5′-GCAUUCGUCCGG-UUGCGCUdTdT-3′
Antisense: 5′-AGCGCAACC-GGACGAAUGCdTdT-3′
Hepatocellular carcinomaHepG2 cellThis nanocomposite can effectively silence the Eg5 and survivin genes and arrest the cell cycle at the G2/M phase. It significantly inhibits tumor growth in HepG2 tumor-bearing mice.[114]
Table 4. Targeted delivery of survivin siRNA by liposomal polymer nanoparticles.
Table 4. Targeted delivery of survivin siRNA by liposomal polymer nanoparticles.
Liposomal Polymer-Based Survivin siRNA Delivery Systems
NanoparticleSurvivin SequenceCancer TypesCell LinesOutcomeRef.
ALPR (HER2/CD44-Targeted Hydrogel Nanobot)Sense: 5′-GCAUUCGUCCGG-UUGCGCUdtdt-3′
Antisense: 5′-AGCGCAACC-GGACGAAUGCdtdt-3′
Breast cancerSKBR-3
MCF-7
MDA-MB-231
ALPR can efficiently deliver Herceptin, peptide, and survivin siRNA to SKBR-3, MDA-MB-231, and MCF-7 cells, demonstrating excellent antitumor effects.[125]
cRGD-PSH-NP/SSense: 5′-mGCAGGUUCCUm-UAUCUGUCATT-3′Hepatocellular carcinomaHepG2 cellcRGD-PSH-NP/S markedly downregulated survivin expression both in vitro and in vivo and exhibited potent tumor inhibition in HepG2-bearing nude mice.[126]
Tf-L-SN38/P/siRNASense: 5′-mGCAGGUUCCUm-UAUCUGUCAdTdT-3′
Antisense:5′-UGAmCAGAm-UAAGGAACCUGmCdTdT-3′
Cervical cancerHeLa cellTf-modified liposomes can specifically bind to transferrin receptors on the surface of tumor cells, increasing drug accumulation in tumor tissues. This enhances the efficacy of chemotherapeutic drugs and inhibits tumor growth.[127]
LNP/survivin siRNASense: 5′-GGACCACCGCAUCUCUACATT-3′
Antisense: 5′-UGUAGAGAU-GCGGUGGUCCTT-3′
Human retinoblastoma cell lineY79 cellDownregulation of survivin via siLNP enhances the cytotoxic effects of carboplatin and melphalan on Y79 cells and primary retinoblastoma cells.[128]
HA/HAase/CS/liposome/survivin-shRNA (HCLR)shRNA: 5′-AATTTGAGGA-AACTGCGGAGA-3′Breast cancerMDA-MB-231 cellHA specifically binds to the CD44 receptor on the surface of tumor cells, promoting tumor targeting and prolonging the retention time of liposomes in the bloodstream. CS undergoes protonation in the tumor microenvironment (pH 6.5) to promote cellular uptake, while pH-triggered HAase release improves the diffusion of the nanocarrier in tumor tissue, enhancing tumor penetration. It exhibits excellent in vivo tumor suppression capabilities.[130]
siRNA-LAPA_LBNPSense: 5′-GUCUGGACCUCA-UGUUGUUdTdT-3′Breast cancerSKBR-3 cellIt has good physical and chemical properties, a high cell uptake rate, and significant cytotoxic effects.[131]
OCA-CC-siRNASense: 5′-GAAUUUGAGGAA-ACUGCGAtt-3′
Antisense: 3′-ttCUUAAACU-CCUUUGACGCU-5′
Breast cancerMCF-7Enhanced cytoplasmic delivery capacity and demonstrated significant antitumor activity in vitro and in vivo through silencing survivin expression.[132]
Table 5. Targeted delivery of survivin siRNA by organic/inorganic hybrid nanomaterials.
Table 5. Targeted delivery of survivin siRNA by organic/inorganic hybrid nanomaterials.
Organic/Inorganic Hybrid Nanomaterials-Based Survivin siRNA Delivery Systems
NanoparticleSurvivin SequenceCancer TypesCell LinesOutcomeRef.
CaP-NPs-siRNASense: 5′-CUAUUGUGACCU-GGACUUATT-3′
Antisense: 5′-UAAGUCCAG-GUCACAAUAGAG-3′
GlioblastomaF98 cellso14PEGMA improves the dispersion stability of nanoparticles, increases cellular uptake efficiency, and reduces the potential cytotoxicity of traditional CaP. Calcium phosphate nanoparticles protect siRNA from degradation and promote its uptake into cells.[134]
CaP-Arg-siRNASense: 5′-GAAGCAGUUUGA-AGAAUUATT-3′
Antisense: 5′-UAAUUCUUC-AAACUGCUUCTT-3′
Lung cancerA549 cellCaP-Arg-siRNAs significantly suppressed the expression of survivin and cyclin B1, resulting in a marked decrease in cell growth and inducing apoptosis drastically.[135]
NDCONH(CH2)2NH-VDGR/survivin siRNASense: 5′-GCATGGGTCCCC-CGACGTTG-3′
Antisense: 5′-GCTCCGGCC-AGAGGCCTCAA-3′
Breast cancerMCF-7 cellReduced survivin expression and displayed a potent inhibitory effect on tumor growth in vitro and in vivo.[136]
CNTs-PS/siRNASense: 5′-CACCGCAUCUCU-ACAUUCATT-3′
Antisense: 5′-UGAAUGUAG-AGAUGCGGUGTT-3′
Cervical cancerHeLa cellThe modified CNTs demonstrated excellent biocompatibility and temperature-sensitive properties. The combination of photothermal therapy and gene therapy caused massive apoptosis in HeLa cells and exhibited strong antitumor activity in vivo.[139]
CMOL@ siRNASUR A:
Sense: 5′-CCUUCCUCACUG-UCAAGAATT-3′
Antisense: 5′-UUCUUGACA-GUGAGGAAGGTT-3′
SUR B:
Sense: 5′-GAGACCAACAAC-AAGCAAATT-3′
Antisense: 5′-UUUGCUUGU-UGUUGGUCUCTT-3′
SUR C:
Sense: 5′-CUACCCGUCAGU-CAAUUGATT-3′
Antisense: 5′-UCAAUUGAC-UGACGGGUAGTT-3′
Colorectal and Breast cancerCT26, 4T1 cellCMOL functions as an effective reactive oxygen species (ROS) generator, and the cationic framework serves as an siRNA cocktail carrier to synergistically downregulate survivin levels. Combined with low-dose X-ray irradiation, it has shown significant antitumor efficacy.[140]
PEG-CPP33@ORI@survivin siRNA@ZIF-90Antisense: 5′-UAAUUCUUC-AAACUGCUUCTT-3′Lung cancerA549 cellPEG-CPP33@NPs significantly improved the cellular uptake efficiency of siRNA in A549 cells and showed stronger tumor accumulation. The strongest antitumor effect in vivo was observed through the co-delivery of ORI and survivin siRNA.[141]
siRNA@MOF-GalNAcSense: 5′-AAGGAGAUCAAC-AUUUUCA-3′
Antisense: 5′-UGAAAAUGU-UGAUCUCCUU-3′
Hepatocellular carcinomaHepG2 cellThe GalNAc-decorated MOF specifically accumulated in HCC tumor tissue and was effectively endocytosed by HCC cells. The protective properties of the MOF increased the stability of siRNA and allowed for significant downregulation of survivin expression in HCC tumors, contributing to tumor inhibition through the suppression of cell proliferation and the induction of apoptosis.[142]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ren, L.; Wang, S.; Li, B.-C.; Ding, G.-B. Research Progress on Polymer-Based Nanocarriers for Tumor-Targeted Delivery of Survivin siRNA. Polymers 2025, 17, 2279. https://doi.org/10.3390/polym17172279

AMA Style

Ren L, Wang S, Li B-C, Ding G-B. Research Progress on Polymer-Based Nanocarriers for Tumor-Targeted Delivery of Survivin siRNA. Polymers. 2025; 17(17):2279. https://doi.org/10.3390/polym17172279

Chicago/Turabian Style

Ren, Luya, Shaoxia Wang, Bin-Chun Li, and Guo-Bin Ding. 2025. "Research Progress on Polymer-Based Nanocarriers for Tumor-Targeted Delivery of Survivin siRNA" Polymers 17, no. 17: 2279. https://doi.org/10.3390/polym17172279

APA Style

Ren, L., Wang, S., Li, B.-C., & Ding, G.-B. (2025). Research Progress on Polymer-Based Nanocarriers for Tumor-Targeted Delivery of Survivin siRNA. Polymers, 17(17), 2279. https://doi.org/10.3390/polym17172279

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