Research Progress of siRNA Systems in Breast Cancer Therapy

Research Article
Open access

Research Progress of siRNA Systems in Breast Cancer Therapy

Hongyan Wang 1*
  • 1 Shandong University    
  • *corresponding author 2950197493@qq.com
Published on 23 October 2025 | https://doi.org/10.54254/2753-8818/2025.AU28321
TNS Vol.144
ISSN (Print): 2753-8818
ISSN (Online): 2753-8826
ISBN (Print): 978-1-80590-441-0
ISBN (Online): 978-1-80590-442-7

Abstract

Breast cancer remains the leading cause of cancer-related deaths among women globally, with treatment facing significant challenges due to drug resistance and metastasis. RNA interference technology, particularly via siRNA, offers a novel strategy for precision therapy. This review comprehensively examines the mechanisms and applications of nanotechnology-mediated siRNA delivery systems. Methodologically, it involves systematic analysis of the design principles underlying polymeric, lipid-based, and metallic nanocarriers to evaluate their efficacy in enhancing cellular uptake, promoting endosomal escape, and protecting siRNA from degradation; additionally, it combines preclinical and clinical data to evaluate these delivery platforms' pharmacodynamic characteristics and therapeutic results. Results demonstrate that siRNA nanocarriers significantly enhance targeted gene-silencing efficiency, exhibiting potent anti-tumor activity in breast cancer models. However, clinical translation is hampered by limitations in specificity, immunogenicity, and evolving regulatory frameworks. To advance clinical application, future research should focus on developing stimuli-responsive nanovectors and combining them with immunotherapy or chemotherapy regimens.

Keywords:

Breast Cancer, siRNA Delivery, Nanocarriers, Clinical Translation

Wang,H. (2025). Research Progress of siRNA Systems in Breast Cancer Therapy. Theoretical and Natural Science,144,62-70.
Export citation

1.  Introduction

Breast cancer remains the leading cause of cancer-related mortality among women worldwide, with over 2.3 million new cases reported in 2022, exacerbated by challenges such as metastasis, drug resistance, and the aggressive nature of triple-negative breast cancer (TNBC) subtypes [1,2]. Traditional cytotoxic chemotherapeutics—including anthracyclines, taxanes, and carboplatin—provide only limited therapeutic benefits while inducing adverse effects such as gastrointestinal reactions, myelosuppression, and liver dysfunction. Immunotherapy drugs such as paclitaxel, cobitinib, atezolizumab and albumin bound paclitaxel are also associated with severe abdominal pain, vaginal bleeding, muscle cramps, paralysis, leukopenia, anemia, cardiotoxicity and other adverse reactions [3-6].

Cells can further evade chemotherapy by activating anti-apoptotic pathways, rendering drug resistance a pervasive and severe issue. This resistance is associated with endocrine resistance, multidrug resistance proteins, breast cancer resistance protein, DNA repair mechanism alterations, and cancer stem cell resistance. These multifaceted challenges underscore the urgent need for innovative therapeutic strategies. In this context, RNA interference (RNAi) technology, particularly through small interfering RNA (siRNA), emerges as a promising avenue for precision therapy. siRNA can specifically silence oncogenes implicated in tumor growth, invasion, and immune evasion, such as Bcl-2, survivin, and PD-L1. However, the clinical translation of siRNA faces significant hurdles, including enzymatic degradation, inefficient cellular uptake, and the need to balance specificity and immunogenicity. To address these challenges, nanotechnology-based delivery systems are being explored to enhance siRNA stability, cellular uptake, and tissue-specific targeting. This review aims to summarize the existing siRNA delivery system schemes for breast cancer treatment, discuss the development and application of these delivery systems, and highlight the current challenges and future directions. Given the broad applicability of these principles, this review, while focusing on breast cancer, also holds relevance for siRNA delivery applications in other diseases.

2.  Molecular mechanisms underlying breast cancer development

Mutations in Key Transcription Factor Genes: Transcription factors play pivotal roles in breast cancer progression. For instance, sustained activation of the STAT3 gene promotes tumor cell proliferation and survival [7]. Additionally, overexpression of the MYC gene drives cell cycle progression, while mutations in the GATA3 gene may impair breast cell differentiation [8,9]. Abnormal activation or suppression of these transcription factors is a major molecular driver in breast cancer initiation and progression. siRNA-based therapeutic strategies targeting these factors via siRNA, such as siSTAT3, siMYC, and siGATA3, may offer novel avenues for breast cancer treatment.

Gene Mutations Promoting Apoptosis Escape: Apoptosis evasion represents another critical step in breast cancer development. Overexpression of the Bcl-2 gene suppresses apoptosis, thereby promoting tumor cell survival [10]. High expression of the Survivin (BIRC5) gene is also associated with tumor cell viability [11]. siRNA therapies targeting these apoptosis-related genes, such as siBcl-2 and siSurvivin, may help restore tumor cell apoptosis sensitivity, thereby inhibiting breast cancer progression.

Gene Mutations Promoting Tumor Metastasis: Tumor metastasis is a key factor in poor breast cancer prognosis. Key molecules in the epithelial-mesenchymal transition (EMT) process, such as Snail, Twist, and ZEB1, along with angiogenesis-related molecules like VEGF-A and VEGFR1/2, all play roles in tumor invasion and metastasis [12-14]. High expression of matrix metalloproteinases (MMPs) is also associated with tumor invasiveness [15]. The integrin signaling pathway plays a vital role in tumor cell adhesion, migration, and invasion. siRNA therapies targeting these metastasis-associated molecules, such as siSnail, siVEGF, and siMMP-9, may help inhibit breast cancer metastasis.

3.  Applications of siRNA in breast cancer therapy

Targeting Dysregulated Transcription Factors: Dysregulated transcription factors represent important therapeutic targets in breast cancer. For instance, siRNA targeting STAT3 and MYC can suppress the activity of these transcription factors, thereby inhibiting tumor cell proliferation and survival [16]. Additionally, siRNA targeting GATA3 may help regulate cell differentiation and inflammatory responses [17]. These siRNA therapeutic strategies may offer more precise treatment options for breast cancer patients.

Targeting Abnormal Apoptosis-Related Genes: Apoptosis evasion is a key survival mechanism for breast cancer cells. siRNA therapies targeting Bcl-2 and Survivin can promote tumor cell apoptosis, thereby inhibiting tumor growth and spread [18,19]. These siRNA therapeutic approaches may help restore tumor cell sensitivity to chemotherapeutic drugs, enhancing treatment efficacy.

Targeting Abnormal Genes Associated with Metastasis: Tumor metastasis poses a major challenge in breast cancer treatment. siRNA therapies targeting metastasis-related molecules such as Snail, Twist, ZEB1, VEGF-A, VEGFR1/2, HIF-1α, MMP-9, and αvβ3 integrin can reduce tumor invasiveness and metastatic potential [15,20,21]. These siRNA-based strategies may help decrease breast cancer recurrence and metastasis, thereby improving patient survival rates.

4.  Research progress in siRNA nanodelivery systems 

4.1.  Types of delivery carriers

Nanocarrier technology has significantly transformed the landscape of siRNA delivery, leading to substantial improvements in molecular stability, cellular uptake, and tissue-specific targeting. Within this broader analytical framework, these advances are largely attributable to the precise engineering of material properties and nanostructural design. Current delivery systems can typically be classified into one of four principal categories.

4.1.1.  Polymeric nanocarriers

Polymeric nanocarriers have increasingly emerged as versatile vehicles for siRNA delivery, primarily due to their favorable biocompatibility, high cargo capacity, and ease of chemical modification. Among these, polyethyleneimine (PEI) and chitosan tend to stand out in the literature. PEI's frequent use is explained by its ability to form stable complexes with siRNA through electrostatic interactions and promote endosomal escape, likely due to the well-documented proton sponge effect [22]. However, PEI’s significant cytotoxicity has driven extensive research into safer alternatives, such as low-molecular-weight crosslinked PEI or PEGylated versions [23].

Additionally, copper sulfide–PEI–siRNA complexes demonstrate the ability to enhance siRNA loading and improve cellular uptake in most metastatic breast cancer models studied [24]. From this particular interpretive perspective, chitosan, on the other hand, appears to be valued for its biodegradability and mucoadhesive properties, but typically needs some form of modification—carboxymethylation, for example—to improve its solubility under physiological conditions. What appears particularly significant about recent developments is that chitosan carriers conjugated with MUC1 aptamers exhibit increased tumor specificity and therapeutic efficacy across several cancer models [25,26].

4.1.2.  Lipid-based nanocarriers

Lipid-based nanocarriers have emerged as the leading platforms for siRNA delivery, with the approval of Patisiran by the FDA underlining their clinical relevance. Their effectiveness is largely attributable to high biocompatibility and their ability to mimic natural membrane fusion processes [27]. In terms of technical approaches, two strategies dominate: first, optimized cationic liposomes composed of DOTAP and DOPE in a 1:1 ratio, which achieve a zeta potential exceeding 50 mV to ensure efficient siRNA complexation; and second, ionizable lipid nanoparticles (LNPs), which utilize pH-sensitive lipids that stay neutral during systemic circulation but become protonated in endosomes, thereby facilitating targeted siRNA release. Both platforms employ conjugated ligands—such as folate or RGD peptides—to enhance tumor targeting specificity [28]. Recent preclinical studies have demonstrated that liposomal formulations co-delivering Bcl-xL siRNA with chemotherapeutics like paclitaxel or crizotinib can induce synergistic apoptosis and achieve a cumulative drug release of over 64% within 12 hours in breast cancer models [2]. Furthermore, hybrid systems incorporating polymers or natural exosomes are being developed to expand the potential for co-delivery of siRNA and chemotherapeutics.

4.1.3.  Inorganic/metal nanocarriers

Within this broader analytical framework, inorganic nanocarriers, which are notable for their distinctive optoelectromagnetic properties, appear to support what could be characterized as the integration of gene silencing therapy, diagnostic imaging, and physical treatment modalities—forming what appear to be genuinely multifunctional siRNA delivery systems. Gold nanoparticles, for instance, can enable precise, spatiotemporally controlled siRNA release in response to external triggers such as laser irradiation, while also appearing to induce photothermal effects that may potentially enhance therapeutic outcomes [29]. These findings demonstrate that graphene oxide-based platforms are highly effective as siRNA carriers, primarily due to their large surface area, which enables substantial adsorption [30,31]. Additionally, further functionalization with cell-penetrating peptides improves tumor targeting—highlighting their significant potential in oncological applications [32-34].

Hollow mesoporous copper sulfide nanoparticles (CuS), from this particular interpretive perspective, seem to exemplify a high degree of multifunctionality. As vectors apparently responsive to near-infrared stimuli, they appear to exhibit a potentially impressive photothermal conversion efficiency. What appears to follow from this analysis is that upon activation, these nanoparticles release Cu²⁺/Cu⁺ ions, which appears to provide evidence that may support the induction of cuproptosis through DLAT oligomerization and concurrently produce reactive oxygen species for chemodynamic therapy [24]. What appears significant in this context, however, is that despite these promising advances, comprehensive biosafety evaluations remain essential, particularly given the multifaceted nature of this evidence and concerns regarding potential metal accumulation and its associated toxicity.

4.1.4.  Co-delivery systems

Integrated co-delivery systems that combine siRNA with immunomodulators or chemotherapeutics are emerging as promising strategies to overcome tumor heterogeneity and drug resistance. For example, cationic liposomes can deliver Bcl-xL-targeted siRNA along with agents like paclitaxel or crizotinib, disrupting microtubule dynamics and blocking anti-apoptotic signaling to exert a dual cytotoxic effect [35]. pH-sensitive micelles can release their therapeutic payloads in response to the acidic tumor microenvironment, enabling sequential delivery for enhanced efficacy. Copper sulfide-silk fibroin nanoparticles loaded with PD-L1 siRNA integrate gene silencing with photothermal therapy, cuproptosis induction, and immunogenic cell death, achieving significant preclinical results such as 60% suppression of PD-L1 expression and 56% primary tumor regression. Biomimetic nanoparticles and exosome-based delivery systems improve drug biodistribution and cellular uptake by leveraging their inherent biocompatibility. However, challenges remain before clinical translation, including precise drug-siRNA ratio control, batch-to-batch reproducibility, and rigorous safety evaluations for regulatory approval and large-scale production.

4.2.  Delivery optimization methods and related challenges

Recent advancements in siRNA nanodelivery have led to the development of highly specialized carriers designed to enhance delivery efficiency and biosafety. For instance, galactosamine conjugation enables siRNA to exploit the asialoglycoprotein receptor pathway, significantly extending its circulation time and enhancing hepatocyte uptake. However, this approach still encounters significant obstacles in penetrating solid tumors, primarily due to the abnormal vasculature and increased interstitial fluid pressure typically found in the tumor microenvironment [36,37].

Consequently, research focus has shifted towards surface modifications with tumor-specific ligands, such as RGD peptides that recognize integrins, or transferrin that binds to overexpressed transferrin receptors on many cancer cells [38,39]. Additionally, the incorporation of polyethylene glycol (PEG) or zwitterionic polymers is critical, as these components can shield nanocarriers from immune detection—enhancing their “stealth” properties and extending circulation time [40]. Considering the complexity of these findings, innovative “smart” nanocarriers have emerged to address the intricacies of the tumor microenvironment. These findings point towards ATP-responsive delivery systems that employ boronic ester linkages, which can react to oxidative stress and acidic pH, as well as polyplexes with thioketal bonds that respond to both pH and reactive oxygen species [41,42]. These systems enable more controlled siRNA release in response to ATP gradients characteristic of metastatic niches. More recently, dual-stimuli sensitive designs have combined triggers like pH with enzymes, or redox with light, to further fine-tune the specificity of siRNA release within tumors [43].

Despite these remarkable developments, several barriers remain before clinical translation can be achieved. Within this framework, it is essential to optimize siRNA chemical modifications—such as 2'-O-methyl and phosphorothioate substitutions—to reduce off-target effects and increase nuclease resistance [44-46]. Additionally, rigorous comparison between different carrier systems is warranted; while solid lipid nanoparticles offer high biocompatibility, metallic vectors like gold nanoshells introduce potential long-term toxicity [47,48]. Moreover, there is a need to systematically address challenges including scalable manufacturing, long-term formulation stability, and the risk of immunogenicity, such as the potential activation of the complement system. Ultimately, the successful translation of siRNA nanomedicines from preclinical models to clinical oncology will likely depend on integrating computational modeling, such as molecular dynamics simulations, and high-throughput screening strategies. These approaches are vital for overcoming multifaceted barriers and realizing the therapeutic potential of siRNA-based interventions in cancer treatment.

5.  Current challenges, limitations, and future development directions

siRNA-mediated RNA interference is widely used in research to silence protein-coding genes with known mRNA sequences. SiRNA has the ability to target any protein, including those difficult to be druggable, and is superior to monoclonal antibodies and small molecule inhibitors in inhibiting druggable pathways, overcoming the limitations faced by traditional targeted the therapeutics [49]. Due to the short length, small molecular weight and high anionization of siRNA, it is easy to be cleared and degraded in the renal system, and the generated by-products will also lead to adverse immune reactions [50]. At the same time, siRNA can also be recognized as an exogenous RNA inducing increased production of interferons or cytokines, which in turn leads to off target effects (that is, siRNA molecules will unexpectedly silence non target genes and interfere with normal cellular processes) or cytotoxicity. Successful delivery of siRNA to target cells also often faces difficulties in being degraded and cleared by nucleases. In addition, due to the hydrophilic and negatively charged nature of siRNA molecules, it is difficult to penetrate the biofilm to be obtained and utilized by target cells, and the cell intake rate is low. Even the delivery system required for siRNA can induce immune responses.

Preclinical research, safety assessment, regulatory approval, and manufacturing processes pose multiple challenges to the development of siRNA therapy. Currently, the preclinical assessment of siRNA is primarily based on human tumor xenograft models and immunodeficient mice. However, these models have clear drawbacks: on the one hand, siRNA cannot fully assess off-target toxicity because it lacks cross-activity on murine proteins due to its targeting of human proteins; On the other hand, the model may inaccurately estimate the actual efficacy of siRNA, because it may not accurately reflect the adaptive immune response triggered by tumor antigen release or the potential regulatory effect of siRNA on immune cells. Although the mouse model with a complete humanized immune system has more advantages in predictive value, its high cost limits its application in routine research.

Through clinical studies, siRNA therapy must methodically assess its long-term consequences and unforeseen biological interactions in order to be considered safe. Comprehensive pharmacokinetic, pharmacodynamic, and toxicological data are necessary for regulatory approval. The approval process is complicated and time-consuming since it is challenging to gather the required data because of the distinct molecular characteristics of siRNA. Furthermore, the secret to achieving commercial application is the scalable production method. However, the widespread promotion of its therapeutic transformation is hampered by the high prices and technological obstacles that still plague the current large-scale production [49].

6.  Conclusion

The application of siRNA-based therapies in breast cancer treatment has shown significant promise, particularly in targeting oncogenes such as Bcl-2, Bcl-xL, and survivin. Nanotechnology-mediated siRNA delivery systems have demonstrated the ability to enhance cellular uptake and endosomal escape, thereby improving the efficiency of targeted gene silencing. These advancements are especially notable in aggressive subtypes like triple-negative breast cancer (TNBC). However, several challenges remain in translating these therapies from preclinical models to clinical practice. Key issues include the specificity of siRNA delivery, the risk of immunogenicity, potential off-target effects that may silence essential genes, inadequate penetration of the blood-tumor barrier, and evolving regulatory frameworks. Future research should focus on developing "smart" stimulus-responsive nanovectors to improve targeting accuracy and reduce side effects. Additionally, combining siRNA therapy with other treatment modalities, such as immunotherapy or chemotherapy, may enhance therapeutic efficacy and overcome drug resistance. Moreover, optimizing siRNA chemical modifications to reduce off-target effects and increase nuclease resistance is crucial. This includes exploring 2'-O-methyl and phosphorothioate substitutions, among others. Systematic comparisons between different carrier systems are also necessary to identify the most effective and safe delivery methods. While solid lipid nanoparticles offer high biocompatibility, metallic vectors like gold nanoshells must be carefully evaluated for potential long-term toxicity. Addressing challenges related to scalable manufacturing, long-term formulation stability, and the risk of immunogenicity will be essential for the successful clinical translation of siRNA therapies. Integrating computational modeling and high-throughput screening strategies will play a pivotal role in overcoming these hurdles and realizing the therapeutic potential of siRNA-based interventions in cancer treatment.


References

[1]. Banerjee, M., & Rajeswari, V. D. (2023). Critical Review on the Different Roles of Exosomes in TNBC and Exosomal-Mediated Delivery of microRNA/siRNA/lncRNA and Drug Targeting Signalling Pathways in Triple-Negative Breast Cancer. Molecules (Basel, Switzerland), 28(4), 1802.

[2]. Mirzaei, S., Paskeh, M. D. A., Entezari, M., Bidooki, S. H., Ghaleh, V. J., Rezaei, S., Hejazi, E. S., Kakavand, A., Behroozaghdam, M., Movafagh, A., Taheriazam, A., Hashemi, M., & Samarghandian, S. (2023). siRNA and targeted delivery systems in breast cancer therapy. Clinical & Translational Oncology: Official Publication of the Federation of Spanish Oncology Societies and of the National Cancer Institute of Mexico, 25(5), 1167–1188.

[3]. Jiang, Y.-Z., et al. (2021). Molecular subtyping and genomic profiling expand precision medicine in refractory metastatic triple-negative breast cancer: The FUTURE trial. Cell Research, 31(2), 178–186.

[4]. Kalra, M., Tong, Y., Jones, D. R., Walsh, T., Danso, M. A., Ma, C. X., Silverman, P., King, M.-C., Badve, S. S., Perkins, S. M., & Miller, K. D. (2021). Cisplatin +/- rucaparib after preoperative chemotherapy in patients with triple-negative or BRCA mutated breast cancer. NPJ Breast Cancer, 7(1), 29.

[5]. Lu, F., et al. (2021). Efficacy and Safety of Platinum-Based Chemotherapy as First-Line Therapy for Metastatic Triple-Negative Breast Cancer: A Meta-Analysis of Randomized Controlled Trials. Technology in Cancer Research & Treatment, 20, 15330338211016369. https: //doi.org/10.1177/15330338211016369

[6]. R, Y., Yy, S., Xh, H., & S, L. (2021). The Impact of Platinum-Containing Chemotherapies in Advanced Triple-Negative Breast Cancer: Meta-Analytical Approach to Evaluating Its Efficacy and Safety. Oncology Research and Treatment, 44(6). https: //doi.org/10.1159/000515353

[7]. Iliopoulos, D., Hirsch, H. A., & Struhl, K. (2009). An epigenetic switch involving NF-κB, Lin28, Let-7 MicroRNA, and IL6 links inflammation to cell transformation. Cell, 139(4), 693-706

[8]. Liang, H., Li, F., et al. (2025). A novel peptide 66CTG stabilizes Myc proto-oncogene protein to promote triple-negative breast cancer growth. Signal Transduction and Targeted Therapy, 10(1), 217.

[9]. Bai, F., Zhang, L. H., Liu, X., Wang, C., Zheng, C., Sun, J., ... & Pei, X. H. (2021). GATA3 functions downstream of BRCA1 to suppress EMT in breast cancer. Theranostics, 11(17), 8218.

[10]. Delbridge, A. R., Grabow, S., Strasser, A., & Vaux, D. L. (2016). Thirty years of BCL-2: translating cell death discoveries into novel cancer therapies. Nature Reviews Cancer, 16(2), 99-109.

[11]. Altieri, D. C. (2008). Survivin, cancer networks and pathway-directed drug discovery. Nature Reviews Cancer, 8(1), 61-70.

[12]. Moody, S. E., Perez, D., Pan, T. C., Sarkisian, C. J., Portocarrero, C. P., Sterner, C. J., ... & Chodosh, L. A. (2005). The transcriptional repressor Snail promotes mammary tumor recurrence. Cancer cell, 8(3), 197-209.

[13]. Yang, J., Mani, S. A., Donaher, J. L., Ramaswamy, S., Itzykson, R. A., Come, C., ... & Weinberg, R. A. (2004). Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. cell, 117(7), 927-939.

[14]. Spaderna, S., et al. (2006). A transient, EMT-linked loss of basement membranes indicates metastasis and poor survival in colorectal cancer. Gastroenterology, 131(3), 830-840.

[15]. Itoh, T., Tanioka, M., Yoshida, H., Yoshioka, T., Nishimoto, H., & Itohara, S. (1998). Reduced angiogenesis and tumor progression in gelatinase A-deficient mice. Cancer research, 58(5), 1048-1051.

[16]. Soucek, L., Whitfield, J., Martins, C. P., Finch, A. J., Murphy, D. J., Sodir, N. M., ... & Evan, G. I. (2008). Modelling Myc inhibition as a cancer therapy. Nature, 455(7213), 679-683.

[17]. Chou, J., Provot, S., & Werb, Z. (2010). GATA3 in development and cancer differentiation: cells GATA have it!. Journal of cellular physiology, 222(1), 42-49.

[18]. Tao, R., Wang, G., Fang, D. D., Zhai, G., Li, Y., Lv, J., ... & Zhai, Y. (2020). Combination of BCL-2/BCL-xL dual inhibitor APG-1252 and chemotherapeutics overcomes resistance to osimertinib in EGFR mutant NSCLC in preclinical models. Cancer Research, 80(16_Supplement), 6223-6223.

[19]. Ryan, B. M., O’Donovan, N., & Duffy, M. J. (2009). Survivin: a new target for anti-cancer therapy. Cancer treatment reviews, 35(7), 553-562.

[20]. Zhang, X., Kon, T., Wang, H., Li, F., Huang, Q., Rabbani, Z. N., ... & Li, C. Y. (2004). Enhancement of hypoxia-induced tumor cell death in vitro and radiation therapy in vivo by use of small interfering RNA targeted to hypoxia-inducible factor-1α. Cancer research, 64(22), 8139-8142.

[21]. Desgrosellier, J. S., Barnes, L. A., Shields, D. J., Huang, M., Lau, S. K., Prévost, N., ... & Cheresh, D. A. (2009). An integrin αvβ3–c-Src oncogenic unit promotes anchorage-independence and tumor progression. Nature medicine, 15(10), 1163-1169.

[22]. Vermeulen, L. M. P., De Smedt, S. C., Remaut, K., & Braeckmans, K. (2018). The proton sponge hypothesis: Fable or fact? European Journal of Pharmaceutics and Biopharmaceutics: Official Journal of Arbeitsgemeinschaft Fur Pharmazeutische Verfahrenstechnik e.V, 129, 184–190.

[23]. Alshaer, W., et al. (2018). Aptamer-guided siRNA-loaded nanomedicines for systemic gene silencing in CD-44 expressing murine triple-negative breast cancer model. Journal of Controlled Release: Official Journal of the Controlled Release Society, 271, 98–106. https: //doi.org/10.1016/j.jconrel.2017.12.022

[24]. Li, Z., Cheng, L., Xu, X., Jia, R., Zhu, S., Zhang, Q., Cheng, G., Wu, B., Liu, Z., Tong, X., Xiao, B., & Dai, F. (2024). Cuproptosis-based layer-by-layer silk fibroin nanoplatform-loaded PD-L1 siRNA combining photothermal and chemodynamic therapy against metastatic breast cancer. Materials Today. Bio, 29, 101298.

[25]. Ferreira, C. S. M., Matthews, C. S., & Missailidis, S. (2006). DNA aptamers that bind to MUC1 tumour marker: Design and characterization of MUC1-binding single-stranded DNA aptamers. Tumour Biology: The Journal of the International Society for Oncodevelopmental Biology and Medicine, 27(6), 289–301.

[26]. Jafari, R., et al. (2019). Anti-Mucin1 Aptamer-Conjugated Chitosan Nanoparticles for Targeted Co-Delivery of Docetaxel and IGF-1R siRNA to SKBR3 Metastatic Breast Cancer Cells. Iranian Biomedical Journal, 23(1), 21–33. https: //doi.org/10.29252/.23.1.21

[27]. Subhan, A., Attia, S. A., & P Torchilin, V. (2022). Targeted siRNA nanotherapeutics against breast and ovarian metastatic cancer: A comprehensive review of the literature. Nanomedicine (London, England), 17(1), 41–64.

[28]. Li, M., et al. (2022). Cationic liposomes co-deliver chemotherapeutics and siRNA for the treatment of breast cancer. European Journal of Medicinal Chemistry, 233, 114198. https: //doi.org/10.1016/j.ejmech.2022.114198

[29]. Asadi, H., et al. (2018). Novel lipid-polymer hybrid nanoparticles for siRNA delivery and IGF-1R gene silencing in breast cancer cells. Journal of Drug Delivery Science and Technology, 48, 96-105.

[30]. Sai, B. M., et al. (2024b). Therapeutic delivery of siRNA for the management of breast cancer and triple-negative breast cancer. Therapeutic Delivery, 15(11), 871–891.

[31]. Yang, Y.-Y., Zhang, W., Liu, H., Jiang, J.-J., Wang, W.-J., & Jia, Z.-Y. (2021). Cell-Penetrating Peptide-Modified Graphene Oxide Nanoparticles Loaded with Rictor siRNA for the Treatment of Triple-Negative Breast Cancer. Drug Design, Development and Therapy, 15, 4961–4972. https: //doi.org/10.2147/DDDT.S330059

[32]. Badparvar, F., Marjani, A. P., Salehi, R., & Ramezani, F. (2024). Dual pH/redox-responsive hyperbranched polymeric nanocarriers with TME-trigger size shrinkage and charge reversible ability for amplified chemotherapy of breast cancer. Scientific Reports, 14(1), 8567. https: //doi.org/10.1038/s41598-024-57296-4

[33]. Bakhtiar, A., Liew, Q. X., Ng, K. Y., & Chowdhury, E. H. (2022). Active targeting via ligand-anchored pH-responsive strontium nanoparticles for efficient nucleic acid delivery into breast cancer cells. Journal of Pharmaceutical Investigation, 52(2), 243–257. https: //doi.org/10.1007/s40005-022-00559-x

[34]. Zhang, C., et al. (2021). Co-delivery of EGFR and BRD4 siRNA by cell-penetrating peptides-modified redox-responsive complex in triple negative breast cancer cells. Life Sciences, 266, 118886.

[35]. Semple, S. C., Leone, R., Barbosa, C. J., Tam, Y. K., & Lin, P. J. C. (2022). Lipid Nanoparticle Delivery Systems to Enable mRNA-Based Therapeutics. Pharmaceutics, 14(2), 398.

[36]. Jain, R. K. (1999). Transport of molecules, particles, and cells in solid tumors. Annual Review of Biomedical Engineering, 1, 241–263. https: //doi.org/10.1146/annurev.bioeng.1.1.241

[37]. Wilhelm, S., et al. (2016). Analysis of nanoparticle delivery to tumours. Nature Reviews Materials, 1(5), 16014.

[38]. Byrne, J. D., Betancourt, T., & Brannon-Peppas, L. (2008). Active targeting schemes for nanoparticle systems in cancer therapeutics. Advanced Drug Delivery Reviews, 60(15), 1615–1626.

[39]. Daniels, T. R., et al. (2012). The transferrin receptor and the targeted delivery of therapeutic agents against cancer. Biochimica Et Biophysica Acta, 1820(3), 291–317.

[40]. Moitra, P., Skrodzki, D., Molinaro, M., Gunaseelan, N., Sar, D., Aditya, T., Dahal, D., Ray, P., & Pan, D. (2024). Context-Responsive Nanoparticle Derived from Synthetic Zwitterionic Ionizable Phospholipids in Targeted CRISPR/Cas9 Therapy for Basal-like Breast Cancer. ACS Nano, 18(12), 9199–9220.

[41]. Li, J., Dirisala, et al. (2017). Therapeutic Vesicular Nanoreactors with Tumor-Specific Activation and Self-Destruction for Synergistic Tumor Ablation. Angewandte Chemie (International Ed. in English), 56(45), 14025–14030.

[42]. Wang, Y., et al. (2014). A nanoparticle-based strategy for the imaging of a broad range of tumours by nonlinear amplification of microenvironment signals. Nature Materials, 13(2), 204–212. https: //doi.org/10.1038/nmat3819

[43]. Mura, S., Nicolas, J., & Couvreur, P. (2013). Stimuli-responsive nanocarriers for drug delivery. Nature Materials, 12(11), 991–1003. https: //doi.org/10.1038/nmat3776

[44]. Anderson, B. R., et al. (2011). Nucleoside modifications in RNA limit activation of 2’-5’-oligoadenylate synthetase and increase resistance to cleavage by RNase L. Nucleic Acids Research, 39(21), 9329–9338.

[45]. Roberts, T. C., Langer, R., & Wood, M. J. A. (2020). Advances in oligonucleotide drug delivery. Nature Reviews. Drug Discovery, 19(10), 673–694. https: //doi.org/10.1038/s41573-020-0075-7

[46]. Zhu, L., Wang, T., Perche, F., Taigind, A., & Torchilin, V. P. (2013). Enhanced anticancer activity of nanopreparation containing an MMP2-sensitive PEG-drug conjugate and cell-penetrating moiety. Proceedings of the National Academy of Sciences of the United States of America, 110(42), 17047–17052.

[47]. Dreaden, E. C., Alkilany, A. M., Huang, X., Murphy, C. J., & El-Sayed, M. A. (2012). The golden age: Gold nanoparticles for biomedicine. Chemical Society Reviews, 41(7), 2740–2779. https: //doi.org/10.1039/c1cs15237h

[48]. Khlebtsov, N., & Dykman, L. (2011). Biodistribution and toxicity of engineered gold nanoparticles: A review of in vitro and in vivo studies. Chemical Society Reviews, 40(3), 1647–1671. https: //doi.org/10.1039/c0cs00018c

[49]. Ngamcherdtrakul, W., & Yantasee, W. (2019). siRNA therapeutics for breast cancer: Recent efforts in targeting metastasis, drug resistance, and immune evasion. Translational Research: The Journal of Laboratory and Clinical Medicine, 214, 105–120. https: //doi.org/10.1016/j.trsl.2019.08.005

[50]. Morad, G., et al. (2019). Tumor-Derived Extracellular Vesicles Breach the Intact Blood-Brain Barrier via Transcytosis. ACS Nano, 13(12), 13853–13865. https: //doi.org/10.1021/acsnano.9b04397


Cite this article

Wang,H. (2025). Research Progress of siRNA Systems in Breast Cancer Therapy. Theoretical and Natural Science,144,62-70.

Data availability

The datasets used and/or analyzed during the current study will be available from the authors upon reasonable request.

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 EWA Publishing and/or the editor(s). EWA Publishing 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.

About volume

Volume title: Proceedings of ICBioMed 2025 Symposium: AI for Healthcare: Advanced Medical Data Analytics and Smart Rehabilitation

ISBN:978-1-80590-441-0(Print) / 978-1-80590-442-7(Online)
Editor:Alan Wang
Conference date: 17 October 2025
Series: Theoretical and Natural Science
Volume number: Vol.144
ISSN:2753-8818(Print) / 2753-8826(Online)

© 2024 by the author(s). Licensee EWA Publishing, Oxford, UK. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license. Authors who publish this series agree to the following terms:
1. Authors retain copyright and grant the series right of first publication with the work simultaneously licensed under a Creative Commons Attribution License that allows others to share the work with an acknowledgment of the work's authorship and initial publication in this series.
2. Authors are able to enter into separate, additional contractual arrangements for the non-exclusive distribution of the series's published version of the work (e.g., post it to an institutional repository or publish it in a book), with an acknowledgment of its initial publication in this series.
3. Authors are permitted and encouraged to post their work online (e.g., in institutional repositories or on their website) prior to and during the submission process, as it can lead to productive exchanges, as well as earlier and greater citation of published work (See Open access policy for details).

References

[1]. Banerjee, M., & Rajeswari, V. D. (2023). Critical Review on the Different Roles of Exosomes in TNBC and Exosomal-Mediated Delivery of microRNA/siRNA/lncRNA and Drug Targeting Signalling Pathways in Triple-Negative Breast Cancer. Molecules (Basel, Switzerland), 28(4), 1802.

[2]. Mirzaei, S., Paskeh, M. D. A., Entezari, M., Bidooki, S. H., Ghaleh, V. J., Rezaei, S., Hejazi, E. S., Kakavand, A., Behroozaghdam, M., Movafagh, A., Taheriazam, A., Hashemi, M., & Samarghandian, S. (2023). siRNA and targeted delivery systems in breast cancer therapy. Clinical & Translational Oncology: Official Publication of the Federation of Spanish Oncology Societies and of the National Cancer Institute of Mexico, 25(5), 1167–1188.

[3]. Jiang, Y.-Z., et al. (2021). Molecular subtyping and genomic profiling expand precision medicine in refractory metastatic triple-negative breast cancer: The FUTURE trial. Cell Research, 31(2), 178–186.

[4]. Kalra, M., Tong, Y., Jones, D. R., Walsh, T., Danso, M. A., Ma, C. X., Silverman, P., King, M.-C., Badve, S. S., Perkins, S. M., & Miller, K. D. (2021). Cisplatin +/- rucaparib after preoperative chemotherapy in patients with triple-negative or BRCA mutated breast cancer. NPJ Breast Cancer, 7(1), 29.

[5]. Lu, F., et al. (2021). Efficacy and Safety of Platinum-Based Chemotherapy as First-Line Therapy for Metastatic Triple-Negative Breast Cancer: A Meta-Analysis of Randomized Controlled Trials. Technology in Cancer Research & Treatment, 20, 15330338211016369. https: //doi.org/10.1177/15330338211016369

[6]. R, Y., Yy, S., Xh, H., & S, L. (2021). The Impact of Platinum-Containing Chemotherapies in Advanced Triple-Negative Breast Cancer: Meta-Analytical Approach to Evaluating Its Efficacy and Safety. Oncology Research and Treatment, 44(6). https: //doi.org/10.1159/000515353

[7]. Iliopoulos, D., Hirsch, H. A., & Struhl, K. (2009). An epigenetic switch involving NF-κB, Lin28, Let-7 MicroRNA, and IL6 links inflammation to cell transformation. Cell, 139(4), 693-706

[8]. Liang, H., Li, F., et al. (2025). A novel peptide 66CTG stabilizes Myc proto-oncogene protein to promote triple-negative breast cancer growth. Signal Transduction and Targeted Therapy, 10(1), 217.

[9]. Bai, F., Zhang, L. H., Liu, X., Wang, C., Zheng, C., Sun, J., ... & Pei, X. H. (2021). GATA3 functions downstream of BRCA1 to suppress EMT in breast cancer. Theranostics, 11(17), 8218.

[10]. Delbridge, A. R., Grabow, S., Strasser, A., & Vaux, D. L. (2016). Thirty years of BCL-2: translating cell death discoveries into novel cancer therapies. Nature Reviews Cancer, 16(2), 99-109.

[11]. Altieri, D. C. (2008). Survivin, cancer networks and pathway-directed drug discovery. Nature Reviews Cancer, 8(1), 61-70.

[12]. Moody, S. E., Perez, D., Pan, T. C., Sarkisian, C. J., Portocarrero, C. P., Sterner, C. J., ... & Chodosh, L. A. (2005). The transcriptional repressor Snail promotes mammary tumor recurrence. Cancer cell, 8(3), 197-209.

[13]. Yang, J., Mani, S. A., Donaher, J. L., Ramaswamy, S., Itzykson, R. A., Come, C., ... & Weinberg, R. A. (2004). Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. cell, 117(7), 927-939.

[14]. Spaderna, S., et al. (2006). A transient, EMT-linked loss of basement membranes indicates metastasis and poor survival in colorectal cancer. Gastroenterology, 131(3), 830-840.

[15]. Itoh, T., Tanioka, M., Yoshida, H., Yoshioka, T., Nishimoto, H., & Itohara, S. (1998). Reduced angiogenesis and tumor progression in gelatinase A-deficient mice. Cancer research, 58(5), 1048-1051.

[16]. Soucek, L., Whitfield, J., Martins, C. P., Finch, A. J., Murphy, D. J., Sodir, N. M., ... & Evan, G. I. (2008). Modelling Myc inhibition as a cancer therapy. Nature, 455(7213), 679-683.

[17]. Chou, J., Provot, S., & Werb, Z. (2010). GATA3 in development and cancer differentiation: cells GATA have it!. Journal of cellular physiology, 222(1), 42-49.

[18]. Tao, R., Wang, G., Fang, D. D., Zhai, G., Li, Y., Lv, J., ... & Zhai, Y. (2020). Combination of BCL-2/BCL-xL dual inhibitor APG-1252 and chemotherapeutics overcomes resistance to osimertinib in EGFR mutant NSCLC in preclinical models. Cancer Research, 80(16_Supplement), 6223-6223.

[19]. Ryan, B. M., O’Donovan, N., & Duffy, M. J. (2009). Survivin: a new target for anti-cancer therapy. Cancer treatment reviews, 35(7), 553-562.

[20]. Zhang, X., Kon, T., Wang, H., Li, F., Huang, Q., Rabbani, Z. N., ... & Li, C. Y. (2004). Enhancement of hypoxia-induced tumor cell death in vitro and radiation therapy in vivo by use of small interfering RNA targeted to hypoxia-inducible factor-1α. Cancer research, 64(22), 8139-8142.

[21]. Desgrosellier, J. S., Barnes, L. A., Shields, D. J., Huang, M., Lau, S. K., Prévost, N., ... & Cheresh, D. A. (2009). An integrin αvβ3–c-Src oncogenic unit promotes anchorage-independence and tumor progression. Nature medicine, 15(10), 1163-1169.

[22]. Vermeulen, L. M. P., De Smedt, S. C., Remaut, K., & Braeckmans, K. (2018). The proton sponge hypothesis: Fable or fact? European Journal of Pharmaceutics and Biopharmaceutics: Official Journal of Arbeitsgemeinschaft Fur Pharmazeutische Verfahrenstechnik e.V, 129, 184–190.

[23]. Alshaer, W., et al. (2018). Aptamer-guided siRNA-loaded nanomedicines for systemic gene silencing in CD-44 expressing murine triple-negative breast cancer model. Journal of Controlled Release: Official Journal of the Controlled Release Society, 271, 98–106. https: //doi.org/10.1016/j.jconrel.2017.12.022

[24]. Li, Z., Cheng, L., Xu, X., Jia, R., Zhu, S., Zhang, Q., Cheng, G., Wu, B., Liu, Z., Tong, X., Xiao, B., & Dai, F. (2024). Cuproptosis-based layer-by-layer silk fibroin nanoplatform-loaded PD-L1 siRNA combining photothermal and chemodynamic therapy against metastatic breast cancer. Materials Today. Bio, 29, 101298.

[25]. Ferreira, C. S. M., Matthews, C. S., & Missailidis, S. (2006). DNA aptamers that bind to MUC1 tumour marker: Design and characterization of MUC1-binding single-stranded DNA aptamers. Tumour Biology: The Journal of the International Society for Oncodevelopmental Biology and Medicine, 27(6), 289–301.

[26]. Jafari, R., et al. (2019). Anti-Mucin1 Aptamer-Conjugated Chitosan Nanoparticles for Targeted Co-Delivery of Docetaxel and IGF-1R siRNA to SKBR3 Metastatic Breast Cancer Cells. Iranian Biomedical Journal, 23(1), 21–33. https: //doi.org/10.29252/.23.1.21

[27]. Subhan, A., Attia, S. A., & P Torchilin, V. (2022). Targeted siRNA nanotherapeutics against breast and ovarian metastatic cancer: A comprehensive review of the literature. Nanomedicine (London, England), 17(1), 41–64.

[28]. Li, M., et al. (2022). Cationic liposomes co-deliver chemotherapeutics and siRNA for the treatment of breast cancer. European Journal of Medicinal Chemistry, 233, 114198. https: //doi.org/10.1016/j.ejmech.2022.114198

[29]. Asadi, H., et al. (2018). Novel lipid-polymer hybrid nanoparticles for siRNA delivery and IGF-1R gene silencing in breast cancer cells. Journal of Drug Delivery Science and Technology, 48, 96-105.

[30]. Sai, B. M., et al. (2024b). Therapeutic delivery of siRNA for the management of breast cancer and triple-negative breast cancer. Therapeutic Delivery, 15(11), 871–891.

[31]. Yang, Y.-Y., Zhang, W., Liu, H., Jiang, J.-J., Wang, W.-J., & Jia, Z.-Y. (2021). Cell-Penetrating Peptide-Modified Graphene Oxide Nanoparticles Loaded with Rictor siRNA for the Treatment of Triple-Negative Breast Cancer. Drug Design, Development and Therapy, 15, 4961–4972. https: //doi.org/10.2147/DDDT.S330059

[32]. Badparvar, F., Marjani, A. P., Salehi, R., & Ramezani, F. (2024). Dual pH/redox-responsive hyperbranched polymeric nanocarriers with TME-trigger size shrinkage and charge reversible ability for amplified chemotherapy of breast cancer. Scientific Reports, 14(1), 8567. https: //doi.org/10.1038/s41598-024-57296-4

[33]. Bakhtiar, A., Liew, Q. X., Ng, K. Y., & Chowdhury, E. H. (2022). Active targeting via ligand-anchored pH-responsive strontium nanoparticles for efficient nucleic acid delivery into breast cancer cells. Journal of Pharmaceutical Investigation, 52(2), 243–257. https: //doi.org/10.1007/s40005-022-00559-x

[34]. Zhang, C., et al. (2021). Co-delivery of EGFR and BRD4 siRNA by cell-penetrating peptides-modified redox-responsive complex in triple negative breast cancer cells. Life Sciences, 266, 118886.

[35]. Semple, S. C., Leone, R., Barbosa, C. J., Tam, Y. K., & Lin, P. J. C. (2022). Lipid Nanoparticle Delivery Systems to Enable mRNA-Based Therapeutics. Pharmaceutics, 14(2), 398.

[36]. Jain, R. K. (1999). Transport of molecules, particles, and cells in solid tumors. Annual Review of Biomedical Engineering, 1, 241–263. https: //doi.org/10.1146/annurev.bioeng.1.1.241

[37]. Wilhelm, S., et al. (2016). Analysis of nanoparticle delivery to tumours. Nature Reviews Materials, 1(5), 16014.

[38]. Byrne, J. D., Betancourt, T., & Brannon-Peppas, L. (2008). Active targeting schemes for nanoparticle systems in cancer therapeutics. Advanced Drug Delivery Reviews, 60(15), 1615–1626.

[39]. Daniels, T. R., et al. (2012). The transferrin receptor and the targeted delivery of therapeutic agents against cancer. Biochimica Et Biophysica Acta, 1820(3), 291–317.

[40]. Moitra, P., Skrodzki, D., Molinaro, M., Gunaseelan, N., Sar, D., Aditya, T., Dahal, D., Ray, P., & Pan, D. (2024). Context-Responsive Nanoparticle Derived from Synthetic Zwitterionic Ionizable Phospholipids in Targeted CRISPR/Cas9 Therapy for Basal-like Breast Cancer. ACS Nano, 18(12), 9199–9220.

[41]. Li, J., Dirisala, et al. (2017). Therapeutic Vesicular Nanoreactors with Tumor-Specific Activation and Self-Destruction for Synergistic Tumor Ablation. Angewandte Chemie (International Ed. in English), 56(45), 14025–14030.

[42]. Wang, Y., et al. (2014). A nanoparticle-based strategy for the imaging of a broad range of tumours by nonlinear amplification of microenvironment signals. Nature Materials, 13(2), 204–212. https: //doi.org/10.1038/nmat3819

[43]. Mura, S., Nicolas, J., & Couvreur, P. (2013). Stimuli-responsive nanocarriers for drug delivery. Nature Materials, 12(11), 991–1003. https: //doi.org/10.1038/nmat3776

[44]. Anderson, B. R., et al. (2011). Nucleoside modifications in RNA limit activation of 2’-5’-oligoadenylate synthetase and increase resistance to cleavage by RNase L. Nucleic Acids Research, 39(21), 9329–9338.

[45]. Roberts, T. C., Langer, R., & Wood, M. J. A. (2020). Advances in oligonucleotide drug delivery. Nature Reviews. Drug Discovery, 19(10), 673–694. https: //doi.org/10.1038/s41573-020-0075-7

[46]. Zhu, L., Wang, T., Perche, F., Taigind, A., & Torchilin, V. P. (2013). Enhanced anticancer activity of nanopreparation containing an MMP2-sensitive PEG-drug conjugate and cell-penetrating moiety. Proceedings of the National Academy of Sciences of the United States of America, 110(42), 17047–17052.

[47]. Dreaden, E. C., Alkilany, A. M., Huang, X., Murphy, C. J., & El-Sayed, M. A. (2012). The golden age: Gold nanoparticles for biomedicine. Chemical Society Reviews, 41(7), 2740–2779. https: //doi.org/10.1039/c1cs15237h

[48]. Khlebtsov, N., & Dykman, L. (2011). Biodistribution and toxicity of engineered gold nanoparticles: A review of in vitro and in vivo studies. Chemical Society Reviews, 40(3), 1647–1671. https: //doi.org/10.1039/c0cs00018c

[49]. Ngamcherdtrakul, W., & Yantasee, W. (2019). siRNA therapeutics for breast cancer: Recent efforts in targeting metastasis, drug resistance, and immune evasion. Translational Research: The Journal of Laboratory and Clinical Medicine, 214, 105–120. https: //doi.org/10.1016/j.trsl.2019.08.005

[50]. Morad, G., et al. (2019). Tumor-Derived Extracellular Vesicles Breach the Intact Blood-Brain Barrier via Transcytosis. ACS Nano, 13(12), 13853–13865. https: //doi.org/10.1021/acsnano.9b04397