Zhenghao
Tao†
,
Haitao
Zhang†
*,
Shang
Wu
,
Jiaheng
Zhang
,
Yao
Cheng
,
Longtianyang
Lei
,
Yang
Qin
,
Hua
Wei
* and
Cui-Yun
Yu
*
Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, 421001, Hengyang, P. R. China. E-mail: zhanghaitao@usc.edu.cn; weih@usc.edu.cn; yucuiyunusc@hotmail.com
First published on 11th January 2024
Gene therapy is a revolutionary treatment approach in the 21st century, offering significant potential for disease prevention and treatment. However, the efficacy of gene delivery is often compromised by the inherent challenges of gene properties and vector-related defects. It is crucial to explore ways to enhance the curative effect of gene drugs and achieve safer, more widespread, and more efficient utilization, which represents a significant challenge in amplification gene therapy advancements. Spherical nucleic acids (SNAs), with their unique physicochemical properties, are considered an innovative solution for scalable gene therapy. This review aims to comprehensively explore the amplifying contributions of SNAs in gene therapy and emphasize the contribution of SNAs to the amplification effect of gene therapy from the aspects of structure, application, and recent clinical translation – an aspect that has been rarely reported or explored thus far. We begin by elucidating the fundamental characteristics and scaling-up properties of SNAs that distinguish them from traditional linear nucleic acids, followed by an analysis of combined therapy treatment strategies, theranostics, and clinical translation amplified by SNAs. We conclude by discussing the challenges of SNAs and provide a prospect on the amplification characteristics. This review seeks to update the current understanding of the use of SNAs in gene therapy amplification and promote further research into their clinical translation and amplification of gene therapy.
In response to this challenge, researchers have explored both viral and non-viral vectors for gene delivery, achieving noteworthy advancements.12 Viral vectors, including adenoviruses, retroviruses, and lentiviruses, facilitate gene transcription by silencing disease-causing genes and integrating targeted gene sequences into the host genome.13 Although viral vectors demonstrate high transfection efficiency, their safety remains a subject of debate. In contrast, non-viral vectors, including liposome complexes, cationic polymers, and inorganic nanoparticles, are favored for their low immunogenicity, ease of quality control, and scalability, thereby presenting significant benefits.14,15 This dichotomy has spurred the development of innovative gene delivery vehicles that optimize gene utilization and enhance quantity, content, and functionality. Numerous gene therapeutic drugs are currently undergoing clinical trials.9,16 However, most of the reported trials, to our knowledge, remain in the early and preliminary clinical stages likely due to the significantly compromised transfection efficiency for clinical treatment. To move toward better clinical translations, it is crucial to develop approaches that can improve the therapeutic efficacy of gene drugs, ensuring safer, more extensive and efficient utilization, which requires sophisticated design and comprehensive consideration in the construction of advanced gene delivery vectors.
Over the past decade, the unique structure of spherical nucleic acids (SNAs) has been unveiled, leading to the development of SNA-based platforms with medical implications. Since Mirkin first combined DNA oligonucleotides with gold nanoparticles to define SNA structures,17 SNAs have been widely recognized as novel nanocarriers consisting of densely packed nucleic acid shells and cores. Owing to their unique three-dimensional structure and highly programmable assembly, SNAs exhibit a range of distinctive physicochemical properties, including reduced degradation risk in the presence of nucleases, efficient transfection of various tissues and cells, and good immunogenicity.18–21 As an innovative nucleic acid delivery system, SNAs possess immense potential as universal transportation nanoplatforms for nucleic acids, drugs, and proteins, with enhanced clinical translation prospects in biomedical applications; therefore, there have been many reviews with a focus on this hot subject of research. For example, Jiang et al. presented thorough elucidation of the selection process concerning the core and densely organized DNA shells within SNAs, and further introduced the applications of SNAs in disease diagnosis and treatment, encompassing in vitro biosensing, intracellular assessment, gene regulation, drug delivery, and immune modulation. Stegh et al. recently summarized the results of the first-in-human clinical trials involving SNAs in the context of solid tumors with an emphasis on the potential of SNAs as innovative gene regulatory and immunostimulatory structures for overcoming drug resistance and immunosuppression in solid tumors. Very recently, our group delineated significant advances in SNA-based precision therapy and immunotherapy for tumors with a focus on the therapeutic insights derived from gene-level precision fluorescence imaging and the utilization of SNAs for meticulous adjuvant and antigen control to achieve optimal immunomodulation.19,20,22,23 Despite many reviews on SNAs, there is a lack of reviews, to our knowledge, summarizing the amplification effect of SNAs, particularly in the context of clinical translations, which is considered to be the most important and noteworthy part of gene therapy amplification.
This review aims to explore the amplification role of SNAs and to summarize the reported notable examples with in-depth discussion and concluding remarks. Briefly, we first elaborate the essential characteristics and scaling-up advantages of SNAs relative to the traditionally used linear nucleic acids. Next, we showcase the amplification strategies of SNAs in diverse therapeutic modalities by summarizing combinatory treatment approaches, along with discussion on the theranostics of SNAs and the presentation of the latest reported clinical translations. Finally, we make instructive concluding remarks on the existing challenges and future prospects in this rapidly developing field of SNAs, including the current obstacles encountered as well as the corresponding emerging solutions. Last but not least, this review offers unique insights into how the amplification characteristics of SNAs can facilitate reasonable design outcomes. The significant role and contribution of SNAs to gene therapy undoubtedly require a timely updated review with a main aim to summarize the amplification attributes as valuable guidance for future clinical translations and rational advanced design of SNA nanoplatforms (Fig. 1).
For example, Liu successfully developed a low-cost and easily producible integrated SNA by assembling hairpin probes on sea urchin-like DNA nanostructures, which significantly reduces the reaction time compared to traditional chain reaction strategies and enhances the detection sensitivity, therefore facilitating the monitoring of low-abundance tumor-related miRNA in living cells (Fig. 2C).34 Another innovative sandwich-type electrochemical immunosensor prepared using an SNA-templated silver nanocluster sensing platform and immobilizing the secondary antibody through host–guest recognition demonstrated the application of spherical nucleic acids in the detection of tumor markers, especially the hepatocellular carcinoma marker alpha-fetoprotein.35 Electrochemical reduction of silver nanoclusters can significantly amplify the immune response signal, which gives the sensor a wide linear range of 0.001 to 100 ng mL−1 and a detection limit of 7.74 fg mL−1, which is comparable or even better performance than conventional methods.
![]() | ||
Fig. 2 Typical functional applications of dense nucleic acid shells. (A) SNA containing ASO and tyrosinase inhibitor prodrugs, enhanced skin permeation and reduced melanin content in B16F10 melanoma cells. Reproduced from ref. 46. Copyright 2021 American Chemical Society. (B) Protein SNAs modified with β-galactosidase-conjugated transferrin receptors (TfR), significantly increasing the accumulation in brain tissues and successfully transporting proteins to the brain and central nervous system. Reproduced from ref. 51. Copyright 2022 American Chemical Society. (C) A low-cost and easily producible integrated SNA that significantly reduces the reaction time and enhances the detection sensitivity, therefore facilitating the monitoring of low-abundance tumor-related miRNA in living cells. Reproduced from ref. 34. Copyright 2023 Elsevier B.V. (D) A study conducted a comprehensive analysis on the stability of nucleic acids in spherical nucleic structures, establishing a correlation between the enzyme-mediated DNA degradation and the oligonucleotide density on nanoparticles. Reproduced from ref. 59 Copyright 2022 American Chemical Society. |
The high affinity and ultra-sensitivity of SNAs not only perform well in the field of cancer, but also have a positive impact on other diseases. For example, Li achieved ultra-sensitive detection of low-abundance myocardial infarction-related miRNAs within 30 minutes by using an electrochemiluminescence strategy, which utilized c-SNA enzymes as nanocatalysts.36 Hu developed a competitive induced fluorescence detection method using sRNA to efficiently identify drinking water standards.37 This highly sensitive detection capability and simplified process contribute to real-time field and other monitoring tasks.
In a word, rapid binding affinity is an essential breakthrough for an ultra-trace or time-consuming study, and SNAs can increase the productivity and efficiency as a powerful tool for potential clinical translation.
Local administration is favored as one of the optimal therapeutic methods due to its convenience and reduced systemic side effects.41,42 However, one of the major challenges in dermatological therapy has long been low skin permeability due to the sebum membrane and stratum corneum constituting a skin barrier to keep out foreign substances.43,44 SNA structures exhibit higher penetration capabilities than traditional linear nucleic acids without the need for disruption or transfection agents. For example, Paller's lipid-based SNAs can locally deliver to the epidermis and inhibit the expression of IL-17A to realize successful psoriasis treatment without any transfection agents.45 Zhang developed a bifunctional oligonucleotide SNA containing antisense oligonucleotides (ASOs) and tyrosinase inhibitor prodrugs, which compared with a single nucleic acid or vehicle enhanced skin permeation and reduced the melanin content in B16F10 melanoma cells (Fig. 2A).46 Song developed hyaluronic acid-based SNAs for the transdermal delivery of the chemotherapeutic drug doxorubicin (Dox) and metalloproteinase inhibitor 1, which overcame the challenge beyond skin permeability and promoted apoptosis in hypertrophic scar cells.47
The blood–brain barrier is a highly regulated barrier that restricts the passive entry of exogenous substances into the central nervous system,48,49 which has been a bottleneck for brain administration for a long time. SNAs can penetrate the brain and offer a potential new approach to addressing this challenge. For instance, Stegh engineered a gold nanoparticle core with a shell of radially oriented siRNA oligonucleotide nanocomplex capable of crossing the blood–brain barrier for silencing the tumor MGMT protein and achieving glioblastoma treatment.50 More importantly, higher active targeting can be achieved through the strategy of SNA modification. Mirkin synthesized protein SNAs modified with β-galactosidase-conjugated transferrin receptors (TfRs), significantly increasing the accumulation in brain tissue and successfully transporting proteins to the brain and central nervous system (Fig. 2B).51 Shi co-encoded the caspase-3 antisense oligonucleotide and a transferrin receptor aptamer into a circle template and developed SNA structures for the treatment of ischemic stroke, which demonstrated a 6.4-fold enhancement in blood–brain barrier penetration capability.52
In short, the efficient uptake and penetration of SNAs make them more extensively employed compared to traditional nucleic acid carriers. This unique property of SNAs enhances the prospects for clinical applications.
The stability of SNAs has been extensively investigated and applied, with recent studies achieving significant breakthroughs. Finn's research team developed a novel class of SNA structures by grafting oligonucleotides onto virus-like particles through copper-catalyzed click chemistry.57 The virus-SNAs possessed robust cellular uptake capabilities and resistance to nucleic acid enzymatic degradation, with their stability being approximately tenfold that of unmodified virus-like particles. Mirkin quantified the enhanced stability of SNAs in enzyme-catalyzed DNA hydrolysis and provided evidence that the negatively charged surface of nanoparticles and the resulting high local salt concentration are the underlying reasons for stability enhancement.58 Additionally, Kanaras's research team conducted a comprehensive analysis of nucleic acid stability in spherical nucleic structures (Fig. 2D).59 They correlated enzyme-mediated DNA degradation with the oligonucleotide density on nanoparticles, revealing that unmodified SNAs are prone to DNA enzyme degradation at low oligonucleotide density levels. In contrast, phosphorothioate-modified oligonucleotides, such as PS-SNAs, demonstrate enhanced resistance to enzyme degradation.
These findings underscore the critical effect of oligonucleotide density on the stability of SNAs, with SNAs retaining their integrity even in a complex environment, offering an indispensable and important index in future clinical translation.
![]() | ||
Fig. 3 Some ingenious examples of SNA with different functional cores in practical applications. (A) A carefully designed SNA based on the programmability and multifunctionality of DNA and gold nanoclusters, which achieved spatiotemporal control of drug delivery. Reproduced from ref. 61. Copyright 2021 American Chemical Society. (B) The introduction of PS- and PO-Ag-SNAs represented a novel antibiotic design strategy suitable for Gram-positive and Gram-negative bacteria. Reproduced from ref. 64. Copyright 2023 Elsevier B.V. (C) A SNA constructed from BP nanosheets and porphyrin zinc metal–organic framework nanosheets to achieve photothermal photodynamic therapeutic effects on tumors. Reproduced from ref. 66. Copyright 1999–2023 John Wiley & Sons, Inc. (D) A functionalized L-SNA was constructed from an FDA-approved 1,2 dioleyl-SN-glycerol-3-phospholipid choline lipid monomer, which promoted broader tissue distribution and longer in vivo circulation times through the characteristics of liposomes. Reproduced from ref. 71. Copyright 2020 American Chemical Society. (E) PLGA nanoparticles employed to build SNA structures to achieve controllable drug release, which not only enabled independent modulation of drug release kinetics, but also preserved nucleic acid stability on nanoparticle surfaces. Reproduced from ref. 78. Copyright 1999–2023 John Wiley & Sons, Inc. (F) A carrier-free anticancer SNA vaccine developed by utilizing a CpG-rich oligonucleotide and VE as the core, with the VE component serving as the hydrophobic driving force for SNA formation. Reproduced from ref. 82. Copyright 2022 American Chemical Society. |
In addition to gold nanoparticles, silver nanoparticles are also valuable choices as an inorganic nanomaterial for SNA construction. Silver nanoparticles can disrupt bacterial membranes and subcellular structures through various mechanisms, introducing antimicrobial properties into SNA structures.62,63 The introduction of PS- and PO-Ag-SNAs by Gryaznov represented a novel antibiotic design strategy suitable for Gram-positive and Gram-negative bacteria and demonstrates the unique advantages of SNA structures in enhancing therapeutic efficacy and stability (Fig. 3B).64
Various inorganic nanoparticles have been widely employed in the preparation of SNAs. The utilization of diverse elements such as Fe, P, Pt, Al, Pd, Cu, Co, In, Ni, and their mixtures enables the preparation of a wider range of SNA types, conferring them with increasingly specialized functions. Iron oxide nanocrystals (Fe3O4 nanoparticles) exhibit magnetism and are extensively used in magnetic structures. In Liu's work, the synthesis of Fe3O4@PDA core–shell NPs offered a novel approach for DNA extraction by using magnetic separation and hybridizing complementary nucleic acids, while demonstrating its potential applications in bioanalytical chemistry.65 What is more, they extended the choice of the core to silicon dioxide and tungsten disulfide materials. Black phosphorus (BP) nanosheets can serve as initiators for photothermal and photodynamic therapies. Wang developed a SNA constructed using BP nanosheets and porphyrin zinc metal–organic framework nanosheets to achieve photothermal photodynamic therapeutic effects on tumors (Fig. 3C).66 SNAs prepared with these inorganic nanomaterials not only possess high biocompatibility but also demonstrate feasibility in biofunctionalization.
Liposomal nanoparticles exhibit excellent biocompatibility.69 For example, Mirkin constructed functionalized liposomal SNAs (L-SNAs) from an FDA-approved 1,2-dioleyl-SN-glycerol-3-phospholipid choline lipid monomer.70 This structure not only preserved the robust internalization capability and stability of metal SNAs, but also promoted broader tissue distribution and longer in vivo circulation times through the characteristics of liposomes, offering novel avenues for therapy (Fig. 3D).71 Furthermore, the structural advantages of L-SNAs were reflected in their delivery efficiency, as evidenced by a significant increase in their distribution across various tissues and circulation within 30 minutes and 24 hours post-injection, highlighting their superior delivery performance compared to linear DNA. Additionally, Mirkin's research successfully reduced the toxicity of siRNA by constructing L-SNAs while retaining their gene-regulatory capacity.72 Compared to gold-core siRNA-SNAs, these novel siRNA-SNAs, resembling hairpins, exhibited enhanced biocompatibility and greater efficacy in cellular uptake and gene silencing due to the L-SNA structures. This offers a new design and synthesis pathway for therapies based on L-SNA structures, expanding their potential applications in the treatment of various diseases, including cancer.
In biology, proteins are the primary executors of cellular functions, and thus regulating proteins can directly control intracellular physiological activities.73 The introduction of protein cores can expand the functionality of SNAs while SNAs enhance protein stability through their outer shell. Mirkin attached oligonucleotides to the surface of a large homotetrameric enzyme (β-galactosidase), establishing a new pathway for oligonucleotides to be internalized by cells and to perform protein functions.74 Another work by Mirkin involved designing SNAs with Cas9 as the core, enabling direct execution of Cas9 functions.75 This design not only demonstrated the direct application of SNAs in gene therapy, but also expanded the potential of SNAs in cellular function manipulation through efficient cellular internalization, endosomal escape, and the nuclear delivery of Cas9 protein SNA. This lays the foundation for the development of protein SNAs as a novel biologically active material, allowing the adjustment of DNA shells and protein cores to amplify functionality.
Using polymer macromolecules as cores to construct SNAs offers a broader space for modification and enhanced loading capacity for controlled drug release.76,77 Mirkin employed PLGA nanoparticles to build SNA structures to achieve controllable drug release (Fig. 3E).78 This strategy not only enabled independent modulation of drug release kinetics, but also preserved nucleic acid stability on nanoparticle surfaces, expanding the application scope of SNAs in therapeutic strategies. Zhang constructed a polymer SNA structure with the SNA shell containing antisense oligonucleotides targeting MC1R and prodrug inhibitors of tyrosinase, which successfully reduced melanin production in melanoma cells.46 Zheng developed a polymer SNA with antisense oligonucleotides and an amphiphilic self-assembling polymer as the core,79 which efficiently loaded nanoprobes and Dox. After treatment, the quantitative average fluorescence intensity was calculated to be 1.51-fold higher than that of nontumoral sites. Meanwhile, the tumor growth rate in the SNA-treated group was 0.15-fold that of the PBS-treated group.
On the other hand, carrier-free nanomedicines have been extensively studied due to their high biocompatibility, high drug loading, simple drug components, and simplified synthesis procedures.80,81 Constructing SNAs with the drug itself as the carrier or using nucleic acids as the main component are current directions in carrier-free nanomedicine research. Zhang demonstrated the potential of a carrier-free anticancer vaccine using a CpG-rich oligonucleotide and vitamin E (VE) as the core. Not only does the VE component serve as the hydrophobic driving force for SNA formation, but also the released VE can also work synergically with CpG DNA to amplify immune responses (Fig. 3F).82 Shi's research demonstrated that the circular template enables the co-encoding of caspase-3 ASO and the TfR aptamer, resulting in the formation of a spherical nucleic acid nanostructure through rolling circle replication. This carrier-free architecture not only simplified the construction, but also mitigated the risk of uncontrolled immune system activation due to the reduced metabolism of natural nucleic acid components.52
In summary, the structure of SNAs with either an inorganic nanomaterial core, an organic nanomaterial core, or a carrier-free nanomedicine core reflects a suitable and valuable application requirement, which broadens the prospect of spherical nucleic acids for various therapeutic applications and breakthroughs in gene therapy to clinical translation.
![]() | ||
Fig. 4 Some representative cases of the application of spherical nucleic acid multimodal therapy. (A) A pH-responsive Dox–SNA conjugate facilitated the delivery of Dox to cancer cells and pH-dependent drug release. Reproduced from ref. 86. Copyright 2023 Royal Society of Chemistry. (B) A SNA conjugate combined with two PTX molecules and an antisense oligonucleotide integrated with fluorouracil, which successfully improved the stability of PTX and effectively achieved a synergistic anti-tumor effect. Reproduced from ref. 91. Copyright 1999–2023 John Wiley & Sons, Inc. (C) pSNAs activated local in situ photothermal effects and reduced side effects of non-specific damage by controllable targeted release capability. Reproduced from ref. 98. Copyright 2023 Royal Society of Chemistry. (D) PSNAs generated reactive oxygen species under near-infrared light exposure, leading to the release of siRNA and pASO and achieving combined photodynamic and gene therapy effects. Reproduced from ref. 101. Copyright 2021 American Chemical Society. (E) A SNA constructed from two CpG immunostimulatory oligonucleotides, termed double CpG-SNA, which enhanced dendritic cell maturation and facilitated a localized immune response against distant tumor cells. Reproduced from ref. 115. Copyright 2020 American Chemical Society. (F) A hydrophobic dodecyl anchored group of liposome SNAs systematically studied, which was found to achieve a strong T cell response after encapsulating its OVA antigen peptide. Reproduced from ref. 119. Copyright 2023 American Chemical Society. |
The poor water solubility and low bioavailability limit the clinical application of paclitaxel (PTX).88 By binding to the SNA shell, the therapeutic efficacy of PTX is enhanced due to its high stability and cellular uptake. Zhang's amphiphilic DNA–PTX coupling exhibited higher stability against nuclease and faster cellular uptake (100 times) than free DNA, resulting in nearly identical cytotoxicity as a free drug.89 Moreover, Zhang used benzyl bromide-modified PTX grafted on the DNA skeleton at the PS modification site and loaded into SNA-like micelle nanoparticles with PTX, which realized a high PTX loading rate (≈53%).90 Subsequently, Zhang used a fluorescent disulfide imine as a linker and combined two PTX molecules with an antisense oligonucleotide integrated with a fluorouracil. This conjugate successfully improved the stability of PTX and effectively inhibited the p-glycoprotein expression, leading to the release of FdU and PTX, resulting in a synergistic anti-tumor effect (Fig. 4B).91
Moreover, numerous drugs including coumarin,78 rapamycin,92 camptothecin,93 BKM120 (a lymphocytic leukemia anticancer drug),94 benzyl 3,4-dihydroxyphenylethylamine (a tyrosine kinase inhibitor),46 and AS1411 (an aptamer targeting nucleolin)95 have demonstrated enhanced uptake and stability when delivered via SNAs. This targeted drug delivery at the site of the lesion has allowed for the maximization of their therapeutic effects. These investigations highlight the diverse applications of SNAs in combined drug therapy, offering various potential advantages and amplifying the therapeutic efficacy of drugs, thereby accelerating their widespread adoption in the field of medicine.
Photothermal therapy is considered one of the most promising cancer treatment methods in recent years, relying on the conversion of light into heat for therapeutic effects.96,97 The structure of SNAs offers enhanced functionality and heightened targeting capabilities for photothermal therapy. For instance, Wang engineered a “sandwich” SNA structure composed of BP nanosheets and zinc porphyrin metal–organic framework nanosheets, which can finely tune the phase-transition temperature by altering the strand length and the proportion of the nucleic acid and base, achieving photothermal-triggered photodynamic therapy under near-infrared laser irradiation.66 Zhao developed programmable self-assembling pSNAs that activated local in situ photothermal effects upon miRNA-21 expression in the MCF-7 tumor and reduced the side effects of non-specific damage by controllable targeted release capability (Fig. 4C).98 Li developed AuNS-ASO to achieve the synergistic ablation of tumor cells through gene therapy and photothermal effects under near-infrared laser irradiation. Remarkably, the AuNS-ASO enables precise in situ delineation of tumor margins with exceptional spatial resolution (<100 μm) in mice tumors, thereby offering intraoperative guidance for optimal tumor resection.99
Photodynamic therapy involves localized irradiation using light of a specific wavelength on applied photosensitizers, generating cytotoxic reactive oxygen species to achieve therapeutic objectives.100 For example, Zhang prepared PSNAs that generated reactive oxygen species under near-infrared light exposure, leading to the release of siRNA and pASO and achieving combined photodynamic and gene therapy effects (Fig. 4D).101 Jiang developed a PSNA nanoplatform for targeted drug release and high-performance cancer treatment, selectively interacting with endogenous ATP to release Dox VE and Ce6 for chemotherapy and photodynamic therapy in synergy.95
Radiotherapy is a crucial method for cancer treatment. However, the challenges of normal tissue intolerance and suboptimal treatment effectiveness have driven significant interest in combination therapies.102,103 Zhou reported a drug delivery strategy that combines radiotherapy with SNA nano-drugs.104 The SNA structure played a dual role in enhancing radiotherapy and acting as an immunomodulator. The combination of radiotherapy with αPD-L1 exhibited potent anti-tumor effects, completely suppressing tumor growth. Bai developed SNAs that were conjugated with PD-L1 aptamers and indocyanine green (ICG) embedded in mesoporous hafnium oxide nanoparticle cores (Hf@ICG-Apt).105 Upon irradiation at the tumor site, the nano-system formed a high tumor-to-background ratio (7.97 ± 0.76 fold) and effectively enhanced radiotherapy to combat cancer.
Toll-like receptors (TLRs) in endolysosomes play a pivotal role in triggering innate immune responses and promoting anti-tumor immunity.108 Reported nucleic acid immune-sensing receptors include TLR3,109 TLR7/8,110 and TLR9,111 which can perceive various nucleic acids from viruses or bacteria, facilitating immune stimulation within the organism. Zhang developed a spherical nucleic acid constructed from CpG polycaprolactones and anti-STAT3 siRNA for synergistic tumor immunotherapy.112 Mirkin engineered thermoresponsive micelle-based SNAs containing stimulatory TLR-9 CpG sequences, exhibiting superior stability and rapid intracellular uptake, thereby serving as effective TLR-9 immunomodulators.113 Mirkin subsequently further developed SNAs comprising RNA with TLR-7/8 selectivity. By anchoring cholesterol-terminated oligonucleotides to the lipid core of LSNA, he significantly enhanced cellular uptake and achieved a higher specificity of TLR receptor activation.114 Moreover, Mirkin developed an SNA constructed from two CpG immunostimulatory oligonucleotides, termed double CpG-SNA, which enhanced dendritic cell maturation (Fig. 4E).115 This advancement is significant due to the ability of a small quantity of this nanomedicine to induce immune system activation. It mitigates off-target toxicity associated with high doses of conventional nano-chemotherapy drugs and facilitates a localized immune response against distant tumor cells, thereby demonstrating the promising potential of SNAs for immune activation.
In addition, several structural designs have co-functionalized antigen peptides with oligonucleotide adjuvants, resulting in the creation of SNAs that activate APC and tumor-targeted T cells. Zhang developed an SNA-based anti-cancer vaccine, which is comprised of phosphodiesterase oligonucleotides and VE. This formulation was designed to deliver OVA antigen, thereby enhancing TLR9 activation and effectively stimulating adaptive immunity.82 Liu designed a liposome SNA containing Dox and CpG response peptide coupling, which can enhance the activation of dendritic cells and promote the amplification of CD8+ and CD4+ T cells.116 Subsequently, Liu constructed a spherical nucleic acid using CpG-ODN and monophosphoryl lipid A double adjuvants, and Dox was radially surrounded as the shell, which can enhance ICD-induced immune responses, achieving synergistic therapeutic effects of chemotherapy and immunotherapy.87 Mirkin et al. enhanced T cell responses by hybridizing antigen peptides onto SNAs.117 They investigated the immunomodulatory activity of liposome spherical nucleic acids by exchanging liposome components and systematically studied the hydrophobic dodecyl anchored group of liposome SNAs, which was found to achieve a strong T-cell response after encapsulating its OVA antigen peptide (Fig. 4F).118,119 In addition, they also developed STING-activated spherical nucleic acids, which can effectively activate the cGAS-STING pathway to generate innate immune responses.120 The development of SNAs that can activate the innate or adaptive immune system and elicit a potent anti-cancer immune response has demonstrated remarkable efficacy.
Innate immunity is one of the weapons in the body's immune defense system, but excessive innate immune responses can lead to autoimmune inflammation.121,122 In such cases, immune modulation is necessary to reduce excessive autoimmune responses. Psoriasis is a T-cell-mediated skin disease with autoimmune characteristics. Tavoosidana designed a hybrid peptide SNA nanoparticle capable of preventing the excessive gene expression and functional activity of T cells, thereby improving psoriatic skin damage.123 Paller locally administered L-SNAs targeting the IL-17 receptor gene, successfully reversing the progression of psoriasis.45 Cui prepared SNAs by regulating the IL-1β mRNA expression in arthritis. They achieved this by downregulating catabolic enzymes in cartilage and upregulating the synthetic part, protecting chondrocytes from degenerative changes and halting the ongoing progression of arthritis.60
In conclusion, these studies demonstrate significant breakthroughs in the realm of integrated cancer therapy and diagnosis using SNAs. They not only provide novel directions for cancer treatment but also lay the foundation for the translation of SNAs into practical tools in clinical practice.
With in-depth research and successful preliminary preclinical experiments on SNAs, biotechnology companies are driving the clinical translation of SNAs. The unique capabilities of SNAs, particularly their ability to penetrate various tissues and cell types, make them potent tools for treating a wide range of diseases. Furthermore, in current clinical trials, certain SNAs have demonstrated promising performance.
The first phase 0 clinical trial (NCT03020017) involving SNA carrying Bcl2L12 siRNA (drug code: NU-0129) was conducted in recurrent glioblastoma.127,128 The trial results demonstrated that the intravenous administration of SNAs at the administered dose, which corresponds to the 1/50th of the not observed adverse-event level, exhibited a favorable safety profile and did not exhibit any treatment-related toxicity.
Using ICP-MS and X-ray fluorescence microscopy, it was observed that SNAs accumulated in the tumors of patients and were associated with cytoplasmic accumulation in tumor cells. Quantitative analysis of the gold element concentration indicated SNA uptake in tumor cells, resulting in reduced Bcl2L12 protein expression and the induction of active caspase-3 and p53 proteins. These results demonstrate that using SNAs is a safe and brain-penetrating precision medical approach, enabling the delivery of siRNA oligonucleotide systems to intracranial tumor sites.
Furthermore, a clinical trial investigating the combined use of SNAs with immunotherapy is underway, focusing on immune checkpoint therapy. The study aims to evaluate the effectiveness of an SNA containing a TLR-9 agonist in immune checkpoint therapy. In the phase I human study, an immune-stimulating SNA compound targeting toll-like receptor 9, known as cavrotolimod (formerly AST-008), demonstrated good tolerance and pharmacological characteristics in healthy participants.129,130 The results indicate that cavrotolimod is an effective innate immune activator, which may exert anti-tumor effects in cancer patients. Based on these findings, cavrotolimod has advanced to phase II clinical trials in combination with PD-1/PD-L1 antibodies for late-stage skin cancer patients.
In studies on psoriasis, a phase I clinical trial involving the topical administration of SNAs carrying siRNA-targeting TNF-α in patients with chronic plaque psoriasis yielded high tolerance and safety. Furthermore, a treatment plan for psoriasis-targeting interleukin-17 receptor alpha is poised to commence phase I clinical trials, marking the clinical therapeutic approach of local SNA administration.131
Exicure Inc., a clinical-stage biotechnology company founded by David Giljohann and Chad Mirkin, is expanding the clinical application of SNAs to treat a variety of diseases, including Angelman syndrome, Huntington's disease, Batten disease, alopecia, motor neuron disease, and spinocerebellar ataxia (Table 1).131 Overall, the translation of SNAs is thriving, and the existing observations will strongly propel the further development of SNA clinical applications, paving the way for future medical advancements.
Disease mode | Drug/target | Phase |
---|---|---|
Melanoma | AST-008 (TLR9) | Clinical phase II |
Head and neck squamous cell carcinoma | AST-008 (TLR9) | Clinical phase II |
Merkel cell carcinoma | AST-008 (TLR9) | Clinical phase II |
Solid tumors | AST-008 (TLR9) | Clinical phase I |
Neoplastic hematologic disorder | AST-008 (TLR9) | Preclinical |
Neuropathic pain | SCN9A Candidate (Nav1.7) | Unknown |
Psoriasis | XCUR-17 (IL-17RA) | Clinical phase I |
Alopecia | XCUR-17 (IL-17RA) | Preclinical |
Angelman syndrome | Oligonucleotide | Preclinical |
Huntington's disease | Oligonucleotide | Preclinical |
Batten disease | Oligonucleotide (CLN3) | Preclinical |
Friedreich's ataxia | XCUR FXN (ATXN) | Preclinical |
Psoriasis | AST-005 (TNF) | Clinical phase I |
Diabetic foot | AST-006 | Drug discovery |
Dermatosis | DMX-102 (SPINK5) | Unknown |
However, there are still challenges to overcome. Firstly, it is crucial to address the impact of the protein corona during circulation and prevent lysosome clearance after cellular uptake. The protein corona refers to the dynamic protein layer that forms around nanoparticles or nucleic acids upon exposure to biological fluids. This corona can significantly alter the interaction of nucleic acids with target cells and tissues, thereby impacting their therapeutic efficacy. It has been reported that the defects of the protein corona can be utilized to actively adsorb functional proteins on the surface of SNAs, leading to less protein loss and stronger anti-degradation ability. Mirkin designed and synthesized SNAs with a predefined protein coronae consisting of functional proteins (monoclonal antibodies targeting human epithelial growth factor receptor 2) immobilized on the oligonucleotide shell. These structures exhibited enhanced stability in buffer and human serum, as well as selectivity for HER2-positive breast cancer cells in mixed cell cultures with HER2-negative breast cancer cells. This strategy of using the protein corona to regulate the in vivo fate of SNAs will improve the design methods of such carriers and enhance the pharmacokinetic characteristics of SNAs.132 This strategy of using the protein corona to regulate the in vivo fate of SNAs will improve the design methods of such carriers and enhance the pharmacokinetic characteristics of SNAs. Lysosomes are cellular organelles that break down and recycle various molecules and particles, including foreign substances like SNAs. Regarding the issue of lysosome degradation, utilizing certain substances to disrupt the lysosomal membrane, thereby causing the release of SNAs into the cytoplasm to exert their effects, is a common approach. It has been reported that photo-induced production of 1O2 can enable lysosome escape.133 Alternatively, the introduction of lanthanide particles binding to the phospholipid head of the endosome membrane has been demonstrated to disrupt the membrane structure and facilitate rapid escape from the lysosome.134 However, the bodily reactions caused by excessive lysosomal disruption still need to be carefully considered.
Secondly, it is necessary to develop optimized synthetic processes that can yield large quantities of SNAs at a reasonable cost for successful commercialization. The first on the list is the synthesis of SNAs without compromising their quality or functionality. This necessitates a fine balance between the use of efficient, cost-effective materials and methods, and the preservation of the structural integrity and bioactivity of SNAs. Advanced techniques in nanotechnology and molecular engineering are key to developing such optimized processes. Notably, recent advancements such as the freeze-anchoring method have been reported as a simple and time-saving alternative to traditional SNA synthesis modes. By reducing the temperature, DNA can be conjugated to AuNPs during freezing without the need for additional reagents, enabling the completion of the conjugation process within a few minutes. Moreover, this method yields a DNA density that is 20–30% higher compared to that achieved using the typical salt-aging approach.135,136 By achieving this objective, SNAs can transition from a promising scientific concept to a widely available commercial product, unlocking new possibilities in science and medicine.
Finally, the in vivo safety of SNAs still requires long-term attention. The core of SNAs is generally composed of metals such as gold, iron, and silver, which can accumulate in the body and lead to poisoning. Additionally, the off-target effects and immunogenicity of nucleic acids themselves are issues that need to be considered when using SNAs. Carrier-free self-delivery strategies have been shown to reduce the defects and maximize the function of the drug due to simplified construction and mitigate the risk of uncontrolled immune system activation. This strategy may become the most ideal way for clinical treatment and an important development direction for SNAs in the future.52,82
In conclusion, the amplification contribution of SNAs for potential clinical translations should not be overlooked. Through an examination of the distinctive amplification structure inherent in SNAs, the distinctive features outlined below undoubtedly strengthen the significant clinical translational potential of SNAs: (i) spherical structure: the spherical structure is chosen due to their minimal potential energy and stability against external forces; furthermore, spheroids, acting as isotropic particles, increase the probability of effective particle collisions, (ii) drug loading form: external binding of high-density nucleic acids for drug loading offers unique opportunities for nucleic acid pairing and amplified SNA binding; moreover, a higher nucleic acid density enhances nanoparticles’ capacity to load various cargoes with greater loading contents, and (iii) flexible and tailor-made compositions: the shell and core components of SNAs can be tailor-made to meet a broad range of different requirements. This adaptability allows for different solutions to be implemented based on distinct disease models.
All in all, the amplification of SNAs is an ongoing process, with the continuous advancement of technology and a deeper understanding of SNAs, more innovations and breakthroughs are expected in the future. This holds the potential to make the treatment of various human diseases a reality in the future.
Footnote |
† These authors contributed equally to this paper. |
This journal is © The Royal Society of Chemistry 2024 |