Skip to main content
MedComm logoLink to MedComm
. 2023 Aug 7;4(4):e339. doi: 10.1002/mco2.339

Lipid‐based nanoparticles as drug delivery systems for cancer immunotherapy

Yang Hao 1,2,3, Zhonghao Ji 1,2, Hengzong Zhou 1, Dongrun Wu 4, Zili Gu 5, Dongxu Wang 1,, Peter ten Dijke 3,
PMCID: PMC10407046  PMID: 37560754

Abstract

Immune checkpoint inhibitors (ICIs) have shown remarkable success in cancer treatment. However, in cancer patients without sufficient antitumor immunity, numerous data indicate that blocking the negative signals elicited by immune checkpoints is ineffective. Drugs that stimulate immune activation‐related pathways are emerging as another route for improving immunotherapy. In addition, the development of nanotechnology presents a promising platform for tissue and cell type‐specific delivery and improved uptake of immunomodulatory agents, ultimately leading to enhanced cancer immunotherapy and reduced side effects. In this review, we summarize and discuss the latest developments in nanoparticles (NPs) for cancer immuno‐oncology therapy with a focus on lipid‐based NPs (lipid‐NPs), including the characteristics and advantages of various types. Using the agonists targeting stimulation of the interferon genes (STING) transmembrane protein as an exemplar, we review the potential of various lipid‐NPs to augment STING agonist therapy. Furthermore, we present recent findings and underlying mechanisms on how STING pathway activation fosters antitumor immunity and regulates the tumor microenvironment and provide a summary of the distinct STING agonists in preclinical studies and clinical trials. Ultimately, we conduct a critical assessment of the obstacles and future directions in the utilization of lipid‐NPs to enhance cancer immunotherapy.

Keywords: lipid‐based nanoparticles, cancer immunotherapy, stimulator of interferon genes, type I interferon, tumor microenvironment


Cancer is a prevalent and lethal ailment that has a considerable social and economic impact globally. Despite notable progress in recent years, immunotherapy poses a challenge due to the limited response of a subset of cancer patients in the clinic. Lipid‐based nanoparticles (lipid‐NPs) have garnered significant interest for their capacity to enhance the effectiveness of cancer immunotherapy while minimizing toxic side effects. This review provides a summary of the physicochemical properties, applications, advantages, and disadvantages of various lipid‐NP types. Moreover, we provide an overview on how lipid‐NPs may broaden the therapeutic scope of cancer immunotherapy, using as example how lipid‐based NPs delivery of agonists targeting stimulators of interferon genes augments immunotherapy.

graphic file with name MCO2-4-e339-g004.jpg

1. INTRODUCTION

Cancer represents a significant public health concern and remains the primary cause of mortality worldwide. Contemporary therapeutic modalities, encompassing conventional interventions such as surgery, chemotherapy, and radiotherapy, as well as novel targeted and photodynamic therapy (PDT), have yielded notable progress in the management of cancer. Notably, the emergence of immunotherapy that attempts to enhance or restore the host's immune system to destroy tumor cells has engendered renewed optimism for cancer patients. 1 Despite the great progress made in recent years, the clinical success of immunotherapeutic drugs has been impeded by various challenges, including primary resistance in that majority of cancers patients do not respond to immunotherapy, immune‐related severe side effects, adaptive resistance resulting from immune escape mechanisms, and acquired resistance leading to cancer recurrence and metastasis. 2 In light of these limitations, the integration of nanoparticles (NPs) in immunotherapy has remarkable potential, especially in the following aspects: antigen delivery and tumor antigen specificity, delivery of immunomodulators, modulation of the tumor microenvironment (TME), and combination therapy strategies. 3

NPs, typically ranging from 100 to 1000 nm in diameter, are composed of various materials with distinct surface modifications and physicochemical properties. Figure 1 presents a schematic representation of the commonly utilized NP types, which can be categorized into inorganic NPs, comprising metals such as gold and silver or silicon colloids, and organic NPs, consisting of lipids, sugars, and biodegradable polymers. 4 While poly (lactic‐co‐glycolic acid) (PLGA) NPs, hydrogels, micelles, and metal NPs have been investigated as potential carriers for anti‐cancer drugs in preclinical studies, only a restricted number of nanomedicines utilizing these formulations have been granted approval by the United States Food and Drug Administration (US FDA) for cancer treatment. 5 An instance of this is Abraxane®, a polymeric NP with an albumin base (with an approximate size of 130 nm) that is commercially available for the administration of chemotherapeutic paclitaxel. It was sanctioned in 2005 for the management of various oncological indications, including breast cancer, non‐small cell lung cancer (NSCLC), pancreatic cancer, and gastric cancer. 6 Furthermore, polymeric micelle‐based NP formulations have been developed as a delivery system for paclitaxel or Docetaxel (Genexol®, Nanoxel®, and Paclical®) to treat advanced cancer types. 7 , 8 , 9 , 10 Herein, we mainly focused on liposomal formulations as delivery platforms in cancer immunotherapy. Lipid‐based nanomaterials (referred to lipid‐NPs), are the most commonly utilized class in clinical settings approved by the US FDA, owing to their unique properties such as formulation simplicity, excellent biocompatibility and safety, and drug loading capacity. 11 Notably, liposomal drugs, such as Doxil®, Myocet®, DaunoXome®, Onivyde®, Vyxeos®, Marqibo®, DepoCyt®, among others, have achieved significant success since their introduction to the market because of effective payload loading efficiency, low toxicity, less immunogenicity, as well as mass producibility and better cost effectiveness. 12 Additionally, lipid‐NPs have demonstrated to serve as delivery systems for immune‐related drugs, which can be loaded with tumor‐associated antigens (TAAs), low‐dose chemotherapeutic agents, photosensitizers/photothermal agents, or other immune adjuvants or antibodies. Lipid‐NPs show the potential to enhance immunotherapeutic drug efficacy, mitigate toxicity, and overcome tumor‐driven immune resistance. 13

FIGURE 1.

FIGURE 1

An overview of commonly used nanoparticles includes polymeric nanoparticles, inorganic nanoparticles and lipid‐based nanoparticles. Figure created with biorender.com.

The primary objective of immunotherapy is to initiate immune response cascades through the innate immune system, which involves the identification of pattern recognition receptors (PRRs), including pathogen‐associated molecular patterns (PAMPs) such as microbial nucleic acids and lipopolysaccharide (LPS), as well as damage‐associated molecular patterns (DAMPs) such as host DNA. 14 , 15 The stimulator of interferon genes (STING) protein, also referred to as TMEM173, MITA, or MPYS, is a recently discovered PRR that recognizes cyclic dinucleotides (CDNs) and activates antigen‐presenting cells (APCs). While initially identified as activated in response to viral infections, subsequent research has demonstrated that the STING pathway also responds to a variety of intra‐ and extracellular stimuli, including DNA damage, metabolic abnormalities, and abnormal cell death. Targeting the STING signaling pathway has emerged as a viable therapeutic strategy in tumor therapy. 16 Recent studies have demonstrated that the activation of the STING pathway by agonists can elicit downstream immune responses, such as the production of interferons (IFNs) and proinflammatory cytokines, and the activation of natural killer (NK) cells and T cells, ultimately leading to the mitigation of cancer progression. 17

Although preclinical studies have demonstrated the effective antitumor effects of multiple STING agonists, their potential therapeutic efficacy in clinical settings is limited by factors such as low cytoplasmic delivery rate, rapid immune clearance rate, noncellular targetability, and systemic inflammatory toxicity. To address these limitations, NPs have been proposed as delivery vehicles for STING agonists, as they can increase stability, prolong blood circulation time, enhance tumor targeting, and promote intracellular uptake thereby maximizing the antitumor immune effect and reducing adverse effects. 18 This approach maximizes the antitumor immune effect while simultaneously minimizing adverse effects. 19 As such, in this review, STING agonists were chosen as the prototypical immunotherapeutic agents to examine the application of NPs for their delivery, with the aim of enhancing the efficacy and safety of intracellular delivery.

This review aims to provide a comprehensive introduction of the diverse lipid‐NPs utilized in cancer immunotherapy, including their characteristics, and applications, advantages, and disadvantages. Furthermore, the review will explore the activation of STING pathway in promoting antitumor immunity and regulating the TME. Additionally, we will summarize the latest therapeutic interventions targeting the STING pathway in clinical trials. Finally, the potential and challenges of utilizing lipid‐based NPs to deliver STING agonists for cancer treatment will be critically evaluated and discussed.

2. LIPID‐BASED NPs AS DRUG DELIVERY SYSTEM IN CANCER IMMUNOTHERAPY

Lipid‐NPs, encompassing liposomes, lipid nanoparticles (LNPs), nanoemulsions (NEs), solid lipid nanoparticles (SLNs) and nanostructured lipid carriers (NLCs), extracellular vesicles (EVs), and lipid polymer hybrid nanoparticles (LPHNPs), and so on, exhibit significant potential for delivering therapies and other applications in theragnostic, cosmetics, and nutrition. 20 The advantages and disadvantages of various lipid‐NPs as drug delivery systems are listed in Table 1.

TABLE 1.

Summary of advantages and disadvantages of main lipid‐NPs.

Particle type Advantages Disadvantages
Liposomes
  • Payload flexibility for hydrophilic and hydrophobic cargo

  • Good biocompatible and biodegradable

  • Drug protection

  • Reduced systemic toxicity

  • Possible surface modification

  • Poor stability

  • Poor drug‐loading efficiency for hydrophobic cargo

  • Problematic immunocompatibility

  • Toxicity of organic solvent

  • Limited storage conditions

  • Limited targeting

  • Possible fast clearance

  • Drug leakage

  • Relative high production costs

Cationic lipid nanoparticles (LNPs)
  • Payload flexibility for hydrophilic and hydrophobic cargo and mRNA products

  • High drug loading efficiency

  • Good biocompatible and biodegradable

  • Drug protection

  • Reduced systemic toxicity

  • Possible surface modification

  • Controlled release

  • Poor stability

  • Toxicity of cationic lipids

Extracellular vesicles (EVs)
  • Payload flexibility for hydrophilic and hydrophobic cargo and mRNA products

  • Good biocompatible and biodegradable

  • Inherited biological and immunological characteristics

  • No toxicity

  • Immunological escape

  • Possible surface modification

  • Ability to cross various biological barrier

  • Poor stability

  • Poor drug‐loading efficiency

  • Heterogeneous physicochemical properties

  • Possible fast clearance

  • No standardized manufacturing methods

  • Difficult to scale‐up and control batch quality

Solid lipid nanoparticles (SLNs)
  • Payload flexibility for hydrophilic and hydrophobic cargo

  • Good drug entrapment efficiency for hydrophobic cargo

  • Good biocompatible and biodegradable

  • Drug protection

  • Low systemic toxicity

  • Possible surface modification

  • High stability

  • Easy sterilization and scale‐up process

  • Relative low production costs

  • Poor drug‐loading efficiency

  • Gelling tendency

  • Possible drug leakage during storage

  • Uneven drug release

  • Difficult to cross biological barriers

Nanostructured lipid carriers (NLCs)
  • All SLN's advantages

  • High drug‐loading efficiency

  • High stability and fewer drug lost during storage

  • Sustainable drug release

  • Complex optimization process of solid/liquid lipids

  • Gelling tendency

  • Difficult to cross biological barriers

Nanoemulsion (NE)
  • Payload flexibility for hydrophilic and hydrophobic cargo

  • Good drug entrapment efficiency for hydrophilic cargo

  • Good stability

  • Good biocompatible and biodegradable

  • Greater absorption by organisms

  • Easy preparation and easy drug loading

  • Relative low production costs

  • Poor drug‐loading efficiency

  • Toxicity of emulsifier

  • Relative fewer surface modification possibility

Lipid polymer hybrid nanoparticles (LPHNPs)
  • Payload flexibility for hydrophilic and hydrophobic cargo and mRNA products

  • Low systemic toxicity

  • Drug protection

  • High encapsulation efficiency (polymer core)

  • Precise control of nanoparticles (NPs) physicochemical properties (polymer shell)

  • Easy surface modification (polymer shell)

  • Good biocompatible and biodegradable (lipid shell)

  • High stability (polymer shell)

  • Poor drug‐loading efficiency (polymer core)

  • Toxicity of cationic lipids (core–shell‐type hollow LPHNPs)

  • Possibility for aggregation and toxicity (polymer shell)

  • Difficult to scale‐up and high production costs (Polymer shell)

  • Difficult to control batch quality (engineered LPHNPs)

Lipid‐NPs possess favorable safety, biocompatibility, degradability, and low toxicity due to their structural characteristics, which resemble those of the plasma membrane of human cells. 20 Additionally, lipid‐NPs can serve as a delivery platform to protect encapsulated cargo from degradation and aggregation during circulation, thereby improving the pharmacokinetic attributes and physicochemical stability of immune agents or combined intervention drugs. 5 Furthermore, lipid‐NPs are widely held to improve the delivery of cargos to tumors by two ways, namely passive and active targeting thereby enhancing intratumor aggregation and reducing ineffective systemic distribution of encapsulated cargos, ultimately resulting in a reduction of unexpected side effects. 21 Passive targeting is also known as the enhanced permeability and retention (EPR) effect, which allows lipid‐NPs to leak preferentially into tumors through permeable tumor vessels and to enable efficient retention within the TME due to reduced lymphatic drainage. 22 Active targeting facilitates the uptake of lipid‐NPs by specific cell types and other compositions in the TME, promoting the cellular uptake and efficiency of therapeutic agents. 23 This can be achieved through the decoration of the lipid‐NPs surfaces with various targeting moieties such as tumor‐specific ligands (e.g., folate, transferrin, granulocyte–macrophage colony‐stimulating factor [GM‐CSF], etc.), peptides (e.g., RGD [Arg‐Gly‐Asp tripeptide] targeting cellular adhesion molecules, NGR [Asn‐Gly‐Arg tripeptide] targeting aminopeptidase N [CD13], etc.), or monoclonal antibodies (mAbs) (e.g., antivascular endothelial growth factor [VEGF]R, anti‐ERB‐B2 receptor tyrosine kinase 2, anticarcinoembryonic antigen, antimucin 1, anti‐CD33, etc.) to enhance their targeting properties. In this way, intratumor aggregation is enhanced and ineffective systemic distribution of encapsulated cargo is reduced, ultimately resulting in the reduction of unexpected side effects. 21 Lipid‐NPs can also selectively target specific immune cells and other compositions in the TME, promoting the cellular uptake of therapeutic agents in APCs or T cells, thereby increasing their immunity‐boosting efficiency. Additionally, lipid‐NPs can achieve controlled drug release in both temporal and spatial dimensions through appropriate design such as magnetic field control, photosensitive control, PH or temperature responsiveness, and chemical substance control. 24 (Detailed descriptions will be given in the Section 2.2.) To date, lipid‐NPs have been extensively studied as carriers for a diverse range of immunotherapeutic agents (e.g., antigens, antibodies, cells, recombinant proteins, and small molecular chemotherapeutic drugs). 24 , 25 , 26 , 27 , 28 In particular, lipid‐based NPs play a crucial role in enhancing the safety and efficacy of immunotherapy (Figure 2).

FIGURE 2.

FIGURE 2

The utilization of lipid‐based nanomaterials in the delivery of immunotherapeutic agents, overcoming challenges, and improving therapeutic efficiency. PK/PD behaviours: pharmacokinetic‐pharmacodynamic behaviours. Figure was created with help of biorender.com.

2.1. General introductions of the early version lipid‐NPs: Liposomes

Liposomes are the archetype version of lipid‐NPs and are defined as sphere‐like vesicles that consist of a hydrophilic core of phospholipid (PL) bilayers with different sizes. 29 , 30 They have the capacity to enclose aqueous drugs within the aqueous core of the vesicle and trap lipophilic drugs in the hydrocarbon chain region of the bilayer, even for macromolecular drugs. 31 Typical liposomes are composed of PLs and contain sterol most often cholesterol to render an optimized structure and improved stability (Figure 3). 32 In addition, it has been observed that the use of cholesterol derivatives, bacterial sterols, phytosterols, and so on can also play an important role in maintaining the mobility, permeability, and stability of PLs. 33 The structure and physicochemical properties (e.g., size, numbers of concentric bilayers, and encapsulation ability) of liposomes depend strongly on their preparation route. The film hydration method and reverse phase evaporation represent the traditional liposome preparation technique (Table 2). 34 , 35 , 36 According to the particle size and lamellarity, liposomes can be classified into unilamellar vesicles (ULVs; one bilayer), oligolamellar vesicles (OLVs; 2–5 bilayers, 100–1000 nm), and multilamellar vesicles (MVLs; >bilayers, >500 nm). The ULV can be further divided into three subclasses based on the particle size, including small unilamellar vesicles (SUVs; diameters 10–100 nm), large unilamellar vesicles (LUVs; diameters >100 nm), and giant unilamellar vesicles (GUVs; diameters >1000 nm). 37 Both SUVs and MLVs can be prepared by controllably adjusting the operating parameters during the film hydration method, especially the hydration conditions, stirring speed, extrusion, and sonication procedure. 34 Moreover, SUVs structure has a long circulation time and the passive target ability to diseased areas, while MLVs structure has an excellent encapsulation efficiency and slow release of lipophilic compounds. 38 Therefore, most of current approved liposomal products are SUVs and MLVs. 39

FIGURE 3.

FIGURE 3

Lipid‐based nanomaterials for immunotherapeutic agents. (A) General schematic illustration of conventional liposomes as spherical vesicles based on a hydrophilic core of phospholipid bilayers and reinforced by cholesterol. (B) Schematic that demonstrates liposomal systems for the encapsulation of hydrophobic drugs into the bilayer membrane and the entrapment of hydrophilic drugs in the aqueous center. (C) Schematic representation of polyethylene glycol (PEG)‐coated liposomes for long circulation. (D) Schematic of multimolecule‐modified liposomes for specific targeting. (E) Schematic of multistimulus‐responsive liposomes for intelligent release. Figure was created with biorender.com.

TABLE 2.

Structural components present in liposomes and preparation method for liposomes.

Classification parameters Comments/methods Specifications Reference
Components for liposomes
Structural components Phospholipids Natural:
  • cardiolipin

  • PA: phosphatidic acid

  • PC: phosphatidylcholine

  • PE: phosphatidylethanolamine

  • PG: phosphatidylglycerol

  • PI: phosphatidylinositol

  • PS: phosphatidylserine

42 , 43 , 44
  • Synthetic:

  • DOPE: 1,2‐dioleoyl‐sn‐glycero‐3‐phosphoethanolamine

  • DLPC: 1,2‐dilauroyl‐sn‐glycero‐3‐phosphocholine

  • DLPE: 1,2‐Dilauroyl‐sn‐glycero‐3‐phosphoethanolamine

  • DLPG: 1,2‐Dilauroyl‐sn‐glycero‐3‐phosphoglycerol

  • DMPC: dimyristoyl phosphatidylcholine

  • DMPE: 1,2‐Dimyristoyl‐sn‐glycero‐3‐phosphoethanolamine

  • DOPC: 1,2‐dioleoyl‐sn‐glycero‐3‐phosphocholine

  • DPPC: dipalmitoyl phosphatidylcholine

  • DPPG: 1,2‐dipalmitoyl‐sn‐glycero‐3‐phosphoglycerol

  • DSPA: 1,2‐distearoyl‐sn‐glycero‐3‐phosphate

  • DSPC: distearoylphosphatidylcholine

  • DSPE: 1,2‐Dioleoyl‐sn‐glycero‐3‐phosphoethanolamine

  • DSPG: 1,2‐distearoyl‐sn‐glycero‐3‐phospho‐(1’‐rac‐glycerol)

  • DSPS: 1,2‐Distearoyl‐sn‐glycero‐3‐phospho‐l‐serine

  • HSPC: hydrogenated soy phosphatidylcholine

  • POPC: palmitoyl‐2‐oleoyl‐sn‐glycero‐3‐phosphocholine

  • SLPC: palmitoyl‐2‐stearoyl(5‐DOXYL)‐sn‐glycero‐3‐phosphocholine

45 , 46
Sterol
  • Cholesterol

  • Progesterone

  • Saponins (ginsenoside Rh2, Rg3; holothurin A, echinoside A, and bivittoside D from sea cucumber, etc.)

  • Phytosterols (β‐sitosterol, retinol, stigmasterol, etc.)

  • Bacterial sterols (ergosterol, lanolin, etc.)

  • Cholesterol derivatives (cholesterol hemisuccinate [CHEMS], lysine‐based cholesterol [CHLYS], cholesteryl arginine ethyl ester [CAE], etc.)

47 , 48 , 49 , 50 , 51 , 52 , 53 , 54 , 55 , 56 , 57 , 58 , 59 , 60 , 61
Additives Hydrophilic polymers
  • PEG

  • PEG derivatives, etc.

20 , 44 , 62 , 63
Ionic agents
  • Stearylamine (SA)

  • Diethyl phosphate (DCP), etc.

Surfactants
  • sodium dodecyl sulfate (SDS)

  • sodium octyl sulfate (SOS)

  • Tween 80, etc.

Preparation methods for liposomes
Passive loading Mechanical dispersion methods
  • The film hydration

  • Microemulsification

  • Ultrasonication method

  • French pressure cell

  • Membrane extrusion

  • Dried reconstituted vesicles

  • High‐pressure homogenization methods

33 , 64
Solvent dispersion method
  • Ethanol injection method

  • Ether injection method

  • De‐emulsification

    • ‐Double‐emulsification

    • ‐Reverse phase evaporation

    • ‐Stable plurilamellar vesicles

65 , 66 , 67
Detergent removal methods
  • Detergent dialysis

  • Column chromatography

  • Dilution

  • Reconstituted Sendai virus enveloped vesicles

68
Size transformation or fusion of vesicles
  • Dehydration–rehydration method

  • Freeze‐drying (lyophilization) method

69 , 70
Active loading Suitable for certain compounds that are both water‐soluble and lipid‐soluble and have ionizable groups. 71 , 72

The physicochemical properties and biological efficacy of liposomes can be influenced by various factors, including the type and size of liposomes, the ratio between different components, the surface charge of lipids, and modifications on the surface of particles. 40 The surface charge of liposomes is typically determined by the hydrophilic polar head group of PLs. PLs, both natural and synthetic, are frequently utilized in the production of lipid‐NPs. Among these, phosphatidylcholine (PC) and phosphatidylethanolamine (PE) derived from plants and animals are the most commonly employed (Table 2). 39 These PLs confer a negative (e.g., phosphatidic acid [PA], phosphatidylserine [PS], phosphatidylglycerol [PG], and cardiolipin), neutral (e.g., PC and PE) charges and positive (e.g. 3β‐[N‐(N′,N′‐dimethylaminoethane)‐carbamoyl]cholesterol (DC‐Chol), N‐[1‐(2,3‐dioleyloxy)propyl]‐N,N,N‐triethylammonium (DOTMA), 1,2‐dioleoyl‐3‐trimethylammoniopropane (DOTAP), 1,2‐dioleoyl‐ sn‐glycero‐3‐ethylphosphocholine (EDOPC), 2,3‐dioleyloxy‐N‐[2‐(sperminecarboxamido)ethyl]‐N,N‐dimethyl‐1‐propanaminium (DOSPA)) charge to liposomes at physiological pH. 32 , 41 Typically, liposomes or particles that are uncharged and possess low charge density exhibit favorable biocompatibility and stability. However, it is imperative to acknowledge the potential for side effects induced by their tendency to aggregate, which may lead to complications such as thrombosis. Section 2.3 provides a comprehensive discussion on cationic lipid (CLD)‐NPs.

2.2. Functionalized liposomes for cancer immunotherapy

As exogenous substances, liposomes may also interact with the immune system (e.g., progenitor cells, monocytes, and macrophages) and have been observed to accumulate to a greater extent in the liver and spleen, which are two major organs of the reticuloendothelial system (RES), compared with other organs. This accumulation has been linked to toxicity and depletion of immune cells, potentially leading to off‐target effects. 73 Numerous approaches have been performed to optimize the formulation of conventional liposomes to reduce the off‐target toxicity, short circulation time, and in vivo instability (as depicted in Figure 3). 74 Of all the approaches, the utilization of polyethylene glycol (PEG)ylated liposomes exhibits considerable potential to improve safety, allow for more efficacious immunotherapy, and is compatible with combination therapies. It has been found that PEGylated liposomes can significantly mitigate the effects of nonmodified liposomes on macrophages and liver function by reducing reticuloendothelial phagocytosis. 75 , 76 PEGylated liposomes that are covalently linked to immunostimulatory agents, specifically anti‐CD137 and interleukin (IL)‐2‐Fc, enables the retention of the robust antitumor effects of immune agonists or antibodies, while simultaneously mitigating the potential for heightened immune‐related adverse events. 77 , 78 This is because PEGylated liposomes create spatial barriers through the formation of “conformational clouds” and hydrated membranes on the liposome surface, thereby augmenting particle stability in circulation and improving EPR effect. This mechanism also prevents the clearance of liposomes by RES and facilitates targeting of the TME. 79

Furthermore, as previously described liposomes can be modified with various targeting moieties to enhance their targeting properties. 32 As an illustration, the internalization of liposomes coated with immunoglobulin M (IgM) by phagocytes was observed to be 100 times greater than that of uncoated liposomes or liposomes coated with unmodified IgM. 80 Additional research group has documented that liposomes modified with a mesothelioma‐targeting human single chain antibody (M1) and radiolabeled with radioactive isotope indium‐111 exhibited a prolonged half‐life in plasma and a significant accumulation in the tumor region 48 h after intravenous administration, in contrast to the minimal uptake observed in tumors treated with untargeted liposomes. Notably, this liposome formulation also demonstrated the ability to contribute to early diagnosis through single photon emission computed tomography (SPECT/CT) imaging. 81 Guo et al. 82 successfully developed liposomes that carry anti‐CD44 antibodies and anti‐IL6R antibodies that specifically target the TME of CD44+ breast cancer cells, thereby inhibiting metastasis in breast cancer mouse models.

Stimulus‐responsive liposomes hold great potential for optimizing the effectiveness of conventional liposomal drug delivery. Through the manipulation of lipid ratios with specific stimulus‐response mechanisms such as changes in temperature or pH and in response to exposure to specific enzymes (e.g., matrix metalloproteinases [MMPs], prostate‐specific antigenases, cathepsin, elastases, hyaluronidase, and Urokinase‐type plasminogen activator, etc.). Therefore, liposomes are enabled to provide heightened selectivity and controlled release of drugs during treatment, leading to decreased systemic toxicity, improved physicochemical properties, and enhanced antitumor efficacy. 83 , 84 A pH‐sensitive liposomal vaccine was synthesized successfully by Yuba et al. 85 through modifying pH‐sensitive dextran derivatives onto the surface of ovalbumin (OVA) molecules‐loaded liposomes. The resultant liposomal vaccine demonstrated stability at physiological pH, but was found to be destructible in the tumor region which has a weakly acidic pH. Further study revealed that the pH‐sensitive OVA vaccine was capable of inducing more effective antigen‐specific immunity and tumor inhibitory efficiency as compared with the unmodified liposomes loaded with OVA. 85 , 86 Moreover, enzyme‐responsive liposomes are an important part of stimulus‐responsive liposomes, which are characterized by triggering drug release via attaching substrates of tumor‐specific enzymes at the response site. 87 In a recent study, a pH and MMPs dual‐responsive liposome was developed by the surface modification of pH‐responsive polymer‐PEG2000 and MMP‐responsive peptide (GPLGVRG), that conjugated with a programmed death‐ligand 1 (PD‐L1) antagonist (hydrolysis resistant D‐peptide NYSKPTDRQYHF). These dual‐responsive liposomes enable improved biodistribution and on‐demand release of immune checkpoint inhibitor (ICI) molecules within tumor regions via specific cleavage of tumor regionally secreted MMP‐2 enzymes following injection. Simultaneously, the accelerated release of doxorubicin (DOX) was observed in the acidic TME. 88

Overall, the unique size and properties of liposomal NPs provide numerous advantages, particularly improved biocompatibility and biodegradability, and increased safety when compared with polymeric and inorganic NPs. Additionally, the lipid/oil solubilizing capacity of these liposomes enhances the solubility of hydrophobic agents. Furthermore, liposomes have the ability to improve bioavailability through the prevention of agent degradation and enhancement of gastro‐intestinal membrane permeability, enhance targetability and controlled drug release. Moreover, multifunctional liposomes also offer apromising and low‐toxicity alternative for a diverse array of drugs that were previously restricted by low therapeutic index or undruggable characteristics. It is reasonable to assert that these liposomes, which possess specific targeting, stimulus‐responsive, combined therapy, and imaging capabilities, will play a pivotal role in significant advancements toward improving the safety and therapeutic benefits for individuals with cancer. 89

2.3. Delivery of therapeutic nucleic acids system by cationic LNPs

Novel RNA‐based immunomodulators, such as mRNA vaccines, antisense, small interfering RNA (siRNA), and mRNA‐based immune drugs, have been developed. 90 Among these, mRNA therapeutics have gained significant attention in cancer immunotherapy due to their ability to induce robust long‐term immune responses. The Pfizer‐BioNTech (BNT162b2) and Moderna (mRNA‐1273) COVID‐19 liposomal vaccines, which utilize mRNA encoding the SARS‐CoV‐2 spike protein as antigens to elicit host immune responses, represent the most compelling successes in this field. 91 , 92 , 93 These vaccines are PEGylated cationic LNPs with a diameter of 80–100 nm. The CLDs used in the BNT162b2 and mRNA‐1273 vaccines are tertiary amines, specifically ALC‐0315 (Pfizer) and SM‐102 (Moderna), respectively. 94 , 95 These vaccines have demonstrated successful efficacy in delivering SARS‐CoV‐2 mRNA for the prevention of COVID‐19 and indicating the huge promise of LNPs as suitable carriers for emerging RNA‐based immunotherapies. 90 Specifically, cationic LNPs have facilitated the feasibility and efficiency of mRNA‐based immunotherapy strategies by safeguarding and delivering new immunotherapy‐related RNA constructs (such as self‐amplifying RNA and circular RNA, microRNA [miRNA] and siRNA for the modification of translation, as well as CRISPR associated protein 9 messenger RNA [Cas9 mRNA] and single guide RNA [sgRNA] used for gene editing) to specific organs or cell types, thereby stimulating the immune response. 96

Cationic LNPs are defined as structures resembling liposomes, with an average size ranging from 80–100 nm, and consisting of four primary constituents: (1) PLs for drug encapsulation, (2) cholesterol for stability maintenance, (3) cationic or ionizable lipids that interact with polyanionic RNA to enhance drug‐loading capacity and facilitate endosomal escape, and (4) lipid‐anchored PEG for improved circulation and reduced immune system recognition. 97 The ionizable CLDs in LNPs are neutrally charged at a physiological pH of 7.4 to minimize the likelihood of aggregation with blood components, while exhibiting a positive charge under acidic conditions (pH∼4) to promote RNA complexation. 98 CLDs have the ability to establish stable complexes with negatively charged mRNA through electrostatic attractions, thereby impeding the binding of mRNA with serum albumin, safeguarding them from degradation by endogenous nucleases, and evading clearance by the host immune system. 99 A considerable synthetic endeavor has led to the discovery of numerous CLDs, including multivalent CLD, multitailed ionizable PLs (iPhos), DC‐chol, DOTMA, dimethyldioctadecylammonium, DOTAP, ethyl PC's, GL67, and DODMA. 100 These CLDs exhibit structural similarities to their natural counterparts, typically comprising a hydrophobic lipid tail and ionizable (cationic) head groups that contain amine moieties, amino moieties, or choline moieties. The caveat here is that the efficacy of permanently charged quaternary amine moieties in the hydrophilic head group region for siRNA delivery has been reported to be inferior to that of other lipids. 101 Further investigation has revealed that ionizable amino lipids with an apparent acid dissociation constant value (pKa) between 6.2 and 6.5 are effective for delivering therapeutic siRNAs. 102 For optimal therapy the goal of the mRNA–lipid complexes is to reach specific target cells and encode sufficient proteins of interest. Associated to this, cationic LNPs tend to bind to negatively charged molecules of the cell membrane via electrostatic interaction, resulting in enhanced permeability and intracellular uptake. 103 Upon internalization, these structural LNPs facilitate efficient delivery of the payload by enhancing intracellular phagocytosis through complexation with the negatively charged endo/lysosomal membrane. This, in turn, accelerates drug release and enables the exertion of its effect in the cytosol. 104

Recent studies have investigated the feasibility of LNPs for packaging mRNA products (e.g., single‐stranded mRNA or engineered circular RNA encoding TAA) as unique cancer neoantigens, for presentation on major histocompatibility complex (MHC) to APCs. 105 , 106 , 107 Zhang et al. 108 revealed that LNPs improve OVA expression in DCs by increased intracellular delivery of OVA‐mRNA, achieving significant beneficial effects in tumor prevention and therapeutic antitumor efficacy. Importantly, a specific 12‐carbon tail lipid endowed this LNP with adjuvant properties through Toll‐like receptors (TLRs)‐4 signaling activation further enhanced the antitumor function of a mRNA vaccine. Additionally, it was established that LNPs can be utilized to load mRNA to enhance the production of immunomodulatory proteins, such as TLR, costimulatory ligands on T cells or B cells, chemokines, and different mAb formats in various cell subpopulations. 45 As an illustration of the latter, intratumoral administration of LNPs, which have an average size of 150–200 nm and contain mRNA encoding IL‐12, IL‐27, and GM‐CSF, were found to elicit anticancer effects. In the mouse B16F10 melanoma xenograft model, repetitive administration of DAL4‐LNP resulted in effective inhibition of tumor growth by inducing adequate infiltration of immune effectors into the tumor without causing systemic toxicity. 109 A separate investigation revealed that the novel DAL4‐LNP, comprising DOPE, cholesterol, PEG, di‐amino lipid materials (DALs), and mRNA, demonstrated the ability of LNPs to safeguard mRNA sequences for effective in vivo administration, leading to significant production of anti‐human epidermal growth factor receptor 2 (HER2) antibodies in serum for a duration of 14 days (45 ± 8.6 μg/mL) following LNPs injection in breast cancer mice. 110 The exploration of LNPs, which carry double‐stranded siRNA for the purpose of knocking down checkpoint inhibitors such as anti‐PD‐L1, indoleamine 2,3‐dioxygenase 1 (IDO), and transcription factor forkhead box P3 (FOXP3), among others, is being investigated for the treatment of cancer, as well as for in vitro engineered DC vaccines. 111 Moreover, the utilization of LNPs for the codelivery of antigen‐encoding‐ and immunomodulatory‐encoding‐mRNA has demonstrated the ability to activate innate immunity through the binding of PRRs expressed by APCs. 112 , 113 , 114 Additionally, the codelivery of immunomodulatory‐encoding mRNA with siRNA for checkpoint inhibitors was shown to counteract the immunosuppressive TME and achieve potent antitumor efficacy. 115 , 116  Advancements in LNP technology, which incorporate CRISPR complexes, have demonstrated their potential as adaptable formulation platforms for the delivery of chimeric antigen receptor (CAR), T cell receptor, and/or circular RNA into lymphocytes, thereby facilitating CAR‐T therapy. 117 , 118 , 119 , 120 , 121 , 122 , 123 , 124 , 125 , 126 Furthermore, recent research has produced multiplexed dendrimer LNPs capable of codelivering siRNA, Cas9 mRNA, and sgRNA, indicating its potential for precise delivery and improved gene‐editing effectiveness. 127 , 128 , 129

While LNPs represent a significant breakthrough for mRNA delivery into cells, it is crucial to consider the potential benefits of targeted delivery of mRNA‐LNP vaccines to lymph nodes (LNs) in terms of improving immune responses and mitigating adverse effects such as hepatic damage. 130 Therefore, the targeted expression of mRNA in specific organs and cells in vivo is widely regarded as the primary challenge for cationic LNPs‐mediated mRNA delivery. To address this challenge, optional bio‐targeting layers, stimulus‐responsive motifs based on temperature, pH, or magnetic fields, and controllable component types could be employed to enable the systematic and accurate therapy or gene editing. 117 , 123 , 124 , 125 , 131 Sakurai et al. 132 reported the accumulation of RGD‐modified LNPs in the vasculature of metastasized cells in a murine model of breast cancer with lung metastasis, but not in healthy cells. It is noteworthy that modifying the structural components, and incorporating supplementary compositions also evince organ or cell targeting to some extent. 118 , 133 , 134 , 135 , 136 In comparison with smaller LNPs that possess a consistent formulation, larger particles containing siRNA or mRNA (>200 nm) exhibited increased cellular uptake in splenic DCs via micropinocytosis. 137 Furthermore, the incorporation of negatively charged 1,2‐dioleoyl‐sn‐glycero‐3‐phosphate (18PA) can function similarly to DOTAP in conferring organ specificity. 118 The ongoing progress in technology such as DNA/RNA barcode or in combination with flow cytometry for the identification of ionizable lipid targeted various organs/tissues/cells suggests significant promise for LNPs‐based mRNA therapeutic applications. 138 We anticipate that targeted LNPs will be developed in the near future to achieve heightened immune responses at reduced RNA dosages.

2.4. EVs: Attractive candidate platforms for next‐generation immune drugs delivery

EVs, including exosomes, microvesicles, and so on, which are PL nanovesicles (NVs), are secreted extracellularly by a wide range of mammalian cell types, including animal cells, plant cells, and microorganisms. Naïve EVs are comprised of cell membranes that bear numerous adhesion proteins on their surface, exhibit multivalency, and possess the ability to target cells. Their inherent physicochemical properties, including appropriate size, hydrophilic shell, low aggregation potential, and cell‐derived antiphagocytic markers, facilitate evasion of RES clearance. 139 The diameter size of EVs typically ranges from 30 to 150 nm, depending on their origin and function. 140 EVs are enriched with various biomolecules, including proteins, lipids, enzymes, transcription factors, and nucleic acids such as DNA and RNA (mRNAs, miRNAs, and long noncoding RNAs [lncRNAs]). 141 They are capable of encapsulating and delivering functional molecules to recipient cells or tissues via body fluids. 142 , 143 Research has substantiated that cancer cells secrete a greater quantity of EVs as compared with nonmalignant cells (e.g. immune cells and epithelial cells). These tumor‐derived EVs (TDEVs) contain molecular components specific to tumors and can be extracted from blood and urine. Considering the important role of TDEVs in the progression of cancer from early stages to metastasis, they have the potential to serve as the next generation of fluid biopsy biomarkers for cancer diagnosis and management. 144 , 145 Furthermore, TDEVs with targeting abilities to transfer functional proteins or nucleic acids to selective cells have demonstrated inspiring preclinical outcomes in cancer nanomedicine and immunotherapy. 146 , 147 But it should be noted that TDEVs as a drug carrier should be engineered by changing the original cargos into therapeutic products or altering modification on the surface of the membrane, thereby preventing the tumor‐promoting effect of TDEVs from counteracting the anticancer efficacy (engineered EVs will be discussed subsequently). 148

Furthermore, the self‐immunomodulatory attribute of exosomes facilitates the progression of novel immunological methodologies in the field of oncology, by activating adaptive and innate effector cell‐mediated immune surveillance. 146 , 149 Certain exosome variants retain the crucial immunostimulatory characteristics of their progenitor cells and can function as cancer vaccines, augmenting the scope and potency of the immune response. 150 For instance, TDEVs enriched with TAA can elicit cross‐priming of cytotoxic T lymphocytes (CTLs). 151 DC‐secreted exosomes possess MHC class I or MHC class II peptide complexes, costimulatory molecules (e.g., CD40, CD80, and CD86), tumor necrosis factor (TNF) superfamily ligands (e.g., TNF, FasL, and TRAIL), and NK group 2, member D (NKG2D) ligands (e.g., UL16 binding protein 1) that facilitate antigen presentation and promote the activation of tumor‐specific CTLs. 152 , 153 , 154 Furthermore, exosomes derived from other immune subpopulations may also possess the capacity to elicit effective antitumor immunity for cancer immunotherapy. 155 An illustration of this phenomenon is the secretion of exosomes by B cells, which contain MHC‐II‐like peptide complexes that stimulate the clonal expansion of CD4+ T cells and modulate cytokine secretion. 156 Additionally, macrophages carrying pathogen‐associated antigens promote dendritic cell maturation and the release of proinflammatory cytokines upon pathogen exposure. M1 macrophages have been observed to release exosomes that effectively reprogram immunosuppressive M2 macrophages to immunoresponsive M1 macrophages. 157 The immunomodulatory capabilities of EVs can be enhanced by directly encapsulating biological cargos such as antigens, immune adjuvants, immune checkpoint molecules, and so on. 146 Generally used immune adjuvants for promoting DC maturation are cytokines, cytosine‐phosphorothioate‐guanine oligonucleotides (CPG ODN), TLR agonists, STING agonists, and LPS derivatives, and so on. 158 Adamus et al. 159 employed a method of encapsulating CpG‐STAT3 antisense oligonucleotide conjugates into EVs derived from neural stem cells (NSC), which resulted in a significant enhancement of the immunoreactivity of natural NSC‐derived exosomes. This approach effectively targeted glioma‐associated microglia and inhibited tumor growth. 159 Furthermore, EVs demonstrated the potential to amplify the antitumor immune effects induced by conventional ablative therapies, such as low‐dose chemotherapy, radiation therapy, and PDT, without causing intolerable systemic toxicity. 160 EVs, therefore, offer a rational platform for the combined administration of ablative therapies. Wang et al. 161 conducted a study wherein EVs were modified to simultaneously transport the secondary generation photosensitizer chlorin e6 (Ce6) for PDT and the TLR agonist R848 for the treatment of prostate cancer. The utilization of EVs in this context effectively addresses the prevailing issues associated with photosensitizers, such as nonspecific accumulation and inadequate water solubility, while also improving antitumor efficacy through the induction of cascade immune responses. 161

In addition, on the basis of directly incorporating biological cargos into EVs, engineered EVs have been designed to further improve targeting, optimize biological distribution, and enhance the immunogenicity and antitumor efficiency of immune drugs. 162 , 163 , 164 Common methods are endogenous engineering of donor cells with homing peptides and appropriately decorating the surface of EVs with antigen‐associated signaling molecules, aptamers, and small molecular drugs. 146 , 165 For example, the cDNA encoding the target OVA homing peptide was inserted into the gene sequence of donor EG7 tumor cells by genetic engineering techniques (transfection), thereby altering the structure of the secreted exosomes to express OVA on their cell membranes. Moreover, the OVA‐pulsed DC‐derived EVs exhibit a stronger stimulatory effect than engineered TDEVs in the induction of OVA‐specific CD8+ T cell responses and antitumor immunity, and both of them showed tumor targeting ability. 166 A variety of transmembrane proteins have been tried to build engineered EVs with homing ability, such as gap junction protein connexin43, lysosome‐associated membrane protein‐2b, fibronectin/tenascin‐C, CD protein (LA), and four transmembrane proteins (CD63, CD9, and CD81). 167 , 168 , 169 Mesenchymal stem cells (MSCs)‐derived exosomes were found to be minimally immunogenic and immunity‐inducible, as well as suitable for low‐cost mass production. 170 In a recent study, a codelivered biological platform was developed that is based on bone marrow MSCs that electroporation‐loaded galectin‐9 siRNA and modified with surface‐bound oxaliplatin prodrugs for the enhanced treatment of pancreatic cancer. The engineering EVs have been found to be effective in inducing immunogenic cell death (ICD), and providing sufficient antigen for antitumor immune responses. Moreover, the EVs have the ability to modulate tumor suppressor macrophage polarization and reduce Tregs' levels, thereby remodeling the TME. 171 Engineered exosomes based on macrophages are effective strategy to modulate the TME in cancer immunotherapy. 172 Gunassekaran et al. 173 extracted exosomes from M1 macrophages, which were subsequently engineered to load nuclear factor kappa‐light‐chain‐enhancer of activated B (NF‐κB) p50 siRNA plus miR‐511‐3p and target the IL4 receptor, named IL4R‐Exo (si/mi). Systemic administration of IL4R‐Exo (si/mi) showed effective tumor targeting by re‐educating TAM into M1‐type macrophages and promoting the activity of immunostimulatory cells to remodel TME, resulting in inhibited tumor growth. 173

Moreover, the biocompatibility, tumor targeting, and controlled release of EVs can be substantially enhanced by physically modifying their surface with liposolubility targeting molecules or PEG coatings. 174 , 175 One example of this is the creation of a genetically engineered multifunctional immunomodulatory exosome (GEMI‐NI‐Exos), which utilizes Expi293F cell‐derived EVs that are encapsulated with PD‐1 and OX40 mAb, and surface‐modified to target CD3 and epidermal growth factor receptor (EGFR). GEMINI‐Exos effectively targets EGFR‐positive breast cancer cells, and its injection induces robust anticancer immunity and efficient tumor suppression in a mouse model of breast cancer. 176 It is noteworthy that PEG coatings may potentially conceal other inherent bioactive surface functionalities of EVs. Furthermore, modification of the molecular structure of EVs film by click chemical method is another engineering strategy. 177 Koh et al. 178 developed a pH‐sensitive engineered exosome by utilizing a dibenzocyclooctyne‐modified antibody to CD47 and SIRPα (aCD47 and aSIRPα) to bind azide‐modified EVs of M1 macrophages. The engineered EVs demonstrated superior RES camouflage, prolonged circulation half‐life, and regulated release, thereby exhibiting the potential for reversing tumor immunosuppressive TME. Additionally, EVs are used to prepare exosomal membrane‐coated NP particles such as PLGA NPs to reduce the immune clearance of NPs and improve their tumor‐specific targeting. 179 Furthermore, membrane fusion methods can be chosen for those EVs which are difficult to modify ligands directly on EVs membranes, also in case for hybrid EVs preparation in which lipids (DOTAP, POPC, DPPC, and POPG) are fused with EVs. 180 , 181 , 182 As such, the outlook on the future of EVs as delivery approach for cancer immunotherapy is promising but researchers should consider that the intricate composition of engineered immune EVs during nanomedicine design phase may pose challenges in their clinical implementation and large‐scale manufacturing.

2.5. SLNs and NLC for the large‐scale production

The emergence of nanotechnology has led to the development of SLNs as a new generation of colloidal NPs, ranging in size from 10 to 1000 nm. 183 Unlike conventional liposomes that contain liquid crystal lipid bilayers, SLNs possess a micelle‐like structure composed of natural or synthetic solid lipids, surfactants, and other coating materials. 184 Common solid lipids include triglycerides, fatty acids, steroids, and waxes, which can solubilize hydrophobic drugs in their melted lipid phase or form a drug‐enriched shell surrounding them. 185 Surfactants are chemical compounds composed of a polar (hydrophilic) head and a nonpolar (hydrophobic/lipophilic) tail. The incorporation of surfactants facilitates the stabilization of SLNs by reducing the interfacial tension (adhesive forces) between two phases. 186 It has been observed that SLNs exhibit superior efficacy in encapsulating hydrophobic drugs in comparison with liposomes. 187 Additionally, SLNs offer greater control over drug release in comparison with other types of lipid‐NPs. The encapsulation of drugs within SLNs can be achieved through physical adsorption, adsorbents, and coprecipitation, while the modulation and optimization of SLNs release are through adjusting formulation into a drug‐enriched core or shell pattern. 188 This is mediated by the rapid diffusion of drugs adsorbed on the surface of SLNs during the initial release phase, while drugs encapsulated in the interior of SLNs exhibit a prolonged release profile due to nonuniform distribution within the lipid matrix. 189

Various production methods, including high‐pressure homogenization, ultrasonic emulsification, thermal melting, and fatty acid emulsification, have been employed to prepare SLNs without the use of organic solvents. 189 The inclusion of solid lipids in SLNs confers superior physical and chemical stability compared with liposomal and polymeric NPs, thereby enabling the production of sterile products. Additionally, these characteristics contribute to a reduction in lipid degradation, which renders SLNs suitable for large‐scale production and reproducibility. 190 The rather straightforward manufacturing process and adaptable carrier properties, including enhanced drug‐loading capacity, improved protein stability, encapsulation efficiency, and precise control, further enhance the clinical potential of SLNs for localized and systemic cancer applications. 191 , 192 , 193 , 194 , 195 Banerjee et al. 196 presented a paclitaxel formulation based on SLNs that was engineered with Tyr‐3‐octreotide (PSM) and demonstrated the ability to induce ICD. This formulation was found to modulate local CD8+ T cell infiltration and systemic immune responses in melanoma mice following administration. 196 Despite these promising results, further research is needed to investigate the incorporation of clinical immunotherapy cargoes into SLNs for cancer treatment.

Upon storage, the solid lipids present in SLNs have a tendency to undergo crystallization, leading to the expulsion of the encapsulated drug into the surrounding medium. This, in turn, results in a decrease in the encapsulation efficiency of SLNs. 186 , 197 To overcome these limitations of SLNs, NLCs have been developed. NLCs are designed to enhance drug loading, prevent drug leakage, and achieve controlled drug release under certain conditions. 198 Unlike SLNs, NLCs are capable of inhibiting drug release during storage by incorporating a small amount of liquid lipids into the solid lipids. This reduces the crystallinity of the lipid core and improves the overall stability of the formulation. 199 Furthermore, the incorporation of liquid lipids has the potential to disturb the lattice arrangement of solid lipids, leading to the formation of anomalous crystalline cores within NLCs, thereby augmenting the drug loading capacity by increasing the available space. 199 , 200 By manipulating the proportion of liquid lipids while preserving the skeletal structure of NLCs can facilitate more accurate regulation of drug release kinetics. 201 Although the field is still young, the deployment of SLNs and NLCs encapsulating widely used cancer immune drugs and some mRNA products hold an encouraging potential for future studies.

2.6. Nanoemulsions

NEs are characterized as biphasic dispersions consisting of two immiscible liquids, namely, a dispersed phase and a continuous phase (water and oil), which are typically stabilized through the use of surfactants and cosurfactants as emulsifiers. 202 The formation of NE is not typically spontaneous, but rather requires the application of energy input through high pressure homogenizers, high shear agitation, or ultrasonic generators. 202 The prevalent forms of emulsions comprise a “water‐in‐oil” (w/o) emulsion, an “oil‐in‐water” (o/w) emulsion, and multiple emulsions, which are contingent upon the proportion of water to oil and the type of emulsifier employed. 203 For instance, emulsifiers that are more soluble in water generally produce w/o emulsions, with their nonpolar tails extending into the oil and their polar head groups oriented toward the water. Conversely, emulsifiers that are more soluble in oil generate o/w emulsions, with their polar heads extending into the water and their nonpolar tails directed toward the oil. 204 The physiochemistry of NE, specifically their drug loading capability and drug release rate, has been found to be dependent on several factors including the water content, types and contents of oils/lipids (typically 5–20 wt% of the NE), and the oil/water partition coefficient of the drug molecule in the NE system. Commonly utilized oils/lipids for NEs consist of long‐chain unsaturated fatty acids, glycerides, vegetable oils, medium‐chain triglycerides, and polyalcohol esters of medium‐chain fatty acids. 205 Consequently, the screening of oils/lipids has been demonstrated to be a crucial aspect of NE development, and novel oils and lipids are currently being developed for use in synthetic NE. One such example is deep‐sea fish oil, which has been demonstrated to be a safer therapeutic option for bleomycin‐induced pulmonary fibrosis in BALB/c mice. 206 , 207 NE exhibit excellent kinetic stability, and the addition of appropriate surfactants can further enhance their stability by reducing the oil–water interfacial area and tension, as well as through electrostatic interactions with the interfacial surface. 208 For instance, oil‐in‐water emulsions containing a soy protein/soy polysaccharide complex as surfactants have been shown to maintain stable oil droplet size. 209

NEs are significant contributors to various diverse fields, including drug carriers, food, cosmetics, and oilfield chemicals. 210 In the context of drug delivery, NEs have exhibited the capacity to efficiently load lipophilic drugs and nutrients, such as fats, carbohydrates, and vitamins, into oil droplets. This process enhances their dispersion and stability in aqueous solutions and facilitates drug release and absorption postdigestion. 207 Furthermore, due to their small droplet size, NEs are considered one of the most promising options for oral drug delivery. 211 The current state of research regarding the use of NEs to facilitate immunotherapy is in its preliminary stages, with a primary focus on the administration of chemotherapeutic drugs such as tamoxifen and dacarbazine, as well as tumor multimodal imaging. 212 , 213 , 214 , 215 , 216 However, initial findings suggest that NEs may hold significant potential in the realm of cancer immunotherapy. Specifically, a tailored NE formulation containing perfluorooctyl bromide and MnO2 NPs has been developed as a theragnostic agent, capable of providing both magnetic resonance imaging (MRI) and computed tomography (CT) dual‐modality imaging, as well as inducing advanced ICD. 217 Furthermore, Zeng et al. 218 reported data indicating the efficacy of a novel NEs platform in promoting the targeting of DCs with TAA or neoepitopes, thereby inducing tumor‐specific immunity with therapeutic potential. Current endeavors aim to explore the potential of NEs in the context of cancer immunotherapy.

2.7. The upgraded version of lipid‐NPs: LPHNPs

The application of lipid‐NPs and polymeric NPs has surfaced as efficacious methodologies to enhance the solubility and stability of anticancer drugs, protract the half‐life of anticancer drugs in plasma, diminish off‐target distribution, mitigate drug toxicity, and foster drug accumulation in disease regions, thereby augmenting the therapeutic efficacy of the loaded drugs. 219 Nevertheless, both liposomal and polymeric NPs exhibit certain limitations, prompting the development of LPHNPs as a novel class of NPs. 220

LPHNPs can be classified into five distinct classes based on their structural composition, including monolithic LPHNPs, polymer core lipid shell NPs, hollow core lipid–polymer–lipid nanoparticles, biomimetic lipid polymer hybrid systems, and polymer caged liposomes. 221 Despite their subtype, LPHNPs offer several advantages, such as enhanced biocompatibility and safety, improved drug loading capacity and controlled release, as well as prolonged circulation time and therapeutic efficacy of drugs. 222 For example, the utilization of CLD‐based cores has demonstrated the ability to incorporate anionic drugs and nucleic acids. 223 This approach can be further enhanced by utilizing lipids or biomimetic lipids, such as those derived from stem cells, platelets, and white blood cells, as a coating layer. This coating layer can reduce the potential for unexpected immune reactions and toxicity, improve biocompatibility, prevent drug excretion, and delay drug release. 224 , 225 Additionally, the incorporation of amphiphilic lipids as a lipid coat can aid in stabilizing the matrix. 226 Furthermore, the incorporation of a polymeric core, such as a PLGA core, in LPHNPs can facilitate the incorporation of both hydrophilic and hydrophobic drugs, preventing their systemic metabolism, and promoting enhanced storage stability. 227 In comparison with SLNs and liposomes, LPHNPs exhibit greater suitability for hydrophilic drugs due to their higher drug encapsulation rate. This is attributed to the electrostatic adsorption effects that enable the formation of drug–polymer complexes, which are subsequently encapsulated in hydrophobic lipids. 227 , 228 Notably, the presence of a PEG layer on the lipid core of LPHNPs confers highly adjustable characteristics and improved physicochemical stability. 229 That is becausethe integration of the lipid–PEG layer into the lipid shell of LPHNPs imparts surface hydrophilicity, prevents absorption by the RES, prolongs circulation time, and improves its steric stability in plasma matrices. 230 Also, this approach has demonstrated success in enabling the traversing of various physiological barriers. 231

In particular, numerous immune agents have undergone testing for delivery efficiency and antitumor therapeutic index by LPHNPs, including refractory chemotherapy drugs, targeted drugs, and mRNA. 232 , 233 One example involves the encapsulation of OVA‐encoding mRNA into hybrid core–shell NPs to create a nanovaccine. This nanovaccine has been shown to enhance vaccination effects specifically in DCs due to macropinocytosis, resulting in improved antitumor growth efficiency and reduced metastases in the lungs of B16‐OVA‐bearing mice following repeated injection. 234 The introduction of additional immune activators, such as various TLR agonists, along with antigens into LPHNPs, has the potential to enhance antigen uptake, improve lysosome escape, increase cytokine production, produce depot effects, and promote trafficking to secondary lymphoid organs. Consequently, these DC‐targeted vaccines can elicit potent and long‐lasting immunity against cancer. 235 , 236 The introduction of additional immune activators, such as various TLR agonists, along with antigens into LPHNPs, has the potential to enhance antigen uptake, improve lysosome escape, increase cytokine production, produce depot effects, and promote trafficking to secondary lymphoid organs. Consequently, these DC‐targeted vaccines can elicit potent and long‐lasting immunity against cancer. 237 These hybridized particles are being considered as a promising platform for adapting combined anticancer modalities that work through boosting host immunity synergistically. For example, various approaches such as image‐guided cancer immunotherapy, chemoimmunotherapy (e.g., CD47 siRNA + etoposide), photoimmunotherapy (e.g., indocyanine green + immune stimulatory recombinant protein FimH; IR‐780 and imatinib; pyropheophorbide‐lipid conjugate [pyrolipid] + oxaliplatin; and recombinant PD‐1 decorated thermal responsive LPHNPs, etc.), multitherapeutic strategy (e.g., Prominin2 siRNA + oxaliplatin + ferroptosis) have been developed. 238 , 239 , 240 , 241 , 242 , 243 In general, LPHNPs, an advanced form of SLP, represent a promising type of nanomaterial with extensive possibilities and potential applications in the areas of RNA therapeutics, targeted immunotherapy, combination immunotherapy, and as an all‐in‐one platform for diagnosis and treatment.

3. cGAS–STING, A PIVOTAL PATHWAY IN CANCER IMMUNOTHERAPY

Under immunosurveillance, the evolution of malignant cells gives rise to tumors capable of evading immune detection or suppressing immune attacks. 244 Immuno‐oncology therapy aims to overcome immunosuppression and reinvigorate the antitumor immunity of cancer patients by blocking immunosuppressive pathways in TME or activating the antitumor immune signals. Cancer immunotherapy has developed rapidly in the past decade, in particular through the success of immune checkpoint‐related therapies. 245 Antibodies that target PD‐1, PD‐L1, lymphocyte‐activation gene 3 (LAG‐3), and CTLA‐4 have been approved by US FDA for the treatment of various solid or advanced tumors and have shown unprecedented efficacy and long‐lasting response. However, clinical data indicate that sole treatment with ICIs to disarm negative signals is ineffective in those cancer patients without sufficient antitumor immunity. 246

Through the in‐depth exploration of tumor immune activation‐related pathways, an increasing number of potentially therapeutic targets have been identified, including the cyclic guanosine monophosphate (GMP)–adenosine monophosphate (AMP) synthase (cGAS)–STING signaling pathway, a widely recognized endogenous sensor of tumors. cGAS is an enzyme that was originally discovered as a cytoplasmic PRR for the detection of pathogen DNA. It can be initiated directly by cytosolic double‐stranded (ds)DNA from invading microbes such as DNA viruses, retroviruses, and intracellular bacteria (Figure 4). 247 In brief, dsDNA enters the cytosol and induces cGAS activation by directly binding to the endoplasmic reticulum (ER). This activated cGAS generates cyclic GMP–AMP (cGAMP), a negatively charged CDN complex in the presence of guanosine‐5'‐triphosphate and adenosine triphosphate. cGAMP functions as a second messenger that binds to the adaptor protein STING in the ER and undergoes dimerization, in turn translocating to the Golgi apparatus and triggering activation of TANK‐binding kinase 1 (TBK1)/IFN regulatory factor 3 (IRF3) signaling. 248 , 249 Alternatively, STING in the form of a dimer can also activate the NF‐κB signaling via IκB kinase‐mediated phosphorylation of IκBα. 248 Thus, STING pathway activation leads to the transcription of type I IFN‐associated genes and genes encoding proinflammatory cytokines (e.g., TNF and IL‐6), which promotes antitumor immunity through the activation of T cells and NK cells. 250 , 251 , 252 Although initially discovered as a PRR for DNA from invading pathogens, under some conditions, cGAS–STING pathway can also be activated by self‐DNA leading to autoimmune disorders. 253 In tumors, DNA or cGAMP from dying tumor cells access the cytosol of APCs and activate the cGAS–STING pathway, leading to the induction of type I IFNs and other immune stimulatory molecules, which are essential in modulating the functions of immune cells. 254 Moreover, cancer treatments that include radiotherapy and chemotherapy which induce DNA damage and genomic instability also showed the ability to activate the cGAS–STING pathway and the development of antitumor immune responses. 255 , 256 The biological effects of STING activation on antitumor immunity will be discussed detail in the next section.

FIGURE 4.

FIGURE 4

Overview of the cyclic GMP–AMP synthase (cGAS)–stimulator of interferon genes (STING) signaling pathway in dendritic cells (DCs). Figure was created using biorender.com.

Given the importance of the cGAS–STING pathway in cancer immunity, considerable effort has been put into targeting cGAS–STING pathway pharmacologically. Numerous agonists for cGAS–STING pathway have been developed and shown effective antitumor effects. In order to upregulate STING expression, STING agonists must penetrate into cells to reach their target. Several CDNs from cGAMP and non‐nucleoside small molecule STING agonists are in different stages of clinical development, which will be covered in detail in Section 4.1. However, one of the major concerns in the clinical usage of STING agonists is that naked STING agonists are poorly internalized by cells. Moreover, potential adverse effects associated with cytokine induction also constrain the application of STING agonists. The development of STING agonists has entered a bottleneck as most novel‐immunodrugs. Therefore, the current investigation focused on delivering STING agonists with diverse vectors. These vectors can be cationic LNPs, liposomes, polymeric NPs and metal NPs. which are the most commonly used carrier system. Among these biomaterials, the lipid‐NPs have advantages of good biocompatibility and safety, low immunogenicity, and biodegradability as described previously. Considering that STING agonists cover the advantages and challenges of the most immune activation adjuvants, and STING agonists are taken as a representative in the subsequent part of this review to discuss the potential of lipid‐NPs application in cancer immunotherapy. The aim of lipid‐NPs for STING agonists is to improve safety and enhance intracellular delivery efficacy, to thereby exploit the full therapeutic potential of STING agonists in immunotherapy.

3.1. Role of cGAS–STING pathway on tumor cells

In light of the bridging role of STING signaling between innate and adaptive immunity in response to DAMPs and PAMPs, a wealth of studies have reported the underlying mechanisms by which the cGAS–STING signaling pathway controls tumor progression. A common conclusion is that cGAS–STING signaling regulates multiple steps in antitumor immunity and ameliorates the immunosuppressive TME‐related factors (Figure 5). 247 , 257 , 258

FIGURE 5.

FIGURE 5

STING pathway activation can convert the tumor microenvironment from an immunosuppressive “cold” state to a proinflammatory “hot” state via the following steps: (A) improved antigen immunogenicity, processing, and presentation, and T‐cell priming, activation, thereby enhancing CD8+ T‐cell immunity; (B) enhanced trafficking and infiltration of T cells into tumor area by induced chemokines due to increased type I IFN; (C) enhanced killing of tumor cells by NK cells and CD8+ T cells; (D) converting to normalized TME including blood vessel normalization, decreased level of hypoxia within the tumor, suppression of immunosuppressive cell types and induction of acute inflammation by inducing the expression of various cytokines and chemokines. Figure was created with biorender.com.

Activation of the STING pathway in tumor cells can directly induce tumor cell senescence or various death modalities (such as apoptosis, necroptosis, ferroptosis, etc.) in a cell‐autonomous manner. This cGAS–STING signaling‐pathway‐mediated tumor cell suppression is thought to be mediated through the regulation of type I IFN (IFN‐α and IFN‐β) and other proinflammatory genes. 259 Evidence suggests that cGAS–STING signaling‐pathway‐mediated autophagic tumor cell death occurs through the formation of micronuclei upon sensing aberrant cell fusion. 260 During the autophagy process, the STING protein acts as both an autophagy substrate and a modulator of autophagy, and a potential mechanism is via autophagosome cargo protein sequestosome 1(SQST)‐mediated signals. 261 Notably, studies suggest that DNA damage under specific conditions triggers the release of DNA (mitochondrial DNA and nuclear DNA) into the cytosol, leading to the activation of STING signaling and subsequent autophagy‐dependent ferroptosis in both mice and human pancreatic cancer cells. These responses have also been found to result in tumor growth control in associated murine pancreatic cancer models. 262 Moreover, the activation of STING signaling is reported to be involved in the induction of tumor cell apoptosis through the IRF3–BCL2‐associated X protein/BCL2‐antagonist/killer 1 pathway and necroptosis by triggering the expression or phosphorylation of mixed‐lineage kinase domain‐like pseudokinase, one of the major regulators of necrosome formation during necroptotic cell death. 263 , 264 Interestingly, in contrast to the proapoptotic effect in tumor cells, activation of the STING pathway in DCs and macrophages is antiapoptotic. 265 , 266

3.2. Role of cGAS–STING pathway on immune cells in TME

Through enhancing the immunogenicity of tumor cells by ICD and promoting the maturation of APCs, thereby initiating CD8+ T‐cell responses, STING activation results in improved therapeutic efficacy. 267 , 268 , 269 , 270 Recent evidence supports the notion that, except for tumor‐cell‐produced single‐stranded DNA (ssDNA) or dsDNA (e.g., induced by genomic instability), DNA damage due to chemotherapy or radiotherapy and abnormal tumor cell proliferation can also directly induce DC maturation and recruitment via STING activation. 271 , 272 In addition, cGAMP amplifies the capacity of neighboring DCs to produce type I IFN and related cytokines by transfer via EVs secretion or gap junctions. 273 Research has demonstrated that the initiation of spontaneous tumor antigen‐specific T cells is reliant on DC activities and the production of type I IFN. 274 Intratumoral injection of STING agonists has been shown to enhance CTLs infiltration into tumors. 275 , 276 Because STING activation and type I IFN secretion within the TME upregulated the expression of genes encoding inflammatory cytokines (IL‐12, TNF‐α, IFN‐γ, etc.) and T cell‐recruiting chemokines (C‐X‐C motif chemokine ligand [CXCL]9 and CXCL10), thereby transforming immunosuppressive “cold” tumors into T‐cell‐inflammatory “hot” tumors. 277 , 278 , 279 Additionally, STING agonists can directly activate T cells, independent of DC‐released type I IFN. However, activation of the STING pathway in T cells may result in impaired T lymphocyte proliferation, reduced memory cell counts, and activation of the T cell emergency and death pathways, ultimately leading to T cell death. 280 , 281

STING activation can also increase the sensitivity of tumor cells to immune NK cells and CTLs. 282 However, careful consideration is still needed to explore the impact of STING activation on NK‐cell function, as one recent publication showed that excessive STING activation in B cells might diminish NK‐cell function. In addition, STING pathway activation can also promote TME normalization by regulating macrophage polarization. 283 Briefly, STING agonists repolarize tumor‐associated macrophages from the M2 type towards the M1 type through NF‐κβ/IRF3 activation and type I IFN upregulation. 284 , 285 It has been reported that systemic or intratumor administration of STING agonists reverses the immunosuppressive TME and contributes to tumor regression. 286

3.3. Role of cGAS–STING pathway on endothelial cells in TME

In addition to immune cells, recent research has identified stromal cells, particularly endothelial cells, as a target of the STING pathway within the TME. 276 , 287 , 288 , 289 The initial STING agonists (DMXAA, 5,6‐dimethylxanthenone‐4‐acetic acid) have demonstrated the ability to act as vascular disruptors, thereby inhibiting tumor growth. 290 Activation of STING within endothelial cells has been shown to promote the secretion of IFN‐β, which in turn leads to T‐cell infiltration into solid tumors. The blockade of IFN signaling using IFNAR antibody or IFNAR ablation can further eliminate T‐cell infiltration. 276 Adhesion to endothelial cells is a crucial step in the process of T‐cell infiltration. 291 Intratumoral injection of STING agonists into tumor‐bearing mice (cGAMP or ADU‐S100) upregulated type I/II IFN genes, vascular stabilizing genes (including Angpt1, Pdgfrb, and Col4a), and adhesion molecules (including Icam, Vcam, and Sell), promoted tumor vascular normalization, reduced tumor hypoxia, and increased tumor‐specific CTL infiltration. 287 However, in VEGF‐rich cancers, VEGF/VEGFR2 signaling might hamper the antitumor efficiency of STING agonists due to its negative regulation of type I IFN signaling activation via ubiquitin‐mediated IFN α and β receptor (IFNAR) degradation. 292 , 293 Additionally, VEGF (and other cytokines) secreted by cancer cells may suppress the expression of molecules that mediate T cell adhesion on endothelial cells or induce the expression of molecules that trigger the death of effector T cells. 294 Related to this, administration of STING agonists together with antiangiogenic/antivascular agents as well as an ICI showed the potential to promote T‐cell infiltration into the TME and prevented tumor recurrence in a colorectal mouse model. 287 Careful consideration needs to be given to the combination of STING agonists with VEGF blockers, as this may inhibit T‐cell infiltration by suppressing the proliferation of endothelial cells within the tumor, leading to the progression of certain cancer types. This latter is because appropriate levels of VEGF are necessary to maintain endothelial cell populations. 295 It can be hypothesized that targeted lipid‐NPs might address this issue through increasing specificity to reduce the uptake of VEGF blockade agents by endothelial cells. However, the effectiveness and possibility of targeted lipid‐NPs for STING and VEGF co‐therapy in VEGF‐rich cancer types still needs to be explored.

4. TARGETING THE cGAS–STING SIGNALING PATHWAY

4.1. Targeting the cGAS–STING signaling pathway in cancer clinical trials

The STING signaling pathway has emerged as a therapeutic target in tumor therapy (Table 3). As the first STING agonist, DMXAA was created and investigated for its anticancer therapeutic benefits. Multiple preclinical studies have indicated that intratumoral or systemic delivery of DMXAA might stimulate the innate immune system, inhibit tumor angiogenesis, and exhibit tumor suppression in various types of cancer (e.g., NSCLC, breast cancer, melanoma, fibrosarcoma, etc.). 285 , 290 , 296 , 297 , 298 However, the clinical outcomes of DMXAA treatment have not been satisfactory, due to the strong affinity of DMXAA for mouse STING protein but not for human STING. 296 , 299 , 300 Inspired by the potent antitumor immunity induction of DMXAA, various agonists targeting human STING signaling, including eukaryotic/prokaryotic STING ligands and synthetic mimics, have been developed: CDNs, non‐CDN small molecules, nanovaccines, antibody–drug conjugates (ADCs), and so on (Figure 6). 301 However, the stability and efficiency of natural STING agonists, such as 2′3′‐cGAMP, 3′3′‐cGAMP, cyclic di‐AMP (CDA), and cyclic di‐GMP (cdGMP), render STING‐targeted therapy a challenge. 302 Additionally, synthetic CDNs have been developed to circumvent the limited half‐life and stability of natural CDNs, and they have shown variable results. For example, ADU‐S100 (ML‐RR‐S2, MIW815) is a synthetic CDN that explicitly targets human STING signaling with enhanced stability and affinity, and has been used in the treatment of advanced and metastatic solid cancer. 285 In clinical settings, the development of ADU‐S100 has been put on hold due to the limited antitumor immunity conferred by single treatment or in combination with various ICIs, including anti‐programmed death‐1 (PD‐1) antibodies and anti‐cluster of differentiation 152 (CTLA‐4) antibodies (clinical trials: NCT02675439, NCT03172936, NCT03937141). A completed Phase 1 study (clinical trial: NCT03010176) showed that 6 of 25 patients responded to the combination therapy of MK‐1454 (a synthetic CDN) and pembrolizumab (anti‐PD‐1 antibodies), whereas no significant response was observed as a result of MK‐1454 monotherapy. 303 Additionally, a self‐assembled CDN, SB11285, has been shown to improve physicochemical properties against enzymatic degradation, and its clinical development as a monotherapy or in combination with atezolizumab is in progress (anti‐PD‐L1 antibodies, clinical trials: NCT04096638).

TABLE 3.

Clinical trials of STING agonists in cancer.

Phase Agonist Cointervention Cancer type Status Clinical trial reference number
Phase I/II CDK‐002/exoSTING (i.t.) Advanced/metastatic, recurrent, injectable solid tumors Completed (plan to into Phase 2 in bladder cancer) NCT04592484
IMSA101 (i.t.) Immune checkpoint inhibitor (ICI) or Immuno‐oncology (IO) therapy Advanced treatment‐refractory malignancies Recruiting NCT04020185
Early Phase I TAK‐676 (CIVO® i.t.) MK‐0482 + pembrolizumab or MK‐4830 + pembrolizumab Solid tumors Recruiting NCT04541108
Phase I SNX281 (i.v.) Pembrolizumab (i.v.) Advanced solid tumors and lymphoma Withdrawn (no enrolment of participants) NCT04609579
E7766 (i.v.) Nonmuscle invasive bladder cancer Withdrawn (no enrolment of participants) NCT04109092
E7766 (i.t.)
  • Advanced solid tumors

  • Lymphomas

Completed (no results posted) NCT04144140
MK‐1454 (i.t.) Pembrolizumab (i.v.)
  • Solid tumors

  • Lymphoma

Completed (results submitted) 310 NCT03010176
MK‐2118 (i.t./s.c.) Pembrolizumab (i.v.)
  • Advanced/metastatic solid tumors or lymphomas

Completed (no results posted) NCT03249792
ADU‐S100 (i.t.) PDR001 (i.v.)
  • Advanced/metastatic solid tumors

  • Lymphomas

Terminated (sponsor's decision) NCT03172936
ADU‐S100 (i.t.) Ipilimumab (i.v.)
  • Advanced/metastatic solid tumors

  • Lymphomas

Terminated (no substantial antitumor activity was observed.) NCT02675439
KL340399 (i.t.) Advanced solid tumors Recruiting NCT05549804
KL340399 (i.v.) Advanced solid tumors Not yet recruiting NCT05387928
TAK‐500 (i.v.) Pembrolizumab (i.v.)
  • Locally advanced

  • Metastatic solid tumors

Recruiting NCT05070247
TAK‐676 (i.v.)

Pembrolizumab (i.v.)

or

Pembrolizumab (i.v.)

Carboplatin/cisplatin (i.v.)

5‐Fluorouracil (i.v.)

Advanced or metastatic solid tumors Recruiting NCT04420884
TAK‐676 (i.v.)

Pembrolizumab (i.v.)

Radiation therapy

  • Non‐small‐cell lung cancer

  • Triple negative breast cancer

  • Head and neck squamous cell carcinoma

Recruiting NCT04879849
GSK3745417 (administration route was not specified) Leukemia Recruiting NCT05424380
Dostarlimab (i.v.) Neoplasms Active, not recruiting NCT03843359
BMS‐986301 (i.m./i.v./i.t.)

Nivolumab (i.v.)

Ipilimumab (i.v.)

Advanced solid cancers Active, not recruiting NCT03956680
SB11285 (i.v.) Atezolizumab (i.v.)
  • Melanoma

  • Head and neck squamous cell carcinoma

  • Solid tumor

Recruiting NCT04096638
BI 1387446 (i.t.) Ezabenlimab (i.v.) Advanced or metastatic cancer Active, not recruiting NCT04147234
SNX281 (i.v.) Pembrolizumab (i.v.)
  • Advanced solid tumors

  • Advanced lymphoma

Recruiting NCT04609579
SYNB1891 (i.t.) Atezolizumab (i.v.)
  • Advanced/metastatic solid tumors

  • Lymphom

Recruiting NCT04167137
ONO‐7914 (administration route was not specified) Nivolumab (i.v.) Solid tumors Recruiting JRCT2031210530
HG381 (administration route was not specified) Advanced solid tumors Recruiting NCT04998422
DN1508052‐01 (s.c.) Neoplasms Completed NCT03843359
KL340399 (administration route was not specified) Advanced solid tumors Not yet recruiting NCT05387928
STING‐dependent Activators (STAVs, i.v.) Dendritic cell vaccine Aggressive relapsed/refractory leukemias Not yet recruiting NCT05321940
Phase II ADU‐S100 (i.t.) Pembrolizumab (i.v.) Head and neck cancer Terminated (no substantial antitumor activity was observed.) NCT03937141
MK‐1454 (i.t.) Pembrolizumab (i.v.) Head and neck squamous cell carcinoma Completed (no results posted) NCT04220866

i.t., intratumorally; i.v., intravenously; s.c., subcutaneously; ND, not described. (Data sources —clinical registration website: www.clinicaltrials.gov)

FIGURE 6.

FIGURE 6

Targeting the cGAS–STING signaling pathway in cancer treatment. Various STING agonists and other forms of therapies targeting the cGAS–STING pathway are depicted. Agonists undergoing clinical trials are indicated with red font, and agonists undergoing preclinical development are indicated with black font. The minus signs in circles above the cell membrane represent negative charge. Figure was created with biorender.com.

Non‐CDN small molecules for STING pathway activation are currently under clinical investigation. E7766, a topologically novel macrocycle‐bridged STING agonist, has been used to treat nonmuscle invasive bladder cancer (NMIBC). 304 Due to insufficient enrolment of participants, this Phase I study was unfortunately terminated in 2020 (clinical trial: NCT04109092). However, another clinical trial, NCT04144140, has since started, aiming to determine the safety profile and efficacy of E7766 as a single agent for intratumoral administration in patients with advanced solid tumors or lymphomas. Both TAK‐676 and MK‐2118 are small‐molecular STING agonists. Currently, both molecules are undergoing clinical assessment as monotherapies or in combination with pembrolizumab for evaluation of their safety and anticancer effects for advanced/metastatic solid tumors (clinical trials: NCT04420884, NCT03249792). HG381 is a non‐CDN small molecule STING agonist developed on the basis of on DNA‐encoded compound library (DEL) technology and small molecule new drug development platform, currently in clinical Phase I (clinical trial: NCT04998422) but no relevant clinical and preclinical data have been released. Another non‐CDN small molecule compound is KL340399, that is presently in Phase I clinical study for the treatment of advanced solid tumor indications through intravenous injection and for superficial or locally advanced, recurrent or metastatic solid tumors. This compound can be administered with the aid of medical imaging instruments via intratumoral injection (clinical trial: NCT05387928).

Multiple ADCs have been developed as therapeutic modalities for cancer with enhanced selectivity to tumor cells and reduced toxicity to normal cells. 305 Inspired by the promising results of the synthetic STING agonist TAK‐676 in Phase 1 clinical trials (NCT04420884, NCT04879849), researchers have designed a novel STING agonist termed TAK500 by linking TAK‐647 to a cysteine–cysteine chemokine receptor type 2 (CCR2) antibody via a protease‐cleavable peptide. 306 , 307 When incorporated into tumor‐infiltrating CCR2+ myeloid cells, this may lead to the activation of STING/IFN signaling and remodel the TME, thereby eliciting antitumor immunity. Currently, TAK‐500 is being evaluated in a Phase I trial for safety, tolerability, and anticancer efficiency, and investigation of its combination with the anti‐PD‐1 blockade antibody for the treatment of late‐stage cancer patients is ongoing (clinical trial: NCT05070247). Another example is the combination of XMT‐2056 (a small‐molecular molecule) and trastuzumab (a mAb targeting human epidermal growth factor receptor [HER] 2 receptor) for boosting host immunity against HER2‐expressing solid tumors; XMT‐2056 has been granted orphan drug status for patients with gastric cancer. A clinical trial of XMT‐2056 is in progress. 308 , 309

4.2. Targeting the cGAS–STING signaling pathway in cancer preclinical studies

In preclinical studies targeting the cGAS–STING signaling pathway, many CDN surrogates have also been developed to enhance antitumor immune responses. For example, various specific metal ions such as manganese ion (Mn2+) and lanthanides have been observed to activate the STING pathway and that Mn2+ self‐assembled into NPs (CDN‐Mn2+ particles, CMP) in concert with CDN STING agonists can initiate potent antitumor immunity at microdoses. 311 , 312 The same STING activation ability has been observed with europium‐based nanovaccines that are composed of GMP, AMP, and coordinating lanthanides. 313 In addition, more non‐CDN small‐molecule STING agonists are being developed. For example, through high‐throughput in vitro screening, a small molecule named G10 (4‐(2‐chloro‐6‐fluorobenzyl)‐N‐(furan‐2‐ylmethyl)‐3‐oxo‐3,4‐dihydro‐2H‐benzo[b][1,4]thiazine‐6‐carboxamide) were screened and categorized as hSTING‐specific agonists that can activate the IFN response via IRF3 in a dependent manner. 314 , 315 Likewise, by using engineered human liver cells HepAD38 in a bioassay that permits the large‐scale screening of compound libraries for activation of cGAS–STING pathway, a bis‐spirocyclic diketopiperazine compound that induces cytokine responses in a manner dependent on functional human STING expression. 316 It has been reported that α‐mangostin, which bears the xanthone skeleton, could also serve as a human STING (hSTING) agonist with the ability to remodel human monocyte‐derived M2 macrophages toward the M1 phenotype. 317 , 318 Similar hSTING specificity was recently observed with bicyclic benzamides and benzothiophene derivatives bearing a benzamide framework and abilities to e ability to inhibit tumor growth by anti‐immunity induced by STING‐activation. 319 Another novel STING agonist can be linked to two amino‐benzimidazole (ABZI) compound (diABZI), which are two symmetry‐related links two symmetry‐based, has the ability to bind with STING protein and exhibits potent and lasted antitumor immunity when injected into syngeneic colon cancer mouse models. 320 , 321

Among those being investigated extensively for preclinical characterization are summarized in Table 4, including bacterial vectors (such as SYNB1891 and STACT‐TREX‐1), ectonucleotide pyrophosphatase/phosphodiesterase (ENPP1) inhibitors, s‐acylthioalkyl ester (SATE)‐based STING‐activation prodrugs selenium‐containing chemicals based on the structure of benzothiophene oxobutanoic acid (MSA‐2), newly developed 7‐(het)aryl 7‐deazapurine CDNs, small‐molecule STING agonist include SR‐001, SR‐012, SR‐717, SR‐301 and SR‐717, as well as novel compound named HB3089, and so on. 322 , 323 , 324 , 325 , 326 These preclinical small molecular STING agonists have been recently extensively reviewed by Garland et al., 327 and will therefore not be discussed in detail in this review.

TABLE 4.

Preclinical studies on STING‐targeting compounds.

Agents Types Target Route of delivery Cancer models Cytokines or chemokines Immune cells References
GSK532 CDN

hSTING

and STING orthologs from cyno, minipig, dog, rat, and mice

i.t. CT26 murine tumor model
  • TNF‐α

  • IL‐6

ND 328
JNJ‐67544412 (JNJ‐4412) CDN

hSTING

mSTING

i.t. Subcutaneous syngeneic murine tumor models
  • IFN‐α

  • IFN‐β

  • IP‐10

  • TNF‐α

  • IL‐6

  • MCP‐1

  • CD8+ T cells

329
3′3′‐cyclic 3′3′‐cAIMP CDN hSTING ND Mutagen diethylnitrosamine (DEN) induced murine hepatocellular carcinoma
  • IFN‐α

  • TNF‐α

  • CCL5

  • CXCL9

  • CXCL10

  • CD3+ T cells

  • CD8+ T cells

330
7‐Deaza Variants CDN

hSTING

ND ND ND ND 324
Ryvu's agonists Non‐CDN small molecule compounds

hSTING

mSTING

i.v. CT26 and EMT6 murine tumor model ND ND 331 , 332
Selvita agonists Non‐CDN small molecule compounds

hSTING

mSTING

Not applicable In vitro
  • IFN‐β

  • TNF‐α

  • DCs

  • Macrophages

333
CRD5500 Non‐CDN small molecule compounds

hSTING

i.t. CT26 murine tumor model
  • IFN‐β

  • TNF‐α

  • DCs

334
i.v. B16F10, PANO2, LL2 and C1498 murine tumor models
  • IFN‐β

  • TNF‐α

  • CXCL10

  • DCs

335
GF3‐002 Non‐CDN small molecule compounds

hSTING

Not applicable In vitro
  • IFN‐β

  • DCs

336
JNJ‐‘6196 Non‐CDN small molecule compounds

hSTING

i.v. Murine tumor models ND
  • DCs

  • Macrophages

  • CD8+ T cells

337
TTI‐10001 Non‐CDN small molecule compounds

hSTING

mSTING

i.t. Multiple syngeneic murine tumor models
  • IFN‐β

  • TNF‐α

  • IL‐6

ND 338

CS‐1018

CS‐1020

CS‐1010

Non‐CDN small molecule compounds

hSTING

mSTING

i.t. B16F10 and MC38 murine tumor models ND ND 339
MSA‐1 Non‐CDN small molecule compounds

hSTING

mSTING

i.t. B16F10, MC38, and CT26 mouse tumor models ND
  • T cells

  • memory T cells

340
MSA‐2 Non‐CDN small molecule compounds

hSTING

Oral administration B16F10, MC38, LL2, and CT26 murine tumor models ND ND 326
VB‐85247 Macrocyclic STING agonists

hSTING

mSTING

Bladder instillation MB49 and NMIBC mouse tumor model
  • type I IFN

  • DCs

341
ALG‐031048 Novel STING agonist, Similar to 2'‐3'‐cGAMP

hSTING

mSTING

i.t.

s.c.

CT26, B16F10, and Hepa1‐6 mouse tumor models
  • IFN‐β

  • CXCL10

  • Macrophages

342
SR‐8541A ENPP1 inhibitor ENPP1 Not applicable In vitro 3D spheroid models and human breast cancer cell line derived organoids; CT‐26 and EMT‐6 mouse models
  • IFN‐β

  • CXCL10

  • NK cells

  • DCs

  • CD3+ T cells

  • CD8+ T cells

343 , 344
SR‐8314 ENPP1 inhibitor ENPP1 i.p. Syngeneic murine tumor model
  • IFN‐β

  • CXCL10

  • CD3+ T cells

  • CD4+/CD8+ T cells

345
MV‐626 ENPP1 inhibitor ENPP1 i.p. Panc02‐SIY and MC38 murine tumor models ND ND 346
AVA‐NP‐695 ENPP1 inhibitor ENPP1 ND 4T1 syngeneic tumor model ND ND 347
ZX‐8177 ENPP1 inhibitor ENPP1 i.p. CT‐26, Panc02 and MC38 murine tumor models
  • IFN‐β

  • IFN‐α

  • CCL5

  • CXCL10

  • Granzyme B+CD8+ T cells

  • NK cells

  • macrophages

348 , 349
LCB33 ENPP1 inhibitor ENPP1 Oral administration CT26 murine tumor model
  • type I IFN

  • T cells

  • Immune cells (not specified)

350
ZXP‐8202 ENPP1 inhibitor ENPP1 ND CT26 murine tumor model
  • IFN‐β

ND 351
SB02024 VPS34 inhibitor VPS34 Oral administration B16F10 mouse tumor model ND ND 352
JAB‐X1800 CD73‐STING iADC

hSTING

mSTING

ND MDA‐MB‐231 xenograft mouse tumor model and hCD73‐MC38 syngeneic mouse tumor model
  • IFN‐β

ND 353
FcγR specific STING‐agonist ADCs FcγR‐STING ADC ND Not applicable In vitro ND FcγRI+ myeloid cells 354
pHLIP‐STINGa pHLIPs (pH‐Low Insertion Peptides) (Var3) linked to a STINGa (diABZI) via a self‐immolating linker hSTING

i.p.

i.v.

CT26, and B16F10 mouse tumor models ND
  • Macrophages

  • Cancer‐associated fibroblasts (CAFs)

  • Myeloid‐derived suppressor cells

  • DCs

  • T cells

  • NK cells

  • Memory T cells

355
ST317 Peptide

hSTING

mSTING

i.v. Syngeneic mouse models
  • type I IFN

  • CXCL10

ND 356
rBCG‐ disA ‐OE Recombinant Bacillus‐Calmette Guerin (BCG) overexpressing CDN

hSTING

mSTING

ND Nonmuscle invasive bladder cancer mouse model
  • TNF‐α

  • IL‐6

  • IL‐1β

  • Nos2

  • CCL2

ND 357
STACT‐TREX1 An inhibitory microRNA to TREX1 was designed and introduced into the STACT strain. TREX1 i.v. CT26, MC38, and B16‐F10 mouse tumor model
  • TNFα

  • IL‐6

ND 358
STINGVAX CDN with GM‐CSF‐producing cellular cancer vaccine

hSTING

mSTING

i.v. CT26, Panc02, and B16‐F10 mouse tumor model
  • IFN ‐α

  • IL‐12

  • IFN‐γ

  • IL‐6

  • CCL2

  • IFN‐β

  • CD4+/CD8+ T cells

  • T helper 1 (TH1) cells

330
ONM‐501 pH sensitive polyvalent PC7A micelles loaded with cGAMP

hSTING

mSTING

i.t.

s.c.

Mice, rats, and primates ND ND 359 , 360

i.t., intratumorally; i.v., intravenously; s.c., subcutaneously; i.p., intraperitoneally; ND, not described.

5. LIPID‐BASED NANOMATERIALS FOR DELIVERY OF STING AGONISTS

As discussed above, although multiple STING agonists have shown effective tumor inhibitory efficiency when given intravenously or intratumorally, their clinical translation has not been materialized. The low cytoplasmic delivery rate is due to the hydrophilic and negative electrical properties of exogenous DNA and CDNs, while the rapid immune clearance and noncellular targeting of natural and synthetic CDNs limit the intracellular activation of the STING pathway. 302 Moreover, relatively high concentrations of STING agonists are needed to elicit sufficient STING signaling activation when administrated intravenously, and probably lead to systemic inflammatory toxicity. 18 To achieve optimal clinical feasibility, the response efficiency of STING agonists still needs to be improved. There is growing evidence indicating that appropriate combination therapy regimens based on STING agonists could expand the therapeutic benefits of STING agonists for cancer patients. Another promising method for improving the efficiency of STING agonists is the use of NPs for the delivery of STING agonists, following the rationale of improving the efficacy and safety of intracellular delivery. 18

5.1. Liposomes for STING agonist delivery

Liposomes, especially PEGylated liposomes, as delivery vehicles for STING agonists or other agents capable of activating cGAS–STING pathway can increase their stability, prolong the blood circulation time, enhance tumor targeting, and promote intracellularization, thus maximizing the antitumor immune effect and reducing adverse effects. 19 As described in a study by Koshy et al., 361 24 h after delivery of non‐PEGylated and PEGylated cGAMP liposomes, cGAMP delivered by PEGylated liposomes aggregated in the whole tumor site rather than within small tumor volumes, as was the case for non‐PEGylated liposome delivery; treatment with PEGylated cGAMP liposomes resulted in a 50% tumor ablation and a complete rejection upon tumor rechallenge in the melanoma mouse model, which was shown unresponsive to non‐PEGylated cationic liposome challenge. In addition, by modifying the percentage of PEG surface modification, liposomes can be fine‐tuned to optimize their serum stability and surface charge properties; the cGAMP liposome formulation with 5 mol% PEG induced a better adaptive immune memory response than the formulation with 10 mol% PEG. 361 The rational structural fine‐tuning through modifications to the liposome surface, such as specific antibodies, peptides, or receptor ligands, can also enhance the biological effects of STING agonists. 362 , 363 , 364 , 365 For instance, mannose‐modified liposomes encapsulating STING ligands (2′‐3′‐cGAMP) with a diameter of 70 nm and a surface charge of about 7 mV were able to specifically target APCs (both DCs and macrophages) and showed increased stimulation of the STING pathway. This approach significantly promoted the transition of the immunosuppressive TME towards an immunostimulatory state as compared with treatment with free cGAMP in the V600E BRAF‐mutated (an activating missense mutation in codon 600 of exon 15 (V600E) of the B‐Raf proto‐oncogene [BRAF] gene) melanoma mouse model. 366 Activation of DCs is a prerequisite for induction of antitumor immunity. Doshi et al. 367 reported a CD103+ DC‐targeted STING‐liposomes, which selectively deliver STING agonists (CDN) into DCs, induce robust antitumor immune responses at a low dose, resulting in improved immunotherapy efficiency and reduced side effects in MC38 and B16F10 tumor models. Additionally, Kocabas et al. 368 examined the immunostimulatory capacity and antitumor potential of pH‐sensitive cationic liposomes codelivered with a STING agonist and other immune adjuvants in a mouse model of melanoma. The liposomes, prepared using DOPE, DC‐chol, PEG‐PE, PC, cholesterol, and cGAMP, had a size of approximately 400–700 nm with stable characteristics under neutral conditions and rapidly destabilized properties in acidic environments (lysosome or late endosome). The cGAMP‐loaded formulation, when codelivered with CpG ODN, induced splenocytes to produce more IFN‐α/β than free cGAMP, and significantly inhibited B16 melanoma growth through re‐education of the TME towards a tumor‐suppressive state and promotion of Th1 immune response. 368 In another study, tumor‐penetrating neutrophils cytopharmaceuticals with STING agonists were developed. In brief, they prepared a cationic liposomal STING agonists (DMXAA) coated with negative HA‐maleimide (HA‐Mal) and conjugated these onto the surface of neutrophils, named NEs@STING‐Mal‐NP. 369 The NEs@STING‐Mal‐NP has the ability of penetrating the tumor vascular endothelium and quick STING agonist release. These characteristics endowed NEs@STING‐Mal‐NP with an increased absorption rate by immune cells and tumor cells, resulting in an antitumor TME stated via macrophages and neutrophils phenotype education and T cell infiltration improvement. 369

5.2. CLD NPs for STING agonist delivery

Cationic LNPs appear more promising for realizing the full potential of STING agonists as they may induce stronger immune responses than anionic or neutral liposomes, due to alkalinizing lysosomal pH and limitation of antigen degradation. 370 , 371 , 372 For example, STING agonists delivered by a system composition of a neutral cytidinyl lipid (DNCA) and a CLD enable robust antitumor effectiveness at a lower dosage, because it can easily enter cells and release STING agonists into the cytoplasm. 19 Moreover, Koshy et al. 361 showed that cholesterol‐stabilized LNPs based on DOTAP, with a size of approximately 160 nm, significantly enhanced the cell association and internalization of 2′3′‐cGAMP in bone‐marrow‐derived dendritic cells (BMDCs) compared with free cGAMP, and enhanced the biological effects of cGAMP on activation of the STING pathway. Unfortunately, intratumoral injection of low doses of cGAMP‐loaded LNPs (0.35 μg × 2 doses) did not show superior antitumor effects in an orthotopic B16‐F10 melanoma model due to limited tumor penetration. Therefore, additional efforts are required to increase the stability and tumor penetration of this formulation. 361 A typical strategy was provided by Nakamura et al., 373 who devised a PEGylated STING agonist LNPs based on YSK12‐C4 (an ionizable CLD with high affinity for immune cells), PEG2000‐DMG, cholesterol, and c‐di‐GMP, which is 170 nm in size and has a 7 mV zeta‐potential. This PEGylated cationic LNPs remained stable at 4°C for up to 9 months, maintaining its ability to stimulate IFN‐β production. Upon intravenous injection, the STING‐LNPs treatment induced a superior antitumor effect in a B16‐F10‐luc2 lung metastatic mouse model. 373 Furthermore, Miyabe et al. 374 tried to effectively control the release of the STING agonist c‐di‐GMP at the tumor site in response to changes in pH; they used a synthetic pH‐sensitive YSK05 lipid with high fusogenic properties, together with palmitoyl‐oleoyl‐phosphoethanolamine, 1,2‐dimyristoyl‐sn‐glycerol (DMG)‐PEG2000, and cholesterol to create a liposomal formulation with a size of approximately 170–180 nm and a potential of around −5 mV. This vehicle showed high fusion and endosomal escape ability, thus leading to a superior activation effect when inducing APC activation and maturation, NK‐cell activation, and T‐cell immune response when compared with free delivery or conventional cationic liposome delivery of c‐di‐GMP. Importantly, c‐di‐GMP/YSK05 liposomes carried with tumor antigens (OVA) exhibited significantly enhanced vaccination effects in a lymphoma mouse model compared with free c‐di‐GMP+OVA, and decreased lung metastasis of melanoma in a B16‐OVA mouse model. 374 , 375

With high‐throughput screening using a combinatorial library of liposomal units (cations, linker molecules, and alkyl chains), Miao et al. 376 recently found that cationic LNPs with a cyclic amine moiety, a dihydroimidazole linker molecule, and a structure of unsaturated lipid chains were able to activate the STING pathway, induce systemic cytokine expression, and enhance antitumor efficacy, indicating the potential of LNPs as an effective STING agonist delivery platform. Additionally, the results showed that when LNPs were further loaded with tumor‐associated OVA mRNA, they could be used to effectively deliver mRNA into lymphoid tissues and stimulate the STING pathway, thus producing a potent antitumor immune response and superior tumor growth inhibition in B16F10‐OVA melanoma mice. 377 This work provides a theoretical basis for the codelivery of multiple antigens and STING agonists by LNPs. More follow‐up studies need to be conducted to clarify the full potential of nanoscale mRNA vaccines based on STING agonists. On the other hand, the selectivity and efficiency of the delivery of STING‐LNPs to different types of immune cells and different TME stages can be adjusted by modifying the targeting ligands on the surface of the LNPs. Covarrubias et al. 377 compared the effects of common targeting ligands (the c(RGDfC) peptide for the targeting of αvβ3 integrins, the CDAEWVDVS peptide for the targeting of P‐selectins, and the CREKA peptide for fibrin targeting) on the selectivity and antitumor efficiency of the 60 nm STING‐LNPs containing various PLs: DPPC, DOPC, DSPE–PEG–ligand, and cdGMP. In a 4T1 mouse tumor model, nontargeted STING‐LNPs were found to be more aggregated in primary tumors and sites of late metastasis, while targeted STING‐LNP variants, especially integrin‐targeting cRGD‐LNPs, accumulated more in early metastases. It might also be interesting to employ more specialized targeting ligands (e.g., CCR2, mannose and intercellular adhesion molecule‐1) for LNP‐based cytoplasmic delivery of STING into APCs. 377

5.3. Cellular‐membrane‐derived vesicles for STING agonist delivery

As discussed earlier, EVs, including exosomes, microvesicles, and so on, have been used and engineered as NVs to carry drugs for cancer therapies because of their ideal safety and stability properties. 378 , 379 , 380 The use of such NVs for STING agonist delivery is an attractive approach, because the unique abilities of cellular vesicles inherited from particular source cell types are able to enhance the selectivity and antitumor efficiency of STING agonists. 146 For example, Rao et al. 381 extracted platelet‐derived NVs, M1 macrophage‐derived NVs, and cancer‐cell‐derived NVs overexpressing high‐affinity signal regulatory protein (SIRP)α variants (SαV‐C‐NVs) and formed hybrid cellular membrane NVs, with a size of 100 nm and encapsulating CDNs (hNVs@cGAMP), through repeated extrusion and ultrasound treatments. The hNVs@cGAMP blocked the antitumor inhibition of the CD47 signaling pathway and promoted the repolarization of tumor‐associated macrophages. Upon intravenous administration, hNVs@cGAMP effectively improved the cytosolic delivery of the drug owing to its specific membrane anchoring and transmembrane‐protein‐mediated endocytosis, and exhibited better inhibition of 4T1 breast tumor growth, recurrence, and lung metastasis compared with hNVs + cGAMP. 381 In addition, exosomes derived from HEK293 cells loaded with a small‐molecule STING agonist (named exoSTING), which is designed to allow delivery directly to APCs, were shown a hundredfold more potent than free STING agonists by delivering drugs into the cytosol, overcoming the bell‐shaped pharmacological properties of STING adjuvants and increasing their tumor retention. 382 Of note, exoSTING is the only liposomal STING agonist so far that is being tested in clinical studies (NCT04592484). Related data has been reported in June 2022 and the company Codiak Biosciences plans to take exoSTING into a Phase 2 study for bladder cancer patients next year (Source: Codiak BioSciences, Inc.; https://ir.codiakbio.com/node/7416/pdf). Similarly, engineered EVs derived from HEK293 cells were used to encapsulate STING agonists ranging in size from about 50 to about 200 nm, and this was found to reduce systemic inflammation and enhance the biological ability of the STING agonists. These data showed that EVs containing STING agonists increased DC and monocyte activation in vitro and improved tumor growth suppression in B16F10, CT26, and EG7‐OVA mouse models with a more than 100‐fold increase in potency compared with free CDNs. 383 Furthermore, a dual‐targeting cGAMP@Exos exhibit antitumor immunity and suppressed immune escape through twice activation of DCs (cGAMP and anti‐CD40) as well as the blocking of PD‐L1 on tumor cells. 174

5.4. Other lipid‐based formulations for STING agonist delivery

Lipid nanodiscs (LNDs) consist of disc‐like lipid bilayers stabilized by amphiphilic biomolecules (termed membrane scaffold proteins) and have attracted extensive attention for the delivery of drugs. 384 , 385 , 386 Recently, Dane et al. 387 conjugated CDN prodrugs with PEG‐PL via thiol‐maleimide coupling (CDN–PEG–lipid), which, upon being internalized by cells, released active STING agonists upon peptidase cleavage. Subsequently, CDN‐functionalized LNDs were self‐assembled with CDN–PEG–lipids, PEGylated lipids (PEG5000lipid), and high Tm (melting temperature) PLs. Compared with the conventional liposome–CDN structure with a diameter of approximately 60 nm, the prepared LND–CDN had more uniform and nanoscale sizes (20–30 nm) and unique structural shapes, leading to improved cellular internalization and superior tumor penetration both in vitro and in vivo. The latter was because LND–CDN easily migrated towards the vessel walls in the blood circulation and presented an increased number of binding areas to the endothelium. 388 After intravenous injection, LND–CDN showed a significantly extended plasma half‐life of 12.6 h as compared with the rapid clearance (half‐life around 1 h) of the free soluble drug and the moderate terminal half‐life of liposome–CDN (7.6 h). 388 Furthermore, LND–CDN demonstrated enhanced CDN delivery to tumor cells and was able to boost T‐cell immune responses through DC priming when compared with both no treatment and the liposome–CDN treatment. At a dose 20 times lower than that of the free STING ligand, a 75% tumor ablation rate was observed in the MC‐38 colorectal mouse model after LND–CDN therapy, while it was almost three times higher than that of the liposome–CDN group. 388 Although LND may be a promising platform for the delivery of STING agonists throughout solid tumors, its translation to clinical application still requires further effort and consideration, such as with respect to optimizing LND storage and reducing the cost of synthesis.

Lipidoids, a class of lipid‐like materials, have also been investigated for the delivery of STING agonists. Chen et al. 389 demonstrated that a 93‐O17S/cGAMP lipidoid consisting of 93‐O17S, cholesterol, DOPE, and STING ligand was able to facilitate cGAMP release within the cytoplasm of APCs by means of the endo‐/lysosomal escape effect and enhanced activation of the STING pathway in the APCs. The lipidoid 93‐O17S was chosen because of its cationic properties, which are favorable for STING agonist encapsulation, and high adjuvant ability after immunization in mice. 390 Another high‐density lipoprotein (LV‐sHDL), loaded with a STING agonist (vadimezan) and a multitargeted tyrosine kinase inhibitor (TKI; Lenvatinib), and consisting of DMPC and cholesterol oleate, appeared as spherical NPs with a diameter of 15 nm and a negatively charged ζ‐potential (about −20 mV). 391 This synthetic dual‐drug lipoproteins LV‐sHDL was able to enhance the tumor accumulation of the encapsulated drugs and the transport of the drugs into the cytosol of the cells via scavenger receptor class B type 1 (SR‐B1) receptors, which are consistently highly expressed on breast cancer cells. 392 Moreover, in vivo data have shown the promising effects of LV‐sHDL on primary tumor growth control and the reduction of lung metastasis in an aggressive 4T1 triple‐negative breast cancer mouse model. The combination of this dual‐drug lipoprotein and anti‐PD‐L1 was further shown to increase its antitumor efficiency. 391

The incorporation of Mn2+ or lanthanides and so on into multifunctional lipid‐NPs can significantly promote the activation of STING pathway and anticancer effects. 393 , 394 , 395 For example, Zhu et al. 396 developed a platform based on a manganese‐containing core with a PEG‐modified PL bilayer shell, where Mn2+ has the ability to optimize MRI and activate cGAS–STING pathway. Another interesting article reported that CDN‐Mn2+ coordination polymers with a PEGylated lipid layer (DOPC: cholesterol: DSPE‐PEG5000), with an average size of 80–160 nm and a neutral surface charge, retained the good tolerance and safety of liposomes while achieving drug dose savings and minimal side effects by amplifying the activation of the STING pathway. 312 Compared with soluble CDNs, CDN‐Mn2+ NP significantly increased CDN uptake and cytoplasmic internalization by BMDCs, synergistically promoted STING pathway activation, and increased IFN‐β and TNF‐α secretion. Intratumoral or intravenous injection of lipidated CDN‐Mn2+ NPs significantly enhanced the tumor accumulation of CDNs and promoted NK‐cell and DC activation in the TME, thereby effectively enhancing the antitumor effects of CDNs in B16F10 and NOOC1 tumor‐bearing mice. 312 Amorphous porous manganese phosphate NPs‐based PL‐coated hybrid NPs encapsulating with DOX have shown a similar enhancement of cGAS/STING activities, as well as improvement of antitumor immune responses and therapeutic potential. 397 Furthermore, the Mn2+ combined with radiosensitization (several radio‐sensitizers such as hafnium, gold, and lanthanide elements, and those high‐Z metals) is a viable strategy to promote synergistic antitumor activity. Dai and coworkers 398 reported a novel pH‐responsive lanthanide‐doped radiosensitizer‐based metal‐phenolic network STING agonist named NaGdF4:Nd@NaLuF4@PEG‐polyphenol/Mn (DSPM), which demonstrated a robust anticancer efficiency via an enhanced STING activation in combination with radiotherapy and immunotherapy.

6. LIPOSOMALLY SUPPORTED STING AGONISTS IN COMBINATION WITH OTHER THERAPIES

6.1. Liposomally supported STING agonists combined with radiotherapy or chemotherapy

Some traditional tumor ablation approaches are highly effective partners for STING agonists. Chemotherapy and radiotherapy have been reported to provide sufficient tumor‐specific neoantigens and TAAs to achieve STING‐agonist‐induced antitumor immunity, which allows the whole tumor to be transformed into a vaccine. 399 A previous report by Jinjin and colleagues 389 showed that a 93‐O17S‐F/cGAMP lipidoid promoted antigen presentation of APCs via electrostatic adsorption of DOX‐pretreatment‐induced antigens, thus producing a strong antitumor immune response and increasing survival rates in a B16F10 tumor model, as well as exhibiting excellent immune memory. Notably, DOX and the 93‐O17S‐F/cGAMP lipidoid were administered sequentially in this current system; additional attempts in future studies could be made to explore integrated NP formulations to simplify the handling procedure and improve the therapeutic benefits. 389

A consensus is that chemotherapy‐ or radiotherapy‐induced cell death can be immunogenic; DAMP or PAMP molecules released from tumor cells after treatments can promote DC activation and maturation. 400 Moreover, the functional cytosolic–DNA–sensing STING pathway is required for immunogenic radiotherapy‐mediated antitumor immunity, and targeting cGAS‐STING pathway has shown a capacity to improve the therapeutic efficiency of radiotherapy. 401 , 402 , 403 It is tempting to speculate that STING agonists in combination with radiotherapy or chemotherapy may produce a better synergistic antitumor effect, and Gu et al. 404 indeed showed that the enchantment of antitumor efficiency through liposomal oxaliplatin in combination with localized STING activation. In accordance with these findings, Liu et al. 405 devised a pH‐sensitive liposomal NP encapsulating cGAMP (liposomal‐NPcGAMP), with an average size of around 120 nm and a negative surface charge of about −40 mV. The liposomal NPcGAMP and tumoral APC‐targeted delivery stimulated STING activation, converting the TME from an immunosuppressive state to a proinflammatory state in lung metastases, and improving host sensitivity to radiotherapy. This liposomal NPcGAMP, combined with radiotherapy (8 Gy × 3), induced potent anticancer immune responses, effectively delayed the growth of the primary tumors, and improved survival with a reduced number of metastatic lung foci on each lung in B16‐OVA and 4T1 tumor‐bearing mice. 405 Furthermore, a recent study reported a bio‐liposome composed of intermingled platelet membranes, liposomes with NK‐activatable target antigen (IgG antibodies) and cisplatin pre‐drug. Bioliposome here protected the cisplatin predrug from degradation, enhanced its RES evade and prolonged the half‐life time, as well as improved tumor accumulation of cisplatin predrug. Most interestingly, the liposomal‐activated NK cells can precisely identify and kill the dying tumor cells induced by chemotherapeutic agents, thus leading to an immune‐responsive TME via STING–IFN pathway that further enhanced the immunotherapy effectiveness. 406

6.2. Liposomally supported STING agonists combined with vaccines/immunostimulatory agonists

The liposomally supported neoadjuvant combinatorial strategy is focused on improving and reprogramming APC functions in the TME via the loading of antigens and/or immunostimulatory adjuvants. The latter can activate specific PRRs in the APCs, which play a crucial role in recognizing PAMPs and DAMPs, thereby enhancing the subsequent activation and expansion of naive T cells in the LNs. 407 A PEGylated liposome consisting of DMPC, DOPC, DOPG, DSPE‐PEG, and cdGMP effectively reduced the systemic (blood) distribution of cdGMP and enhanced its accumulation in LNs (especially in APCs) 15‐fold compared with the levels observed 24 h after cdGMP‐free delivery following subcutaneous injection. 408 Moreover, in this study, PEGylated liposomes were used as trans‐adjuvants for tumor antigens (OVA) through simultaneous delivery rather than encapsulation in one platform, showing superior synergistic cellular and humoral immune effects to antigen vaccine versus free soluble drugs against cancer. 408 Atukorale et al. 400 synthesized a dual immunostimulatory agonist (monophosphoryl‐Lipid A [MPLA] and cdGMP)‐encapsulating neutral LNPs (referred to as immuno‐NPs), with a size of 50 nm, consisting of DOPC, DPPC, 3 mol% mPEG2000‐DSPE, and chloroform. MPLA is a TLR4 agonist that is currently used as a vaccine adjuvant due to its potent modulation of innate immunity. 409 On the one hand, this systemic liposomal delivery promoted selective and increased uptake of agonists by tumor‐resident APCs in a variety of aggressive mouse tumor models (B16F10, 4T1, and D2.A1), resulting in improved potency and safety profiles of the agents compared with free agonist delivery. On the other hand, however, the dual‐agonist immuno‐NPs increased production of IFN‐β (75‐fold) and TNFα (16‐fold) by APCs compared with single‐agonist NPs as a result of the synergistic effect on the activation of innate immunity through nonoverlapping PRR pathways (the STING pathway and the TLR4 pathway, respectively). 400 The combination of STING agonists and other immunomodulatory adjuvants through liposomal recipes can induce a more potent innate immune response and re‐educate the immunosuppressive TME. 400 These findings indicate the effectiveness and safety of liposomal vaccines based on STING agonists, with special potential for the future translation of human cancer vaccines.

6.3. Liposomally supported STING agonists combined with mAbs or ICIs

Despite that single STING therapy showed unsatisfactory therapeutic results, combining STING agonists with ICI therapies have shown great potential for inducing synergistic therapeutic responses (Table 3). 410 The basic concept of STING‐agonist‐based combination strategies is to transform the TME from a noninflammatory, immune‐excluded “cold” state to an inflamed, immunostimulatory “hot” state by enhancing the antitumor responses in the cancer‐immune cycle or reducing the evasive effect towards immune defenses. 411 , 412 STING and Type I IFN play a key role in enhancing the cross‐presentation by DCs and subsequential T cell migration and chemotaxis. Importantly, adequate adaptive immune cells, in particular CTLs, are a requisite for effective ICI. In addition, STING pathway‐activation‐mediated IFN secretion can alter tumor immunogenicity by upregulating PD‐L1, IDO, MHC I and calreticulin in tumor cells. 413 , 414 , 415 However, it is imperative to acknowledge that while STING plays a beneficial role in the immune response against cancer, its activation inappropriately may hinder the antitumor immune response. 416 Upregulated PD‐L1 or IDO expression may lead to immunosuppression and STING treatment failure, especially those with strong tolerogenic responses to DNA and low tumor antigenicity. 417 Thus, the combination of STING agonists and ICIs is a promising avenue to explore to improve monotherapy in multiple ICI resistance models. 418 , 419 For example, combination therapy of STING agonist and anti‐transforming growth factor β/PD‐L1 bispecific antibody YM101 produced robust innate and adaptive antitumor responses in immunotherapy‐resistant cancer models by recruiting immune cells and stimulating their function. 418

Liposome‐based STING agonists in combination with ICI therapy achieve better anticancer effects by trafficking diverse immune cells into the tumor, reverting the immunosuppressive microenvironment, and overcoming ICI resistance with negligible systemic side effects. 373 , 377 , 400 , 420 , 421 , 422 For instance, the previously described cationic liposome composed by Nakamura et al. 373 was able to overcome anti‐PD‐1 resistance in mice with metastatic melanoma by increasing the expression of PD‐L1 in cancer cells. This increase in PD‐L1 in cancer cells was due to the production of IFN‐γ by activated systemic NK cells, which were initiated from liver macrophages containing the internalized STING liposomes. 373 Similarly, a synergistic antitumor effect and improved curative responses were observed in B16F10‐ and BRAF‐mutated murine melanoma models with liposome‐based STING‐PD‐L1 combination therapy, emphasizing the critical role of “exhausted” tumor‐infiltrating CD8+ T cells in antitumor action. 421 Furthermore, a simultaneous combination treatment of anti‐PD‐1 and anti‐CTLA‐4 with STING‐agonist‐containing liposomes effectively reduced the number of pulmonary metastatic nodules in a mouse melanoma lung metastasis model, whereas free cGAMP + anti‐PD‐1 + anti‐CTLA‐4 did not. 420 Related to this, the delivery protocol needs to be carefully designed, setting parameters of interval time and treatment cycle for STING‐LNP and ICI therapy in order to achieve the best antitumor effects. 423 Additional studies will be needed to analyze whether the liposomal formulation as a delivery platform for combinational STING therapies can be expanded to other therapeutic approaches that are aimed at amplifying T‐cell function, such as agonistic mAbs against various receptors (e.g., 4‐1BB (CD137), OX40 (CD134), CD28, CD27, etc.), cytokines (IL‐2 and IL‐12), and inhibitors of other immunosuppressive molecules (LAG‐3, T‐cell Ig and mucin‐domain containing‐3, IDO). Taken together, future studies need to explore if liposomal‐delivery‐based combination therapies rather than STING monotherapies can achieve improved therapeutic efficiency.

7. CONCLUDING REMARKS AND FUTURE OUTLOOK

In this review, we summarized the latest concepts of various types of lipid‐based nanomaterials for cancer vaccination and TME immunomodulation. Ideal lipid‐NP formulations for immunopharmaceuticals must possess characteristics such as biocompatibility, biodegradability, nonimmunogenicity, and targeted delivery to the TME. Lipid‐NPs‐supported cancer vaccines are more effective than “free” drugs to produce a longer‐lasting and broader immune response, inhibit tumor growth and prevent tumor metastasis or recurrence. In addition, TME modulation supported by lipid‐NPs offers more possibilities to remodel the immune profile of TME and assist tumor‐infiltrating immune cells to be effective within the tumor. Specifically, lipid‐NPs act as material carriers effectively protects immunotherapeutic proteins and nucleic acids from degradation, increase the targeting and bioavailability of encapsulated cytokines and genes, achieve spatiotemporally controlled release of immunotherapeutic agents, and effectively reduce systemic side effects of mAbs. In addition, lipid‐NPs are generally considered to be more safe and less immunogenic compared with other delivery vehicles such as polymeric NPs and inorganic NPs. This can be explained by their similarity in composition to cell membranes, which makes them more “natural” to the immune system and less likely to elicit significant immune responses. Therefore, the introduction of lipid‐NPs can amplify immunotherapeutic efficacy while overcoming the current drawbacks of immunotherapy (e.g., poor delivery efficiency, lack of tumor targeting, and severe side effects) to a certain extent. The above‐mentioned advantages have made lipid‐NPs as drug carriers more attractive for cancer immunotherapy.

Furthermore, the STING pathway, which serves as a sensor for initiating the innate immune response, has been identified as a promising target in the treatment of cancer. More basic research with the aim of more deeply understanding the mechanisms of STING signaling and its interaction with TME‐associated factors is necessary to amplify the anticancer potential of STING pathway targeting. It is of note that STING pathway‐activation‐induced local inflammation and antitumor immunity contribute to tumor control in most cases. Still, it is unclear whether chronic inflammation due to long‐term STING agonist exposure promotes tumor growth and metastasis, indicating the importance of balance between STING‐agonist‐induced immune response and inflammation. Therefore, a comprehensive understanding of the biphasic role of STING pathway activation in curbing cancer progression is necessary, and clearer treatment protocols and availability of patient‐selective biomarkers for individual tumor types able to indicate which those patients for whom STING activation will be therapeutically beneficial are still needed.

By discussing the application and potential of lipid‐NPs for STING agonists, we summarized the future perspectives focused on the following points to further promote the application of lipid‐NPs in cancer immunotherapy: (1) in certain instances, lipid‐NPs may trigger an inflammatory response, particularly when CLDs interact with serum proteins and the degradation of biomaterials like LPHNPs. Thus, it is crucial to comprehend the regulation of toxicity and inflammatory responses of lipid‐NPs on immune cells and to enhance immunotherapeutic efficacy and prevent immunotoxicity. (2) The highly heterogeneous and changeable TME is another major challenge that needs to be overcome. Thus, depending on the patient's immune status and tumor characteristics, it would be informative to rationally design lipid‐NPs to carry patient‐specific antigens, appropriate immunotherapeutic agents or combinations thereof; as well as optimize the lipid‐NP parameters (e.g., material composition, geometry, surface chemistry, charge, and mechanical properties) to achieve higher encapsulation rates and controlled release abilities and prompt the lipid‐NPs to cross biological obstacles. (3) Nanopharmaceutical formulations depending on EPR effects remain insignificant effectiveness and exists a potential risk of off‐target effects. Therefore, there should be continued exploration of specific cell–drug interactions in TME to improve the target effects of lipid‐NPs. The combination of immune lipid‐NPs with ablative therapies (chemotherapy, PDT, and radiation therapy) that transiently enhance the permeability of tumor tissue should be strengthened for the enhancement of the intratumor accumulation of NPs to synergistically augment anticancer efficacy. (4) There exists an urgent requirement to continuously identify tumor‐specific and immunogenic neoantigens, innovate and improve existing immunotherapeutic modalities within the domain of fundamental medicine and biology. (5) Further research domain of immune lipid‐NPs can be all‐in‐one platform for diagnosis, treatment and real‐time monitoring through the integration of appropriate monitoring biosensors, enabling the adjustment of drug dosage as necessary. (6) Accurate drug release control and multifunction of complex NPs structures are required, leading to expensive assembly costs for scale‐up production and slow approval from US FDA. Thus, the ratio between efficacy gains and overall cost‐effectiveness including preparation and storage maintenance of the lipid‐NPs must be evaluated. Additionally, programmed preparation processes remain a crucial aspect to be addressed remains a crucial aspect to be addressed to ensure sufficient quantities for clinical applications and consistency between production batches and the safety and quality control of LNP manufacturing.

In conclusion, research on lipid‐NPs‐based cancer immunotherapy is flourishing but there are still some challenges remaining for transition of lipid‐NPs from experimental studies to clinical applications. This review discusses the characteristics of lipid‐NPs subtypes, their applications, as well as their specific strengths and weaknesses. It also takes STING agonists as an example to illustrate the utilization and obstacles of lipid‐NPs in the field, underscoring the imperative for additional investigation to effectively regulate and refine the manipulation of these NPs for improved cancer immunotherapy.

AUTHOR CONTRIBUTION

Conceptualization: H. Y.; writing—original draft preparation: H. Y.; writing—review and editing: H. Y., Z. G., Z. H. Z., and W. D. R.; figure—original draft preparation: H. Y. and J. Z. H.; figure—visualization: H. Y.; supervision: W. D. X. and P. t. D.; final editing: P. t. D. All authors have read and approved the final manuscript.

CONFLICT OF INTEREST STATEMENT

The authors declare no conflict of interest.

ETHICS STATEMENT

Not applicable.

ACKNOWLEDGMENTS

H. Y. was funded by Jilin Province Chinese Postdoctoral International Exchange Program (Grant No. YJ20220406).

Hao Y, Ji Z, Zhou H, et al. Lipid‐based nanoparticles as drug delivery systems for cancer immunotherapy. MedComm. 2023;4:e339. 10.1002/mco2.339

Contributor Information

Dongxu Wang, Email: wang_dong_xu@jlu.edu.cn.

Peter ten Dijke, Email: p.ten_dijke@lumc.nl.

DATA AVAILABILITY STATEMENT

Not applicable.

REFERENCES

  • 1. Waldman AD, Fritz JM, Lenardo MJ. A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat Rev Immunol. 2020;20(11):651‐668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Arruebo M, Vilaboa N, Saez‐Gutierrez B, et al. Assessment of the evolution of cancer treatment therapies. Cancers (Basel). 2011;3(3):3279‐3330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Riley RS, June CH, Langer R, Mitchell MJ. Delivery technologies for cancer immunotherapy. Nat Rev Drug Discov. 2019;18(3):175‐196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Jeevanandam J, Barhoum A, Chan YS, Dufresne A, Danquah MK. Review on nanoparticles and nanostructured materials: history, sources, toxicity and regulations. Beilstein J Nanotechnol. 2018;9:1050‐1074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Mitchell MJ, Billingsley MM, Haley RM, Wechsler ME, Peppas NA, Langer R. Engineering precision nanoparticles for drug delivery. Nat Rev Drug Discov. 2021;20(2):101‐124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Kundranda MN, Niu J. Albumin‐bound paclitaxel in solid tumors: clinical development and future directions. Drug Des Devel Ther. 2015;9:3767‐3777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Ranade AA, Joshi DA, Phadke GK, et al. Clinical and economic implications of the use of nanoparticle paclitaxel (Nanoxel) in India. Ann Oncol. 2013;24(Suppl 5):v6‐12. [DOI] [PubMed] [Google Scholar]
  • 8. Kim TY, Kim DW, Chung JY, et al. Phase I and pharmacokinetic study of Genexol‐PM, a cremophor‐free, polymeric micelle‐formulated paclitaxel, in patients with advanced malignancies. Clin Cancer Res. 2004;10(11):3708‐3716. [DOI] [PubMed] [Google Scholar]
  • 9. Northfelt DW, Allred JB, Liu H, et al. Phase 2 trial of paclitaxel polyglumex with capecitabine for metastatic breast cancer. Am J Clin Oncol. 2014;37(2):167‐171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Bernabeu E, Cagel M, Lagomarsino E, Moretton M, Chiappetta DA. Paclitaxel: what has been done and the challenges remain ahead. Int J Pharm. 2017;526(1‐2):474‐495. [DOI] [PubMed] [Google Scholar]
  • 11. Bobo D, Robinson KJ, Islam J, Thurecht KJ, SR Corrie. Nanoparticle‐based medicines: a review of FDA‐approved materials and clinical trials to date. Pharm Res. 2016;33(10):2373‐2387. [DOI] [PubMed] [Google Scholar]
  • 12. Anselmo AC, Mitragotri S. Nanoparticles in the clinic: an update. Bioeng Transl Med. 2019;4(3):e10143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Gabizon A, Goren D, Cohen R, Barenholz Y. Development of liposomal anthracyclines: from basics to clinical applications. J Control Release. 1998;53(1‐3):275‐279. [DOI] [PubMed] [Google Scholar]
  • 14. Mogensen TH. Pathogen recognition and inflammatory signaling in innate immune defenses. Clin Microbiol Rev. 2009;22(2):240‐273, Table of Contents. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Kumar H, Kawai T, Akira S. Pathogen recognition by the innate immune system. Int Rev Immunol. 2011;30(1):16‐34. [DOI] [PubMed] [Google Scholar]
  • 16. Flood BA, Higgs EF, Li S, Luke JJ, Gajewski TF. STING pathway agonism as a cancer therapeutic. Immunol Rev. 2019;290(1):24‐38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Kwon J, Bakhoum SF. The cytosolic DNA‐sensing cGAS‐STING pathway in cancer. Cancer Discov. 2020;10(1):26‐39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Wehbe M, Wang‐Bishop L, Becker KW, et al. Nanoparticle delivery improves the pharmacokinetic properties of cyclic dinucleotide STING agonists to open a therapeutic window for intravenous administration. J Control Release. 2021;330:1118‐1129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Hofheinz RD, Gnad‐Vogt SU, Beyer U, Hochhaus A. Liposomal encapsulated anti‐cancer drugs. Anti‐Cancer Drug. 2005;16(7):691‐707. [DOI] [PubMed] [Google Scholar]
  • 20. Immordino ML, Dosio F, Cattel L. Stealth liposomes: review of the basic science, rationale, and clinical applications, existing and potential. Int J Nanomedicine. 2006;1(3):297‐315. [PMC free article] [PubMed] [Google Scholar]
  • 21. Lowery A, Onishko H, Hallahan DE, Han Z. Tumor‐targeted delivery of liposome‐encapsulated doxorubicin by use of a peptide that selectively binds to irradiated tumors. J Control Release. 2011;150(1):117‐124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Nakamura Y, Mochida A, Choyke PL, Kobayashi H. Nanodrug delivery: is the enhanced permeability and retention effect sufficient for curing cancer? Bioconjug Chem. 2016;27(10):2225‐2238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Attia MF, Anton N, Wallyn J, Omran Z, Vandamme TF. An overview of active and passive targeting strategies to improve the nanocarriers efficiency to tumour sites. J Pharm Pharmacol. 2019;71(8):1185‐1198. [DOI] [PubMed] [Google Scholar]
  • 24. Gu Z, Da Silva CG, Van der Maaden K, Ossendorp F, Cruz LJ. Liposome‐based drug delivery systems in cancer immunotherapy. Pharmaceutics. 2020;12(11):1054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Ou W, Jiang L, Gu Y, et al. Regulatory T cells tailored with pH‐responsive liposomes shape an immuno‐antitumor milieu against tumors. ACS Appl Mater Interfaces. 2019;11(40):36333‐36346. [DOI] [PubMed] [Google Scholar]
  • 26. Yuba E, Uesugi S, Yamaguchi A, Harada A, Kono K. pH‐sensitive polysaccharide derivatives‐modified liposomes as antigen delivery vehicles for cancer immunotherapy. J Control Release. 2015;213:e30. [DOI] [PubMed] [Google Scholar]
  • 27. Yuba E, Yamaguchi A, Yoshizaki Y, Harada A, Kono K. Bioactive polysaccharide‐based pH‐sensitive polymers for cytoplasmic delivery of antigen and activation of antigen‐specific immunity. Biomaterials. 2017;120:32‐45. [DOI] [PubMed] [Google Scholar]
  • 28. Li X, Luo Y, Huang Z, Wang Y, Wu J, Zhou S. Multifunctional liposomes remodeling tumor immune microenvironment for tumor chemoimmunotherapy. Small Methods. 2023;7:e2201327. [DOI] [PubMed] [Google Scholar]
  • 29. Sharma A, Sharma US. Liposomes in drug delivery: progress and limitations. Int J Pharmaceut. 1997;154(2):123‐140. [Google Scholar]
  • 30. Nakhaei P, Margiana R, Bokov DO, et al. Liposomes: structure, biomedical applications, and stability parameters with emphasis on cholesterol. Front Bioeng Biotechnol. 2021;9:705886. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 31. Patra JK, Das G, Fraceto LF, et al. Nano based drug delivery systems: recent developments and future prospects. J Nanobiotechnology. 2018;16(1):71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Tenchov R, Bird R, Curtze AE, Zhou Q. Lipid nanoparticles horizontal line from liposomes to mRNA vaccine delivery, a landscape of research diversity and advancement. ACS Nano. 2021;15(11):16982‐17015. [DOI] [PubMed] [Google Scholar]
  • 33. Akbarzadeh A, Rezaei‐Sadabady R, Davaran S, et al. Liposome: classification, preparation, and applications. Nanoscale Res Lett. 2013;8(1):102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Has C, Sunthar P. A comprehensive review on recent preparation techniques of liposomes. J Liposome Res. 2020;30(4):336‐365. [DOI] [PubMed] [Google Scholar]
  • 35. Patil YP, Jadhav S. Novel methods for liposome preparation. Chem Phys Lipids. 2014;177:8‐18. [DOI] [PubMed] [Google Scholar]
  • 36. Koynova R, Tenchov B. Recent progress in liposome production, relevance to drug delivery and nanomedicine. Recent Pat Nanotechnol. 2015;9(2):86‐93. [DOI] [PubMed] [Google Scholar]
  • 37. Yingchoncharoen P, Kalinowski DS, Richardson DR. Lipid‐based drug delivery systems in cancer therapy: what is available and what is yet to come. Pharmacol Rev. 2016;68(3):701‐787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Large DE, Abdelmessih RG, Fink EA, Auguste DT. Liposome composition in drug delivery design, synthesis, characterization, and clinical application. Adv Drug Deliv Rev. 2021;176:113851. [DOI] [PubMed] [Google Scholar]
  • 39. Liu P, Chen G, Zhang J. A review of liposomes as a drug delivery system: current status of approved products, regulatory environments, and future perspectives. Molecules. 2022;27(4):1372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Shah S, Dhawan V, Holm R, Nagarsenker MS, Perrie Y. Liposomes: advancements and innovation in the manufacturing process. Adv Drug Deliv Rev. 2020;154‐155:102‐122. [DOI] [PubMed] [Google Scholar]
  • 41. Kohli AG, Kierstead PH, Venditto VJ, Walsh CL, Szoka FC. Designer lipids for drug delivery: from heads to tails. J Control Release. 2014;190:274‐287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Samad A, Sultana Y, Aqil M. Liposomal drug delivery systems: an update review. Curr Drug Deliv. 2007;4(4):297‐305. [DOI] [PubMed] [Google Scholar]
  • 43. Vemuri S, Rhodes CT. Preparation and characterization of liposomes as therapeutic delivery systems: a review. Pharm Acta Helv. 1995;70(2):95‐111. [DOI] [PubMed] [Google Scholar]
  • 44. Monteiro N, Martins A, Reis RL, Neves NM. Liposomes in tissue engineering and regenerative medicine. J R Soc Interface. 2014;11(101):20140459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Hou X, Zaks T, Langer R, Dong Y. Lipid nanoparticles for mRNA delivery. Nat Rev Mater. 2021;6(12):1078‐1094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. van Hoogevest P, Wendel A. The use of natural and synthetic phospholipids as pharmaceutical excipients. Eur J Lipid Sci Technol. 2014;116(9):1088‐1107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Silva C, Aranda FJ, Ortiz A, Martinez V, Carvajal M, Teruel JA. Molecular aspects of the interaction between plants sterols and DPPC bilayers: an experimental and theoretical approach. J Colloid Interface Sci. 2011;358(1):192‐201. [DOI] [PubMed] [Google Scholar]
  • 48. Zhu Y, Wang A, Zhang S, et al. Paclitaxel‐loaded ginsenoside Rg3 liposomes for drug‐resistant cancer therapy by dual targeting of the tumor microenvironment and cancer cells. J Adv Res. 2023;49:159‐173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Xia J, Zhang S, Zhang R, et al. Targeting therapy and tumor microenvironment remodeling of triple‐negative breast cancer by ginsenoside Rg3 based liposomes. J Nanobiotechnol. 2022;20(1):414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Hong C, Liang J, Xia J, et al. One stone four birds: a novel liposomal delivery system multi‐functionalized with ginsenoside Rh2 for tumor targeting therapy. Nanomicro Lett. 2020;12(1):129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Wang H, Zheng Y, Sun Q, et al. Ginsenosides emerging as both bifunctional drugs and nanocarriers for enhanced antitumor therapies. J Nanobiotechnol. 2021;19(1):322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Zhu Y, Liang J, Gao C, et al. Multifunctional ginsenoside Rg3‐based liposomes for glioma targeting therapy. J Control Release. 2021;330:641‐657. [DOI] [PubMed] [Google Scholar]
  • 53. Yu BS, Jo IH. Interaction of sea cucumber saponins with multilamellar liposomes. Chem Biol Interact. 1984;52(2):185‐202. [DOI] [PubMed] [Google Scholar]
  • 54. Tai K, Rappolt M, He X, et al. Effect of beta‐sitosterol on the curcumin‐loaded liposomes: vesicle characteristics, physicochemical stability, in vitro release and bioavailability. Food Chem. 2019;293:92‐102. [DOI] [PubMed] [Google Scholar]
  • 55. Lee DU, Park HW, Lee SC. Comparing the stability of retinol in liposomes with cholesterol, beta‐sitosterol, and stigmasterol. Food Sci Biotechnol. 2021;30(3):389‐394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Lopez C, David‐Briand E, Lollier V, et al. Solubilization of free beta‐sitosterol in milk sphingomyelin and polar lipid vesicles as carriers: structural characterization of the membranes and sphingosome morphology. Food Res Int. 2023;165:112496. [DOI] [PubMed] [Google Scholar]
  • 57. Han C, Yang C, Li X, Liu E, Meng X, Liu B. DHA loaded nanoliposomes stabilized by beta‐sitosterol: preparation, characterization and release in vitro and vivo. Food Chem. 2022;368:130859. [DOI] [PubMed] [Google Scholar]
  • 58. Park S, Kim HK. Development of skin‐permeable flexible liposome using ergosterol esters containing unsaturated fatty acids. Chem Phys Lipids. 2023;250:105270. [DOI] [PubMed] [Google Scholar]
  • 59. Carrer V, Guzman B, Marti M, Alonso C, Coderch L. Lanolin‐based synthetic membranes as percutaneous absorption models for transdermal drug delivery. Pharmaceutics. 2018;10(3):73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Nie Y, Ji L, Ding H, et al. Cholesterol derivatives based charged liposomes for doxorubicin delivery: preparation, in vitro and in vivo characterization. Theranostics. 2012;2(11):1092‐1103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Monpara J, Kanthou C, Tozer GM, Vavia PR. Rational design of cholesterol derivative for improved stability of paclitaxel cationic liposomes. Pharm Res. 2018;35(4):90. [DOI] [PubMed] [Google Scholar]
  • 62. Shafiei M, Ansari MNM, Razak SIA, Khan MUA. A comprehensive review on the applications of exosomes and liposomes in regenerative medicine and tissue engineering. Polymers (Basel). 2021;13(15):2529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Singh S, Vardhan H, Kotla NG, Maddiboyina B, Sharma D, Webster TJ. The role of surfactants in the formulation of elastic liposomal gels containing a synthetic opioid analgesic. Int J Nanomedicine. 2016;11:1475‐1482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Sturm L, Poklar UN. Basic methods for preparation of liposomes and studying their interactions with different compounds, with the emphasis on polyphenols. Int J Mol Sci. 2021;22(12):6547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Cortesi R, Esposito E, Gambarin S, Telloli P, Menegatti E, Nastruzzi C. Preparation of liposomes by reverse‐phase evaporation using alternative organic solvents. J Microencapsul. 1999;16(2):251‐256. [DOI] [PubMed] [Google Scholar]
  • 66. Shaker S, Gardouh AR, Ghorab MM. Factors affecting liposomes particle size prepared by ethanol injection method. Res Pharm Sci. 2017;12(5):346‐352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Deamer DW. Preparation and properties of ether‐injection liposomes. Ann N Y Acad Sci. 1978;308:250‐258. [DOI] [PubMed] [Google Scholar]
  • 68. Schubert R. Liposome preparation by detergent removal. Methods Enzymol. 2003;367:46‐70. [DOI] [PubMed] [Google Scholar]
  • 69. Seltzer SE, Gregoriadis G, Dick R. Evaluation of the dehydration‐rehydration method for production of contrast‐carrying liposomes. Invest Radiol. 1988;23(2):131‐138. [DOI] [PubMed] [Google Scholar]
  • 70. Costa AP, Xu X, Burgess DJ. Freeze‐anneal‐thaw cycling of unilamellar liposomes: effect on encapsulation efficiency. Pharm Res. 2014;31(1):97‐103. [DOI] [PubMed] [Google Scholar]
  • 71. Sur S, Fries AC, Kinzler KW, Zhou S, Vogelstein B. Remote loading of preencapsulated drugs into stealth liposomes. Proc Natl Acad Sci USA. 2014;111(6):2283‐2288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Gubernator J. Active methods of drug loading into liposomes: recent strategies for stable drug entrapment and increased in vivo activity. Expert Opin Drug Deliv. 2011;8(5):565‐580. [DOI] [PubMed] [Google Scholar]
  • 73. Inglut CT, Sorrin AJ, Kuruppu T, et al. Immunological and toxicological considerations for the design of liposomes. Nanomaterials (Basel). 2020;10(2):190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Sercombe L, Veerati T, Moheimani F, Wu SY, Sood AK, Hua S. Advances and challenges of liposome assisted drug delivery. Front Pharmacol. 2015;6:286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Allen TM, Hansen C, Martin F, Redemann C, Yauyoung A. Liposomes containing synthetic lipid derivatives of poly(ethylene glycol) show prolonged circulation half‐lives invivo. Biochim Biophys Acta. 1991;1066(1):29‐36. [DOI] [PubMed] [Google Scholar]
  • 76. Allen C, Dos Santos N, Gallagher R, et al. Controlling the physical behavior and biological performance of liposome formulations through use of surface grafted poly(ethylene glycol). Bioscience Rep. 2002;22(2):225‐250. [DOI] [PubMed] [Google Scholar]
  • 77. Zhang Y, Li N, Suh H, Irvine DJ. Nanoparticle anchoring targets immune agonists to tumors enabling anti‐cancer immunity without systemic toxicity. Nat Commun. 2018;9(1):6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Kwong B, Gai SA, Elkhader J, Wittrup KD, Irvine DJ. Localized immunotherapy via liposome‐anchored Anti‐CD137 + IL‐2 prevents lethal toxicity and elicits local and systemic antitumor immunity. Cancer Res. 2013;73(5):1547‐1558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Caliceti P, Veronese FM. Pharmacokinetic and biodistribution properties of poly(ethylene glycol)‐protein conjugates. Adv Drug Deliver Rev. 2003;55(10):1261‐1277. [DOI] [PubMed] [Google Scholar]
  • 80. Weissmann G, Bloomgarden D, Kaplan R, et al. A general method for the introduction of enzymes, by means of immunoglobulin‐coated liposomes, into lysosomes of deficient cells. Proc Natl Acad Sci USA. 1975;72(1):88‐92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Iyer AK, Su Y, Feng J, et al. The effect of internalizing human single chain antibody fragment on liposome targeting to epithelioid and sarcomatoid mesothelioma. Biomaterials. 2011;32(10):2605‐2613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Guo C, Chen Y, Gao W, et al. Liposomal nanoparticles carrying anti‐IL6R antibody to the tumour microenvironment inhibit metastasis in two molecular subtypes of breast cancer mouse models. Theranostics. 2017;7(3):775‐788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Lee Y, Thompson DH. Stimuli‐responsive liposomes for drug delivery. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2017;9(5). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Zangabad PS, Mirkiani S, Shahsavari S, et al. Stimulus‐responsive liposomes as smart nanoplatforms for drug delivery applications. Nanotechnol Rev. 2018;7(1):95‐122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Yuba E, Tajima N, Yoshizaki Y, Harada A, Hayashi H, Kono K. Dextran derivative‐based pH‐sensitive liposomes for cancer immunotherapy. Biomaterials. 2014;35(9):3091‐3101. [DOI] [PubMed] [Google Scholar]
  • 86. Yanagihara S, Kasho N, Sasaki K, et al. pH‐Sensitive branched beta‐glucan‐modified liposomes for activation of antigen presenting cells and induction of antitumor immunity. J Mater Chem B. 2021;9(37):7713‐7724. [DOI] [PubMed] [Google Scholar]
  • 87. Rahim MA, Jan N, Khan S, et al. Recent advancements in stimuli responsive drug delivery platforms for active and passive cancer targeting. Cancers (Basel). 2021;13(4):670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Liu Y, Chen XG, Yang PP, Qiao ZY, Wang H. Tumor microenvironmental pH and enzyme dual responsive polymer‐liposomes for synergistic treatment of cancer immuno‐chemotherapy. Biomacromolecules. 2019;20(2):882‐892. [DOI] [PubMed] [Google Scholar]
  • 89. Fan Y, Marioli M, Zhang K. Analytical characterization of liposomes and other lipid nanoparticles for drug delivery. J Pharm Biomed Anal. 2021;192:113642. [DOI] [PubMed] [Google Scholar]
  • 90. Wang C, Zhang Y, Dong Y. Lipid nanoparticle‐mRNA formulations for therapeutic applications. Acc Chem Res. 2021;54(23):4283‐4293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Mohamed K, Rzymski P, Islam MS, et al. COVID‐19 vaccinations: the unknowns, challenges, and hopes. J Med Virol. 2022;94(4):1336‐1349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Khan I, Ahmed Z, Sarwar A, Jamil A, Anwer F. The potential vaccine component for COVID‐19: a comprehensive review of global vaccine development efforts. Cureus. 2020;12(6):e8871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Li Y, Tenchov R, Smoot J, Liu C, Watkins S, Zhou Q. A comprehensive review of the global efforts on COVID‐19 vaccine development. ACS Cent Sci. 2021;7(4):512‐533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Sabnis S, Kumarasinghe ES, Salerno T, et al. A novel amino lipid series for mRNA delivery: improved endosomal escape and sustained pharmacology and safety in non‐human primates. Mol Ther. 2018;26(6):1509‐1519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Schoenmaker L, Witzigmann D, Kulkarni JA, et al. mRNA‐lipid nanoparticle COVID‐19 vaccines: structure and stability. Int J Pharm. 2021;601:120586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Miao L, Zhang Y, Huang L. mRNA vaccine for cancer immunotherapy. Mol Cancer. 2021;20(1):41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Kon E, Elia U, Peer D. Principles for designing an optimal mRNA lipid nanoparticle vaccine. Curr Opin Biotechnol. 2022;73:329‐336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Kulkarni JA, Darjuan MM, Mercer JE, et al. On the formation and morphology of lipid nanoparticles containing ionizable cationic lipids and siRNA. ACS Nano. 2018;12(5):4787‐4795. [DOI] [PubMed] [Google Scholar]
  • 99. Samaridou E, Heyes J, Lutwyche P. Lipid nanoparticles for nucleic acid delivery: current perspectives. Adv Drug Deliv Rev. 2020;154‐155:37‐63. [DOI] [PubMed] [Google Scholar]
  • 100. Liu S, Cheng Q, Wei T, et al. Membrane‐destabilizing ionizable phospholipids for organ‐selective mRNA delivery and CRISPR‐Cas gene editing. Nat Mater. 2021;20(5):701‐710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Semple SC, Akinc A, Chen J, et al. Rational design of cationic lipids for siRNA delivery. Nat Biotechnol. 2010;28(2):172‐176. [DOI] [PubMed] [Google Scholar]
  • 102. Jayaraman M, Ansell SM, Mui BL, et al. Maximizing the potency of siRNA lipid nanoparticles for hepatic gene silencing in vivo. Angew Chem Int Ed Engl. 2012;51(34):8529‐8533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Swingle KL, Hamilton AG, Mitchell MJ. Lipid nanoparticle‐mediated delivery of mRNA therapeutics and vaccines. Trends Mol Med. 2021;27(6):616‐617. [DOI] [PubMed] [Google Scholar]
  • 104. Zhang Y, Sun C, Wang C, Jankovic KE, Dong Y. Lipids and lipid derivatives for RNA delivery. Chem Rev. 2021;121(20):12181‐12277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Beck JD, Reidenbach D, Salomon N, et al. mRNA therapeutics in cancer immunotherapy. Mol Cancer. 2021;20(1):69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Chen R, Wang SK, Belk JA, et al. Engineering circular RNA for enhanced protein production. Nat Biotechnol. 2023;41(2):262‐272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Amaya L, Grigoryan L, Li Z, et al. Circular RNA vaccine induces potent T cell responses. Proc Natl Acad Sci USA. 2023;120(20):e2302191120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Zhang H, You X, Wang X, et al. Delivery of mRNA vaccine with a lipid‐like material potentiates antitumor efficacy through toll‐like receptor 4 signaling. Proc Natl Acad Sci USA. 2021;118(6):e2005191118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Liu JQ, Zhang C, Zhang X, et al. Intratumoral delivery of IL‐12 and IL‐27 mRNA using lipid nanoparticles for cancer immunotherapy. J Control Release. 2022;345:306‐313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Rybakova Y, Kowalski PS, Huang Y, et al. mRNA delivery for therapeutic anti‐HER2 antibody expression in vivo. Mol Ther. 2019;27(8):1415‐1423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Hobo W, Novobrantseva TI, Fredrix H, et al. Improving dendritic cell vaccine immunogenicity by silencing PD‐1 ligands using siRNA‐lipid nanoparticles combined with antigen mRNA electroporation. Cancer Immunol Immunother. 2013;62(2):285‐297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Fan N, Chen K, Zhu R, et al. Manganese‐coordinated mRNA vaccines with enhanced mRNA expression and immunogenicity induce robust immune responses against SARS‐CoV‐2 variants. Sci Adv. 2022;8(51):eabq3500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Zhang Y, Yan J, Hou X, et al. STING Agonist‐derived LNP‐mRNA vaccine enhances protective immunity against SARS‐CoV‐2. Nano Lett. 2023;23(7):2593‐2600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Pan L, Zhang L, Deng W, et al. Spleen‐selective co‐delivery of mRNA and TLR4 agonist‐loaded LNPs for synergistic immunostimulation and Th1 immune responses. J Control Release. 2023;357:133‐148. [DOI] [PubMed] [Google Scholar]
  • 115. Lee K, Kim SY, Seo Y, Kim MH, Chang J, Lee H. Adjuvant incorporated lipid nanoparticles for enhanced mRNA‐mediated cancer immunotherapy. Biomater Sci. 2020;8(4):1101‐1105. [DOI] [PubMed] [Google Scholar]
  • 116. Raimondo TM, Reed K, Shi D, Langer R, Anderson DG. Delivering the next generation of cancer immunotherapies with RNA. Cell. 2023;186(8):1535‐1540. [DOI] [PubMed] [Google Scholar]
  • 117. Rosenblum D, Gutkin A, Kedmi R, et al. CRISPR‐Cas9 genome editing using targeted lipid nanoparticles for cancer therapy. Sci Adv. 2020;6(47):eabc9450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Cheng Q, Wei T, Farbiak L, Johnson LT, Dilliard SA, Siegwart DJ. Selective organ targeting (SORT) nanoparticles for tissue‐specific mRNA delivery and CRISPR‐Cas gene editing. Nat Nanotechnol. 2020;15(4):313‐320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Han JP, Kim M, Choi BS, et al. In vivo delivery of CRISPR‐Cas9 using lipid nanoparticles enables antithrombin gene editing for sustainable hemophilia A and B therapy. Sci Adv. 2022;8(3):eabj6901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Finn JD, Smith AR, Patel MC, et al. A single administration of CRISPR/Cas9 lipid nanoparticles achieves robust and persistent in vivo genome editing. Cell Rep. 2018;22(9):2227‐2235. [DOI] [PubMed] [Google Scholar]
  • 121. Qiu M, Glass Z, Chen J, et al. Lipid nanoparticle‐mediated codelivery of Cas9 mRNA and single‐guide RNA achieves liver‐specific in vivo genome editing of Angptl3. Proc Natl Acad Sci USA. 2021;118(10):e2020401118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Kenjo E, Hozumi H, Makita Y, et al. Low immunogenicity of LNP allows repeated administrations of CRISPR‐Cas9 mRNA into skeletal muscle in mice. Nat Commun. 2021;12(1):7101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Tang Q, Liu J, Jiang Y, Zhang M, Mao L, Wang M. Cell‐selective messenger RNA delivery and CRISPR/Cas9 genome editing by modulating the interface of phenylboronic acid‐derived lipid nanoparticles and cellular surface sialic acid. ACS Appl Mater Interfaces. 2019;11(50):46585‐46590. [DOI] [PubMed] [Google Scholar]
  • 124. Gao K, Li J, Song H, et al. In utero delivery of mRNA to the heart, diaphragm and muscle with lipid nanoparticles. Bioact Mater. 2023;25:387‐398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Shi D, Toyonaga S, Anderson DG. In vivo RNA delivery to hematopoietic stem and progenitor cells via targeted lipid nanoparticles. Nano Lett. 2023;23(7):2938‐2944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Yi J, Lei X, Guo F, et al. Co‐delivery of Cas9 mRNA and guide RNAs edits hepatitis B virus episomal and integration DNA in mouse and tree shrew models. Antiviral Res. 2023;215:105618. [DOI] [PubMed] [Google Scholar]
  • 127. Zhang D, Wang G, Yu X, et al. Enhancing CRISPR/Cas gene editing through modulating cellular mechanical properties for cancer therapy. Nat Nanotechnol. 2022;17(7):777‐787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Li B, Manan RS, Liang SQ, et al. Combinatorial design of nanoparticles for pulmonary mRNA delivery and genome editing. Nat Biotechnol. 2023; [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Farbiak L, Cheng Q, Wei T, et al. All‐in‐one dendrimer‐based lipid nanoparticles enable precise HDR‐mediated gene editing in vivo. Adv Mater. 2021;33(30):e2006619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Chen J, Ye Z, Huang C, et al. Lipid nanoparticle‐mediated lymph node‐targeting delivery of mRNA cancer vaccine elicits robust CD8(+) T cell response. Proc Natl Acad Sci USA. 2022;119(34):e2207841119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Lokugamage MP, Sago CD, Gan Z, Krupczak BR, Dahlman JE. Constrained nanoparticles deliver siRNA and sgRNA to T cells in vivo without targeting ligands. Adv Mater. 2019;31(41):e1902251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Sakurai Y, Hada T, Kato A, Hagino Y, Mizumura W, Harashima H. Effective therapy using a liposomal siRNA that targets the tumor vasculature in a model murine breast cancer with lung metastasis. Mol Ther Oncolytics. 2018;11:102‐108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Dilliard SA, Cheng Q, Siegwart DJ. On the mechanism of tissue‐specific mRNA delivery by selective organ targeting nanoparticles. Proc Natl Acad Sci USA. 2021;118(52):e2109256118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Wang X, Liu S, Sun Y, et al. Preparation of selective organ‐targeting (SORT) lipid nanoparticles (LNPs) using multiple technical methods for tissue‐specific mRNA delivery. Nat Protoc. 2023;18(1):265‐291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Sakurai Y, Mizumura W, Ito K, et al. Improved stability of siRNA‐loaded lipid nanoparticles prepared with a PEG‐monoacyl fatty acid facilitates ligand‐mediated siRNA delivery. Mol Pharm. 2020;17(4):1397‐1404. [DOI] [PubMed] [Google Scholar]
  • 136. Bevers S, Kooijmans SAA, Van de Velde E, et al. mRNA‐LNP vaccines tuned for systemic immunization induce strong antitumor immunity by engaging splenic immune cells. Mol Ther. 2022;30(9):3078‐3094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Sasaki K, Sato Y, Okuda K, Iwakawa K, Harashima H. mRNA‐loaded lipid nanoparticles targeting dendritic cells for cancer immunotherapy. Pharmaceutics. 2022;14(8):1572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Mukai H, Ogawa K, Kato N, Kawakami S. Recent advances in lipid nanoparticles for delivery of nucleic acid, mRNA, and gene editing‐based therapeutics. Drug Metab Pharmacokinet. 2022;44:100450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Xu Z, Zeng S, Gong Z, Yan Y. Exosome‐based immunotherapy: a promising approach for cancer treatment. Mol Cancer. 2020;19(1):160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Thakur A, Parra DC, Motallebnejad P, Brocchi M, Chen HJ. Exosomes: small vesicles with big roles in cancer, vaccine development, and therapeutics. Bioact Mater. 2022;10:281‐294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Xu R, Rai A, Chen M, Suwakulsiri W, Greening DW, Simpson RJ. Extracellular vesicles in cancer ‐ implications for future improvements in cancer care. Nat Rev Clin Oncol. 2018;15(10):617‐638. [DOI] [PubMed] [Google Scholar]
  • 142. Li M, Cai H, Deng R, Cheng J, Shi Y. Effects of exosomes on tumor immunomodulation and their potential clinical applications (Review). Int J Oncol. 2022;61(6):147. [DOI] [PubMed] [Google Scholar]
  • 143. Thakur A, Xu C, Li WK, et al. In vivo liquid biopsy for glioblastoma malignancy by the AFM and LSPR based sensing of exosomal CD44 and CD133 in a mouse model. Biosens Bioelectron. 2021;191:113476. [DOI] [PubMed] [Google Scholar]
  • 144. Mukherjee A, Bisht B, Dutta S, Paul MK. Current advances in the use of exosomes, liposomes, and bioengineered hybrid nanovesicles in cancer detection and therapy. Acta Pharmacol Sin. 2022;43(11):2759‐2776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Wortzel I, Dror S, Kenific CM, Lyden D. Exosome‐mediated metastasis: communication from a distance. Dev Cell. 2019;49(3):347‐360. [DOI] [PubMed] [Google Scholar]
  • 146. Syn NL, Wang L, Chow EK, Lim CT, Goh BC. Exosomes in cancer nanomedicine and immunotherapy: prospects and challenges. Trends Biotechnol. 2017;35(7):665‐676. [DOI] [PubMed] [Google Scholar]
  • 147. Tarasov VV, Svistunov AA, Chubarev VN, et al. Extracellular vesicles in cancer nanomedicine. Semin Cancer Biol. 2021;69:212‐225. [DOI] [PubMed] [Google Scholar]
  • 148. Kahroba H, Hejazi MS, Samadi N. Exosomes: from carcinogenesis and metastasis to diagnosis and treatment of gastric cancer. Cell Mol Life Sci. 2019;76(9):1747‐1758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Moore C, Kosgodage U, Lange S, Inal JM. The emerging role of exosome and microvesicle‐ (EMV‐) based cancer therapeutics and immunotherapy. Int J Cancer. 2017;141(3):428‐436. [DOI] [PubMed] [Google Scholar]
  • 150. Zitvogel L, Regnault A, Lozier A, et al. Eradication of established murine tumors using a novel cell‐free vaccine: dendritic cell‐derived exosomes. Nat Med. 1998;4(5):594‐600. [DOI] [PubMed] [Google Scholar]
  • 151. Wolfers J, Lozier A, Raposo G, et al. Tumor‐derived exosomes are a source of shared tumor rejection antigens for CTL cross‐priming. Nat Med. 2001;7(3):297‐303. [DOI] [PubMed] [Google Scholar]
  • 152. Lu Z, Zuo B, Jing R, et al. Dendritic cell‐derived exosomes elicit tumor regression in autochthonous hepatocellular carcinoma mouse models. J Hepatol. 2017;67(4):739‐748. [DOI] [PubMed] [Google Scholar]
  • 153. Thery C, Duban L, Segura E, Veron P, Lantz O, Amigorena S. Indirect activation of naive CD4+ T cells by dendritic cell‐derived exosomes. Nat Immunol. 2002;3(12):1156‐1162. [DOI] [PubMed] [Google Scholar]
  • 154. Pitt JM, Andre F, Amigorena S, et al. Dendritic cell‐derived exosomes for cancer therapy. J Clin Invest. 2016;126(4):1224‐1232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Zhang L, Yu D. Exosomes in cancer development, metastasis, and immunity. Biochim Biophys Acta Rev Cancer. 2019;1871(2):455‐468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Raposo G, Nijman HW, Stoorvogel W, et al. B lymphocytes secrete antigen‐presenting vesicles. J Exp Med. 1996;183(3):1161‐1172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Cheng L, Wang Y, Huang L. Exosomes from M1‐polarized macrophages potentiate the cancer vaccine by creating a pro‐inflammatory microenvironment in the lymph node. Mol Ther. 2017;25(7):1665‐1675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Sarkar I, Garg R, van Drunen Littel‐van den Hurk S. Selection of adjuvants for vaccines targeting specific pathogens. Expert Rev Vaccines. 2019;18(5):505‐521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Adamus T, Hung CY, Yu C, et al. Glioma‐targeted delivery of exosome‐encapsulated antisense oligonucleotides using neural stem cells. Mol Ther Nucleic Acids. 2022;27:611‐620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Jang Y, Kim H, Yoon S, et al. Exosome‐based photoacoustic imaging guided photodynamic and immunotherapy for the treatment of pancreatic cancer. J Control Release. 2021;330:293‐304. [DOI] [PubMed] [Google Scholar]
  • 161. Wang D, Wan Z, Yang Q, et al. Sonodynamical reversion of immunosuppressive microenvironment in prostate cancer via engineered exosomes. Drug Deliv. 2022;29(1):702‐713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. Saari H, Lazaro‐Ibanez E, Viitala T, Vuorimaa‐Laukkanen E, Siljander P, Yliperttula M. Microvesicle‐ and exosome‐mediated drug delivery enhances the cytotoxicity of paclitaxel in autologous prostate cancer cells. J Control Release. 2015;220(Pt B):727‐737. [DOI] [PubMed] [Google Scholar]
  • 163. Toffoli G, Hadla M, Corona G, et al. Exosomal doxorubicin reduces the cardiac toxicity of doxorubicin. Nanomedicine (Lond). 2015;10(19):2963‐2971. [DOI] [PubMed] [Google Scholar]
  • 164. Wu P, Zhang B, Ocansey DKW, Xu W, Qian H. Extracellular vesicles: a bright star of nanomedicine. Biomaterials. 2021;269:120467. [DOI] [PubMed] [Google Scholar]
  • 165. Richter M, Vader P, Fuhrmann G. Approaches to surface engineering of extracellular vesicles. Adv Drug Deliv Rev. 2021;173:416‐426. [DOI] [PubMed] [Google Scholar]
  • 166. Hao S, Bai O, Yuan J, Qureshi M, Xiang J. Dendritic cell‐derived exosomes stimulate stronger CD8+ CTL responses and antitumor immunity than tumor cell‐derived exosomes. Cell Mol Immunol. 2006;3(3):205‐211. [PubMed] [Google Scholar]
  • 167. Martins‐Marques T, Pinho MJ, Zuzarte M, et al. Presence of Cx43 in extracellular vesicles reduces the cardiotoxicity of the anti‐tumour therapeutic approach with doxorubicin. J Extracell Vesicles. 2016;5:32538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Fordjour FK, Guo C, Ai Y, Daaboul GG, Gould SJ. A shared, stochastic pathway mediates exosome protein budding along plasma and endosome membranes. J Biol Chem. 2022;298(10):102394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Kondo E, Iioka H, Saito K. Tumor‐homing peptide and its utility for advanced cancer medicine. Cancer Sci. 2021;112(6):2118‐2125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170. Elahi FM, Farwell DG, Nolta JA, Anderson JD. Preclinical translation of exosomes derived from mesenchymal stem/stromal cells. Stem Cells. 2020;38(1):15‐21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171. Zhou W, Zhou Y, Chen X, et al. Pancreatic cancer‐targeting exosomes for enhancing immunotherapy and reprogramming tumor microenvironment. Biomaterials. 2021;268:120546. [DOI] [PubMed] [Google Scholar]
  • 172. Xia Y, Rao L, Yao H, Wang Z, Ning P, Chen X. Engineering macrophages for cancer immunotherapy and drug delivery. Adv Mater. 2020;32(40):e2002054. [DOI] [PubMed] [Google Scholar]
  • 173. Gunassekaran GR, Poongkavithai Vadevoo SM, Baek MC, Lee B. M1 macrophage exosomes engineered to foster M1 polarization and target the IL‐4 receptor inhibit tumor growth by reprogramming tumor‐associated macrophages into M1‐like macrophages. Biomaterials. 2021;278:121137. [DOI] [PubMed] [Google Scholar]
  • 174. Fan Y, Zhou Y, Lu M, Si H, Li L, Tang B. Responsive dual‐targeting exosome as a drug carrier for combination cancer immunotherapy. Research (Wash D C). 2021;2021:9862876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Alvarez‐Erviti L, Seow Y, Yin H, Betts C, Lakhal S, Wood MJ. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol. 2011;29(4):341‐345. [DOI] [PubMed] [Google Scholar]
  • 176. Cheng Q, Dai Z, Smbatyan G, Epstein AL, Lenz HJ, Zhang Y. Eliciting anti‐cancer immunity by genetically engineered multifunctional exosomes. Mol Ther. 2022;30(9):3066‐3077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Luan X, Sansanaphongpricha K, Myers I, Chen H, Yuan H, Sun D. Engineering exosomes as refined biological nanoplatforms for drug delivery. Acta Pharmacol Sin. 2017;38(6):754‐763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Koh E, Lee EJ, Nam GH, et al. Exosome‐SIRPalpha, a CD47 blockade increases cancer cell phagocytosis. Biomaterials. 2017;121:121‐129. [DOI] [PubMed] [Google Scholar]
  • 179. Liu C, Zhang W, Li Y, et al. Microfluidic sonication to assemble exosome membrane‐coated nanoparticles for immune evasion‐mediated targeting. Nano Lett. 2019;19(11):7836‐7844. [DOI] [PubMed] [Google Scholar]
  • 180. Jhan YY, Prasca‐Chamorro D, Palou Zuniga G, et al. Engineered extracellular vesicles with synthetic lipids via membrane fusion to establish efficient gene delivery. Int J Pharm. 2020;573:118802. [DOI] [PubMed] [Google Scholar]
  • 181. Armstrong JP, Holme MN, Stevens MM. Re‐engineering extracellular vesicles as smart nanoscale therapeutics. ACS Nano. 2017;11(1):69‐83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182. Xu Y, Feng K, Zhao H, Di L, Wang L, Wang R. Tumor‐derived extracellular vesicles as messengers of natural products in cancer treatment. Theranostics. 2022;12(4):1683‐1714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Mishra V, Bansal KK, Verma A, et al. Solid lipid nanoparticles: emerging colloidal nano drug delivery systems. Pharmaceutics. 2018;10(4):191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184. Muller RH, Mader K, Gohla S. Solid lipid nanoparticles (SLN) for controlled drug delivery—a review of the state of the art. Eur J Pharm Biopharm. 2000;50(1):161‐177. [DOI] [PubMed] [Google Scholar]
  • 185. Chaudhuri A, Kumar DN, Shaik RA, et al. Lipid‐based nanoparticles as a pivotal delivery approach in triple negative breast cancer (TNBC) therapy. Int J Mol Sci. 2022;23(17):10068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186. Duan Y, Dhar A, Patel C, et al. A brief review on solid lipid nanoparticles: part and parcel of contemporary drug delivery systems. RSC Adv. 2020;10(45):26777‐26791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187. Jacob S, Nair AB, Shah J, et al. Lipid nanoparticles as a promising drug delivery carrier for topical ocular therapy—an overview on recent advances. Pharmaceutics. 2022;14(3):533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188. Parhi R, Suresh P. Preparation and characterization of solid lipid nanoparticles—a review. Curr Drug Discov Technol. 2012;9(1):2‐16. [DOI] [PubMed] [Google Scholar]
  • 189. Mehnert W, Mader K. Solid lipid nanoparticles: production, characterization and applications. Adv Drug Deliv Rev. 2001;47(2‐3):165‐196. [DOI] [PubMed] [Google Scholar]
  • 190. Del Pozo‐Rodriguez A, Solinis MA, Rodriguez‐Gascon A. Applications of lipid nanoparticles in gene therapy. Eur J Pharm Biopharm. 2016;109:184‐193. [DOI] [PubMed] [Google Scholar]
  • 191. Cai S, Yang Q, Bagby TR, Forrest ML. Lymphatic drug delivery using engineered liposomes and solid lipid nanoparticles. Adv Drug Deliv Rev. 2011;63(10‐11):901‐908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192. Serini S, Cassano R, Corsetto PA, Rizzo AM, Calviello G, Trombino S. Omega‐3 PUFA loaded in resveratrol‐based solid lipid nanoparticles: physicochemical properties and antineoplastic activities in human colorectal cancer cells in vitro. Int J Mol Sci. 2018;19(2):586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193. Almeida AJ, Souto E. Solid lipid nanoparticles as a drug delivery system for peptides and proteins. Adv Drug Deliv Rev. 2007;59(6):478‐490. [DOI] [PubMed] [Google Scholar]
  • 194. Pandey V, Gajbhiye KR, Soni V. Lactoferrin‐appended solid lipid nanoparticles of paclitaxel for effective management of bronchogenic carcinoma. Drug Deliv. 2015;22(2):199‐205. [DOI] [PubMed] [Google Scholar]
  • 195. Jain A, Kesharwani P, Garg NK, et al. Galactose engineered solid lipid nanoparticles for targeted delivery of doxorubicin. Colloids Surf B Biointerfaces. 2015;134:47‐58. [DOI] [PubMed] [Google Scholar]
  • 196. Banerjee I, De M, Dey G, et al. A peptide‐modified solid lipid nanoparticle formulation of paclitaxel modulates immunity and outperforms dacarbazine in a murine melanoma model. Biomater Sci. 2019;7(3):1161‐1178. [DOI] [PubMed] [Google Scholar]
  • 197. Uner M, Yener G. Importance of solid lipid nanoparticles (SLN) in various administration routes and future perspectives. Int J Nanomedicine. 2007;2(3):289‐300. [PMC free article] [PubMed] [Google Scholar]
  • 198. Das S, Ng WK, Tan RB. Are nanostructured lipid carriers (NLCs) better than solid lipid nanoparticles (SLNs): development, characterizations and comparative evaluations of clotrimazole‐loaded SLNs and NLCs? Eur J Pharm Sci. 2012;47(1):139‐151. [DOI] [PubMed] [Google Scholar]
  • 199. Weber S, Zimmer A, Pardeike J. Solid lipid nanoparticles (SLN) and nanostructured lipid carriers (NLC) for pulmonary application: a review of the state of the art. Eur J Pharm Biopharm. 2014;86(1):7‐22. [DOI] [PubMed] [Google Scholar]
  • 200. Gordillo‐Galeano A, Mora‐Huertas CE. Solid lipid nanoparticles and nanostructured lipid carriers: a review emphasizing on particle structure and drug release. Eur J Pharm Biopharm. 2018;133:285‐308. [DOI] [PubMed] [Google Scholar]
  • 201. Khan MS, Baskoy SA, Yang C, et al. Lipid‐based colloidal nanoparticles for applications in targeted vaccine delivery. Nanoscale Adv. 2023;5(7):1853‐1869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202. Lam RS, Nickerson MT. Food proteins: a review on their emulsifying properties using a structure‐function approach. Food Chem. 2013;141(2):975‐984. [DOI] [PubMed] [Google Scholar]
  • 203. Khan AY, Talegaonkar S, Iqbal Z, Ahmed FJ, Khar RK. Multiple emulsions: an overview. Curr Drug Deliv. 2006;3(4):429‐443. [DOI] [PubMed] [Google Scholar]
  • 204. McClements DJ, Jafari SM. Improving emulsion formation, stability and performance using mixed emulsifiers: a review. Adv Colloid Interface Sci. 2018;251:55‐79. [DOI] [PubMed] [Google Scholar]
  • 205. Jaiswal M, Dudhe R, Sharma PK. Nanoemulsion: an advanced mode of drug delivery system. 3 Biotech. 2015;5(2):123‐127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206. Galdino de Souza D, Santos DS, Simon KS, et al. Fish oil nanoemulsion supplementation attenuates bleomycin‐induced pulmonary fibrosis BALB/c mice. Nanomaterials (Basel). 2022;12(10):1683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207. Salvia‐Trujillo L, Martin‐Belloso O, McClements DJ. Excipient nanoemulsions for improving oral bioavailability of bioactives. Nanomaterials (Basel). 2016;6(1):1683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208. Sole I, Solans C, Maestro A, Gonzalez C, Gutierrez JM. Study of nano‐emulsion formation by dilution of microemulsions. J Colloid Interface Sci. 2012;376(1):133‐139. [DOI] [PubMed] [Google Scholar]
  • 209. Yin B, Deng W, Xu K, Huang L, Yao P. Stable nano‐sized emulsions produced from soy protein and soy polysaccharide complexes. J Colloid Interface Sci. 2012;380(1):51‐59. [DOI] [PubMed] [Google Scholar]
  • 210. Ashaolu TJ. Nanoemulsions for health, food, and cosmetics: a review. Environ Chem Lett. 2021;19(4):3381‐3395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211. Hormann K, Zimmer A. Drug delivery and drug targeting with parenteral lipid nanoemulsions—a review. J Control Release. 2016;223:85‐98. [DOI] [PubMed] [Google Scholar]
  • 212. Tagne JB, Kakumanu S, Ortiz D, Shea T, Nicolosi RJ. A nanoemulsion formulation of tamoxifen increases its efficacy in a breast cancer cell line. Mol Pharm. 2008;5(2):280‐286. [DOI] [PubMed] [Google Scholar]
  • 213. Tagne JB, Kakumanu S, Nicolosi RJ. Nanoemulsion preparations of the anticancer drug dacarbazine significantly increase its efficacy in a xenograft mouse melanoma model. Mol Pharm. 2008;5(6):1055‐1063. [DOI] [PubMed] [Google Scholar]
  • 214. Kakumanu S, Tagne JB, Wilson TA, Nicolosi RJ. A nanoemulsion formulation of dacarbazine reduces tumor size in a xenograft mouse epidermoid carcinoma model compared to dacarbazine suspension. Nanomedicine. 2011;7(3):277‐283. [DOI] [PubMed] [Google Scholar]
  • 215. Chapelin F, Capitini CM, Ahrens ET. Fluorine‐19 MRI for detection and quantification of immune cell therapy for cancer. J Immunother Cancer. 2018;6(1):105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216. Janjic JM, Ahrens ET. Fluorine‐containing nanoemulsions for MRI cell tracking. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2009;1(5):492‐501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217. Kuai X, Zhu Y, Yuan Z, et al. Perfluorooctyl bromide nanoemulsions holding MnO(2) nanoparticles with dual‐modality imaging and glutathione depletion enhanced HIFU‐eliciting tumor immunogenic cell death. Acta Pharm Sin B. 2022;12(2):967‐981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218. Zeng B, Middelberg AP, Gemiarto A, et al. Self‐adjuvanting nanoemulsion targeting dendritic cell receptor Clec9A enables antigen‐specific immunotherapy. J Clin Invest. 2018;128(5):1971‐1984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219. Misra R, Acharya S, Sahoo SK. Cancer nanotechnology: application of nanotechnology in cancer therapy. Drug Discov Today. 2010;15(19‐20):842‐850. [DOI] [PubMed] [Google Scholar]
  • 220. Alavi S, Haeri A, Dadashzadeh S. Utilization of chitosan‐caged liposomes to push the boundaries of therapeutic delivery. Carbohydr Polym. 2017;157:991‐1012. [DOI] [PubMed] [Google Scholar]
  • 221. Mohanty A, Uthaman S, Park IK. Utilization of polymer‐lipid hybrid nanoparticles for targeted anti‐cancer therapy. Molecules. 2020;25(19):4377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222. Shah S, PF, RSR , SBS, SS . Lipid polymer hybrid nanocarriers: insights into synthesis aspects, characterization, release mechanisms, surface functionalization and potential implications. Colloid Interface Sci Commun. 2022;46:100570. [Google Scholar]
  • 223. Zhao X, Li F, Li Y, et al. Co‐delivery of HIF1alpha siRNA and gemcitabine via biocompatible lipid‐polymer hybrid nanoparticles for effective treatment of pancreatic cancer. Biomaterials. 2015;46:13‐25. [DOI] [PubMed] [Google Scholar]
  • 224. Zhang M, Du Y, Wang S, Chen B. A review of biomimetic nanoparticle drug delivery systems based on cell membranes. Drug Des Devel Ther. 2020;14:5495‐5503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225. Liu Y, Xie X, Chen H, et al. Advances in next‐generation lipid‐polymer hybrid nanocarriers with emphasis on polymer‐modified functional liposomes and cell‐based‐biomimetic nanocarriers for active ingredients and fractions from Chinese medicine delivery. Nanomedicine. 2020;29:102237. [DOI] [PubMed] [Google Scholar]
  • 226. Wu XY. Strategies for optimizing polymer‐lipid hybrid nanoparticle‐mediated drug delivery. Expert Opin Drug Deliv. 2016;13(5):609‐612. [DOI] [PubMed] [Google Scholar]
  • 227. Sivadasan D, Sultan MH, Madkhali O, Almoshari Y, Thangavel N. Polymeric lipid hybrid nanoparticles (PLNs) as emerging drug delivery platform‐A comprehensive review of their properties, preparation methods, and therapeutic applications. Pharmaceutics. 2021;13(8):1291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228. Tahir N, Madni A, Correia A, et al. Lipid‐polymer hybrid nanoparticles for controlled delivery of hydrophilic and lipophilic doxorubicin for breast cancer therapy. Int J Nanomed. 2019;14:4961‐4974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229. Hadinoto K, Sundaresan A, Cheow WS. Lipid‐polymer hybrid nanoparticles as a new generation therapeutic delivery platform: a review. Eur J Pharm Biopharm. 2013;85(3 Pt A):427‐443. [DOI] [PubMed] [Google Scholar]
  • 230. Bose RJC, Ravikumar R, Karuppagounder V, Bennet D, Rangasamy S, Thandavarayan RA. Lipid‐polymer hybrid nanoparticle‐mediated therapeutics delivery: advances and challenges. Drug Discov Today. 2017;22(8):1258‐1265. [DOI] [PubMed] [Google Scholar]
  • 231. Thomas OS, Weber W. Overcoming physiological barriers to nanoparticle delivery‐are we there yet? Front Bioeng Biotechnol. 2019;7:415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232. Garg NK, Tandel N, Jadon RS, Tyagi RK, Katare OP. Lipid‐polymer hybrid nanocarrier‐mediated cancer therapeutics: current status and future directions. Drug Discov Today. 2018;23(9):1610‐1621. [DOI] [PubMed] [Google Scholar]
  • 233. Zhong Y, Du S, Dong Y. mRNA delivery in cancer immunotherapy. Acta Pharm Sin B. 2023;13(4):1348‐1357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234. Persano S, Guevara ML, Li Z, et al. Lipopolyplex potentiates anti‐tumor immunity of mRNA‐based vaccination. Biomaterials. 2017;125:81‐89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235. Wang N, Zuo Y, Wu S, Huang C, Zhang L, Zhu D. Spatio‐temporal delivery of both intra‐ and extracellular toll‐like receptor agonists for enhancing antigen‐specific immune responses. Acta Pharm Sin B. 2022;12(12):4486‐4500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236. Zhang L, Wu S, Qin Y, et al. Targeted codelivery of an antigen and dual agonists by hybrid nanoparticles for enhanced cancer immunotherapy. Nano Lett. 2019;19(7):4237‐4249. [DOI] [PubMed] [Google Scholar]
  • 237. Ou W, Thapa RK, Jiang L, et al. Regulatory T cell‐targeted hybrid nanoparticles combined with immuno‐checkpoint blockage for cancer immunotherapy. J Control Release. 2018;281:84‐96. [DOI] [PubMed] [Google Scholar]
  • 238. Abdel‐Bar HM, Walters AA, Wang JT, KT Al‐Jamal. Combinatory delivery of etoposide and siCD47 in a lipid polymer hybrid delays lung tumor growth in an experimental melanoma lung metastatic model. Adv Healthc Mater. 2021;10(7):e2001853. [DOI] [PubMed] [Google Scholar]
  • 239. Hwang J, Zhang W, Park HB, Yadav D, Jeon YH, Jin JO. Escherichia coli adhesin protein‐conjugated thermal responsive hybrid nanoparticles for photothermal and immunotherapy against cancer and its metastasis. J Immunother Cancer. 2021;9(7):e002666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240. Ou W, Jiang L, Thapa RK, et al. Combination of NIR therapy and regulatory T cell modulation using layer‐by‐layer hybrid nanoparticles for effective cancer photoimmunotherapy. Theranostics. 2018;8(17):4574‐4590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241. He C, Duan X, Guo N, et al. Core‐shell nanoscale coordination polymers combine chemotherapy and photodynamic therapy to potentiate checkpoint blockade cancer immunotherapy. Nat Commun. 2016;7:12499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242. Li X, Younis MH, Wei W, Cai W. PD‐L1 ‐ targeted magnetic fluorescent hybrid nanoparticles: illuminating the path of image‐guided cancer immunotherapy. Eur J Nucl Med Mol Imaging. 2023; [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243. Wang Y, Chen Q, Song H, et al. A triple therapeutic strategy with antiexosomal iron efflux for enhanced ferroptosis therapy and immunotherapy. Small. 2022;18(41):e2201704. [DOI] [PubMed] [Google Scholar]
  • 244. Chen DS, Mellman I. Oncology meets immunology: the cancer‐immunity cycle. Immunity. 2013;39(1):1‐10. [DOI] [PubMed] [Google Scholar]
  • 245. Zhang Y, Zhang Z. The history and advances in cancer immunotherapy: understanding the characteristics of tumor‐infiltrating immune cells and their therapeutic implications. Cell Mol Immunol. 2020;17(8):807‐821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246. Bagchi S, Yuan R, Engleman EG. Immune checkpoint inhibitors for the treatment of cancer: clinical impact and mechanisms of response and resistance. Annu Rev Pathol. 2021;16:223‐249. [DOI] [PubMed] [Google Scholar]
  • 247. Jiang ML, Chen PX, Wang L, et al. cGAS‐STING, an important pathway in cancer immunotherapy. J Hematol Oncol. 2020;13(1):81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248. Cai X, Chiu YH, Chen ZJJ. The cGAS‐cGAMP‐STING pathway of cytosolic DNA sensing and signaling. Mol Cell. 2014;54(2):289‐296. [DOI] [PubMed] [Google Scholar]
  • 249. Zhang CG, Shang GJ, Gui X, Zhang XW, Bai XC, Chen ZJJ. Structural basis of STING binding with and phosphorylation by TBK1. Nature. 2019;567(7748):394‐+. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250. Li XD, Wu JX, Gao DX, Wang H, Sun LJ, Chen ZJJ. Pivotal roles of cGAS‐cGAMP signaling in antiviral defense and immune adjuvant effects. Science. 2013;341(6152):1390‐1394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251. Ivashkiv LB, Donlin LT. Regulation of type I interferon responses. Nat Rev Immunol. 2014;14(1):36‐49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252. Fuertes MB, Woo SR, Burnett B, Fu YX, Gajewski TF. Type I interferon response and innate immune sensing of cancer. Trends Immunol. 2013;34(2):67‐73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253. Gao D, Li T, Li XD, et al. Activation of cyclic GMP‐AMP synthase by self‐DNA causes autoimmune diseases. Proc Natl Acad Sci USA. 2015;112(42):E5699‐5705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254. Woo SR, Fuertes MB, Corrales L, et al. STING‐dependent cytosolic DNA sensing mediates innate immune recognition of immunogenic tumors. Immunity. 2014;41(5):830‐842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255. Deng L, Liang H, Xu M, et al. STING‐Dependent Cytosolic DNA sensing promotes radiation‐induced type I interferon‐dependent antitumor immunity in immunogenic tumors. Immunity. 2014;41(5):843‐852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256. Yum S, Li M, Chen ZJ. Old dogs, new trick: classic cancer therapies activate cGAS. Cell Res. 2020;30(8):639‐648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257. Pepin G, Gantier MP. cGAS‐STING activation in the tumor microenvironment and its role in cancer immunity (vol 1024, pg 175, 2017). Adv Exp Med Biol. 2017;1024:E1‐E1. [DOI] [PubMed] [Google Scholar]
  • 258. Hines JB, Kacew AJ, Sweis RF. The development of STING agonists and emerging results as a cancer immunotherapy. Curr Oncol Rep. 2023;25(3):189‐199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259. Kato Y, Park J, Takamatsu H, et al. Apoptosis‐derived membrane vesicles drive the cGAS‐STING pathway and enhance type I IFN production in systemic lupus erythematosus. Ann Rheum Dis. 2018;77(10):1507‐1515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260. Ku JWK, Chen Y, Lim BJW, Gasser S, Crasta KC, Gan YH. Bacterial‐induced cell fusion is a danger signal triggering cGAS‐STING pathway via micronuclei formation. Proc Natl Acad Sci USA. 2020;117(27):15923‐15934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261. Zhang R, Kang R, Tang D. The STING1 network regulates autophagy and cell death. Signal Transduct Target Ther. 2021;6(1):208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262. Li C, Zhang Y, Liu J, Kang R, Klionsky DJ, Tang D. Mitochondrial DNA stress triggers autophagy‐dependent ferroptotic death. Autophagy. 2021;17(4):948‐960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263. Wang H, Sun L, Su L, et al. Mixed lineage kinase domain‐like protein MLKL causes necrotic membrane disruption upon phosphorylation by RIP3. Mol Cell. 2014;54(1):133‐146. [DOI] [PubMed] [Google Scholar]
  • 264. Liu D, Wu H, Wang C, et al. STING directly activates autophagy to tune the innate immune response. Cell Death Differ. 2019;26(9):1735‐1749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265. Gulen MF, Koch U, Haag SM, et al. Signalling strength determines proapoptotic functions of STING. Nat Commun. 2017;8(1):427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266. Zhou Y, Fei M, Zhang G, et al. Blockade of the phagocytic receptor MerTK on tumor‐associated macrophages enhances P2×7R‐dependent STING activation by tumor‐derived cGAMP. Immunity. 2020;52(2):357‐373 e9. [DOI] [PubMed] [Google Scholar]
  • 267. Chattopadhyay S, Liu YH, Fang ZS, et al. Synthetic immunogenic cell death mediated by intracellular delivery of STING agonist nanoshells enhances anticancer chemo‐immunotherapy. Nano Lett. 2020;20(4):2246‐2256. [DOI] [PubMed] [Google Scholar]
  • 268. Wang‐Bishop L, Wehbe M, Shae D, et al. Potent STING activation stimulates immunogenic cell death to enhance antitumor immunity in neuroblastoma. J Immunother Cancer. 2020;8(1):e000282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269. Klarquist J, Hennies CM, Lehn MA, Reboulet RA, Feau S, Janssen EM. STING‐mediated DNA sensing promotes antitumor and autoimmune responses to dying cells. J Immunol. 2014;193(12):6124‐6134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270. Ding L, Wang Q, Martincuks A, et al. STING agonism overcomes STAT3‐mediated immunosuppression and adaptive resistance to PARP inhibition in ovarian cancer. J Immunother Cancer. 2023;11(1):e005627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271. Wang YF, Liu ZG, Yuan HF, et al. The reciprocity between radiotherapy and cancer immunotherapy. Clin Cancer Res. 2019;25(6):1709‐1717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272. Sabatte J, Maggini J, Nahmod K, et al. Interplay of pathogens, cytokines and other stress signals in the regulation of dendritic cell function. Cytokine Growth F R. 2007;18(1‐2):5‐17. [DOI] [PubMed] [Google Scholar]
  • 273. Chen Q, Boire A, Jin X, et al. Carcinoma‐astrocyte gap junctions promote brain metastasis by cGAMP transfer. Nature. 2016;533(7604):493‐+. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274. Diamond MS, Kinder M, Matsushita H, et al. Type I interferon is selectively required by dendritic cells for immune rejection of tumors. J Exp Med. 2011;208(10):1989‐2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275. Ohkuri T, Kosaka A, Ishibashi K, et al. Intratumoral administration of cGAMP transiently accumulates potent macrophages for anti‐tumor immunity at a mouse tumor site. Cancer Immunol Immunother. 2017;66(6):705‐716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276. Demaria O, De Gassart A, Coso S, et al. STING activation of tumor endothelial cells initiates spontaneous and therapeutic antitumor immunity. Proc Natl Acad Sci USA. 2015;112(50):15408‐15413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277. Gan Y, Li XY, Han SZ, et al. The cGAS/STING pathway: a novel target for cancer therapy. Front Immunol. 2022;12:795401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278. Zlotnik A, Yoshie O. The chemokine superfamily revisited. Immunity. 2012;36(5):705‐716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279. Viola A, Sarukhan A, Bronte V, Molon B. The pros and cons of chemokines in tumor immunology. Trends Immunol. 2012;33(10):496‐504. [DOI] [PubMed] [Google Scholar]
  • 280. Larkin B, Ilyukha V, Sorokin M, Buzdin A, Vannier E, Poltorak A. Cutting edge: activation of STING in T cells induces type I IFN responses and cell death. J Immunol. 2017;199(2):397‐402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281. Cerboni S, Jeremiah N, Gentili M, et al. Intrinsic antiproliferative activity of the innate sensor STING in T lymphocytes. J Exp Med. 2017;214(6):1769‐1785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282. Tan YS, Sansanaphongpricha K, Xie YY, et al. Mitigating SOX2‐potentiated immune escape of head and neck squamous cell carcinoma with a STING‐inducing nanosatellite vaccine. Clin Cancer Res. 2018;24(17):4242‐4255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 283. Wang QW, Bergholz JS, Ding LY, et al. STING agonism reprograms tumor‐associated macrophages and overcomes resistance to PARP inhibition in BRCA1‐deficient models of breast cancer. Nat Commun. 2022;13(1):3022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 284. Ma RH, Ji TT, Chen DG, et al. Tumor cell‐derived microparticles polarize M2 tumor‐associated macrophages for tumor progression. Oncoimmunology. 2016;5(4):e1118599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285. Corrales L, Glickman LH, McWhirter SM, et al. Direct activation of STING in the tumor microenvironment leads to potent and systemic tumor regression and immunity. Cell Rep. 2015;11(7):1018‐1030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286. Jing W, McAllister D, Vonderhaar EP, et al. STING agonist inflames the pancreatic cancer immune microenvironment and reduces tumor burden in mouse models. J Immunother Cancer. 2019;7(1):115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287. Yang H, Lee WS, Kong SJ, et al. STING activation reprograms tumor vasculatures and synergizes with VEGFR2 blockade. J Clin Invest. 2019;129(10):4350‐4364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 288. Lee SJ, Yang H, Kim WR, et al. STING activation normalizes the intraperitoneal vascular‐immune microenvironment and suppresses peritoneal carcinomatosis of colon cancer. J Immunother Cancer. 2021;9(6):e002195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 289. Chelvanambi M, Fecek R, Taylor J, Storkus W. Sting agonist‐based treatment promotes vascular normalization and tertiary lymphoid structure formation in the therapeutic melanoma microenvironment. J Immunother Cancer. 2020;8:A359‐A360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 290. Downey CM, Aghaei M, Schwendener RA, Jirik FR. DMXAA causes tumor site‐specific vascular disruption in murine non‐small cell lung cancer, and like the endogenous non‐canonical cyclic dinucleotide STING agonist, 2′3′‐cGAMP, induces M2 macrophage repolarization. PLoS One. 2014;9(6):e99988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291. Carman CV, Martinelli R. T lymphocyte‐endothelial interactions: emerging understanding of trafficking and antigen‐specific immunity. Front Immunol. 2015;6:603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292. Zheng H, Qian J, Carbone CJ, Leu NA, Baker DP, Fuchs SY. Vascular endothelial growth factor‐induced elimination of the type 1 interferon receptor is required for efficient angiogenesis. Blood. 2011;118(14):4003‐4006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 293. Sidky YA, Borden EC. Inhibition of angiogenesis by interferons: effects on tumor‐ and lymphocyte‐induced vascular responses. Cancer Res. 1987;47(19):5155‐5161. [PubMed] [Google Scholar]
  • 294. Bourhis M, Palle J, Galy‐Fauroux I, Terme M. Direct and indirect modulation of T cells by VEGF—a counteracted by anti‐angiogenic treatment. Front Immunol. 2021;12:616837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295. Mulligan JK, Rosenzweig SA, Young MR. Tumor secretion of VEGF induces endothelial cells to suppress T cell functions through the production of PGE2. J Immunother. 2010;33(2):126‐135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296. Kim S, Li L, Maliga Z, Yin Q, Wu H, Mitchison TJ. Anticancer flavonoids are mouse‐selective STING agonists. ACS Chem Biol. 2013;8(7):1396‐1401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 297. Philpott M, Baguley BC, Ching LM. Induction of tumour necrosis factor‐alpha by single and repeated doses of the antitumour agent 5,6‐dimethylxanthenone‐4‐acetic acid. Cancer Chemoth Pharm. 1995;36(2):143‐148. [DOI] [PubMed] [Google Scholar]
  • 298. Baguley BC, Ching LM. Immunomodulatory actions of xanthenone anticancer agents. BioDrugs. 1997;8(2):119‐127. [DOI] [PubMed] [Google Scholar]
  • 299. Conlon J, Burdette DL, Sharma S, et al. Mouse, but not human STING, binds and signals in response to the vascular disrupting agent 5,6‐dimethylxanthenone‐4‐acetic acid. J Immunol. 2013;190(10):5216‐5225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 300. Gao P, Ascano M, Zillinger T, et al. Structure‐function analysis of STING activation by c[G(2′,5′) pA(3′,5′)p] and targeting by antiviral DMXAA. Cell. 2013;154(4):748‐762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 301. Amouzegar A, Chelvanambi M, Filderman JN, Storkus WJ, Luke JJ. STING agonists as cancer therapeutics. Cancers. 2021;13(11):2695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 302. Aval LM, Pease JE, Sharma R, Pinato DJ. Challenges and opportunities in the clinical development of STING agonists for cancer immunotherapy. J Clin Med. 2020;9(10):3323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 303. Harrington KJ, Brody J, Ingham M, et al. Preliminary results of the first‐in‐human (FIH) study of MK‐1454, an agonist of stimulator of interferon genes (STING), as monotherapy or in combination with pembrolizumab (pembro) in patients with advanced solid tumors or lymphomas. Ann Oncol. 2018;29:712‐712. [Google Scholar]
  • 304. Endo A, Kim DS, Huang KC, et al. Discovery of E7766: a representative of a novel class of macrocycle‐bridged STING agonists (MBSAs) with superior potency and pan‐genotypic activity. Cancer Res. 2019;79(13):1740‐1743.30952631 [Google Scholar]
  • 305. Khongorzul P, Ling CJ, Khan FU, Ihsan AU, Zhang J. Antibody‐drug conjugates: a comprehensive review. Mol Cancer Res. 2020;18(1):3‐19. [DOI] [PubMed] [Google Scholar]
  • 306. Diamond JR, Henry JT, Falchook GS, et al. Phase 1a/1b study design of the novel STING agonist, immune‐stimulating antibody‐conjugate (ISAC) TAK‐500, with or without pembrolizumab in patients with advanced solid tumors. J Clin Oncol. 2022;40(16_suppl):TPS2690‐TPS2690. [Google Scholar]
  • 307. Diamond JR, Henry JT, Falchook GS, et al. Abstract CT249: first‐in‐human study of TAK‐500, a novel STING agonist immune stimulating antibody conjugate (ISAC), alone and in combination with pembrolizumab in patients with select advanced solid tumors. Cancer Res. 2022;82(12_Supplement):CT249‐CT249. [Google Scholar]
  • 308. Duvall JR, Bukhalid RA, Cetinbas NM, et al. XMT‐2056, a well‐tolerated, Immunosynthen‐based STING‐agonist antibody‐drug conjugate which induces anti‐tumor immune activity. Cancer Res. 2021;81(13):1738. [Google Scholar]
  • 309. Duvall JR, Bukhalid RA, Cetinbas NM, et al. XMT‐2056, a HER2‐targeted Immunosynthen STING‐agonist antibody‐drug conjugate, binds a novel epitope of HER2 and shows increased anti‐tumor activity in combination with trastuzumab and pertuzumab. Cancer Res. 2022;82(12_Supplement):3503‐3503. [Google Scholar]
  • 310. Gogoi H, Mansouri S, Jin L. The age of cyclic dinucleotide vaccine adjuvants. Vaccines (Basel). 2020;8(3):453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 311. Lv M, Chen M, Zhang R, et al. Manganese is critical for antitumor immune responses via cGAS‐STING and improves the efficacy of clinical immunotherapy. Cell Res. 2020;30(11):966‐979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 312. Sun X, Zhang Y, Li J, et al. Amplifying STING activation by cyclic dinucleotide‐manganese particles for local and systemic cancer metalloimmunotherapy. Nat Nanotechnol. 2021;16(11):1260‐1270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 313. Luo Z, Liang X, He T, et al. Lanthanide‐nucleotide coordination nanoparticles for STING activation. J Am Chem Soc. 2022;144(36):16366‐16377. [DOI] [PubMed] [Google Scholar]
  • 314. Sali TM, Pryke KM, Abraham J, et al. Characterization of a novel human‐specific STING agonist that elicits antiviral activity against emerging alphaviruses. PLoS Pathog. 2015;11(12):e1005324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 315. Banerjee M, Middya S, Shrivastava R, et al. G10 is a direct activator of human STING. PLoS One. 2020;15(9):e0237743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 316. Liu B, Tang L, Zhang X, et al. A cell‐based high throughput screening assay for the discovery of cGAS‐STING pathway agonists. Antiviral Res. 2017;147:37‐46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 317. Cavlar T, Deimling T, Ablasser A, Hopfner KP, Hornung V. Species‐specific detection of the antiviral small‐molecule compound CMA by STING. EMBO J. 2013;32(10):1440‐1450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 318. Zhang Y, Sun Z, Pei J, et al. Identification of alpha‐mangostin as an agonist of human STING. ChemMedChem. 2018;13(19):2057‐2064. [DOI] [PubMed] [Google Scholar]
  • 319. Ding C, Song Z, Shen A, Chen T, Zhang A. Small molecules targeting the innate immune cGAS‒STING‒TBK1 signaling pathway. Acta Pharm Sin B. 2020;10(12):2272‐2298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 320. Ramanjulu JM, Pesiridis GS, Yang J, et al. Author correction: design of amidobenzimidazole STING receptor agonists with systemic activity. Nature. 2019;570(7761):E53. [DOI] [PubMed] [Google Scholar]
  • 321. Ramanjulu JM, Pesiridis GS, Yang J, et al. Design of amidobenzimidazole STING receptor agonists with systemic activity. Nature. 2018;564(7736):439‐443. [DOI] [PubMed] [Google Scholar]
  • 322. Xie Z, Wang Z, Fan F, et al. Structural insights into a shared mechanism of human STING activation by a potent agonist and an autoimmune disease‐associated mutation. Cell Discov. 2022;8(1):133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 323. Feng X, Pan L, Qian Z, et al. Discovery of selenium‐containing STING agonists as orally available antitumor agents. J Med Chem. 2022;65(22):15048‐15065. [DOI] [PubMed] [Google Scholar]
  • 324. Vavrina Z, Perlikova P, Milisavljevic N, et al. Design, synthesis, and biochemical and biological evaluation of novel 7‐deazapurine cyclic dinucleotide analogues as STING receptor agonists. J Med Chem. 2022;65(20):14082‐14103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 325. Xie Z, Lu L, Wang Z, et al. S‐acylthioalkyl ester (SATE)‐based prodrugs of deoxyribose cyclic dinucleotides (dCDNs) as the STING agonist for antitumor immunotherapy. Eur J Med Chem. 2022;243:114796. [DOI] [PubMed] [Google Scholar]
  • 326. Pan BS, Perera SA, Piesvaux JA, et al. An orally available non‐nucleotide STING agonist with antitumor activity. Science. 2020;369(6506):eaba6098. [DOI] [PubMed] [Google Scholar]
  • 327. Garland KM, Sheehy TL, Wilson JT. Chemical and biomolecular strategies for STING pathway activation in cancer immunotherapy. Chem Rev. 2022;122(6):5977‐6039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 328. Yang J, Adam M, Clemens J, et al. Preclinical characterization of GSK532, a novel STING agonist with potent anti‐tumor activity. Cancer Res. 2018;78(13_Supplement):5554‐5554. [Google Scholar]
  • 329. Smith M, Chin D, Chan S, et al. In vivo administration of the STING agonist, JNJ‐67544412, leads to complete regression of established murine subcutaneous tumors. Cancer Res. 2020;80(16_Supplement):5567‐5567. [Google Scholar]
  • 330. Thomsen MK, Skouboe MK, Boularan C, et al. The cGAS‐STING pathway is a therapeutic target in a preclinical model of hepatocellular carcinoma. Oncogene. 2020;39(8):1652‐1664. [DOI] [PubMed] [Google Scholar]
  • 331. Rogacki M, Chmielewski S, Zawadzka M, et al. 601 Development of improved small molecule STING agonists suitable for systemic administration. BMJ Specialist J. 2020;8(3). [Google Scholar]
  • 332. Chmielewski S, Zawadzka M, Mazurek J, et al. Abstract 4532A: development of selective small molecule STING agonists suitable for systemic administration. Cancer Res. 2020;80(16_Supplement):4532A‐4532A. [Google Scholar]
  • 333. Dobrzańska M, Chmielewski S, Zawadzka M, et al. Discovery and characterization of next‐generation small molecule direct STING agonists. Cancer Res. 2019;79(13_Supplement):4983‐4983. [Google Scholar]
  • 334. Banerjee M, Basu S, Middya S, et al. Abstract LB‐061: CRD5500: a versatile small molecule STING agonist amenable to bioconjugation as an ADC. Cancer Res. 2019;79(13_Supplement):LB‐061‐LB‐061. [Google Scholar]
  • 335. Banerjee M, Basu S, Middya S, et al. Intravenous administration of the small molecule STING agonist CRD5500 elicits potent anti‐tumor immune responses in cold tumors. Cancer Res. 2022;82(12_Supplement):1996‐1996. [Google Scholar]
  • 336. Binder GA, Gambino CS, Kharitonova A, Metcalf RS, Daniel KG, Guida WC. Computationally assisted target screening of STING agonist for immunologic therapy. Cancer Res. 2019;79(13_Supplement):6‐6. [Google Scholar]
  • 337. Chan SR, Bignan G, Pierson E, et al. Abstract 5567A: JNJ‐‘6196: a next generation STING agonist with potent preclinical activity by the IV route. Cancer Res. 2020;80(16_Supplement):5567A‐5567A. [Google Scholar]
  • 338. Wang Z, Dove P, Rosa D, et al. Preclinical characterization of a novel non‐cyclic dinucleotide small molecule STING agonist with potent antitumor activity in mice. Cancer Res. 2019;79(13_Supplement):3854‐3854. [Google Scholar]
  • 339. Li A, Song Y, Dong C, Chen X, Yang J. Discovery of novel STING agonists with robust anti‐tumor activity. Cancer Res. 2020;80(16_Supplement):3317‐3317. [Google Scholar]
  • 340. Perera SA, Kopinja JE, Ma Y, et al. Combining STING agonists with an anti‐PD‐1 antagonist results in marked antitumor activity in immune‐excluded tumors. Cancer Res. 2018;78(13_Supplement):4721‐4721. [Google Scholar]
  • 341. Prabagar M, Bommireddy V, Siegel R, et al. The novel STING agonist VB‐85247 induces robust durable antitumor immune responses by intravesical administration in a non‐muscle invasive bladder cancer model. Cancer Res. 2021;81(13_Supplement):524‐524. [Google Scholar]
  • 342. Jekle A, Thatikonda S, Stevens S, et al. Preclinical characterization of ALG‐031048, a novel STING agonist with potent anti‐tumor activity in mice. Cancer Res. 2020;80(16_Supplement):4520‐4520. [Google Scholar]
  • 343. Weston AS, Thode TG, Rodriguez del Villar R, et al. Abstract LB‐118: SR8541A is a potent inhibitor of ENPP1 and exhibits dendritic cell mediated antitumor activity. Cancer Res. 2020;80(16_Supplement):LB‐118‐LB‐118. [Google Scholar]
  • 344. Weston AS, Thode T, Ng S, et al. Abstract LB323: inhibition of ENPP1 using small molecule, SR‐8541A, enhances the effect of checkpoint inhibition in cancer models. Cancer Res. 2023;83(8_Supplement):LB323‐LB323. [Google Scholar]
  • 345. Weston A, Thode T, Munoz R, et al. Preclinical studies of SR‐8314, a highly selective ENPP1 inhibitor and an activator of STING pathway. Cancer Res. 2019;79(13_Supplement):3077‐3077. [Google Scholar]
  • 346. Baird J, Dietsch G, Florio V, et al. MV‐626, a potent and selective inhibitor of ENPP1 enhances STING activation and augments T‐cell mediated anti‐tumor activity in vivo (Annual Meeting). 2018.
  • 347. Kulkarni A, Goswami A, Goyal S, Khurana P, Ghoshal I. AVA‐NP‐695, a potent and selective ENPP1 inhibitor, abrogates tumor metastasis in 4T1 syngeneic tumor model and demonstrates strong tumor regression when combined with radiation. Cancer Res. 2023;83(7_Supplement):3244‐3244. [Google Scholar]
  • 348. Qin X, Li Y‐y, Sun S, et al. ENPP1 inhibitor ZX‐8177 in combination with chemotherapy or radiation exhibits synergistic anti‐tumor effects via STING‐mediated response. Cancer Res. 2023;83(7_Supplement):577‐577. [Google Scholar]
  • 349. Li Y‐y, Peng Z, Sun S, et al. ENPP1 inhibitor ZX‐8177 enhances anti‐tumor activity of conventional therapies by modulating tumor microenvironment. Cancer Res. 2022;82(12_Supplement):5486‐5486. [Google Scholar]
  • 350. Yoon PO, Kim J, Lee A‐R, et al. A novel small molecule inhibitor of ENPP1 promotes T and NK cell activation and enhances anti‐tumor efficacy in combination with immune checkpoint blockade therapy. Cancer Res. 2023;83(7_Supplement):702‐702. [Google Scholar]
  • 351. Peng Z, Guo K, Li Y‐Y, et al. Potent ENPP1 inhibitors activating STING pathway in tumor microenvironment. Cancer Res. 2021;81(13_Supplement):1275‐1275. [Google Scholar]
  • 352. Cetinbas NM, Catcott KC, Monnell T, et al. Tumor cell‐targeted STING‐agonist antibody‐drug conjugates achieve potent anti‐tumor activity by delivering STING agonist specifically to tumor cells andFcγRI‐expressing subset of myeloid cells. Cancer Res. 2022;82(12_Supplement):2114‐2114. [Google Scholar]
  • 353. Ye Y, Yang G, Wang H, et al. JAB‐X1800: a potent immunostimulatory antibody‐drug conjugate (iADC) targeting CD73. JAB. 1800;4:1. [Google Scholar]
  • 354. Wu YT, Fang Y, Wei Q, et al. Tumor‐targeted delivery of a STING agonist improvescancer immunotherapy. Proc Natl Acad Sci USA. 2022;119(49):e2214278119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 355. Engelman DM, Reshetnyak YK, Andreev OA. Durable eradication of tumors by single injections of a pHLIP‐STING agonist. Cancer Res. 2023;83(7_Supplement):1804‐1804. [Google Scholar]
  • 356. Olesen CE, Axelgaard E, Nilsson E, et al. Systemic delivery of ST317, a cell penetrating STING pathway sensitizer, results in strong anti‐tumor activity. Cancer Res. 2023;83(7_Supplement):1852‐1852. [Google Scholar]
  • 357. Singh A, Praharaj M, Joice G, et al. MP57‐02 Next‐gen sting‐agonist like Bcg confers enhanced immunogenicity and antitumor efficacy in vitro and in vivo. J Urol. 2019;201(Supplement 4):e813‐e813. [Google Scholar]
  • 358. Makarova AM, Iannello A, Rae CS, et al. STACT‐TREX1: a systemically‐administered STING pathway agonist targets tumor‐resident myeloid cells and induces adaptive anti‐tumor immunity in multiple preclinical models. Cancer Res. 2019;79(13_Supplement):5016‐5016. [Google Scholar]
  • 359. Chen Z, Huang G, Torres K, et al. Abstract LB245: ONM‐501, a dual‐activating polyvalent STING agonist, enhances tumor retention and demonstrates favorable preclinical safety profile. Cancer Res. 2023;83(8_Supplement):LB245‐LB245. [Google Scholar]
  • 360. Li S, Wang J, Wilhelm J, et al. ONM‐501: a polyvalent STING agonist for oncology immunotherapy. Cancer Res. 2022;82(12 Supplement):4234.36112059 [Google Scholar]
  • 361. Koshy ST, Cheung AS, Gu L, Graveline AR, Mooney DJ. Liposomal delivery enhances immune activation by STING agonists for cancer immunotherapy. Adv Biosyst. 2017;1(1‐2):1600013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 362. Yu MK, Park J, Jon S. Targeting strategies for multifunctional nanoparticles in cancer imaging and therapy. Theranostics. 2012;2(1):3‐44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 363. de Menezes DEL, Kirchmeier MJ, Gagne JF, Pilarski LM, Allen TM. Cellular trafficking and cytotoxicity of anti‐CD19‐targeted liposomal doxorubicin in B lymphoma cells. J Liposome Res. 1999;9(2):199‐228. [Google Scholar]
  • 364. Sapra P, Allen TM. Ligand‐targeted liposomal anticancer drugs. Prog Lipid Res. 2003;42(5):439‐462. [DOI] [PubMed] [Google Scholar]
  • 365. Medina OP, Zhu Y, Kairemo K. Targeted liposomal drug delivery in cancer. Curr Pharm Design. 2004;10(24):2981‐2989. [DOI] [PubMed] [Google Scholar]
  • 366. Li K, Ye Y, Liu L, et al. The lipid platform increases the activity of STING agonists to synergize checkpoint blockade therapy against melanoma. Biomater Sci. 2021;9(3):765‐773. [DOI] [PubMed] [Google Scholar]
  • 367. Doshi AS, Cantin S, Prickett LB, Mele DA, Amiji M. Systemic nano‐delivery of low‐dose STING agonist targeted to CD103+ dendritic cells for cancer immunotherapy. J Control Release. 2022;345:721‐733. [DOI] [PubMed] [Google Scholar]
  • 368. Kocabas BB, Almacioglu K, Bulut EA, et al. Dual‐adjuvant effect of pH‐sensitive liposomes loaded with STING and TLR9 agonists regress tumor development by enhancing Th1 immune response. J Control Release. 2020;328:587‐595. [DOI] [PubMed] [Google Scholar]
  • 369. Hao M, Zhu L, Hou S, et al. Sensitizing tumors to immune checkpoint blockage via STING agonists delivered by tumor‐penetrating neutrophil cytopharmaceuticals. ACS Nano. 2023. [DOI] [PubMed] [Google Scholar]
  • 370. Nakanishi T, Kunisawa J, Hayashi A, et al. Positively charged liposome functions as an efficient immunoadjuvant in inducing cell‐mediated immune response to soluble proteins. J Control Release. 1999;61(1‐2):233‐240. [DOI] [PubMed] [Google Scholar]
  • 371. Zolnik BS, Gonzalez‐Fernandez A, Sadrieh N, Dobrovolskaia MA. Minireview: nanoparticles and the immune system. Endocrinology. 2010;151(2):458‐465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 372. Yan W, Chen W, Huang L. Reactive oxygen species play a central role in the activity of cationic liposome based cancer vaccine. J Control Release. 2008;130(1):22‐28. [DOI] [PubMed] [Google Scholar]
  • 373. Nakamura T, Sato T, Endo R, et al. STING agonist loaded lipid nanoparticles overcome anti‐PD‐1 resistance in melanoma lung metastasis via NK cell activation. J Immunother Cancer. 2021;9(7):e002852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 374. Miyabe H, Hyodo M, Nakamura T, Sato Y, Hayakawa Y, Harashima H. A new adjuvant delivery system ‘cyclic di‐GMP/YSK05 liposome’ for cancer immunotherapy. J Control Release. 2014;184:20‐27. [DOI] [PubMed] [Google Scholar]
  • 375. Nakamura T, Miyabe H, Hyodo M, Sato Y, Hayakawa Y, Harashima H. Liposomes loaded with a STING pathway ligand, cyclic di‐GMP, enhance cancer immunotherapy against metastatic melanoma. J Control Release. 2015;216:149‐157. [DOI] [PubMed] [Google Scholar]
  • 376. Miao L, Li L, Huang Y, et al. Delivery of mRNA vaccines with heterocyclic lipids increases anti‐tumor efficacy by STING‐mediated immune cell activation. Nat Biotechnol. 2019;37(10):1174‐1185. [DOI] [PubMed] [Google Scholar]
  • 377. Covarrubias G, Moon TJ, Loutrianakis G, et al. Comparison of the uptake of untargeted and targeted immunostimulatory nanoparticles by immune cells in the microenvironment of metastatic breast cancer. J Mater Chem B. 2022;10(2):224‐235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 378. Batrakova EV, Kim MS. Using exosomes, naturally‐equipped nanocarriers, for drug delivery. J Control Release. 2015;219:396‐405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 379. van Dommelen SM, Vader P, Lakhal S, et al. Microvesicles and exosomes: opportunities for cell‐derived membrane vesicles in drug delivery. J Control Release. 2012;161(2):635‐644. [DOI] [PubMed] [Google Scholar]
  • 380. van der Meel R, Fens MHAM, Vader P, van Solinge WW, Eniola‐Adefeso O, Schiffelers RM. Extracellular vesicles as drug delivery systems: lessons from the liposome field. J Control Release. 2014;195:72‐85. [DOI] [PubMed] [Google Scholar]
  • 381. Rao L, Wu L, Liu Z, et al. Hybrid cellular membrane nanovesicles amplify macrophage immune responses against cancer recurrence and metastasis. Nat Commun. 2020;11(1):4909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 382. Jang SC, Economides KD, Moniz RJ, et al. ExoSTING, an extracellular vesicle loaded with STING agonists, promotes tumor immune surveillance. Commun Biol. 2021;4(1):497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 383. McAndrews KM, Che SPY, LeBleu VS, Kalluri R. Effective delivery of STING agonist using exosomes suppresses tumor growth and enhances antitumor immunity. J Biol Chem. 2021;296 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 384. Denisov IG, Sligar SG. Nanodiscs for structural and functional studies of membrane proteins. Nat Struct Mol Biol. 2016;23(6):481‐486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 385. Bayburt TH, Grinkova YV, Sligar SG. Self‐assembly of discoidal phospholipid bilayer nanoparticles with membrane scaffold proteins. Nano Lett. 2002;2(8):853‐856. [Google Scholar]
  • 386. Denisov IG, Grinkova YV, Lazarides AA, Sligar SG. Directed self‐assembly of monodisperse phospholipid bilayer nanodiscs with controlled size. J Am Chem Soc. 2004;126(11):3477‐3487. [DOI] [PubMed] [Google Scholar]
  • 387. Dane EL, Belessiotis‐Richards A, Backlund C, et al. STING agonist delivery by tumour‐penetrating PEG‐lipid nanodiscs primes robust anticancer immunity. Nat Mater. 2022;21(6):710‐+. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 388. Bariwal J, Ma HR, Altenberg GA, Liang HJ. Nanodiscs: a versatile nanocarrier platform for cancer diagnosis and treatment. Chem Soc Rev. 2022;51(5):1702‐1728. [DOI] [PubMed] [Google Scholar]
  • 389. Chen JJ, Qiu M, Ye ZF, et al. In situ cancer vaccination using lipidoid nanoparticles. Sci Adv. 2021;7(19). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 390. Cookenham T, Lanzer KG, Gage E, et al. Vaccination of aged mice with adjuvanted recombinant influenza nucleoprotein enhances protective immunity. Vaccine. 2020;38(33):5256‐5267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 391. Zheng C, Zhang W, Wang J, et al. Lenvatinib‐ and vadimezan‐loaded synthetic high‐density lipoprotein for combinational immunochemotherapy of metastatic triple‐negative breast cancer. Acta Pharm Sin B. 2022;12(9):3726‐3738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 392. Yuan BY, Wu CS, Wang XW, et al. High scavenger receptor class B type I expression is related to tumor aggressiveness and poor prognosis in breast cancer. Tumor Biol. 2016;37(3):3581‐3588. [DOI] [PubMed] [Google Scholar]
  • 393. Huang A, Zhou W. Mn‐based cGAS‐STING activation for tumor therapy. Chin J Cancer Res. 2023;35(1):19‐43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 394. Wang C, Sun Z, Zhao C, et al. Maintaining manganese in tumor to activate cGAS‐STING pathway evokes a robust abscopal anti‐tumor effect. J Control Release. 2021;331:480‐490. [DOI] [PubMed] [Google Scholar]
  • 395. Zheng C, Song Q, Zhao H, et al. A nanoplatform to boost multi‐phases of cancer‐immunity‐cycle for enhancing immunotherapy. J Control Release. 2021;339:403‐415. [DOI] [PubMed] [Google Scholar]
  • 396. Zhu C, Ma Q, Gong L, et al. Manganese‐based multifunctional nanoplatform for dual‐modal imaging and synergistic therapy of breast cancer. Acta Biomater. 2022;141:429‐439. [DOI] [PubMed] [Google Scholar]
  • 397. Hou L, Tian C, Yan Y, Zhang L, Zhang H, Zhang Z. Manganese‐based nanoactivator optimizes cancer immunotherapy via enhancing innate immunity. ACS Nano. 2020;14(4):3927‐3940. [DOI] [PubMed] [Google Scholar]
  • 398. Yan J, Wang G, Xie L, et al. Engineering radiosensitizer‐based metal‐phenolic networks potentiate STING pathway activation for advanced radiotherapy. Adv Mater. 2022;34(10):e2105783. [DOI] [PubMed] [Google Scholar]
  • 399. Paston SJ, Brentville VA, Symonds P, Durrant LG. Cancer vaccines, adjuvants, and delivery systems. Front Immunol. 2021;12:627932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 400. Atukorale PU, Moon TJ, Bokatch AR, et al. Dual agonist immunostimulatory nanoparticles combine with PD1 blockade for curative neoadjuvant immunotherapy of aggressive cancers. Nanoscale. 2022;14(4):1144‐1159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 401. Deng LF, Liang H, Xu M, et al. STING‐dependent cytosolic DNA sensing promotes radiation‐induced type I interferon‐dependent antitumor immunity in immunogenic tumors. Immunity. 2014;41(5):843‐852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 402. Park CG, Hartl CA, Schmid D, Carmona EM, Kim HJ, Goldberg MS. Extended release of perioperative immunotherapy prevents tumor recurrence and eliminates metastases. Sci Transl Med. 2018;10(433):eaar1916. [DOI] [PubMed] [Google Scholar]
  • 403. Baird JR, Friedman D, Cottam B, et al. Radiotherapy combined with novel STING‐targeting oligonucleotides results in regression of established tumors. Cancer Res. 2016;76(1):50‐61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 404. Gu Z, Hao Y, Schomann T, Ossendorp F, Ten Dijke P, Cruz LJ. Enhancing anti‐tumor immunity through liposomal oxaliplatin and localized immunotherapy via STING activation. J Control Release. 2023;357:531‐544. [DOI] [PubMed] [Google Scholar]
  • 405. Liu Y, Crowe WN, Wang LL, et al. An inhalable nanoparticulate STING agonist synergizes with radiotherapy to confer long‐term control of lung metastases. Nat Commun. 2019;10:5108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 406. Yang X, Bian J, Wang Z, et al. A bio‐liposome activating natural killer cell by illuminating tumor homogenization antigen properties. Adv Sci (Weinh). 2023;10(12):e2205449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 407. Pulendran B, Arunachalam PS, O'Hagan DT. Emerging concepts in the science of vaccine adjuvants. Nat Rev Drug Discov. 2021;20(6):454‐475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 408. Hanson MC, Crespo MP, Abraham W, et al. Nanoparticulate STING agonists are potent lymph node‐targeted vaccine adjuvants. J Clin Invest. 2015;125(6):2532‐2546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 409. Romero CD, Varma TK, Hobbs JB, Reyes A, Driver B, Sherwood ER. The Toll‐like receptor 4 agonist monophosphoryl lipid a augments innate host resistance to systemic bacterial infection. Infect Immun. 2011;79(9):3576‐3587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 410. Robert C. A decade of immune‐checkpoint inhibitors in cancer therapy. Nat Commun. 2020;11(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 411. Corrales L, McWhirter SM, Dubensky TW, Gajewski TF. The host STING pathway at the interface of cancer and immunity. J Clin Invest. 2016;126(7):2404‐2411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 412. Li AP, Yi M, Qin S, Song YP, Chu Q, Wu KM. Activating cGAS‐STING pathway for the optimal effect of cancer immunotherapy. J Hematol Oncol. 2019;12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 413. Fu J, Kanne DB, Leong M, et al. STING agonist formulated cancer vaccines can cure established tumors resistant to PD‐1 blockade. Sci Transl Med. 2015;7(283):283ra52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 414. Moore E, Clavijo PE, Davis R, et al. Established T cell‐inflamed tumors rejected after adaptive resistance was reversed by combination STING activation and PD‐1 pathway blockade. Cancer Immunol Res. 2016;4(12):1061‐1071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 415. Grabosch S, Bulatovic M, Zeng F, et al. Cisplatin‐induced immune modulation in ovarian cancer mouse models with distinct inflammation profiles. Oncogene. 2019;38(13):2380‐2393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 416. Zhu Y, An X, Zhang X, Qiao Y, Zheng T, Li X. STING: a master regulator in the cancer‐immunity cycle. Mol Cancer. 2019;18(1):152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 417. Lemos H, Mohamed E, Huang L, et al. STING promotes the growth of tumors characterized by low antigenicity via IDO activation. Cancer Res. 2016;76(8):2076‐2081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 418. Yi M, Niu M, Wu Y, et al. Combination of oral STING agonist MSA‐2 and anti‐TGF‐beta/PD‐L1 bispecific antibody YM101: a novel immune cocktail therapy for non‐inflamed tumors. J Hematol Oncol. 2022;15(1):142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 419. Jiang M, Chen P, Wang L, et al. cGAS‐STING, an important pathway in cancer immunotherapy. J Hematol Oncol. 2020;13(1):81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 420. Koshy ST, Cheung AS, Gu L, Graveline AR, Mooney DJ. Liposomal delivery enhances immune activation by STING agonists for cancer immunotherapy. Advanced Biosystems. 2017;1(1‐2):1600013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 421. Li KS, Ye YY, Liu LQ, et al. The lipid platform increases the activity of STING agonists to synergize checkpoint blockade therapy against melanoma. Biomater Sci UK. 2021;9(3):765‐773. [DOI] [PubMed] [Google Scholar]
  • 422. Cheng N, Watkins‐Schulz R, Junkins RD, et al. A nanoparticle‐incorporated STING activator enhances antitumor immunity in PD‐L1‐insensitive models of triple‐negative breast cancer. JCI Insight. 2018;3(22):e120638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 423. Khalifa AM, Nakamura T, Sato Y, et al. Interval‐ and cycle‐dependent combined effect of STING agonist loaded lipid nanoparticles and a PD‐1 antibody. Int J Pharm. 2022;624:122034. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Not applicable.


Articles from MedComm are provided here courtesy of Wiley

RESOURCES