2,572
Views
0
CrossRef citations to date
0
Altmetric
Review

Lipid Nanovesicle Platforms for Hepatocellular Carcinoma Precision Medicine Therapeutics: Progress and Perspectives

ORCID Icon & ORCID Icon
Article: 2313696 | Received 06 Jul 2023, Accepted 30 Jan 2024, Published online: 15 Feb 2024

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related mortality globally. HCC is highly heterogenous with diverse etiologies leading to different driver mutations potentiating unique tumor immune microenvironments. Current therapeutic options, including immune checkpoint inhibitors and combinations, have achieved limited objective response rates for the majority of patients. Thus, a precision medicine approach is needed to tailor specific treatment options for molecular subsets of HCC patients. Lipid nanovesicle platforms, either liposome- (synthetic) or extracellular vesicle (natural)-derived present are improved drug delivery vehicles which may be modified to contain specific cargos for targeting specific tumor sites, with a natural affinity for liver with limited toxicity. This mini-review provides updates on the applications of novel lipid nanovesicle-based therapeutics for HCC precision medicine and the challenges associated with translating this therapeutic subclass from preclinical models to the clinic.

Introduction

Hepatocellular Carcinoma (HCC) is a growing global public health burden. HCC is the sixth most common cancer globally, with > 900,000 cases each year, and the third highest in cancer-related mortality, with > 800,000 deaths each year.Citation1 HCC typically follows a sequalae of chronic liver disease, with the main etiologies including Hepatitis B and C virus (HBV & HCV) infection, alcohol-related disease, steatotic liver diseases (SLD) (e.g., metabolic dysfunction associated SLD [MASLD], diabetes mellitus, obesity), and toxin exposure (e.g., cigarette smoke, aflatoxin, liver fluke).Citation2 HCC has a dismal prognosis with a 5-year overall survival (OS) rate of ~ 15–20% and <18 months median survival with current therapeutic paradigms.Citation3 Very few patients are diagnosed at early stages where surgical resection/transplantation is feasible and nearly curative.Citation4 In fact, the vast majority of patients are diagnosed with advanced disease, limiting their options to systemic agents, including tyrosine kinase inhibitors (TKIs) and, more recently, immune checkpoint inhibitors (ICIs). Despite ICIs demonstrating improved OS of roughly six months over TKIs, such as Sorafenib, the benefit is still marginal with only 25–30% response rates in patients.Citation5–7

Therefore, novel targeted therapies used in conjunction with immunotherapy, in a precision-medicine based approach, may overcome HCC therapeutic resistance to ICIs in molecular subsets of patients. Lipid nanovesicle platforms, either liposome- (synthetic) or extracellular vesicle (natural)-derived, have demonstrated promise as drug delivery vehicles to the liver and for targeted cancer agents. These nanocarriers are ideal drug delivery vehicles which may be functionalized to harbor specific cargo molecules and “home” to specific tumor sites, with native affinity for liver with limited toxicity.Citation8,Citation9 This mini-review discusses HCC molecular subclasses and current treatment paradigms, along with applications of novel lipid nanovesicle-based therapeutics for HCC precision medicine with a focus on naturally-derived nanovesicle formulations, and the challenges associated with translating this new therapeutic subclass from preclinical models to the clinic.

Genetic heterogeneity and immunologic landscape

Over the last decade, next-generation sequencing technologies have been utilized to profile genetic drivers of HCC, guiding the path toward precision medicine therapeutics. Our current understanding of the HCC genomic landscape includes major somatic mutations in TERT (~50%; telomere maintenance; promoter mutation and gain-of-function [GOF]), TP53 (~30%; cell cycle control; missense/nonsense; loss-of-function [LOF]), CTNNB1 (~30%; Wnt/β-catenin signaling; missense; GOF), ARID1A (~10%; chromatin remodeling; truncating/missense; LOF), and TSC2 (~10%; cell growth; deletions; LOF).Citation10,Citation11 Less common molecular drivers include FGF19 (~10%), AXIN1 (~6%), MYC (~6%), APC (~5%), and MET (~2%).Citation10,Citation11 Some of these mutations may not be mutually exclusive; however, mutations in Wnt/β-catenin pathway members and TP53 tend to be mutually exclusive events.Citation12 This dichotomy also forms the foundation for defining the various molecular subclasses of HCC described here.

The two main molecular classification systems proposed are the G1-G6 system by Boyault et al.Citation13 and the S1-S3 subgroups by Hoshida et al.Citation14 Briefly, G1-G3 and S1-S2 subclasses represent proliferative/poorly differentiated tumors associated with chromosomal instability, high HBV viral load, and TP53 mutations, while G5-G6 and S3 subclasses represent non-proliferative/well-differentiated tumors associated with chromosomal stability, alcohol/HCV/NASH-driven HCC, and CTNNB1 mutations.Citation13,Citation14 More recently, HCC can be classified into inflamed (Hoshida S1-S2 subgroups) or non-inflamed (Hoshida S3) subgroups.Citation14,Citation15 The inflamed class of HCC (~25% of patients) demonstrates increased expression of gene signatures related to immune infiltration (i.e., cytotoxic T cells, tertiary lymphoid structures [TLS], IFN alpha and gamma signaling, and chemokines CXCL9, CXCL10), high immune checkpoint immunohistochemical expression, CTNNB1-mutated depleted, and enrichment of amplification in q13 locus (CCND1, FGF19).Citation15 Additionally, the inflamed class can be further subdivided into either immune-active or immune-exhausted, with the immune-active subclass representing high adaptive immunity gene expression with improved survival and reduced rates of recurrence. The immune exhausted subclass demonstrates activated stroma and immunosuppressive gene set signatures.Citation16 Overall, these classification systems illustrate how tumor genetics drive both tumoral heterogeneity and specific tumor microenvironments, which may be differentially susceptible to various systemic agents, and thus, may require tailored treatment options for patients informed by tissue and/or liquid biopsy.Citation17

Current treatment modalities and patient selection

For advanced HCC, current standard of care has shifted from the use of TKIs toward ICIs in the last decade. ICIs are monoclonal antibodies which block the interaction between immune checkpoint molecules (e.g., programmed death-ligand 1 [PDL1] on tumor cells interacting with programmed cell death protein 1 [PD1] on T cells) potentiating cytotoxic CD8+ T cell mediated tumor cell killing.Citation18 The IMbrave 150 trial demonstrated 19.2 months median survival with atezolizumab (anti-PDL1 antibody) plus bevacizumab (anti-VEGF antibody) compared to 13.4 months median survival with sorafenib (TKI).Citation19 Also, the HIMALAYA trial demonstrated 16.4 months median survival with the ICI combination of tremelimumab (anti-CTLA4 antibody) plus durvalumab (anti-PDL1 antibody) compared to 13.7 months median survival with sorafenib.Citation7 Moreover, the CARES-310 trial demonstrated 22.1 months median survival with camrelizumab (anti-PD1 antibody) plus the VEGFR2-targeted TKI rivoceranib compared to 15.2 months median survival with sorafenib.Citation20 However, despite the improved OS in ICI treated patients, response rates overall remain relatively low with only 25–30% of patients achieving objective response rates (ORR). Low ORRs are poorly understood but have been linked to patient tumor microenvironments with low tumor-infiltrating effector T lymphocyte density, high regulatory T cell density, and high expression of oncofetal genes.Citation21 Thus, to improve these response rates, an individualized treatment approach is warranted to guide therapeutic selection based on underlying genetic alterations. This may be aided by tissue or liquid biopsy for key drivers of HCC tumorigenesis.Citation17 However, an improved understanding of which genetic drivers influence the immune microenvironment resistant to ICI response is warranted for screening, along with needing an expanded arsenal of drugs targeting these underlying pathways to be used in conjunction with ICIs.

On a molecular basis, the Wnt/β-catenin pathway has been the most prominently studied pathway to evaluate ICI resistance, yet controversy remains whether all mutations in the pathway decrease immune infiltration to the same degree, and thus ICI resistance.Citation16, Citation21, Citation22 Moreover, despite studies demonstrating the feasibility of prospective tissue genotyping to identify clinically actionable driver mutations, very few patients receive personalized therapeutic intervention.Citation10 The major driver mutations in HCC are currently not actionableCitation23; therefore, efforts should be made to identify and stratify patients which may respond to current druggable targets, including FGF19/FGFR4, VEGF, TSC1/2, and MET inhibitors.Citation24,Citation25 Although none of these targets have shown clinical responses, these molecular events may be co-occurring in the background of strong drivers (e.g., TP53, CTNNB1), and thus a combination of therapeutics may need to be eventually employed. Thus, further studies are needed in clinically relevant animal models to determine the differential response of ICIs in combination with targeted therapy approaches in unique molecular subsets of HCC.

Synthetic lipid nanovesicle drug delivery platforms for HCC

Synthetic lipid nanovesicles have conventionally been nanoliposome-based formulations containing distinct molecular entities, including either RNA interference (RNAi) technologies or chemotherapeutic drugs. Nanoliposomes typically size range between 10 nm to 200 nm in diameter and are composed of a phospholipid bilayer with or without cholesterol, resulting in an aqueous interior and an outer hydrophobic exterior.Citation26 The main types of nanoliposomes include small unilamellar vesicles (<100 nm), large unilamellar (>100 nm), and multilamellar vesicles (>500 nm), with the former two more typically used for nanomedicine applications.Citation27 Excellent reviews elsewhere discuss preparation methodologies (e.g., reverse-phase evaporation, freeze-thaw method, vaporization technique, and others) of nanoliposome formulations.Citation28,Citation29 Briefly, the phospholipid characteristics (e.g., degrees of unsaturation, quantity of fatty acid moieties, and others) and the number of cholesterol molecules can affect the membrane configuration.Citation30,Citation31 Further modifications to the nanoliposomal structure include the addition of either polyethylene glycolCitation32 or surface ligands,Citation33 which avoids host immune system elimination and improves cellular targeting, respectively. For cellular uptake, nanovesicles are internalized typically through endocytosis or phagocytosis, with nanovesicle structure influencing which mechanistic process.Citation34 Efficient perfusion of the liver through its dual blood supply mediates optimal delivery, and lipid nanovesicle uptake is augmented due to its fenestrated endothelium. Additionally, opsonization by ApoE facilitates low-density lipoprotein (LDL) receptor (LDLR)-mediated uptake into hepatocytes (“endogenous targeting”), while engineering N-acetylgalactosamine (GalNAc)-PEG-lipid on the nanovesicle surface can target the asialoglycoprotein receptor (ASGPR) on hepatocytes (“exogenous targeting”), with both options providing efficient delivery to the liver.Citation35,Citation36 This well-characterized ApoE-LDLR endogenous hepatocyte targeting mechanism is the route by which Patisiran, the first FDA approved siRNA-based drug, facilitates its end-organ targeting to the liver and mechanism of action.Citation37 The remainder of this section will discuss the applications of nanoliposomes as targeted drug delivery vehicles in various preclinical models of HCC as potential precision medicine therapeutic platforms ().

Figure 1. Schematic representation of nanoliposome and extracellular vesicle loading strategies, cellular uptake mechanisms of these drug delivery vehicles, and clinical parameters to monitor for toxicity in patients. Figure made in BioRender.

Figure 1. Schematic representation of nanoliposome and extracellular vesicle loading strategies, cellular uptake mechanisms of these drug delivery vehicles, and clinical parameters to monitor for toxicity in patients. Figure made in BioRender.

The realization of using lipid nanovesicles as a targeted therapy delivery vehicle for liver cancer in humans was first achieved in 2013 by Tabernero and colleagues in their phase I study. This lipid nanovesicle (ALN-VSP) encapsulated siRNAs targeting vascular endothelial growth factor (VEGF) and kinesin spindle protein (KSP) to treat patients with liver metastases.Citation38 Tumor regression was achieved in nearly 50% of the patients in the trial. These results, demonstrating the safety, tolerability, ability to achieve target downregulation in the liver, and short-term clinical responses underscore the importance and potential of using lipid nanovesicles for HCC therapy.

Nanoliposomes encapsulating RNA interference (RNAi) platforms, such as small interfering RNAs (siRNAs), microRNAs (miRNAs), or messenger RNAs (mRNAs) have been administered as drug delivery systems in preclinical models of HCC with considerable success in terms of safety, tolerability, and treatment response. Various groups have attempted to use RNAi to either target oncogenic genes involved in cell cycle regulation and cell proliferation/death pathways, or directly inhibit driver mutations deemed to be traditionally “undruggable.” For reviews on how RNAi platforms are processed following cellular uptake, we refer the interested reader to the following reviews.Citation39–42 An example of directly targeting oncogenic factors is illustrated by work from Younis colleagues where they encapsulated both a siRNA to midkine (MK; a gene involved in many cellular pathways including apoptosis and angiogenesisCitation43 and the chemotherapeutic, sorafenib, into a nanoliposome functionalized to contain 3 components: 1) SP94 peptide (specific to HCC cells), 2) YSK05 lipid (increased cytotoxic effects and limited endosomal escape), and 3) specific phosphatidylcholine/cholesterol ratio (improves liposome stability).Citation44 They demonstrated both in vitro and in vivo that their nanoliposome had specific uptake to HCC cells over normal hepatocytes, potentiated sorafenib’s effects, and resulted in profound tumor regressions (~70%).Citation44,Citation45 Additionally, Woitok et al. delivered siRNA targeting Jun N-terminal kinase-2 (Jnk2), known to affect fibrosis progression, in lipid nanovesicle to mice with chronic liver disease and demonstrated decreased HCC premalignant nodules and a shift in the immune microenvironment of the diseased liver.Citation46 Moreover, targeting key cellular pathways in HCC with siRNAs has also been feasible as demonstrated by the work from Fitamant and colleagues.Citation47 They delivered nanovesicles containing siRNA to Yes-associated protein 1 (YAP), a key downstream transcriptional co-activator of Hippo signaling, resulting in tumor regression through directing hepatocyte differentiation to normal hepatocyte-like cells. Other groups have also delivered nanoliposomes containing siRNAs targeting PD-L1,Citation48 T cell immunoglobulin mucin-3Citation49 (Tim-3; immune checkpoint molecule), vascular endothelial growth factorCitation50 (VEGF; angiogenic factor), alpha-fetoproteinCitation51 (AFP; biomarker for HCC), cyclo-oxygenase-2Citation52 (COX-2; important for prostaglandin synthesis in inflammatory processes), hypoxia inducible factor 1 subunit alphaCitation53 (HIF1a), or RNA NCitation6methyladenosine (mCitation6A) reader protein YTHDF1Citation54 either alone or in combination with chemotherapeutics. Moreover, miRNAs can be packaged into nanoliposomes to target specific cellular pathways. For example, Zhao et al. loaded miR-375 and sorafenib in nanoliposomes to hinder autophagic processes and reduce tumor burden.Citation55 Lastly, mRNAs may also be packaged into nanovesicles for HCC therapy. Lai et al. demonstrated that delivery of IL-12 mRNA in nanovesicles reduced tumor burden and prolonged survival of transgenic MYC-induced HCC mice.Citation56 This effect was also associated with a shift toward a more anti-tumor immune microenvironment with increases in T helper cells and IFNγ expression.Citation56 Similar effects were seen with mRNA for OX40L encapsulated nanovesicles.Citation57 Overall, lipid nanoparticles provide an efficient platform to deliver both chemotherapeutics and gene therapy at subtoxic doses with high efficiency and stability.Citation44,Citation53

As previously discussed, modifying the outer shell of the nanoliposome can improve the delivery efficiency and targeting to the desired end organ. For targeting HCC cells specifically, various groups have functionalized nanoliposomes to target CXCR4 high expressing cells given its sorafenib resistance mechanisms. These studies have demonstrated reduced toxicity with targeted nanoparticles and synergistic effects when combined with chemotherapies, such as a sorafenib.Citation58–60 Additionally, GalNAc-conjugated nanovesicles have demonstrated considerable success in highly relevant animal models of molecular subsets of HCC with the nanoliposomes encapsulating siRNAs to oncogenic drivers, such as CTNNB1.Citation61,Citation62 Also, the lipid configuration and inclusion of PEG/mannose into the membrane can also affect targeting to different liver cell types.Citation63 Therefore, using targeting molecules on nanoliposome surface can improve the efficiency of tumor cell transfection and diminish off-target effects.

Moreover, another strategy is modifying the lipid composition of the liposome for controllable release of chemotherapeutic agents through stimuli responses. Examples of this include using either temperature sensitive,Citation64 pH responsive,Citation65,Citation66 photo-sensitive, magnetic-sensitive, or ultrasound-guided lipids.Citation67 In terms of temperature-sensitive lipids, Peng et al.Citation64 utilized PF127 (copolymer) which has temperature-sensitive properties and aids in degrading the nanoliposome following photothermal conversion of IR-780 (a near-infrared [NIR] dye) also contained on the nanoliposome surface. This combination of PF127 and IR-780 allowed for efficient doxorubicin and sorafenib release at the tumor site in vivo. Also, as illustrated by Li et al.,Citation65 interchanging the nanoliposome bilayer to include the cationic lipid (2E)-4-(dioleostearin)-amino-4-carbonyl-2-butenonic (DC), can allow for direct tumor cell internalization upon conformational change in the acidic tumor microenvironment, and subsequently release its cargo in the acidified endosome. This allowed for reduced drug toxicity and targeting of tumor cells over normal hepatocytes. Overall, the lipid composition can allow for improved pharmacokinetics and tumor cell internalization.

Extracellular vesicle-based drug delivery platforms for HCC

Extracellular vesicles (EVs) are lipid nanovesicles (50 nm to >2000 nm) which are spontaneously produced by nearly all mammalian cells and released into extracellular fluid as part of autocrine, paracrine, and endocrine cell-to-cell signaling circuits.Citation68 There are various EV subclasses, including exosomes (derived from endosomal membrane trafficking machinery), microvesicles (outward plasma membrane blebbings), and apoptotic bodies (from apoptotic processes). All EVs contain cargos comprising various membrane and soluble proteins, nucleic acid species, and metabolites, which are specific to their cell of origin. Once released into the extracellular milieu, EVs travel systemically until they make contact with and fuse with their target cell plasma membrane through various endocytic or phagocytic mechanisms.Citation69 The natural ability for EVs to avoid immune system clearance, systemically travel to end organs, and package cargos within lipid bilayers has made them an attractive tool for drug delivery. Through the use of nanomedicine platforms, EV mimetics are being translated to the clinic as novel drug delivery vehicles. Various researchers have developed different EV mimetic technologies, either through modifying parental cells (e.g., stem cells, fibroblasts, immune cells) and isolating their EVs for delivery, or ex vivo loading of cargo components into EVs. This section will explore applications of EV mimetics for HCC precision medicine in preclinical models (), and we refer to the reader to excellent reviews detailing techniques used for preparation of EV-based therapeutics, including their isolation and purification.Citation70–74

The main class of EV mimetics utilized for HCC targeted therapy are siRNA- encapsulated EVs, which target specific mRNAs encoding oncogenic signaling proteins. Various groups have identified target genes, which when suppressed, may synergize with ICIs. One target is CD38, a transmembrane protein which is aberrantly expressed in many tumors and associated with a pro-inflammatory tumor microenvironment, and has been shown to be associated ICI response.Citation75,Citation76 EVs isolated from bone marrow mesenchymal stem cells packaged with siRNA to CD38 (via electroporation) reduced HCC tumor burden, metastatic potential, repolarized macrophages from M2 (immunosuppressive) to M1 (pro-inflammatory) phenotype, and improved ICI response.Citation75 Other genes/pathways identified which have been targeted with siRNAs packaged in EVs, include components of the ferroptosis pathway (GPX4 and DHODH),Citation77 cell cycle regulation (CDK1),Citation78 JAK/STAT pathway (STAT6),Citation79 and NFkB pathway (p50 subunit).Citation80 Rather than directly targeting translation of molecules displayed on tumor cell surface mediating immunosuppression, another approach is targeting the underlying genetic mutation of the tumor cell. Matusda and colleagues designed an siRNA targeting CTNNB1 delivered within EVs.Citation81 Using the Met/β-catenin mouse model (which represents ~ 10% of human HCC), they remarkably demonstrated that delivery of milk-derived EVs encapsulating siRNA to CTNNB1 (using transfection techniques) reduced tumor burden, in part through reversing the immunosuppressive tumor microenvironment driven by β-catenin, which allowed for synergy with ICIs. Another group utilized a similar platform, but functionalized the EVs to target EpCAM-positive HCC cells.Citation82 These studies along with others previously mentionedCitation61, Citation62, Citation82 provide direct evidence that therapeutically targeting oncogenic mutations with siRNAs are effective approaches to treat HCC. And, using EVs may have improved RNA delivery efficiency, unique targeting capabilities, and enhanced biocompatibility compared to synthetic nanovesicle platforms.Citation83–85

Similar to siRNAs, miRNAs packaged into EVs offer another platform to target actively proliferating cancer cells. Many miRNAs have been implicated in HCC pathogenesis, including miR-21, miR-125b, miR-155, and miR-221/222.Citation86 Particularly, miR-125b down-regulation is associated with worse overall survival.Citation87 Baldari and colleagues isolated EVs (via polymer-based methods) from adipose-derived stromal cells (ADSCs) engineered to express miR-125b with a unique “ExoMotif” sequence that increases release of miR-125b into EVs.Citation88 These EVs were delivered in vitro to HepG2 and HuH-7 cells and reduced cell proliferation, along with expression of p53 signaling pathway components.Citation88 In another study, Mahati and colleagues loaded mesenchymal stem cell (MSC)-derived EVs with miR-26a (via electroporation) and observed impaired cell proliferation and migration in vitro, along with reduced tumor burden in subcutaneous HCC models.Citation89 Lastly, Ellipilli and colleagues demonstrated that combined Paclitaxel and miR-122 (liver specific miRNA; reduced levels shown in HCC) administration within GalNAc-EVs reduced tumor burden in multiple mice xenograft HCC models.Citation90 Complementary to RNAi, another strategy for EV therapeutics includes exogenous or endogenous small molecule and protein loading. Exogenous protein loading of EVs has been excellently reviewed elsewhere, but includes techniques such as mixing, sonication, electroporation, freeze-thaw cycles, and extrusion, with sonication and extrusion being the most efficient.Citation91–94 Monoclonal antibodies, nanobodies, and various cytokines can even be packaged into EVs to target specific immune checkpoint molecules to induce native immune activity.Citation95,Citation96 However, these techniques may damage the membrane integrity of EVs.Citation92,Citation97 Endogenous protein loading into EVs is a novel technique which hijacks cell signaling cascades to load particular payloads into EVs, which can be isolated, and subsequently administered as therapeutics. Different groups have utilized the ability of FK506 binding protein (FKBP) and FKBP12–rapamycin-binding (FRB) domain to heterodimerize following rapamycin administration.Citation98,Citation99 The FRB domain is fused to the protein of interest via a GGSGG linker, and the FKBP domain is fused to a canonical EV protein (e.g., CD81 or CD63) via the N-myristoylation sequence to facilitate protein entry into EVs. Cell lines can be modified to express these fusion proteins and EVs can be isolated and delivered in vivo for effective protein delivery.Citation98,Citation99 Small molecule/chemotherapeutic agent packaging into EVs have demonstrated potential, including the use of doxorubicin,Citation100 norcantharidin,Citation101 and sorafenib.Citation102 Additionally, Cas9 ribonucleoprotein can be packaged into EVs and delivered in vivo to liver, offering avenues for HCC gene therapy.Citation103–105 Overall, these methods of protein/small molecule packaging are appealing options for therapeutic delivery to liver.

In the last two decades, recombinant Adeno-associated viruses (AAVs) have been explored as gene delivery vehicles for cancer due to their ability to target many cell types and long-lasting gene expression.Citation106 More recently, EVs have been shown to be associate with isolated AAVs (termed “vexosomes”) during virus isolation from cell-culture media. These vexosomes have become an alternate gene delivery vehicle.Citation107,Citation108 Moreover, vexosomes protect AAVs from antibody neutralization, a major issue for AAV in vivo translation.Citation109 Khan et al. isolated AAV6-derived vexosomes (via ultracentrifugation) containing an inducible caspase 9 (iCasp9), which upon delivery with a prodrug (AP20187), results in impaired HCC cell proliferation in vitro and tumor cell death in vivo via apoptosis.Citation110 Overall, vexosomes are another gene therapy-based EV mimetic technology which are highly efficient delivery vehicles, require lower therapeutic doses than AAVs, and are not cumbersome to manufacture.

Lastly, EVs isolated from allogeneic or autologous cell sources are another therapeutic option for HCC. Kim and colleagues have demonstrated that EVs isolated from natural killer (NK) cells, which contain proteins important for mediating immunogenic cell death, can functionally impair HCC growth in vitro and in vivo.Citation111 These NK-EVs (isolated via ultracentrifugation) express granzyme B, FasL, and TRAIL and mediate apoptosis through inducing caspase-3, 7, 8, and 9 upon internalization in tumor cells. Additionally, another cell type with promise as a therapeutic source of EVs are ADSCs. Wu and colleagues revealed that ADSC-EVs (isolated via ultracentrifugation of culture media) decreased hepatic fibrosis and glutamine synthetase levels, suggesting that this may have therapeutic potential in subsets of HCC.Citation112 Moreover, another cell type which has demonstrated promise are dendritic cell (DC)-derived EVs. The pathogenesis of CTNNB1-mutated HCC involves defective recruitment of DCs,Citation113 likely making DC-EVs an interesting platform as an HCC therapeutic. Lu and colleagues systemically administered DC-EVs in three different HCC models and observed shifts in the tumor microenvironment such as increases in cytotoxic CD8 T-cells and fewer immunosuppressive T regulatory cells, which associated with tumor regression.Citation114 Lastly, M1 macrophages-derived EVs loaded with docosahexaenoic acid have been shown to induce ferroptosis and reduce tumor burden in orthotopic HCC models.Citation115 Therefore, EVs isolated from allogeneic sources have intrinsic capabilities to alter tumor cell survival and growth. However, autologous-derived EVs may have improved tumor targeting properties. Villa et al. illustrated that EVs derived from blood plasma of cancer patients had selective uptake into associated patient-derived xenograft (PDX) mouse models.Citation116 Therefore, autologous EV sources may be another pipeline for manufacture with improved tumor-specific targeting properties.

Challenges in good manufacturing practices for nanovesicle therapeutics

Many of the challenges of translating nanovesicle therapeutics are shared between synthetic and natural platforms; however, this section will focus on the nuances associated with translating EV-based therapeutics. The first consideration is isolation purity. Current clinical Good Manufacturing Processes (GMP) of therapeutic EVs may lead to downstream isolation of contaminants (e.g., viral) from cell culture supernatants.Citation117 For regulatory agency approval of EVs, a complete biochemical characterization is required for biologics, which remains incomplete due to technological limitations and EV isolation best practices.Citation117 Additionally, given EVs are a cell-free therapy, the mechanisms of cellular uptake/targeting, cargo delivery/release, and an understanding of the precise bioactive and nonactive components are unclear.Citation117,Citation118 Whether the membrane lipids/proteins, or the proteins/nucleic acids in the lumen, or both, contribute to the intended therapeutic effect is not determined. Therefore, extensive functional assays, “–omic,” and imaging platforms are needed to fully elucidate and differentiate the physiochemical properties and bioactivity of EVs. The International Society for Extracellular Vesicles (ISEV) has established guidelines for clinical GMP of therapeutic EVs.Citation119

The second consideration is cellular source and cell culture ecosystems of therapeutic EVs. As discussed in the previous section, cellular sources of therapeutic EVs for cancer can include either stem cells, immune cells, or nonparenchymal/stromal cells. Each of these cell types require different culture methods and release differing quantities of EVs. Additionally, cell culture practices of these cell types typically include utilizing fetal bovine serum (FBS) as a culture media supplement, which presents challenges due to introducing FBS-derived EVs into the pool of cell culture-derived EVs.Citation120 Simply, this contamination means that upon isolation of EVs from the cell culture supernatant, the final EV fraction will contain both EVs from the FBS and the cultured cells.Citation120 To circumvent these issues, the use of EV-depleted FBS or serum-free culture conditions have been proposed, with each providing their own inherent limitations, including cell death, incomplete elimination of FBS-derived EVs, and changes to cellular differentiation/state.Citation120,Citation121 Moreover, when culturing cells, the passage number, cell seeding density, and timing of media harvest can contribute to heterogeneity in cultured cells, and thus EVs isolated.Citation117,Citation122

The third consideration is the scale of manufacturing. For mass production of EVs, unique culture systems are needed, such as stacked culture vessels or bioreactors.Citation117,Citation118 Also, each EV isolation protocol (e.g., ultracentrifugation, precipitation, size-exclusion chromatography, and filtration) present differences in efficiency, quantity, purity, and quality of final EV formulations.Citation123 For example, although centrifugation-based approaches improve EV purity, this is at the expense of cost and time.Citation120 Lastly, with high-volume manufacturing, evaluating differences in batches is also important to consider.Citation117

Oncology clinical trials implementing nanovesicle platforms

The translation of lipid nanoparticles and EVs to clinical practice as HCC therapies has not moved swiftly. Currently, EVs are being studied as diagnostic biomarkersCitation124 for HCC to detect initial diagnosis, response to therapy, and disease recurrenceCitation125 using DNA mutationsCitation126, Citation127 or methylationCitation128 patterns, mRNACitation129/miRNA signatures,Citation130 or proteinsCitation131, Citation132 encapsulated in their lumen. This section will briefly cover in-human studies in oncology which has successfully translated nanovesicle therapeutic platforms to the clinic. To investigate whether lipid nanovesicles were actively being translated into clinical trials, we surveyed the clinicaltrials.gov website to search for active or terminated trials. A review of the clinicaltrials.gov website for clinical trials related to “cancer” and “exosomes” yielded 132 studies, with 7 unique studies focusing on therapeutic applications (). Additionally, a review for clinical trials related to “cancer” and “nanovesicle” yielded 12 studies, with 7 unique studies focusing on therapeutic applications (). Overall, there are few trials investigating the therapeutic potential of lipid nanovesicle platforms in HCC space. Notably, Omega Therapeutics is leading their phase I/II MYCHELANGELO™ trial (NCT05497453) evaluating OTX-2002 as monotherapy or in combination with HCC standard of care (TKIs or ICIs), which is an mRNA therapeutic encapsulated in lipid nanovesicle which decreases c-MYC gene expression through modifying the c-Myc transcript via epigenetic modulation.Citation133 They most recently (September 2023) have described preliminary results in 8 patients and observed on-target effects with associated decreases in c-MYC gene expression.Citation134 This signals the transition of siRNA/mRNA lipid nanovesicle therapeutics from the preclinical to clinical realm to target traditionally “undruggable” oncogenic drivers to be used in conjunction with standard of care agents (i.e., TKIs or ICIs).

Table 1. Clinical trials registered on clinicaltrials.Gov website for use of lipid nanovesicles and extracellular vesicles in oncology.

Conclusions and future perspectives

Lipid nanovesicles are next-generation drug delivery vehicles swiftly becoming part of the oncologist armamentarium. Compared to the administration of “naked” drug, encapsulated drug within lipid nanovesicles allows for reduced toxicity, improved biocompatibility, and improved in vivo efficacy through enhanced delivery to end-organ and target cell internalization. Several studies have illuminated the potential of lipid nanovesicles, both synthetic and natural, as drug delivery platforms in preclinical models and in patients, with several companies licensing these technologies from academia and translating their products to the clinic. These platforms are ideal drug delivery vehicles for treating various liver pathologies, including cancer, due to the liver’s inherent dual blood supply and fenestrated endothelium to allow for efficient systemic administration and hepatocyte delivery, respectively. Also, these nanovesicles are opsonized by ApoE and recognized by the hepatocyte LDLR for efficient targeting. Or functionalization of the nanovesicle may allow for directed cell-type specificity.Citation135

There are distinct advantages and disadvantages of each platform (). To improve the translation of this new EV class of biologics to the clinic, there are several technical challenges, including improving isolation techniques, component characterization, and manufacturing.Citation70,Citation117 Additionally, an enhanced understanding of the factors lending toward high biocompatibility of EVs may augment the development and translation of synthetic nanovesicles.Citation117 Despite these challenges, the future is bright for nanovesicle therapeutic applications in oncology, particularly EVs, and as technology advances, these roadblocks will only become surpassed and push these biologics toward clinical practice. For translation of EV therapeutics, lessons may be learned from some of the hurdles overcome by those involved in translating nanoliposome formulations.Citation136 For example, for nanoliposomes, great detail was undertaken to understand how the composition of ionizable lipids, various active drug loading techniques, and the cholesterol composition in the membrane affected drug stability, and thus enhanced in vivo activity.Citation137 Additionally, the size of the nanovesicle plays an important role in the ability to target the liver (and specific cell-type), with studies concluding <100 nm is ideal for hepatocyte delivery.Citation136,Citation138 Interrogation of all these different tunable characteristics of nanovesicles for EV-based drug delivery vehicles will ultimately improve their translatability to the clinic.

Table 2. Advantages and disadvantages of different nanovesicle platforms for liver cancer.

Acknowledgments

The authors would like to acknowledge the University of Pittsburgh School of Medicine Cell Therapy Course directed by Dr. Alejandro Soto-Gutierrez and Dr. Diana Metes. The authors would also like to acknowledge support from Dr. Satdarshan P. Monga.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Additional information

Funding

This work was supported by the National Institutes of Health under Grants 1T32EB001026 & 1F30CA284540 to BML and 1K22CA258677 to ERD.

References

  • Rumgay H, Arnold M, Ferlay J, Lesi O, Cabasag CJ, Vignat J, Laversanne M, McGlynn KA, Soerjomataram I. Global burden of primary liver cancer in 2020 and predictions to 2040. J Hepatol. 2022;77(6):1598–17. doi:10.1016/j.jhep.2022.08.021.
  • Singal AG, Lampertico P, Nahon P. Epidemiology and surveillance for hepatocellular carcinoma: new trends. J Hepatol. 2020;72(2):250–61. doi:10.1016/j.jhep.2019.08.025.
  • Llovet JM, Kelley RK, Villanueva A, Singal AG, Pikarsky E, Roayaie S, Lencioni R, Koike K, Zucman-Rossi J, Finn RS. et al. Hepatocellular carcinoma. Nat Rev Dis Primers. 2021;7(1):6. doi:10.1038/s41572-020-00240-3.
  • Galle PR, Forner A, Llovet JM, Mazzaferro V, Piscaglia F, Raoul J-L, Schirmacher P, Vilgrain V, European Association for the Study of the Liver. Electronic address eee, European association for the study of the L. EASL clinical practice guidelines: management of hepatocellular carcinoma. J Hepatol. 2018;69(1):182–236. doi:10.1016/j.jhep.2018.03.019.
  • Cheng AL, Qin S, Ikeda M, Galle PR, Ducreux M, Kim TY, Lim HY, Kudo M, Breder V, Merle P. et al. Updated efficacy and safety data from IMbrave150: Atezolizumab plus bevacizumab vs. sorafenib for unresectable hepatocellular carcinoma. J Hepatol. 2022;76(4):862–73. doi:10.1016/j.jhep.2021.11.030.
  • Bejjani AC, Finn RS. Hepatocellular carcinoma: pick the winner—tyrosine kinase inhibitor versus immuno-oncology agent–based combinations. J Clin Oncol. 2022;40(24):2763–73. doi:10.1200/JCO.21.02605.
  • Abou-Alfa GK, Lau G, Kudo M, Chan SL, Kelley RK, Furuse J, Sukeepaisarnjaroen W, Kang Y-K, Van Dao T, De Toni EN. et al. Tremelimumab plus durvalumab in unresectable hepatocellular carcinoma. NEJM Evidence. 2022;1(8):EVIDoa2100070. doi:10.1056/EVIDoa2100070.
  • Zhang G, Huang X, Xiu H, Sun Y, Chen J, Cheng G, Song Z, Peng Y, Shen Y, Wang J. et al. Extracellular vesicles: natural liver-accumulating drug delivery vehicles for the treatment of liver diseases. J Extracell Vesicles. 2020;10(2):e12030. doi:10.1002/jev2.12030.
  • Zhu Y, Shen R, Vuong I, Reynolds RA, Shears MJ, Yao ZC, Hu Y, Cho WJ, Kong J, Reddy SK. et al. Multi-step screening of DNA/lipid nanoparticles and co-delivery with siRNA to enhance and prolong gene expression. Nat Commun. 2022;13(1):4282. doi:10.1038/s41467-022-31993-y.
  • Harding JJ, Nandakumar S, Armenia J, Khalil DN, Albano M, Ly M, Shia J, Hechtman JF, Kundra R, El Dika I. et al. Prospective genotyping of hepatocellular carcinoma: clinical implications of next-generation sequencing for matching patients to targeted and immune therapies. Clin Cancer Res. 2019;25(7):2116–26. doi:10.1158/1078-0432.CCR-18-2293.
  • Rebouissou S, Nault JC. Advances in molecular classification and precision oncology in hepatocellular carcinoma. J Hepatol. 2020;72(2):215–29. doi:10.1016/j.jhep.2019.08.017.
  • Guichard C, Amaddeo G, Imbeaud S, Ladeiro Y, Pelletier L, Maad IB, Calderaro J, Bioulac-Sage P, Letexier M, Degos F. et al. Integrated analysis of somatic mutations and focal copy-number changes identifies key genes and pathways in hepatocellular carcinoma. Nat Genet. 2012;44(6):694–8. doi:10.1038/ng.2256.
  • Boyault S, Rickman DS, de Reynies A, Balabaud C, Rebouissou S, Jeannot E, Hérault A, Saric J, Belghiti J, Franco D. et al. Transcriptome classification of HCC is related to gene alterations and to new therapeutic targets. Hepatology. 2007;45(1):42–52. doi:10.1002/hep.21467.
  • Hoshida Y, Nijman SM, Kobayashi M, Chan JA, Brunet JP, Chiang DY, Villanueva A, Newell P, Ikeda K, Hashimoto M. et al. Integrative transcriptome analysis reveals common molecular subclasses of human hepatocellular carcinoma. Cancer Res. 2009;69(18):7385–92. doi:10.1158/0008-5472.CAN-09-1089.
  • Sia D, Jiao Y, Martinez-Quetglas I, Kuchuk O, Villacorta-Martin C, Castro de Moura M, Putra J, Camprecios G, Bassaganyas L, Akers N. et al. Identification of an Immune-specific Class of Hepatocellular Carcinoma, Based on Molecular Features. Gastroenterology. 2017;153(3):812–26. doi:10.1053/j.gastro.2017.06.007.
  • Montironi C, Castet F, Haber PK, Pinyol R, Torres-Martin M, Torrens L, Mesropian A, Wang H, Puigvehi M, Maeda M. et al. Inflamed and non-inflamed classes of HCC: a revised immunogenomic classification. Gut. 2023;72(1):129–40. doi:10.1136/gutjnl-2021-325918.
  • Lehrich BM, Zhang J, Monga SP, Dhanasekaran R. Battle of the biopsies: role of tissue and liquid biopsy in hepatocellular carcinoma. J Hepatol. 2023. doi:10.1016/j.jhep.2023.11.030.
  • 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(1):223–49. doi:10.1146/annurev-pathol-042020-042741.
  • Finn RS, Qin S, Ikeda M, Galle PR, Ducreux M, Kim TY, Kudo M, Breder V, Merle P, Kaseb AO. et al. Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma. N Engl J Med. 2020;382(20):1894–905. doi:10.1056/NEJMoa1915745.
  • Qin S, Chan SL, Gu S, Bai Y, Ren Z, Lin X, Chen Z, Jia W, Jin Y, Guo Y. et al. Camrelizumab plus rivoceranib versus sorafenib as first-line therapy for unresectable hepatocellular carcinoma (CARES-310): a randomised, open-label, international phase 3 study. Lancet. 2023;402(10408):1133–46. doi:10.1016/S0140-6736(23)00961-3.
  • Zhu AX, Abbas AR, de Galarreta MR, Guan Y, Lu S, Koeppen H, Zhang W, Hsu C-H, He AR, Ryoo B-Y. et al. Molecular correlates of clinical response and resistance to atezolizumab in combination with bevacizumab in advanced hepatocellular carcinoma. Nat Med. 2022;28(8):1599–611. doi:10.1038/s41591-022-01868-2.
  • Chen L, Zhou Q, Liu J, Zhang W. CTNNB1 alternation is a potential biomarker for immunotherapy prognosis in patients with hepatocellular carcinoma. Front Immunol. 2021;12:759565. doi:10.3389/fimmu.2021.759565.
  • Zucman-Rossi J, Villanueva A, Nault JC, Llovet JM. Genetic landscape and biomarkers of hepatocellular carcinoma. Gastroenterology. 2015;149(5):1226–39 e4. doi:10.1053/j.gastro.2015.05.061.
  • Schulze K, Imbeaud S, Letouze E, Alexandrov LB, Calderaro J, Rebouissou S, Couchy G, Meiller C, Shinde J, Soysouvanh F. et al. Exome sequencing of hepatocellular carcinomas identifies new mutational signatures and potential therapeutic targets. Nat Genet. 2015;47(5):505–11. doi:10.1038/ng.3252.
  • Llovet JM, Montal R, Sia D, Finn RS. Molecular therapies and precision medicine for hepatocellular carcinoma. Nat Rev Clin Oncol. 2018;15(10):599–616. doi:10.1038/s41571-018-0073-4.
  • Torchilin VP. Recent advances with liposomes as pharmaceutical carriers. Nat Rev Drug Discov. 2005;4(2):145–60. doi:10.1038/nrd1632.
  • Samad A, Sultana Y, Aqil M. Liposomal drug delivery systems: an update review. Curr Drug Deliv. 2007;4(4):297–305. doi:10.2174/156720107782151269.
  • Vemuri S, Rhodes CT. Preparation and characterization of liposomes as therapeutic delivery systems: a review. Pharm Acta Helv. 1995;70(2):95–111. doi:10.1016/0031-6865(95)00010-7.
  • Mozafari MR. Liposomes: an overview of manufacturing techniques. Cell Mol Biol Lett. 2005;10:711–19.
  • Maruyama K, Kennel SJ, Huang L. Lipid composition is important for highly efficient target binding and retention of immunoliposomes. Proc Natl Acad Sci U S A. 1990;87(15):5744–8. doi:10.1073/pnas.87.15.5744.
  • Immordino ML, Dosio F, Cattel L. Stealth liposomes: review of the basic science, rationale, and clinical applications, existing and potential. Int J Nanomedicine. 2006;1:297–315.
  • Torchilin VP, Omelyanenko VG, Papisov MI, Bogdanov AA Jr., Trubetskoy VS, Herron JN, Gentry CA. Poly(ethylene glycol) on the liposome surface: on the mechanism of polymer-coated liposome longevity. Biochimica et Biophysica Acta (BBA) - Biomembranes. 1994;1195(1):11–20. doi:10.1016/0005-2736(94)90003-5.
  • Noble GT, Stefanick JF, Ashley JD, Kiziltepe T, Bilgicer B. Ligand-targeted liposome design: challenges and fundamental considerations. Trends Biotechnol. 2014;32(1):32–45. doi:10.1016/j.tibtech.2013.09.007.
  • Nagayasu A, Uchiyama K, Kiwada H. The size of liposomes: a factor which affects their targeting efficiency to tumors and therapeutic activity of liposomal antitumor drugs. Adv Drug Deliv Rev. 1999;40(1–2):75–87. doi:10.1016/S0169-409X(99)00041-1.
  • Akinc A, Querbes W, De S, Qin J, Frank-Kamenetsky M, Jayaprakash KN, Jayaraman M, Rajeev KG, Cantley WL, Dorkin JR. et al. Targeted delivery of RNAi therapeutics with endogenous and exogenous ligand-based mechanisms. Mol Ther. 2010;18(7):1357–64. doi:10.1038/mt.2010.85.
  • Kasiewicz LN, Biswas S, Beach A, Ren H, Dutta C, Mazzola AM, Rohde E, Chadwick A, Cheng C, Garcia SP. et al. GalNAc-lipid nanoparticles enable non-LDLR dependent hepatic delivery of a CRISPR base editing therapy. Nat Commun. 2023;14(1):2776. doi:10.1038/s41467-023-37465-1.
  • Suhr OB, Coelho T, Buades J, Pouget J, Conceicao I, Berk J, Schmidt H, Waddington-Cruz M, Campistol JM, Bettencourt BR. et al. Efficacy and safety of patisiran for familial amyloidotic polyneuropathy: a phase II multi-dose study. Orphanet J Rare Dis. 2015;10(1):109. doi:10.1186/s13023-015-0326-6.
  • Tabernero J, Shapiro GI, LoRusso PM, Cervantes A, Schwartz GK, Weiss GJ, Paz-Ares L, Cho DC, Infante JR, Alsina M. et al. First-in-humans trial of an RNA interference therapeutic targeting VEGF and KSP in cancer patients with liver involvement. Cancer Discov. 2013;3(4):406–17. doi:10.1158/2159-8290.CD-12-0429.
  • Wilson RC, Doudna JA. Molecular mechanisms of RNA interference. Annu Rev Biophys. 2013;42(1):217–39. doi:10.1146/annurev-biophys-083012-130404.
  • Castanotto D, Rossi JJ. The promises and pitfalls of RNA-interference-based therapeutics. Nature. 2009;457(7228):426–33. doi:10.1038/nature07758.
  • Pai SI, Lin YY, Macaes B, Meneshian A, Hung CF, Wu TC. Prospects of RNA interference therapy for cancer. Gene Ther. 2006;13(6):464–77. doi:10.1038/sj.gt.3302694.
  • Hou B, Qin L, Huang L. Liver cancer cells as the model for developing liver-targeted RNAi therapeutics. Biochem Biophys Res Commun. 2023;644:85–94. doi:10.1016/j.bbrc.2023.01.007.
  • Zhong J, Huang HL, Li J, Qian FC, Li LQ, Niu PP, Dai L-C. Development of hybrid-type modified chitosan derivative nanoparticles for the intracellular delivery of midkine-siRNA in hepatocellular carcinoma cells. Hepatobiliary Pancreat Dis Int. 2015;14(1):82–89. doi:10.1016/S1499-3872(15)60336-8.
  • Younis MA, Khalil IA, Abd Elwakil MM, Harashima H. A multifunctional lipid-based nanodevice for the highly specific codelivery of Sorafenib and Midkine siRNA to hepatic cancer cells. Mol Pharm. 2019;16(9):4031–44. doi:10.1021/acs.molpharmaceut.9b00738.
  • Younis MA, Khalil IA, Elewa YHA, Kon Y, Harashima H. Ultra-small lipid nanoparticles encapsulating sorafenib and midkine-siRNA selectively-eradicate sorafenib-resistant hepatocellular carcinoma in vivo. J Control Release. 2021;331:335–49. doi:10.1016/j.jconrel.2021.01.021.
  • Woitok MM, Zoubek ME, Doleschel D, Bartneck M, Mohamed MR, Kiessling F, Lederle W, Trautwein C, Cubero FJ. Lipid-encapsulated siRNA for hepatocyte-directed treatment of advanced liver disease. Cell Death Disease. 2020;11(5):343. doi:10.1038/s41419-020-2571-4.
  • Fitamant J, Kottakis F, Benhamouche S, Tian HS, Chuvin N, Parachoniak CA, Nagle J, Perera R, Lapouge M, Deshpande V. et al. YAP inhibition restores hepatocyte differentiation in advanced HCC, leading to tumor Regression. Cell Rep. 2015;10(10):1692–707. doi:10.1016/j.celrep.2015.02.027.
  • Huang KW, Hsu FF, Qiu JT, Chern GJ, Lee YA, Chang CC, Huang Y-T, Sung Y-C, Chiang C-C, Huang R-L. et al. Highly efficient and tumor-selective nanoparticles for dual-targeted immunogene therapy against cancer. Sci Adv. 2020;6(3):eaax5032. doi:10.1126/sciadv.aax5032.
  • Song C, Zhang J, Wen R, Li Q, Zhou J, Xiaoli L, Wu Z, Lv Y, Wu R. Improved anti-hepatocellular carcinoma effect by enhanced Co-delivery of Tim-3 siRNA and sorafenib via multiple pH triggered drug-eluting nanoparticles. Mater Today Bio. 2022;16:100350. doi:10.1016/j.mtbio.2022.100350.
  • Yao Y, Wang T, Liu Y, Zhang N. Co-delivery of sorafenib and VEGF-siRNA via pH-sensitive liposomes for the synergistic treatment of hepatocellular carcinoma. Artif Cells, Nanomed Biotechnol. 2019;47(1):1374–83. doi:10.1080/21691401.2019.1596943.
  • Punuch K, Wongwan C, Jantana S, Somboonyosdech C, Rodponthukwaji K, Kunwong N, Nguyen KT, Sirivatanauksorn V, Sirivatanauksorn Y, Srisawat C. et al. Study of siRNA delivery via polymeric nanoparticles in combination with angiogenesis inhibitor for the treatment of AFP-Related liver cancer. Int J Mol Sci. 2022;23(20):23. doi:10.3390/ijms232012666.
  • Jamal F, Ahmed G, Farazuddin M, Altaf I, Farheen S, Zia Q, Azhar A, Ahmad H, Khan AA, Somavarapu S. et al. Potential of siRNA-bearing subtilosomes in the treatment of diethylnitrosamine-induced hepatocellular carcinoma. Molecules. 2023;28(5):28. doi:10.3390/molecules28052191.
  • Chen Y, Zhao D, Xiao F, Li X, Li J, Su Z, Jiang X. Microfluidics-enabled Serial Assembly of Lipid-siRNA-sorafenib Nanoparticles for Synergetic Hepatocellular Carcinoma Therapy. Adv Mater. 2023;35(13):e2209672. doi:10.1002/adma.202209672.
  • Wang L, Zhu L, Liang C, Huang X, Liu Z, Huo J, Zhang Y, Zhang Y, Chen L, Xu H. et al. Targeting N6-methyladenosine reader YTHDF1 with siRNA boosts antitumor immunity in NASH-HCC by inhibiting EZH2-IL-6 axis. J Hepatol. 2023;79(5):1185–200. doi:10.1016/j.jhep.2023.06.021.
  • Zhao P, Li M, Wang Y, Chen Y, He C, Zhang X, Yang T, Lu Y, You J, Lee RJ. et al. Enhancing anti-tumor efficiency in hepatocellular carcinoma through the autophagy inhibition by miR-375/sorafenib in lipid-coated calcium carbonate nanoparticles. Acta Biomater. 2018;72:248–55. doi:10.1016/j.actbio.2018.03.022.
  • Lai I, Swaminathan S, Baylot V, Mosley A, Dhanasekaran R, Gabay M, Felsher DW. Lipid nanoparticles that deliver IL-12 messenger RNA suppress tumorigenesis in MYC oncogene-driven hepatocellular carcinoma. J Immunother Cancer. 2018;6(1):125. doi:10.1186/s40425-018-0431-x.
  • Deng Z, Yang H, Tian Y, Liu Z, Sun F, Yang P. An OX40L mRNA vaccine inhibits the growth of hepatocellular carcinoma. Front Oncol. 2022;12:975408. doi:10.3389/fonc.2022.975408.
  • Liu JY, Chiang T, Liu CH, Chern GG, Lin TT, Gao DY, Chen Y. Delivery of siRNA using CXCR4-targeted nanoparticles modulates tumor microenvironment and achieves a potent antitumor response in liver cancer. Mol Ther. 2015;23(11):1772–82. doi:10.1038/mt.2015.147.
  • Gao DY, Lin Ts T, Sung YC, Liu YC, Chiang WH, Chang CC, Liu J-Y, Chen Y. CXCR4-targeted lipid-coated PLGA nanoparticles deliver sorafenib and overcome acquired drug resistance in liver cancer. Biomaterials. 2015;67:194–203. doi:10.1016/j.biomaterials.2015.07.035.
  • Zheng N, Liu W, Li B, Nie H, Liu J, Cheng Y, Wang J, Dong H, Jia L. Co-delivery of sorafenib and metapristone encapsulated by CXCR4-targeted PLGA-PEG nanoparticles overcomes hepatocellular carcinoma resistance to sorafenib. J Exp Clin Cancer Res. 2019;38(1):232. doi:10.1186/s13046-019-1216-x.
  • Ganesh S, Koser ML, Cyr WA, Chopda GR, Tao J, Shui X, Ying B, Chen D, Pandya P, Chipumuro E. et al. Direct pharmacological inhibition of β-catenin by RNA interference in tumors of diverse origin. Mol Cancer Ther. 2016;15(9):2143–54. doi:10.1158/1535-7163.MCT-16-0309.
  • Tao J, Zhang R, Singh S, Poddar M, Xu E, Oertel M, Chen X, Ganesh S, Abrams M, Monga SP. et al. Targeting β-catenin in hepatocellular cancers induced by coexpression of mutant β-catenin and K-Ras in mice. Hepatology. 2017;65(5):1581–99. doi:10.1002/hep.28975.
  • Kim M, Jeong M, Hur S, Cho Y, Park J, Jung H, Seo Y, Woo HA, Nam KT, Lee K. et al. Engineered ionizable lipid nanoparticles for targeted delivery of RNA therapeutics into different types of cells in the liver. Sci Adv. 2021;7(9). doi:10.1126/sciadv.abf4398.
  • Peng Y, Su Z, Wang X, Wu T, Xiao H, Shuai X, Lan Y. Near-infrared light laser-triggered release of doxorubicin and sorafenib from TemperatureSensitive liposomes for synergistic therapy of hepatocellular carcinoma. J Biomed Nanotechnol. 2020;16(9):1381–93. doi:10.1166/jbn.2020.2975.
  • Li Y, Miao Y, Chen M, Chen X, Li F, Zhang X, Gan Y. Stepwise targeting and responsive lipid-coated nanoparticles for enhanced tumor cell sensitivity and hepatocellular carcinoma therapy. Theranostics. 2020;10(8):3722–36. doi:10.7150/thno.42008.
  • Su Y, Zhang Z, Lee LTO, Peng L, Lu L, He X, Zhang X. Amphiphilic dendrimer doping enhanced pH-sensitivity of liposomal vesicle for effective Co-delivery toward synergistic ferroptosis–apoptosis therapy of hepatocellular carcinoma. Adv Healthcare Mater. 2023;12(6):e2202663. doi:10.1002/adhm.202202663.
  • Mahmoud K, Swidan S, El-Nabarawi M, Teaima M. Lipid based nanoparticles as a novel treatment modality for hepatocellular carcinoma: a comprehensive review on targeting and recent advances. J Nanobiotechnology. 2022;20(1):109. doi:10.1186/s12951-022-01309-9.
  • van Niel G, D’Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 2018;19(4):213–28. doi:10.1038/nrm.2017.125.
  • Mulcahy LA, Pink RC, Carter DR. Routes and mechanisms of extracellular vesicle uptake. J Extracell Vesicles. 2014;3(1). doi:10.3402/jev.v3.24641.
  • Cheng L, Hill AF. Therapeutically harnessing extracellular vesicles. Nat Rev Drug Discov. 2022;21(5):379–99. doi:10.1038/s41573-022-00410-w.
  • Murphy DE, de Jong OG, Brouwer M, Wood MJ, Lavieu G, Schiffelers RM, Vader P. Extracellular vesicle-based therapeutics: natural versus engineered targeting and trafficking. Experimental & Molecular Medicine. 2019;51(3):1–12. doi:10.1038/s12276-019-0223-5.
  • Claridge B, Lozano J, Poh QH, Greening DW. Development of extracellular vesicle therapeutics: challenges, considerations, and opportunities. Front Cell Dev Biol. 2021;9:734720. doi:10.3389/fcell.2021.734720.
  • Esmaeili A, Alini M, Baghaban Eslaminejad M, Hosseini S. Engineering strategies for customizing extracellular vesicle uptake in a therapeutic context. Stem Cell Res Ther. 2022;13(1):129. doi:10.1186/s13287-022-02806-2.
  • Nedeva C, Mathivanan S. Engineering Extracellular Vesicles for Cancer Therapy. Subcell Biochem. 2021;97:375–92.
  • Deng J, Ke H. Overcoming the resistance of hepatocellular carcinoma to PD-1/PD-L1 inhibitor and the resultant immunosuppression by CD38 siRNA-loaded extracellular vesicles. Oncoimmunology. 2023;12(1):2152635. doi:10.1080/2162402X.2022.2152635.
  • HHM N, Lee RY, Goh S, Tay ISY, Lim X, Lee B, Chew V, Li H, Tan B, Lim S. et al. Immunohistochemical scoring of CD38 in the tumor microenvironment predicts responsiveness to anti-PD-1/PD-L1 immunotherapy in hepatocellular carcinoma. J Immunother Cancer. 2020;8(2):e000987. doi:10.1136/jitc-2020-000987.
  • Li X, Yu Q, Zhao R, Guo X, Liu C, Zhang K, Zhang W, Liu J, Yu J, Wang S. et al. Designer exosomes for targeted delivery of a novel therapeutic cargo to enhance sorafenib-mediated ferroptosis in hepatocellular carcinoma. Front Oncol. 2022;12:898156. doi:10.3389/fonc.2022.898156.
  • Zhou X, Miao Y, Wang Y, He S, Guo L, Mao J, Chen M, Yang Y, Zhang X, Gan Y. et al. Tumour-derived extracellular vesicle membrane hybrid lipid nanovesicles enhance siRNA delivery by tumour-homing and intracellular freeway transportation. J Extracell Vesicles. 2022;11(3):e12198. doi:10.1002/jev2.12198.
  • Kamerkar S, Leng C, Burenkova O, Jang SC, McCoy C, Zhang K, Dooley K, Kasera S, Zi T, Sisó S. et al. Exosome-mediated genetic reprogramming of tumor-associated macrophages by exoASO-STAT6 leads to potent monotherapy antitumor activity. Sci Adv. 2022;8(7):eabj7002. doi:10.1126/sciadv.abj7002.
  • 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:10.1016/j.biomaterials.2021.121137.
  • Matsuda A, Ishiguro K, Yan IK, Patel T. Extracellular Vesicle-Based Therapeutic Targeting of β-Catenin to Modulate Anticancer Immune Responses in Hepatocellular Cancer. Hepatol Commun. 2019;3(4):525–41. doi:10.1002/hep4.1311.
  • Ishiguro K, Yan IK, Lewis-Tuffin L, Patel T. Targeting liver cancer stem cells using engineered biological nanoparticles for the treatment of hepatocellular cancer. Hepatol Commun. 2020;4(2):298–313. doi:10.1002/hep4.1462.
  • Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, Lotvall JO. Exosome-mediated transfer of mRnas and microRnas is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9(6):654–9. doi:10.1038/ncb1596.
  • Skog J, Wurdinger T, van Rijn S, Meijer DH, Gainche L, Sena-Esteves M, Carter BS, Krichevsky AM, Breakefield XO. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 2008;10(12):1470–76. doi:10.1038/ncb1800.
  • Reshke R, Taylor JA, Savard A, Guo H, Rhym LH, Kowalski PS, Trung MT, Campbell C, Little W, Anderson DG. et al. Reduction of the therapeutic dose of silencing RNA by packaging it in extracellular vesicles via a pre-microRNA backbone. Nat Biomed Eng. 2020;4(1):52–68. doi:10.1038/s41551-019-0502-4.
  • Morishita A, Oura K, Tadokoro T, Fujita K, Tani J, Masaki T. MicroRNAs in the pathogenesis of hepatocellular carcinoma: a review. Cancers Basel. 2021;13(3):514. doi:10.3390/cancers13030514.
  • Li W, Xie L, He X, Li J, Tu K, Wei L, Wu J, Guo Y, Ma X, Zhang P. et al. Diagnostic and prognostic implications of microRNAs in human hepatocellular carcinoma. Int J Cancer. 2008;123(7):1616–22. doi:10.1002/ijc.23693.
  • Baldari S, Di Rocco G, Magenta A, Picozza M, Toietta G. Extracellular vesicles–encapsulated MicroRNA-125b produced in genetically modified mesenchymal stromal cells inhibits hepatocellular carcinoma cell proliferation. Cells. 2019;8(12):1560. doi:10.3390/cells8121560.
  • Mahati S, Fu X, Ma X, Zhang H, Xiao L. Delivery of miR-26a using an exosomes-based nanosystem inhibited proliferation of hepatocellular carcinoma. Front Mol Biosci. 2021;8:738219. doi:10.3389/fmolb.2021.738219.
  • Ellipilli S, Wang H, Binzel DW, Shu D, Guo P. Ligand-displaying-exosomes using RNA nanotechnology for targeted delivery of multi-specific drugs for liver cancer regression. Nanomedicine. 2023;50:102667. doi:10.1016/j.nano.2023.102667.
  • Walker S, Busatto S, Pham A, Tian M, Suh A, Carson K, Quintero A, Lafrence M, Malik H, Santana MX. et al. Extracellular vesicle-based drug delivery systems for cancer treatment. Theranostics. 2019;9(26):8001–17. doi:10.7150/thno.37097.
  • Haney MJ, Klyachko NL, Zhao Y, Gupta R, Plotnikova EG, He Z, Patel T, Piroyan A, Sokolsky M, Kabanov AV. et al. Exosomes as drug delivery vehicles for Parkinson’s disease therapy. J Control Release. 2015;207:18–30. doi:10.1016/j.jconrel.2015.03.033.
  • Busatto S, Iannotta D, Walker SA, Di Marzio L, Wolfram J. A simple and quick method for loading proteins in extracellular vesicles. Pharm (Basel). 2021;14(4):14. doi:10.3390/ph14040356.
  • Han Y, Jones TW, Dutta S, Zhu Y, Wang X, Narayanan SP, Fagan SC, Zhang D. Overview and update on methods for cargo loading into extracellular vesicles. Processes (Basel). 2021;9(2):356. doi:10.3390/pr9020356.
  • 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):3066–77. doi:10.1016/j.ymthe.2022.06.013.
  • Zhang J, Song H, Dong Y, Li G, Li J, Cai Q, Yuan S, Wang Y, Song H. Surface engineering of HEK293 cell-derived extracellular vesicles for improved pharmacokinetic profile and targeted delivery of IL-12 for the treatment of hepatocellular carcinoma. Int J Nanomedicine. 2023;18:209–23. doi:10.2147/IJN.S388916.
  • Kooijmans SAA, Stremersch S, Braeckmans K, de Smedt SC, Hendrix A, Wood MJA, Schiffelers RM, Raemdonck K, Vader P. Electroporation-induced siRNA precipitation obscures the efficiency of siRNA loading into extracellular vesicles. J Control Release. 2013;172(1):229–38. doi:10.1016/j.jconrel.2013.08.014.
  • Ilahibaks NF, Ardisasmita AI, Xie S, Gunnarsson A, Brealey J, Vader P, de Jong OG, de Jager S, Dekker N, Peacock B. et al. TOP-EVs: technology of protein delivery through extracellular vesicles is a versatile platform for intracellular protein delivery. J Control Release. 2023;355:579–92. doi:10.1016/j.jconrel.2023.02.003.
  • Somiya M, Kuroda S. Engineering of extracellular vesicles for small molecule-regulated cargo loading and cytoplasmic delivery of bioactive proteins. Mol Pharm. 2022;19(7):2495–505. doi:10.1021/acs.molpharmaceut.2c00192.
  • Yang C, Guan Z, Pang X, Tan Z, Yang X, Li X, Guan F. Desialylated mesenchymal stem cells-derived extracellular vesicles loaded with doxorubicin for targeted inhibition of hepatocellular carcinoma. Cells. 2022;11(17):2642. doi:10.3390/cells11172642.
  • Liang L, Zhao L, Wang Y, Wang Y. Treatment for hepatocellular carcinoma is enhanced when norcantharidin is encapsulated in exosomes derived from bone marrow mesenchymal stem cells. Mol Pharm. 2021;18(3):1003–13. doi:10.1021/acs.molpharmaceut.0c00976.
  • Mo J, Da X, Li Q, Huang J, Lu L, Lu H. The study of exosomes-encapsulated mPEG-PLGA polymer drug-loaded particles for targeted therapy of liver cancer. J Oncol. 2022;2022:4234116. doi:10.1155/2022/4234116.
  • Wan T, Zhong J, Pan Q, Zhou T, Ping Y, Liu X. Exosome-mediated delivery of Cas9 ribonucleoprotein complexes for tissue-specific gene therapy of liver diseases. Sci Adv. 2022;8(37):eabp9435. doi:10.1126/sciadv.abp9435.
  • Zhuang J, Tan J, Wu C, Zhang J, Liu T, Fan C, Li J, Zhang Y. Extracellular vesicles engineered with valency-controlled DNA nanostructures deliver CRISPR/Cas9 system for gene therapy. Nucleic Acids Res. 2020;48(16):8870–82. doi:10.1093/nar/gkaa683.
  • Chen R, Huang H, Liu H, Xi J, Ning J, Zeng W, Shen C, Zhang T, Yu G, Xu Q. et al. Friend or foe? Evidence indicates endogenous exosomes can deliver functional gRNA and Cas9 protein. Small. 2019;15(38):e1902686. doi:10.1002/smll.201902686.
  • Rolling F, Samulski RJ. AAV as a viral vector for human gene therapy. Generation of recombinant virus. Mol Biotechnol. 1995;3(1):9–15. doi:10.1007/BF02821330.
  • Hudry E, Martin C, Gandhi S, Gyorgy B, Scheffer DI, Mu D, Merkel SF, Mingozzi F, Fitzpatrick Z, Dimant H. et al. Exosome-associated AAV vector as a robust and convenient neuroscience tool. Gene Ther. 2016;23(4):380–92. doi:10.1038/gt.2016.11.
  • Gyorgy B, Fitzpatrick Z, Crommentuijn MH, Mu D, Maguire CA. Naturally enveloped AAV vectors for shielding neutralizing antibodies and robust gene delivery in vivo. Biomaterials. 2014;35(26):7598–609. doi:10.1016/j.biomaterials.2014.05.032.
  • Liu B, Li Z, Huang S, Yan B, He S, Chen F, Liang Y. AAV-Containing exosomes as a novel vector for improved gene delivery to lung cancer cells. Front Cell Dev Biol. 2021;9:707607. doi:10.3389/fcell.2021.707607.
  • Khan N, Maurya S, Bammidi S, Jayandharan GR. AAV6 Vexosomes Mediate Robust Suicide Gene Delivery in a Murine Model of Hepatocellular Carcinoma. Mol Ther Methods Clin Dev. 2020;17:497–504. doi:10.1016/j.omtm.2020.03.006.
  • Kim HY, Min HK, Song HW, Yoo A, Lee S, Kim KP, Park J-O, Choi YH, Choi E. Delivery of human natural killer cell-derived exosomes for liver cancer therapy: an in vivo study in subcutaneous and orthotopic animal models. Drug Deliv. 2022;29(1):2897–911. doi:10.1080/10717544.2022.2118898.
  • Wu B, Feng J, Guo J, Wang J, Xiu G, Xu J, Ning K, Ling B, Fu Q, Xu J. et al. Adscs-derived exosomes ameliorate hepatic fibrosis by suppressing stellate cell activation and remodeling hepatocellular glutamine synthetase-mediated glutamine and ammonia homeostasis. Stem Cell Res Ther. 2022;13(1):494. doi:10.1186/s13287-022-03049-x.
  • de Galarreta M R, Bresnahan E, Molina-Sanchez P, Lindblad KE, Maier B, Sia D, Puigvehi M, Miguela V, Casanova-Acebes M, Dhainaut M. et al. β-Catenin Activation Promotes Immune Escape and Resistance to Anti–PD-1 Therapy in Hepatocellular Carcinoma. Cancer Discov. 2019;9(8):1124–41. doi:10.1158/2159-8290.CD-19-0074.
  • Lu Z, Zuo B, Jing R, Gao X, Rao Q, Liu Z, Qi H, Guo H, Yin H. Dendritic cell-derived exosomes elicit tumor regression in autochthonous hepatocellular carcinoma mouse models. J Hepatol. 2017;67(4):739–48. doi:10.1016/j.jhep.2017.05.019.
  • Meng M, Zhang X, Li Q, Han J, Chen Y, Qiao H, Yang Y, Huang X. Engineering M1-derived nanovesicles loading with docosahexaenoic acid synergizes ferroptosis and immune activation for treating hepatocellular carcinoma. Cancer Nanotechnol. 2023;14(1):17. doi:10.1186/s12645-023-00166-x.
  • Villa A, Garofalo M, Crescenti D, Rizzi N, Brunialti E, Vingiani A, Belotti P, Sposito C, Franzè S, Cilurzo F. et al. Transplantation of autologous extracellular vesicles for cancer-specific targeting. Theranostics. 2021;11(5):2034–47. doi:10.7150/thno.51344.
  • Herrmann IK, Wood MJA, Fuhrmann G. Extracellular vesicles as a next-generation drug delivery platform. Nat Nanotechnol. 2021;16(7):748–59. doi:10.1038/s41565-021-00931-2.
  • Rohde E, Pachler K, Gimona M. Manufacturing and characterization of extracellular vesicles from umbilical cord–derived mesenchymal stromal cells for clinical testing. Cytotherapy. 2019;21(6):581–92. doi:10.1016/j.jcyt.2018.12.006.
  • Lener T, Gimona M, Aigner L, Borger V, Buzas E, Camussi G, Chaput N, Chatterjee D, Court FA, Del Portillo HA. et al. Applying extracellular vesicles based therapeutics in clinical trials – an ISEV position paper. J Extracell Vesicles. 2015;4(1):30087. doi:10.3402/jev.v4.30087.
  • Lehrich BM, Liang Y, Fiandaca MS. Foetal bovine serum influence on in vitro extracellular vesicle analyses. J Extracell Vesicles. 2021;10(3):e12061. doi:10.1002/jev2.12061.
  • Lehrich BM, Liang Y, Khosravi P, Federoff HJ, Fiandaca MS. Fetal Bovine Serum-Derived Extracellular Vesicles Persist within Vesicle-Depleted Culture Media. Int J Mol Sci. 2018;19(11):3538. doi:10.3390/ijms19113538.
  • Patel DB, Gray KM, Santharam Y, Lamichhane TN, Stroka KM, Jay SM. Impact of cell culture parameters on production and vascularization bioactivity of mesenchymal stem cell-derived extracellular vesicles. Bioeng Transl Med. 2017;2(2):170–79. doi:10.1002/btm2.10065.
  • Lee YXF, Johansson H, Wood MJA, El Andaloussi S. Considerations and implications in the purification of extracellular vesicles - a cautionary tale. Front Neurosci. 2019;13:1067. doi:10.3389/fnins.2019.01067.
  • Liang Y, Lehrich BM, Zheng S, Lu M. Emerging methods in biomarker identification for extracellular vesicle-based liquid biopsy. J Extracell Vesicles. 2021;10(7):e12090. doi:10.1002/jev2.12090.
  • Chen VL, Xu D, Wicha MS, Lok AS, Parikh ND. Utility of liquid biopsy analysis in detection of hepatocellular Carcinoma, determination of prognosis, and disease monitoring: a systematic review. Clin Gastroenterol Hepatol. 2020;18(13):2879–902 e9. doi:10.1016/j.cgh.2020.04.019.
  • Li Y, Wu J, Li E, Xiao Z, Lei J, Zhou F, Yin X, Hu D, Mao Y, Wu L. et al. TP53 mutation detected in circulating exosomal DNA is associated with prognosis of patients with hepatocellular carcinoma. Cancer Biol Ther. 2022;23(1):439–45. doi:10.1080/15384047.2022.2094666.
  • Wang P, Song Q, Ren J, Zhang W, Wang Y, Zhou L, Wang D, Chen K, Jiang L, Zhang B. et al. Simultaneous analysis of mutations and methylations in circulating cell-free DNA for hepatocellular carcinoma detection. Sci Transl Med. 2022;14(672):eabp8704. doi:10.1126/scitranslmed.abp8704.
  • Phan TH, Chi Nguyen VT, Thi Pham TT, Nguyen VC, Ho TD, Quynh Pham TM, Tran T-H, Nguyen TD, Khang Le ND, Nguyen T-H. et al. Circulating DNA methylation profile improves the accuracy of serum biomarkers for the detection of nonmetastatic hepatocellular carcinoma. Future Oncol. 2022;18(39):4399–413. doi:10.2217/fon-2022-1218.
  • Sun N, Lee YT, Zhang RY, Kao R, Teng PC, Yang Y, Yang P, Wang JJ, Smalley M, Chen P-J. et al. Purification of HCC-specific extracellular vesicles on nanosubstrates for early HCC detection by digital scoring. Nat Commun. 2020;11(1):4489. doi:10.1038/s41467-020-18311-0.
  • Kim SS, Baek GO, Son JA, Ahn HR, Yoon MK, Cho HJ, Yoon JH, Nam SW, Cheong JY, Eun JW. et al. Early detection of hepatocellular carcinoma via liquid biopsy: panel of small extracellular vesicle-derived long noncoding RNAs identified as markers. Mol Oncol. 2021;15(10):2715–31. doi:10.1002/1878-0261.13049.
  • Shuen TWH, Alunni-Fabbroni M, Ocal E, Malfertheiner P, Wildgruber M, Schinner R, Pech M, Benckert J, Sangro B, Kuhl C. et al. Extracellular vesicles may predict response to radioembolization and Sorafenib treatment in advanced hepatocellular carcinoma: an exploratory analysis from the SORAMIC trial. Clin Cancer Res. 2022;28(17):3890–901. doi:10.1158/1078-0432.CCR-22-0569.
  • Sun N, Zhang C, Lee YT, Tran BV, Wang J, Kim H, Lee J, Zhang RY, Wang JJ, Hu J. et al. HCC EV ECG score: an extracellular vesicle-based protein assay for detection of early-stage hepatocellular carcinoma. Hepatology. 2023;77(3):774–88. doi:10.1002/hep.32692.
  • Rodriguez-Rivera II, Wu T-H, Ciotti R, Senapedis W, Sullivan K, Gao JZ, Palakurthi S, McCauley T, Moore Y. A phase 1/2 open-label study to evaluate the safety, tolerability, pharmacokinetics, pharmacodynamics, and preliminary antitumor activity of OTX-2002 as a single agent and in combination with standard of care in patients with hepatocellular carcinoma and other solid tumor types known for association with the MYC oncogene (MYCHELANGELO I). J Clin Oncol. 2023;41:TPS627–TPS.
  • CancerNetwork. OTX-2002 shows encouraging safety in small hepatocellular carcinoma cohort. 2023.
  • Bottger R, Pauli G, Chao PH, Al Fayez N, Hohenwarter L, Li SD. Lipid-based nanoparticle technologies for liver targeting. Adv Drug Deliv Rev. 2020;154-155:79–101. doi:10.1016/j.addr.2020.06.017.
  • Akinc A, Maier MA, Manoharan M, Fitzgerald K, Jayaraman M, Barros S, Ansell S, Du X, Hope MJ, Madden TD. et al. The Onpattro story and the clinical translation of nanomedicines containing nucleic acid-based drugs. Nat Nanotechnol. 2019;14(12):1084–7. doi:10.1038/s41565-019-0591-y.
  • 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):565–80. doi:10.1517/17425247.2011.566552.
  • Kalita T, Dezfouli SA, Pandey LM, Uludag H. siRNA functionalized lipid nanoparticles (LNPs) in management of diseases. Pharmaceutics. 2022;14(11):14. doi:10.3390/pharmaceutics14112520.