Next Article in Journal
SKAP1 Is a Novel Biomarker and Therapeutic Target for Gastric Cancer: Evidence from Expression, Functional, and Bioinformatic Analyses
Next Article in Special Issue
Delivery of Immunostimulatory Cargos in Nanocarriers Enhances Anti-Tumoral Nanovaccine Efficacy
Previous Article in Journal
Sarcospan Deficiency Increases Oxidative Stress and Arrhythmias in Hearts after Acute Ischemia-Reperfusion Injury
Previous Article in Special Issue
Exploring a Nuclear-Selective Radioisotope Delivery System for Efficient Targeted Alpha Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Liver Cell Type-Specific Targeting by Nanoformulations for Therapeutic Applications

1
I. Department of Medicine, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
2
Children’s Hospital, University Medical Center, Langenbeckstrasse 1, 55131 Mainz, Germany
3
Department of Dermatology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(14), 11869; https://doi.org/10.3390/ijms241411869
Submission received: 5 May 2023 / Revised: 21 June 2023 / Accepted: 21 July 2023 / Published: 24 July 2023
(This article belongs to the Special Issue Recent Advances of Targeted Drug Delivery and Nanocarriers)

Abstract

:
Hepatocytes exert pivotal roles in metabolism, protein synthesis and detoxification. Non-parenchymal liver cells (NPCs), largely comprising macrophages, dendritic cells, hepatic stellate cells and liver sinusoidal cells (LSECs), serve to induce immunological tolerance. Therefore, the liver is an important target for therapeutic approaches, in case of both (inflammatory) metabolic diseases and immunological disorders. This review aims to summarize current preclinical nanodrug-based approaches for the treatment of liver disorders. So far, nano-vaccines that aim to induce hepatitis virus-specific immune responses and nanoformulated adjuvants to overcome the default tolerogenic state of liver NPCs for the treatment of chronic hepatitis have been tested. Moreover, liver cancer may be treated using nanodrugs which specifically target and kill tumor cells. Alternatively, nanodrugs may target and reprogram or deplete immunosuppressive cells of the tumor microenvironment, such as tumor-associated macrophages. Here, combination therapies have been demonstrated to yield synergistic effects. In the case of autoimmune hepatitis and other inflammatory liver diseases, anti-inflammatory agents can be encapsulated into nanoparticles to dampen inflammatory processes specifically in the liver. Finally, the tolerance-promoting activity especially of LSECs has been exploited to induce antigen-specific tolerance for the treatment of allergic and autoimmune diseases.

1. Introduction

The liver constitutes one of the largest organs of the body and exerts numerous essential functions associated with digestion, metabolism, protein synthesis and the detoxification of xenobiotics [1]. Hence, the liver is a primary site of various metabolic disorders [2]. Further, chronic hepatitis [3] and metabolic disorders [4] may result in liver fibrosis or even cirrhosis. Cirrhosis is considered a premalignant state as 80% of hepatocellular carcinoma (HCC), the most frequent type of primary liver cancer, develop in cirrhotic livers [5]. In addition, due to anatomical reasons, the liver is also a primary organ of tumor metastasis [6]. The therapy of liver tumors is complicated by the default tolerogenic state of liver-resident non-parenchymal cells (NPCs) [7]. Hence, the liver, with its different specialized cell types, is a highly interesting target organ for cell-type-addressing nanoformulations, thereby avoiding adverse side effects, such as systemic toxicity [8]. However, as outlined in this review, the structure of the liver has proven a challenge for nano-based cell-targeting approaches.
On the one hand, the liver acts as a biological barrier and may sequestrate the vast majority of administered nanoparticles (NPs) from the bloodstream, when particles (>6 nm) are not eliminated by the kidney [9]. However, on the other hand, within the liver NPs need to pass several biological barriers hindering their successful uptake by the different liver cell populations [10]. In this regard, NPs intended to address hepatocytes need to cross the sinusoidal fenestrations of liver sinusoidal endothelial cells (LSECs) [11,12]. Furthermore, Kupffer cells (KCs), that constitute the major liver-resident macrophage population [13], and LSECs express various types of Fc and scavenger receptors [14] for the efficient uptake of larger KC [13] and smaller LSEC [15] macromolecules, and consequently also NPs [9]. Therefore, hepatocyte-targeting NPs need to circumvent unwanted uptake by the aforementioned liver NPC populations.
This review aims to summarize the strategies currently evaluated in preclinical in vivo models for the treatment of liver diseases with nanoformulations that may allow the poor solubility of drugs to be overcome by encapsulation and enable the codelivery of distinct compounds that act on different signaling pathways or effector molecules into the same cell to yield synergistic effects [16,17]. Consequently, nanoformulations are also intended to prevent systemic toxicity, as frequently observed, e.g., for systemically applied anti-cancer drugs [18,19]. Therefore, nanoformulations need to address their cellular target either via passive or active targeting [20]. Whereas the former is determined by the intrinsic properties of the NP-like size and charge, active targeting is achieved by the attachment of receptor-binding moieties on the NP surface. For the treatment of liver-associated disorders, nanoformulations need to exert disease-specific biological effects; for instance, in the case of virus-induced hepatitis nano-vaccines aim to evoke antiviral immune responses [21,22], whereas in the case of autoimmune hepatitis nanodrugs are intended to dampen inflammation [23].
Liver fibrosis, i.e., the accumulation of extracellular matrix (ECM) proteins due to the activation of hepatic stellate cells (HSCs), is commonly observed in the course of chronic liver diseases, such as non-alcoholic fatty liver disease [24]. Extensive liver fibrosis may result in cirrhosis, which, in turn, predisposes the liver to HCC [25]. Hence, a number of nanoformulations that revert HSC activation are under development. Further, in the case of liver cancer, nanodrugs may either directly address tumor cells to yield tumor cell-restricted cytotoxicity [26], or aim to reprogram tumor-associated cells that support tumor immune evasion [27]. In the case of liver anti-tumor therapy, the efficacy of nanoformulations has been assayed in several cases in combination with an immune checkpoint blockade (ICB) that aims to enhance T cell responses to improve therapeutic effects [28].
Finally, we will also discuss how the common default tolerance-inducing role of the various liver NPC populations (KCs [29]; LSECs [30]; dendritic cells (DCs) [31]) has been exploited to induce antigen-specific regulatory T cells (Tregs) for the treatment of allergies [32] and autoimmune diseases [33].

2. Nanodrugs for the Treatment of Liver Disease

2.1. Viral Hepatitis

Hepatitis constitutes a state of liver inflammation that is induced by a variety of noxae, such as viral infections, alcohol abuse and autoimmune reactions [34]. The pathogenesis of liver damage in hepatitis B and autoimmune hepatitis, as the most frequent kinds of hepatitis, shows certain similarities [35]. In both diseases, cells of the immune system cause inflammatory reactions that damage the liver. In chronic hepatitis B, virus antigen-specific T cells are considered to be the main driver [36]. These virus-specific lymphocytes are not capable of eradicating the virus but recognize virus-specific proteins in the liver which cause hepatocyte damage [37]. Under these inflammatory conditions, KCs promote activation the of HSCs, which transform into myofibroblasts that produce large amounts of ECM proteins, which replace the parenchymal tissue [38]. However, a plethora of other NPCs (e.g., liver-resident macrophages) contribute to fibrogenesis in the liver. A comprehensive review of the underlying mechanisms and involved cell types was recently published [39]. When fibrogenesis persists, liver cirrhosis, as an end-stage of virtually every chronic liver disease, can occur [40]. Portal hypertension is a consequence of cirrhosis and is responsible for the most severe clinical complications, such as bleeding from gastro-esophageal varices and hepatic encephalopathy [41].
Viral hepatitis can be caused by five different hepatitis viruses, which have different transmission routes and variable clinical courses [42]. The pathogenicity of hepatitis G for humans is unclear and is therefore not further discussed. All of them are single-stranded RNA viruses, except hepatitis B (HBV), which contains its genetic material as stable DNA [43]. Hepatitis D virus (HDV) is a satellite virus, as it can propagate only in the presence of HBV because it requires HBV surface antigens in its viral envelope in order to replicate [44]. All types of hepatitis viruses can lead to fulminant hepatitis, while only HBV, hepatitis C virus (HCV) and, in rare cases, hepatitis E virus (HEV) can cause chronic infections, constituting a risk of cirrhosis and HCC [45,46]. Co-infection of HBV and HDV is considered the most serious type of viral hepatitis, which leads to a complicated clinical course with fast progression to cirrhosis [47].
Hepatitis A virus (HAV) and HEV are transmitted by the fecal–oral route. HBV infections occur as a result of exposure to contaminated blood and body fluids [48]. In practice, HBV is mostly transmitted vertically (mother-to-child). HCV is transmitted by contaminated blood (e.g., needle sharing, contaminated blood transfusion) or, in rare cases, genital secretions (mostly reported for men who have sex with men) [49].
The clinical course of the infection depends on the type of hepatitis virus. HAV and HEV infections play a minor role in clinics because most patients spontaneously recover, and only at-risk groups may develop acute liver failure [50]. The treatment of hepatitis C dramatically improved in 2014 when effective antiviral regimes were clinically approved [51]. Oral tablet regimes achieve very high curing rates and novel drug loaded β-cyclodextrin NPs may even improve the safety and tolerability of treatments [52]. Accordingly, the World Health Organization is confident that hepatitis C may one day be eradicated [53]. In contrast, there is a risk of hepatitis B reactivation even in cured patients [54]. One of the main reasons for the refractory nature of hepatitis B towards treatment is the covalently closed circular DNA (ccc DNA) of HBV, which serves as a template for all transcripts and persists in the nucleus of hepatocytes [55]. The reactivation of HBV can occur under immunosuppression and is a potentially life-threatening condition when liver failure occurs [56].
Since HBV cannot be completely eliminated by the immune system, prophylactic vaccination is a pillar of all prevention strategies [57]. One strategy to achieve effective protection is vaccines that include adjuvants which induce a strong immune response. [58]. Traditional hepatitis B vaccines use aluminum compounds as an adjuvant to induce a robust humoral response [59]. However, vaccines with this kind of adjuvant cannot yield an adequate cellular immune response to effectively recognize and eliminate HBV-infected cells [60]. Therefore, novel adjuvants which increase not only humoral but also cellular immune responses are needed. For example, Pu Shan and coworkers developed a saponin-based nano-adjuvant which induced stronger humoral and cellular immune responses than aluminum-based adjuvants in a murine hepatitis model [61]. Further, Qiao et al. generated nano-vaccines composed of chitosan plus heparin that encapsulated the HBV surface and core antigen, respectively, and contained immunostimulatory CpG oligo in addition [62]. In a mouse model of chronic hepatitis B infection these two types of nano-vaccines were coapplied. After subcutaneous injection, the two nanovaccines targeted the draining lymph nodes, achieving the seroclearance of HBV surface antigens in most mice. In addition, vaccination induced long-term immune memory and protected the mice from HBV reinfection. Interferon-α (IFN-α), either in its soluble form or conjugated to polyethylene glycol (PEG), is used to treat chronic hepatitis B but only 20–40% of patients respond well, and the treatment is associated with side effects due to its systemic administration [63]. Fayes and coworkers demonstrated the liver-restricted induction of IFN-α by the toll-like receptor 7 (ligand imiquimod (IMQ)) when delivered as a nanoformulation [64]. To this end, IMQ was encapsulated into liposomal vesicles, which conferred hepatocyte-specific uptake, and into the anionic liposomes that mediated KC-restricted internalization. In a mouse model of chronic hepatitis B, both types of NPs yielded liver-specific IFN-α production and concomitantly reduced HBV DNA serum levels.

2.2. Autoimmune Hepatitis

The exact etiology of autoimmune hepatitis is not known [65]. A genetic predisposition in combination with environmental factors, viral infection and drugs may trigger this disorder. Hepatitis is considered to be of autoimmune origin only when viral hepatitis can be excluded, reflecting the high degree of histological and clinical similarities between viral and autoimmune hepatitis [35]. In autoimmune hepatitis, elevated titers of autoantibodies (e.g., antinuclear and anti-smooth muscle antibodies) and increased immunoglobulin G levels are observed, which are normally not present in viral hepatitis and, thus, can be used in differential diagnosis [66].
The mainstay of treatment is immunosuppressive drugs, such as glucocorticoids, azathioprine or methotrexate and mycophenolate in severe cases [67]. However, the systemic use of immunosuppressive drugs provokes the risk of side effects, such as Cushing’s syndrome in the case of glucocorticoids [68], or opportunistic infections when applying azathioprine, methotrexate and mycophenolate, respectively [69]. An interesting concept to circumvent the systemic effects of glucocorticoids is dexamethasone-loaded NPs [23], which display an enhanced liver tropism when composed of avidin and nucleic acid [70,71]. Upon intraperitoneal administration, these NPs primarily addressed the liver and high levels of dexamethasone were detected in livers but not in sera [70]. Another interesting application of immunosuppressive NPs is to alleviate the side effects of ICB in cancer therapy [72]. Poly(L-lactic-co-glycolic acid) (PLGA) NPs coated with the anti-programmed cell death protein (PD-)1 antibody did not induce the hepatotoxicity that may occur in the course of ICB therapy [73] without affecting the antitumor effect of ICB in a murine tumor model [72].

2.3. Non-Alcoholic Fatty Liver Disease (NAFLD)

With an estimated global prevalence of almost ~25%, non-alcoholic fatty liver disease (NAFLD) is a leading cause of liver disease worldwide [74]. NAFLD ranges from steatosis without or with mild inflammation [75] to advanced stage non-alcoholic steatohepatitis (NASH), which is characterized by the necroinflammation and ballooning of hepatocytes [76]. NASH can progress to fibrosis and, ultimately, cirrhosis with the risk of HCC or intrahepatic cholangiocellular carcinoma and death [77]. The risk factors for NAFLD are obesity, type 2 diabetes mellitus and metabolic syndrome.
Despite great research efforts, no drug has been approved yet for NAFLD or NASH treatment. However, several clinical studies are ongoing to evaluate potential drug candidates. For instance, the anti-diabetic drug pioglitazone and vitamin E (i.e., δ-tocotrienol and α-tocopherol) demonstrated a therapeutic effect (e.g., an improvement in hepatic steatosis) in patients with NAFLD. Obeticholic acid, a farnesoid X nuclear receptor, improved the histological features of NASH but long-term results are still pending [78]. However, weight loss and dietary modifications are currently the therapeutic mainstay for these liver disorders [79]. Plant-derived celastrol (CEL) is a promising anti-inflammatory and anti-obesity drug but its low oral bioavailability hampers its clinical use [80]. Albumin-based NPs were synthesized to facilitate the liver-specific delivery of CEL [81]. To this end, CEL was encapsulated into lactosylated bovine serum albumin using high pressure homogenization. CEL-NP showed improved uptake into hepatocytes and enhanced hepatic deposition compared to free CEL. Further, encapsulated CEL outperformed free CEL in reducing lipid deposition, ameliorating liver function and enhancing insulin sensitivity in a murine model of diet-induced NAFLD without causing side effects. Besides, the genes for lipogenesis and lipid transport were upregulated by treatment.
Reducing calorie uptake in overweight patients may be an additional application of NPs. α-glucosidase is an approved drug for limiting the absorption of polysaccharides and disaccharides but it is ineffective for monosaccharides [82]. However, the boronic acid-containing polymer nanocomplex (Nano-Poly-BA) absorbed all types of saccharides and, thus, could be used to avoid their intestinal uptake [83]. Nano-Poly-BA showed remarkable after-meal blood glucose reductions in type 1 and type 2 diabetic mouse models when mice were fed with coke, blueberry jam or porridge. Orally administered Nano-Poly-BA proved to be non-absorbable and non-toxic. The authors concluded that Nano-Poly-BA may help diabetic, overweight and even healthy people to manage their sugar intake. In the methionine-choline-deficient diet mouse model the administration of a liposomal formulation equipped with a sphingosine receptor-targeting moiety for HSC targeting [84] and containing short chain C6-ceramide normalized dysregulated lipid homeostasis [85].
In the course of fibrosis-inducing events, activated LSECs close fenestration gaps and are no longer able to keep HSCs in a quiescent state; they therefore transdifferentiate to myofibroblasts and produce ECM proteins to a large extent [86]. HSCs store large amounts of vitamin A in the liver [87]. In line, several studies demonstrated the vitamin A-mediated targeting of various types of NPs to HSCs, which inhibited HSC activation-dependent fibrosis in various rodent models (Figure 1). For example, Perri et al. employed silver NPs that contained a nitric oxide donor [88] to trigger soluble guanylyl cyclase signaling [89]. Liposomes encapsulating sterol regulatory element-binding protein 2-specific small interfering (si)RNA and anti-miR-33a [90] prevented TGF-β-mediated HSC activation [91]. Qiao and coworkers [92] developed core-shell polymeric micelles that codelivered the antioxidant silibinin [93] and collagen1a1-specific siRNA [94], thereby attenuating ECM formation.
As an alternative approach, Zhang et al. applied liposomes with hyaluronic acid, previously reported to target LSECs and HSCs [95] (Table 1), and containing simvastatin to revert LSEC capillarization [96]. Subsequently, a second liposomal nano-carrier was employed to deliver collagen 1-specific siRNA to HSCs and, thereby, attenuate ECM formation.
In addition, untargeted nanohydrogel particles, which efficiently accumulate in the liver, hold promise for antifibrotic treatment. Two intravenous injections of anti-collagen 1-specific siRNA-loaded nanohydrogel particles (2 mg/kg siRNA) reduced the hepatic collagen load to the levels of healthy control mice [97,98].
Figure 1. Nanoformulations for the inhibition of liver cirrhosis. LSEC serve to keep HSC in a quiescent state (blocking arrow). Various stress factors result in the loss of LSEC fenestration, termed capillarization, and the loss of LSEC-mediated HSC inactivation (dashed blocking arrow). Activated HSCs transdifferentiate to myofibroblasts and generate ECM proteins, resulting in fibrosis, cirrhosis and, ultimately, HCC. Fibrosis-counteracting nanoformulations target LSECs, e.g., via mannose [99] and hyaluronic acid [96], and deliver simvastatin to revert capillarization and to reestablish HSC-inhibition by LSECs. Alternatively, nanodrugs may target HSCs especially via vitamin A, and inhibit ECM production, e.g., by the delivery of drugs that inhibit collagen expression at various levels [88,90,92,96,100]. ECM, extracellular matrix; HA, hyaluronic acid; HCC, hepatocellular carcinoma; HSC, hepatic stellate cell; LNP, lipid nanoparticle; LSEC, liver sinusoidal endothelial cell; miR, micro RNA; NP, nanoparticle; PLGA, poly(L-lactic-co-glycolic acid); siRNA, small interfering RNA. Created with BioRender.com (4 May 2023).
Figure 1. Nanoformulations for the inhibition of liver cirrhosis. LSEC serve to keep HSC in a quiescent state (blocking arrow). Various stress factors result in the loss of LSEC fenestration, termed capillarization, and the loss of LSEC-mediated HSC inactivation (dashed blocking arrow). Activated HSCs transdifferentiate to myofibroblasts and generate ECM proteins, resulting in fibrosis, cirrhosis and, ultimately, HCC. Fibrosis-counteracting nanoformulations target LSECs, e.g., via mannose [99] and hyaluronic acid [96], and deliver simvastatin to revert capillarization and to reestablish HSC-inhibition by LSECs. Alternatively, nanodrugs may target HSCs especially via vitamin A, and inhibit ECM production, e.g., by the delivery of drugs that inhibit collagen expression at various levels [88,90,92,96,100]. ECM, extracellular matrix; HA, hyaluronic acid; HCC, hepatocellular carcinoma; HSC, hepatic stellate cell; LNP, lipid nanoparticle; LSEC, liver sinusoidal endothelial cell; miR, micro RNA; NP, nanoparticle; PLGA, poly(L-lactic-co-glycolic acid); siRNA, small interfering RNA. Created with BioRender.com (4 May 2023).
Ijms 24 11869 g001
Table 1. Target receptors and receptor-binding moieties for liver cell type-specific addressing by NPs.
Table 1. Target receptors and receptor-binding moieties for liver cell type-specific addressing by NPs.
Cell TypeTarget ReceptorTargeting MoietyRef.
HCCαvβ3 integrin receptor [101]Cyclic RGD peptide [102][103,104]
Asialoglycoprotein receptor [105]N-acetylgalactosamine[106,107,108]
CD44 [109]Hyaluronic acid[110,111,112]
CXC motif chemokine receptor 4 (CXCR4) [113]CTCE-9908 peptide [102][114]
CD147 [115]Anti-CD147 antibody[116,117,118]
EGFR [119]GE11 peptide [120][121]
Folate receptor [122]Folate[123,124,125]
Glycyrrhetinic acid receptor [126]Glycyrrhizic acid[9,127,128,129]
Glypican3 (GPC3) [130]Membrane of GPC3-specific CAR-T cells [131]
Anti-GPC3 antibody[132]
Nucleolin receptor [133]AS1411 aptamer [134][135]
Transferin receptor [136],Transferin,[137]
UnknownTLS11a aptamer [138], SP94 peptide [139], [140,141]
HSCMannose-6-phosphate receptor [142]Mannose-6-phosphate[143,144,145]
Retinol-binding protein receptor [146,147] Retinol-binding protein[148,149]
Collagen type VI receptor [150]Cyclic arginine-glycine-aspartate peptide[150,151]
CD44Hyaluronic acid[95,129,152]
Platelet-derived growth factor beta receptor [153]pPB peptide [154][155,156,157]
KCCD163 [158]Anti-CD163 antibody[159,160]
CD206 [158]Mannose[137,161,162,163]
LSECCD44Hyaluronic acid[95]
CD206 [164]Mannose[99,165,166]
Stabilin-1, -2 [167,168]Cholesteryl oleate, ApoB peptide[32,165,169,170]

2.4. Hepatic Tumors/Metastasis

Liver cancer has been ranked as the third leading cause of cancer deaths worldwide and its incidence is further increasing [5]. HCC is the most prevalent primary liver cancer in adults, which accounts for up to 90% of cases, followed by cholangiocarcinoma (CCC) [171]. Only the minority are hepatocellular cholangiocarcinoma (a mixed form of HCC and CCC), angiosarcoma and hepatoblastoma. HCC originates predominantly from hepatocytes and transformed activated hepatic progenitor cells, while CCC is a bile duct neoplasm [172,173]. The predominant risk factor for HCC is parenchymal damage. Patients with cirrhosis have a high annual risk of ~1–6% of developing HCC [174]. Besides cirrhosis, an important risk factor for CCC is autoimmune disease of the bile duct system (e.g., primary biliary cholangitis or primary sclerosing cholangitis) [175,176]. The mainstay of treatment is a combination of the immune checkpoint programmed cell death-1 ligand (PD-L1)-blocking antibody, atezolizumab [177], with the anti-angiogenic vascular endothelial growth factor (VEGF)-A-neutralizing antibody, bevacizumab [178], in patients with unresectable HCC, reaching an overall survival of 13.2 months [179]. For unresectable CCC, chemotherapy combined with the PD-L1 immune checkpoint inhibitor, durvalumab [177], could improve overall survival, progression-free survival and objective response rates compared to chemotherapy alone, while the estimated 24-month survival rates remained low (24%) [180]. Besides primary cancer, the liver is susceptible to hematogenous metastasis due to its high blood perfusion [6]. Hence, numerous cancer patients present with liver metastasis in the course of disease progression, for example, colorectal cancer patients show liver metastasis in up to half of all cases [181]. LSECs play an important role in liver metastasis by expressing adhesion receptors, such as E-selectin, thereby engaging extravasating tumor cells [182,183].
Liver NPCs and infiltrating immunoregulatory cells promote the growth of HCC and liver metastasis by the release of mitogenic factors, such as hepatocyte growth factor (HGF) [184] and proangiogenic factors, such as VEGF [185]. In addition, anti-inflammatory cytokines, such as interleukin (IL)-10 [186] and transforming growth factor (TGF)-β [187] contribute to establish an immunosuppressive tumor microenvironment (TME) [188,189] that consists on cellular level of immune cells (see below), endothelial cells, HSCs and cancer-associated fibroblasts (CAFs) [190]. Interestingly, CAFs originate from diverse cell types, including mesenchymal stem cells, fibroblasts, endothelial and epithelial cells, as well as hepatocytes that (trans)differentiate under the influence of TME-derived cues [191], but the predominant cellular source is activated HSCs [192]. CAFs support tumor growth through the release of various factors, such as TGF-β [193], HGF [194] and IL-6 [195], but also chemokines, such as CC-chemokine ligand (CCL)2 [196], which attracts monocytes that may polarize towards tumor-associated macrophages (TAMs) [197]. Chemokines also attract myeloid-derived suppressor cells (MDSCs) [198] that arise in the bone marrow under the influence of TME-derived mediators, such as IL-1β and IL-6 from granulocytic and monocytic progenitors, respectively [199]. Polymorphonuclear neutrophilic leukocyte (PMN) may acquire an immunosuppressive state under the influence of the TME as well, similar to PMN-like MDSCs [200]. Besides immunoregulatory cells of myeloid origin, Treg cells are also attracted towards the tumor site, e.g., by CCL22 [201].
So far, the vast majority of preclinically evaluated nanoformulations for the treatment of liver cancer have focused on the delivery of cytotoxic agents into the tumor (Figure 2). In most cases these nanoformulations passively target the liver as demonstrated, for example, by gold NPs [202], micelles composed of diblock copolymers of methoxy poly (ethylene glycol)-poly(L-lactic acid) (mPEG-PLA) [203] and β-tricalcium phosphate NPs [204].
However, a growing number of studies have been aimed at targeting active tumor cells by decorating nanocarriers with moieties that address receptors highly expressed by tumor cells [205], such as, for example RGD peptides [103] that engage integrin αvβ3 [206], epidermal growth factor receptor binding peptides [121], the folate receptor [123] and cluster of differentiation (CD)44- [110] and CD147- [207] specific antibody fragments [116]. In the case of the latter study, additional decoration with a cell-penetrating peptide [208] improved cellular uptake [116]. The HCC receptors and the concordant ligands used to achieve HCC-specific NP delivery are listed in Table 1. As an alternative approach for HCC targeting, in screening studies, aptamers with an intrinsic HCC-binding affinity to as-yet undefined receptors have been identified and used for HCC targeting by NP [138,139].
Figure 2. Nanodrugs for liver tumor therapy. Tumors establish an immunosuppressive tumor microenvironment (TME) by attracting immune cells that are reprogrammed to exert immunoinhibitory effects. Nanoformulations may either passively ([202,203,204]) or actively ([103,116,121,162]) target tumor cells to deliver cytotoxic drugs alone ([106,162,209]) or in combination with signaling inhibiting agents ([210,211]). As an alternative, nanoformulations may target components of the TME and cause their differentiation to acquire anti-tumor activity ([162,212]). Ab, antibody; CCL, CC-chemokine ligand; EGFR, epidermal growth factor receptor; IL, interleukin; LNP, lipid nanoparticle; mMDSCs, monocytic myeloid-derived suppressor cells; miRNA, micro RNA; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; NP, nanoparticle; PD-1, programmed cell death protein 1; PEG, polyethylene glycol; PLA, poly(L-lactic acid); PMN, polymorphonuclear neutrophilic leukocyte; siRNA, small interfering RNA; TAM, tumor-associated macrophage; VEGF, vascular endothelial growth factor. Created with BioRender.com (4 May 2023).
Figure 2. Nanodrugs for liver tumor therapy. Tumors establish an immunosuppressive tumor microenvironment (TME) by attracting immune cells that are reprogrammed to exert immunoinhibitory effects. Nanoformulations may either passively ([202,203,204]) or actively ([103,116,121,162]) target tumor cells to deliver cytotoxic drugs alone ([106,162,209]) or in combination with signaling inhibiting agents ([210,211]). As an alternative, nanoformulations may target components of the TME and cause their differentiation to acquire anti-tumor activity ([162,212]). Ab, antibody; CCL, CC-chemokine ligand; EGFR, epidermal growth factor receptor; IL, interleukin; LNP, lipid nanoparticle; mMDSCs, monocytic myeloid-derived suppressor cells; miRNA, micro RNA; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; NP, nanoparticle; PD-1, programmed cell death protein 1; PEG, polyethylene glycol; PLA, poly(L-lactic acid); PMN, polymorphonuclear neutrophilic leukocyte; siRNA, small interfering RNA; TAM, tumor-associated macrophage; VEGF, vascular endothelial growth factor. Created with BioRender.com (4 May 2023).
Ijms 24 11869 g002
In most cases, tumor-targeting nanoformulations aimed to improve the tumor-specific effects of chemotherapeutics by attenuating systemic toxicity [213]. However, in a growing number of studies the potential of nanocarriers to codeliver different agents with distinct molecular targets to achieve synergistic effects has been demonstrated. In many cases, a chemotherapeutic drug (e.g., doxorubicin or oxaliplatin) is codelivered with a signal transduction inhibitor (e.g., sorafenib) that inhibits the pathways upregulated in cancer [214], such as Raf kinases [106] or phosphoinositide 3-kinases (PI3Ks)/mechanistic targets of rapamycin (mTORs) [209]. In further combination approaches, cytotoxic agents were delivered with metal-containing nano-carriers to enable chemo-photothermal therapy [215,216]. As a rather novel approach, cytotoxic drugs have been codelivered with small interfering (si)RNA to inhibit, e.g., VEGF-mediated neoangiogenesis [210], and with micro (mi)RNA to inhibit the components of the PI3K signaling pathway [211].
Furthermore, the TME has been targeted by nanoformulations with the aim of decreasing its immunosuppressive character. Hypoxia is a hallmark of larger solid tumors which promotes the immunosuppressive state of the TME, both by reprograming, e.g., macrophages towards TAMs [217] and by directly inhibiting the activity of tumor-infiltrating leukocytes, e.g., via metabolic cues [218]. Furthermore, hypoxia drives the expression of proangiogenic genes, such as VEGFs, resulting in the neovascularization of the tumor [219]. Chang et al. generated NPs that consisted of an MnO2 core to generate oxygen from hydrogen peroxide decomposition and a lipid–PLGA shell containing sorafenib as an angiogenesis inhibitor [220]. HCC targeting was achieved by conjugating the HCC-targeting SP94 peptide [139]. This composite nanoformulation inhibited tumor growth, which was associated with the repolarization of TAMs towards M1-like macrophages, and elevated the tumor infiltration of CD8+ cytotoxic T lymphocytes (CTLs), as well as impairing tumor vascularization. Wang and coworkers observed that so-called high-density lipoproteins displayed intrinsic HCC- and macrophage-targeting properties [221]. In the same study, the codelivery of vadimezan to disrupt newly formed blood vessels within the tumor [222] and of the cytotoxic drug, gemcitabine [223], caused a release of danger-associated molecules which, in turn, also resulted in a shift of TAMs towards M1-like macrophages and increased CTL infiltration.
In a TAM-focused approach, Wang and coworkers [162] demonstrated the synergistic anti-tumor effects of sorafenib-loaded cationic lipid (L)NPs for direct HCC killing and coapplied cationic LNPs coated with mannose for TAM targeting that were loaded with the nuclear factor kappa-light-chain-enhancer of activated B-cells (NF-κB) inhibitor, IMD-0354 [224], to achieve TAM reprograming towards M1-like macrophages exerting anti-tumor activity [225]. Likewise, NPs composed of cationic chitosan and anionic poly-glutamic acid and loaded with doxorubicin for tumor cell killing and IL-12 to reprogram TAMs towards M1 macrophages were demonstrated to accumulate in the liver and to exert therapeutic efficacy in an HCC model [226]. In the case of HCCs, TAMs are the major PD-L1-expressing cell type within the TME, and limit T cell activity by the engagement of PD-1 [227]. PD-L1-binding immune checkpoint inhibitors were reported to reduce the number of TAMs and to improve tumor infiltration by CD4+ T helper cells (Th) and CTLs [228]. As an alternative to ICB treatment, which has been associated with adverse side effects [229], LNPs were loaded with a plasmid DNA that encoded a solubilized PD-1 receptor to bind to PD-L1 [230]. These NPs accumulated in the liver and exerted therapeutic activity in HCC models, which was associated with decreased TAM numbers and the attenuated infiltration of PMN-related MDSCs but enhanced CTL infiltration. In a combination approach, LNPs coated with mannose for TAM targeting were co-loaded with a NF-κB inhibitor to reprogram TAMs and with a PD-1-blocking antibody to enhance T effector cell proliferation [212] into nanogels that released their content in a matrix metalloproteinase 2-responsive manner at the tumor site [163]. Further strategies to reprogram and deplete TAMs in HCC have recently been reviewed [231].
In HCC both the loss of LSEC fenestration and the accumulating ECM may prevent drug delivery to the tumor site. Simvastatin has been demonstrated to restore LSEC capillarization by activating the transcription factor Kruppel-like factor (KLF)2 [232]. KLF2, in turn, activated endothelial nitric oxide synthase and the derived NO triggered soluble guanylyl cyclase-dependent signaling [233]. Yu and coworkers demonstrated in a murine HCC model that PLGA-based NPs loaded with simvastatin and coated with mannose to target LSECs reverted LSEC capillarization and deactivated HSCs [99]. Moreover, simvastatin induced the production of the chemokine CXCL16 by LSECs, which attracted natural killer T cells that exerted anti-tumor activity. Lipid calcium-phosphate NPs, which were conjugated with aminoethyl anisamide suggested to engage the sigma-1 receptor found to be highly expressed by activated HSCs [234], delivered plasmid DNA encoding the anti-fibrotic peptide, relaxin [100], thereby reverting fibrosis within the tumor lesion [235]. Synergistic anti-tumor effects were achieved when combining this approach with a PD-L1 blockade. Further studies on nanodrug delivery to HSCs and the relevance of these strategies to address closely related CAFs are discussed by Kaps and Schuppan [236].

2.5. Tolerance Induction

Tolerance towards self-antigens is conferred largely in the thymus, resulting in the deletion of T cells whose antigen-recognizing T cell receptor displays a high affinity towards a self-protein-derived peptide antigen, and, in case of intermediate affinity, promotes their differentiation towards Tregs [237]. In the periphery, T cell tolerance towards self-proteins, as well as harmless environmental antigens, is induced and maintained by immature and tolerogenic DCs that anergize reactive T cells or convert them into Tregs [238]. The loss of tolerance towards self-antigens results in autoimmune diseases [239] and, in the case of environmental antigens, in allergies [240]. The only established therapeutic treatment option for allergies is based on the subcutaneous or sublingual delivery of the allergen to (immature) cutaneous DCs to reestablish tolerance [241]. However, this approach requires several years of treatment and is not always successful. For autoimmune diseases no established therapy is available yet [242]. Ongoing clinical trials aim to reestablish self-tolerance by tolerizing in vitro monocyte-derived DCs, e.g., with anti-inflammatory cytokines, such as IL-10, or drugs, such as glucocorticoids, followed by the reinfusion of these DCs into the patient [243,244]. In many cases, these DCs are loaded beforehand with relevant autoantigens. Preclinical trials aim to address DCs in vivo by employing nano-vaccines that deliver an auto-antigen or allergen, respectively, and in many cases also contain a tolerance-promoting agent [245].
More recently, several studies have aimed to exploit the intrinsic tolerance-promoting activity of liver NPCs [246] as an alternative to DC tolerization, since the former are known to induce Tregs [247] and to inhibit T effector cells [248] in an antigen-specific manner. Through comparative biodistribution experiments, Carambio and coworkers identified the ability of iron oxide-based NPs coated with poly(maleic acid-alt-1-octadecene) [249] to passively target LSECs in vivo after systemic administration [250]. In a mouse model of multiple sclerosis, experimental autoimmune encephalomyelitis (EAE) was conducted. These NPs, when coated with the relevant autoantigen used for EAE induction, inhibited the onset of disease when applied one day after EAE induction in myelin basic protein-immunized B10.PL mice and delayed the onset of EAE in myelin oligodendrocyte glycoprotein-immunized C7BL/6 mice. Inhibitory effects in C7BL/6 mice were also apparent in a therapeutic setting. In either case, LSEC-induced CD4+ Tregs played a major role in EAE inhibition. In a subsequent study, Carambio et al. demonstrated the efficacy of this tolerization approach in a model of CD8+ T cell-driven autoimmune cholangitis by inhibiting CD8+ T cell cytotoxicity and inflammatory cytokine production [33].
Concerning allergy models, Xu and coworkers generated LNPs [166] that were decorated with mannose to predominantly address LSECs [166], which display high mannose receptor density [14,251]. The administration of mannosylated LNPs loaded with peanut allergen (Ara h2) peptide-encoding mRNA resulted in the generation of IL-10 producing Foxp3+ Tregs, and largely prevented anaphylactic effects, including IgE production, mast cell activation and a drop in body temperature, both when applied prior to or after sensitization [166]. Liu and coworkers demonstrated that PLGA-based NPs decorated with mannose or an apolipoprotein (Apo) B-derived peptide after intravenous application into mice predominantly addressed LSECs via the mannose and the stabilin-2 receptor, respectively [165]. Both receptors are highly expressed by LSECs [14]. In allergy models, the prophylactic administration of NPs containing the model antigen ovalbumin (OVA) and coated with ApoB peptide resulted in the induction of OVA-specific Tregs that generated the anti-inflammatory cytokines, IL-10 and TGF-β [165]. In accordance, this vaccination approach prevented both anaphylactic and asthmatic reactions in the relevant models by preventing a Th2 response and concomitant IgE production. In a subsequent study the same LSEC-focused delivery approach, using the peanut allergen, Ara h2, as a cargo, yielded tolerogenic effects in a peanut anaphylaxis model [240].
In a follow-up study [32], the aforementioned LSEC-targeting tolerization approach using OVA as a model allergen showed similar efficacy as using PLGA-based NPs containing the mTOR inhibitory drug, rapamycin (immTOR), previously reported to passively target and tolerize DCs, thereby inducing Treg and attenuating B cell responses in mice [252] and cynomolgus monkeys [253]. However, a previous study demonstrated that immTOR NPs accumulated not only in the spleen, but also in the liver [252]. Ilyinskii et al. showed that immTOR addressed hepatocytes as well as all types of liver NPs [254]. Further, immTOR administration tolerized KCs, DCs and LSECs, as deduced from the downregulation of MHCII and costimulatory receptors, accompanied by the increased expression of the coinhibitory receptor, PD-L1. Further, hepatic T cell numbers were diminished, and the remaining population presented with elevated PD-1 levels. LSECs isolated from immTOR-injected mice inhibited T cell proliferation. In accordance with the broad anti-inflammatory effects of immTOR, pretreated mice displayed attenuated inflammation after the application of concavalin A.
In light of the general finding that many types of NPs accumulate in the liver [9], the study by Ilyinski and coworkers [254] underscores that nano-carriers previously shown to tolerize DCs in the spleen or lymph nodes [252,253] may also have a considerable impact on liver NPC populations. Further studies are required to revisit such nano-carriers in order to assess the potential synergistic effects of DCs and potentially tolerized liver NPCs, often not taken into account in previous ex vivo analyses, with regard to systemic tolerance induction.

3. Conclusions

The liver constitutes a highly suitable target organ for nanoformulations that enable drug delivery in a cell type-specific manner to reduce the adverse effects often observed in cases of systemic application. Based on detailed knowledge of cell type-specific surface receptors and intracellular signaling pathways targeting and reprogramming both hepatocytes for the treatment of metabolic diseases and liver NPCs to counteract fibrosis as a preliminary stage of cirrhosis/HCC, as well as of immunological traits, is now possible. Taking into account the extensive crosstalk between liver cell types, e.g., in HCC and TME combination therapies that co-target distinct cell types with appropriate nanodrugs and with clinically approved agents, such as immune checkpoint inhibitors, respectively, may achieve synergistic therapeutic effects. Moreover, besides the treatment of liver-restricted diseases the default protolerogenic state of liver NPCs, and here especially of LSECs, may be exploited to imprint systemic antigen-specific tolerance for the treatment of allergies and autoimmune diseases. Such approaches may be combined with nano-vaccines that target DCs in the periphery.

Author Contributions

Conceptualization, M.B.; original draft writing, L.K., M.J.L., P.S., M.S., Y.Z., S.F., M.L.C., S.G., P.R.G. and M.B.; supervision, L.K., S.G. and M.B. All authors have read and agreed to the published version of the manuscript.

Funding

L.K., S.G. and M.B. are funded by Deutsche Forschungsgemeinschaft (SFB1066), grant numbers B15 (S.G., M.B.) and B17 (L.K.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank the Research Center for Immunotherapy (FZI; University Medical Center, Mainz, Germany) for continuous support.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

CAFCancer-associated fibroblast
CCCCholangiocarcinoma
CCLCC-chemokine ligand
CELCelastrol
CDCluster of differentiation
CTLCytotoxic T lymphocyte
DCDendritic cell
DC-SIGNDC-specific intercellular adhesion molecule-3-grabbing non-integrin
DSPCDistearoylphosphatidylcholine
EAEExperimental autoimmune encephalomyelitits
ECMExtracellular matrix
EGFREpidermal growth factor receptor
HAVHepatitis virus A
HBVHepatitis virus B
HCCHepatocellular carcinoma
HCVHepatitis virus C
HDVHepatitis virus D
HEVHepatitis virus E
HGFHepatocyte growth factor
HSCHepatic stellate cell
ICBImmune checkpoint blockade
IFN-αInterferon alpha
ILInterleukin
IMQImiquimod
immTORPLGA NP containing mTOR
KCKupffer cell
KLF2Kruppel-like factor 2
Lac-BSALactosylated bovine serum albumin
LNPLipid NP
MDSCMyeloid-derived suppressor cell
mTORMechanistic target of rapamycin
NAFLDNon-alcoholic fatty liver disease
Nano-Poly-BABoronic acid-containing polymer nanocomplex
NASHNon-alcoholic steatohepatitis
NF-κBNuclear factor kappa-light-chain-enhancer of activated B cells
NPNanoparticle
NPCNon-parenchymal cell
OVAOvalbumin
PD-1Programmed cell death protein 1
PD-L1Programmed cell death-1 ligand 1
PEGPolyethylene glycol
PI3KPhosphoinositide 3-kinase
PLAPoly(L-lactic acid)
PLGAPoly(L-lactic-co-glycolic acid)
PMNPolymorphonuclear neutrophilic leukocyte
siRNASmall interfering RNA
TAMTumor-associated macrophage
ThT helper cell
TMETumor microenvironment
TregRegulatory T cell
VEGFVascular endothelial growth factor

References

  1. Trefts, E.; Gannon, M.; Wasserman, D.H. The liver. Curr. Biol. 2017, 27, R1147–R1151. [Google Scholar] [CrossRef] [PubMed]
  2. Yang, Q.; Zhang, S.; Ma, J.; Liu, S.; Chen, S. In Search of Zonation Markers to Identify Liver Functional Disorders. Oxidative Med. Cell. Longev. 2020, 2020, 9374896. [Google Scholar] [CrossRef] [PubMed]
  3. Rizzo, G.E.M.; Cabibbo, G.; Craxì, A. Hepatitis B Virus-Associated Hepatocellular Carcinoma. Viruses 2022, 14, 986. [Google Scholar] [CrossRef] [PubMed]
  4. Ghidini, M.; Ramai, D.; Facciorusso, A.; Singh, J.; Tai, W.; Rijavec, E.; Galassi, B.; Grossi, F.; Indini, A. Metabolic disorders and the risk of cholangiocarcinoma. Expert Rev. Gastroenterol. Hepatol. 2021, 15, 999–1007. [Google Scholar] [CrossRef]
  5. Llovet, J.M.; Kelley, R.K.; Villanueva, A.; Singal, A.G.; Pikarsky, E.; Roayaie, S.; Lencioni, R.; Koike, K.; Zucman-Rossi, J.; Finn, R.S. Hepatocellular carcinoma. Nat. Rev. Dis. Primers 2021, 7, 6. [Google Scholar] [CrossRef]
  6. Tsilimigras, D.I.; Brodt, P.; Clavien, P.A.; Muschel, R.J.; D’Angelica, M.I.; Endo, I.; Parks, R.W.; Doyle, M.; de Santibañes, E.; Pawlik, T.M. Liver metastases. Nat. Rev. Dis. Primers 2021, 7, 27. [Google Scholar] [CrossRef]
  7. Zheng, M.; Tian, Z. Liver-Mediated Adaptive Immune Tolerance. Front. Immunol. 2019, 10, 2525. [Google Scholar] [CrossRef] [Green Version]
  8. George, A.; Shah, P.A.; Shrivastav, P.S. Natural biodegradable polymers based nano-formulations for drug delivery: A review. Int. J. Pharm. 2019, 561, 244–264. [Google Scholar] [CrossRef]
  9. Li, J.; Chen, C.; Xia, T. Understanding Nanomaterial-Liver Interactions to Facilitate the Development of Safer Nanoapplications. Adv. Mater. 2022, 34, e2106456. [Google Scholar] [CrossRef]
  10. Böttger, R.; Pauli, G.; Chao, P.H.; Al Fayez, N.; Hohenwarter, L.; Li, S.D. Lipid-based nanoparticle technologies for liver targeting. Adv. Drug Deliv. Rev. 2020, 154–155, 79–101. [Google Scholar] [CrossRef]
  11. Szafranska, K.; Kruse, L.D.; Holte, C.F.; McCourt, P.; Zapotoczny, B. The whole Story About Fenestrations in LSEC. Front. Physiol. 2021, 12, 735573. [Google Scholar] [CrossRef]
  12. Jacobs, F.; Wisse, E.; De Geest, B. The role of liver sinusoidal cells in hepatocyte-directed gene transfer. Am. J. Pathol. 2010, 176, 14–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Dou, L.; Shi, X.; He, X.; Gao, Y. Macrophage Phenotype and Function in Liver Disorder. Front. Immunol. 2019, 10, 3112. [Google Scholar] [CrossRef] [Green Version]
  14. Bhandari, S.; Li, R.; Simón-Santamaría, J.; McCourt, P.; Johansen, S.D.; Smedsrød, B.; Martinez-Zubiaurre, I.; Sørensen, K.K. Transcriptome and proteome profiling reveal complementary scavenger and immune features of rat liver sinusoidal endothelial cells and liver macrophages. BMC Mol. Cell Biol. 2020, 21, 85. [Google Scholar] [CrossRef] [PubMed]
  15. Bhandari, S.; Larsen, A.K.; McCourt, P.; Smedsrød, B.; Sørensen, K.K. The Scavenger Function of Liver Sinusoidal Endothelial Cells in Health and Disease. Front. Physiol. 2021, 12, 757469. [Google Scholar] [CrossRef] [PubMed]
  16. Adepu, S.; Ramakrishna, S. Controlled Drug Delivery Systems: Current Status and Future Directions. Molecules 2021, 26, 5905. [Google Scholar] [CrossRef]
  17. Meng, Q.Y.; Cong, H.L.; Hu, H.; Xu, F.J. Rational design and latest advances of codelivery systems for cancer therapy. Mater. Today Bio 2020, 7, 100056. [Google Scholar] [CrossRef]
  18. Homan, M.; Warrier, G.; Lao, C.D.; Yentz, S.; Kraft, S.; Fecher, L.A. Treatment related toxicities with combination BRAF and MEK inhibitor therapy in resected stage III melanoma. Front. Oncol. 2022, 12, 855794. [Google Scholar] [CrossRef] [PubMed]
  19. De Martin, E.; Michot, J.M.; Papouin, B.; Champiat, S.; Mateus, C.; Lambotte, O.; Roche, B.; Antonini, T.M.; Coilly, A.; Laghouati, S.; et al. Characterization of liver injury induced by cancer immunotherapy using immune checkpoint inhibitors. J. Hepatol. 2018, 68, 1181–1190. [Google Scholar] [CrossRef]
  20. Katopodi, T.; Petanidis, S.; Tsavlis, D.; Anestakis, D.; Charalampidis, C.; Chatziprodromidou, I.; Eskitzis, P.; Zarogoulidis, P.; Kosmidis, C.; Matthaios, D.; et al. Engineered multifunctional nanocarriers for controlled drug delivery in tumor immunotherapy. Front. Oncol. 2022, 12, 1042125. [Google Scholar] [CrossRef] [PubMed]
  21. Fung, S.; Choi, H.S.J.; Gehring, A.; Janssen, H.L.A. Getting to HBV cure: The promising paths forward. Hepatology 2022, 76, 233–250. [Google Scholar] [CrossRef]
  22. Gehring, S.; Pietrzak-Nguyen, A.; Fichter, M.; Landfester, K. Novel strategies in vaccine design: Can nanocapsules help prevent and treat hepatitis B? Nanomedicine 2017, 12, 1205–1207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Medina-Montano, C.; Rivero Berti, I.; Gambaro, R.C.; Limeres, M.J.; Svensson, M.; Padula, G.; Chain, C.Y.; Cisneros, J.S.; Castro, G.R.; Grabbe, S.; et al. Nanostructured Lipid Carriers Loaded with Dexamethasone Prevent Inflammatory Responses in Primary Non-Parenchymal Liver Cells. Pharmaceutics 2022, 14, 1611. [Google Scholar] [CrossRef]
  24. Kisseleva, T.; Brenner, D. Molecular and cellular mechanisms of liver fibrosis and its regression. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 151–166. [Google Scholar] [CrossRef] [PubMed]
  25. Dhar, D.; Baglieri, J.; Kisseleva, T.; Brenner, D.A. Mechanisms of liver fibrosis and its role in liver cancer. Exp. Biol. Med. 2020, 245, 96–108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Cheng, C.; Li, C.; Zhu, X.; Han, W.; Li, J.; Lv, Y. Doxorubicin-loaded Fe3O4-ZIF-8 nano-composites for hepatocellular carcinoma therapy. J. Biomater. Appl. 2019, 33, 1373–1381. [Google Scholar] [CrossRef] [PubMed]
  27. Huang, Y.; Wang, T.; Yang, J.; Wu, X.; Fan, W.; Chen, J. Current Strategies for the Treatment of Hepatocellular Carcinoma by Modulating the Tumor Microenvironment via Nano-Delivery Systems: A Review. Int. J. Nanomed. 2022, 17, 2335–2352. [Google Scholar] [CrossRef]
  28. Xu, J.; Zheng, Q.; Cheng, X.; Hu, S.; Zhang, C.; Zhou, X.; Sun, P.; Wang, W.; Su, Z.; Zou, T.; et al. Chemo-photodynamic therapy with light-triggered disassembly of theranostic nanoplatform in combination with checkpoint blockade for immunotherapy of hepatocellular carcinoma. J. Nanobiotechnol. 2021, 19, 355. [Google Scholar] [CrossRef]
  29. Breous, E.; Somanathan, S.; Vandenberghe, L.H.; Wilson, J.M. Hepatic regulatory T cells and Kupffer cells are crucial mediators of systemic T cell tolerance to antigens targeting murine liver. Hepatology 2009, 50, 612–621. [Google Scholar] [CrossRef] [Green Version]
  30. Kruse, N.; Neumann, K.; Schrage, A.; Derkow, K.; Schott, E.; Erben, U.; Kühl, A.; Loddenkemper, C.; Zeitz, M.; Hamann, A.; et al. Priming of CD4+ T cells by liver sinusoidal endothelial cells induces CD25low forkhead box protein 3-regulatory T cells suppressing autoimmune hepatitis. Hepatology 2009, 50, 1904–1913. [Google Scholar] [CrossRef]
  31. Bamboat, Z.M.; Stableford, J.A.; Plitas, G.; Burt, B.M.; Nguyen, H.M.; Welles, A.P.; Gonen, M.; Young, J.W.; DeMatteo, R.P. Human liver dendritic cells promote T cell hyporesponsiveness. J. Immunol. 2009, 182, 1901–1911. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Liu, Q.; Wang, X.; Liu, X.; Liao, Y.P.; Chang, C.H.; Mei, K.C.; Jiang, J.; Tseng, S.; Gochman, G.; Huang, M.; et al. Antigen- and Epitope-Delivering Nanoparticles Targeting Liver Induce Comparable Immunotolerance in Allergic Airway Disease and Anaphylaxis as Nanoparticle-Delivering Pharmaceuticals. ACS Nano 2021, 15, 1608–1626. [Google Scholar] [CrossRef] [PubMed]
  33. Carambia, A.; Gottwick, C.; Schwinge, D.; Stein, S.; Digigow, R.; Şeleci, M.; Mungalpara, D.; Heine, M.; Schuran, F.A.; Corban, C.; et al. Nanoparticle-mediated targeting of autoantigen peptide to cross-presenting liver sinusoidal endothelial cells protects from CD8 T-cell-driven autoimmune cholangitis. Immunology 2021, 162, 452–463. [Google Scholar] [CrossRef] [PubMed]
  34. Mehta, P.; Reddivari, A.K.R. Hepatitis. In StatPearls; StatPearls Publishing: St. Petersburg, FL, USA, 2022. [Google Scholar]
  35. Maya, R.; Gershwin, M.E.; Shoenfeld, Y. Hepatitis B virus (HBV) and autoimmune disease. Clin. Rev. Allergy Immunol. 2008, 34, 85–102. [Google Scholar] [CrossRef]
  36. Buschow, S.I.; Jansen, D. CD4+ T Cells in Chronic Hepatitis B and T Cell-Directed Immunotherapy. Cells 2021, 10, 1114. [Google Scholar] [CrossRef]
  37. Maini, M.K.; Boni, C.; Lee, C.K.; Larrubia, J.R.; Reignat, S.; Ogg, G.S.; King, A.S.; Herberg, J.; Gilson, R.; Alisa, A.; et al. The role of virus-specific CD8(+) cells in liver damage and viral control during persistent hepatitis B virus infection. J. Exp. Med. 2000, 191, 1269–1280. [Google Scholar] [CrossRef]
  38. Xu, J.; Liu, X.; Koyama, Y.; Wang, P.; Lan, T.; Kim, I.G.; Kim, I.H.; Ma, H.Y.; Kisseleva, T. The types of hepatic myofibroblasts contributing to liver fibrosis of different etiologies. Front. Pharmacol. 2014, 5, 167. [Google Scholar] [CrossRef] [Green Version]
  39. Lurje, I.; Gaisa, N.T.; Weiskirchen, R.; Tacke, F. Mechanisms of organ fibrosis: Emerging concepts and implications for novel treatment strategies. Mol. Asp. Med. 2023, 92, 101191. [Google Scholar] [CrossRef]
  40. Quaglia, A.; Alves, V.A.; Balabaud, C.; Bhathal, P.S.; Bioulac-Sage, P.; Crawford, J.M.; Dhillon, A.P.; Ferrell, L.; Guido, M.; Hytiroglou, P.; et al. Role of aetiology in the progression, regression, and parenchymal remodelling of liver disease: Implications for liver biopsy interpretation. Histopathology 2016, 68, 953–967. [Google Scholar] [CrossRef]
  41. De Franchis, R.; Bosch, J.; Garcia-Tsao, G.; Reiberger, T.; Ripoll, C. Baveno VII—Renewing consensus in portal hypertension. J. Hepatol. 2022, 76, 959–974. [Google Scholar] [CrossRef]
  42. Wiktor, S.Z. Viral Hepatitis. In Major Infectious Diseases; Holmes, K.K., Bertozzi, S., Bloom, B.R., Jha, P., Eds.; The International Bank for Reconstruction and Development, The World Bank: Washington, DC, USA, 2017. [Google Scholar]
  43. Tsukuda, S.; Watashi, K. Hepatitis B virus biology and life cycle. Antivir. Res. 2020, 182, 104925. [Google Scholar] [CrossRef] [PubMed]
  44. Netter, H.J.; Barrios, M.H.; Littlejohn, M.; Yuen, L.K.W. Hepatitis Delta Virus (HDV) and Delta-Like Agents: Insights Into Their Origin. Front. Microbiol. 2021, 12, 652962. [Google Scholar] [CrossRef] [PubMed]
  45. Castaneda, D.; Gonzalez, A.J.; Alomari, M.; Tandon, K.; Zervos, X.B. From hepatitis A to E: A critical review of viral hepatitis. World J. Gastroenterol. 2021, 27, 1691–1715. [Google Scholar] [CrossRef]
  46. Klöhn, M.; Schrader, J.A.; Brüggemann, Y.; Todt, D.; Steinmann, E. Beyond the Usual Suspects: Hepatitis E Virus and Its Implications in Hepatocellular Carcinoma. Cancers 2021, 13, 5867. [Google Scholar] [CrossRef] [PubMed]
  47. Krause, A.; Haberkorn, U.; Mier, W. Strategies for the treatment of HBV/HDV. Eur. J. Pharmacol. 2018, 833, 379–391. [Google Scholar] [CrossRef] [PubMed]
  48. EASL 2017 Clinical Practice Guidelines on the management of hepatitis B virus infection. J. Hepatol. 2017, 67, 370–398. [CrossRef] [Green Version]
  49. EASL recommendations on treatment of hepatitis C: Final update of the series(☆). J. Hepatol. 2020, 73, 1170–1218. [CrossRef]
  50. Joon, A.; Rao, P.; Shenoy, S.M.; Baliga, S. Prevalence of Hepatitis A virus (HAV) and Hepatitis E virus (HEV) in the patients presenting with acute viral hepatitis. Indian J. Med. Microbiol. 2015, 33, S102–S105. [Google Scholar] [CrossRef]
  51. Pawlotsky, J.M. Interferon-Free Hepatitis C Virus Therapy. Cold Spring Harb. Perspect. Med. 2020, 10, a036855. [Google Scholar] [CrossRef]
  52. El-Shafai, N.M.; Masoud, M.S.; Ibrahim, M.M.; Ramadan, M.S.; Mersal, G.A.M.; El-Mehasseb, I.M. Drug delivery of sofosbuvir drug capsulated with the β-cyclodextrin basket loaded on chitosan nanoparticle surface for anti-hepatitis C virus (HCV). Int. J. Biol. Macromol. 2022, 207, 402–413. [Google Scholar] [CrossRef]
  53. Kouroumalis, E.; Voumvouraki, A. Hepatitis C virus: A critical approach to who really needs treatment. World J. Hepatol. 2022, 14, 1. [Google Scholar] [CrossRef] [PubMed]
  54. Hu, J.; Cheng, J.; Tang, L.; Hu, Z.; Luo, Y.; Li, Y.; Zhou, T.; Chang, J.; Guo, J.T. Virological Basis for the Cure of Chronic Hepatitis B. ACS Infect. Dis. 2019, 5, 659–674. [Google Scholar] [CrossRef] [PubMed]
  55. Testoni, B.; Lebossé, F.; Scholtes, C.; Berby, F.; Miaglia, C.; Subic, M.; Loglio, A.; Facchetti, F.; Lampertico, P.; Levrero, M.; et al. Serum hepatitis B core-related antigen (HBcrAg) correlates with covalently closed circular DNA transcriptional activity in chronic hepatitis B patients. J. Hepatol. 2019, 70, 615–625. [Google Scholar] [CrossRef]
  56. Cao, X.; Wang, Y.; Li, P.; Huang, W.; Lu, X.; Lu, H. HBV Reactivation During the Treatment of Non-Hodgkin Lymphoma and Management Strategies. Front. Oncol. 2021, 11, 685706. [Google Scholar] [CrossRef] [PubMed]
  57. Pattyn, J.; Hendrickx, G.; Vorsters, A.; Van Damme, P. Hepatitis B Vaccines. J. Infect. Dis. 2021, 224, S343–S351. [Google Scholar] [CrossRef]
  58. Firdaus, F.Z.; Skwarczynski, M.; Toth, I. Developments in Vaccine Adjuvants. Methods Mol. Biol. 2022, 2412, 145–178. [Google Scholar] [CrossRef]
  59. Oelschlager, K.A.; Termini, M.S.; Stevenson, C. Preventing Hepatitis B Virus Infection Among U.S. Military Personnel: Potential Impact of a 2-Dose Versus 3-Dose Vaccine on Medical Readiness. Mil. Med. 2022, 188, e2067–e2073. [Google Scholar] [CrossRef]
  60. Das, S.; Ramakrishnan, K.; Behera, S.K.; Ganesapandian, M.; Xavier, A.S.; Selvarajan, S. Hepatitis B Vaccine and Immunoglobulin: Key Concepts. J. Clin. Transl. Hepatol. 2019, 7, 165–171. [Google Scholar] [CrossRef] [Green Version]
  61. Shan, P.; Wang, Z.; Li, J.; Wei, D.; Zhang, Z.; Hao, S.; Hou, Y.; Wang, Y.; Li, S.; Wang, X.; et al. A New Nano Adjuvant of PF3 Used for an Enhanced Hepatitis B Vaccine. Front. Bioeng. Biotechnol. 2022, 10, 903424. [Google Scholar] [CrossRef]
  62. Qiao, D.; Chen, Y.; Liu, L. Engineered therapeutic nanovaccine against chronic hepatitis B virus infection. Biomaterials 2021, 269, 120674. [Google Scholar] [CrossRef]
  63. Ye, J.; Chen, J. Interferon and Hepatitis B: Current and Future Perspectives. Front. Immunol. 2021, 12, 733364. [Google Scholar] [CrossRef]
  64. Al Fayez, N.; Rouhollahi, E.; Ong, C.Y.; Wu, J.; Nguyen, A.; Böttger, R.; Cullis, P.R.; Witzigmann, D.; Li, S.D. Hepatocyte-targeted delivery of imiquimod reduces hepatitis B virus surface antigen. J. Control. Release 2022, 350, 630–641. [Google Scholar] [CrossRef]
  65. Olivas, I.; Rodríguez-Tajes, S.; Londoño, M.C. Autoimmune hepatitis: Challenges and novelties. Med. Clin. 2022, 159, 289–298. [Google Scholar] [CrossRef]
  66. Komori, A. Recent updates on the management of autoimmune hepatitis. Clin. Mol. Hepatol. 2021, 27, 58–69. [Google Scholar] [CrossRef]
  67. Terziroli Beretta-Piccoli, B.; Mieli-Vergani, G.; Vergani, D. Autoimmune hepatitis: Standard treatment and systematic review of alternative treatments. World J. Gastroenterol. 2017, 23, 6030–6048. [Google Scholar] [CrossRef]
  68. Nieman, L.K. Cushing’s syndrome: Update on signs, symptoms and biochemical screening. Eur. J. Endocrinol. 2015, 173, M33–M38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Jothimani, D.; Cramp, M.E.; Mitchell, J.D.; Cross, T.J. Treatment of autoimmune hepatitis: A review of current and evolving therapies. J. Gastroenterol. Hepatol. 2011, 26, 619–627. [Google Scholar] [CrossRef] [PubMed]
  70. Violatto, M.B.; Casarin, E.; Talamini, L.; Russo, L.; Baldan, S.; Tondello, C.; Messmer, M.; Hintermann, E.; Rossi, A.; Passoni, A.; et al. Dexamethasone Conjugation to Biodegradable Avidin-Nucleic-Acid-Nano-Assemblies Promotes Selective Liver Targeting and Improves Therapeutic Efficacy in an Autoimmune Hepatitis Murine Model. ACS Nano 2019, 13, 4410–4423. [Google Scholar] [CrossRef]
  71. Ongaro, A.; Violatto, M.B.; Casarin, E.; Pellerani, I.; Marchini, G.; Ribaudo, G.; Salmona, M.; Carbone, M.; Passoni, A.; Gnodi, E.; et al. The mode of dexamethasone decoration influences avidin-nucleic-acid-nano-assembly organ biodistribution and in vivo drug persistence. Nanomed. Nanotechnol. Biol. Med. 2022, 40, 102497. [Google Scholar] [CrossRef] [PubMed]
  72. Shen, S.; Dai, H.; Fei, Z.; Chai, Y.; Hao, Y.; Fan, Q.; Dong, Z.; Zhu, Y.; Xu, J.; Ma, Q.; et al. Immunosuppressive Nanoparticles for Management of Immune-Related Adverse Events in Liver. ACS Nano 2021, 15, 9111–9125. [Google Scholar] [CrossRef]
  73. Jennings, J.J.; Mandaliya, R.; Nakshabandi, A.; Lewis, J.H. Hepatotoxicity induced by immune checkpoint inhibitors: A comprehensive review including current and alternative management strategies. Expert Opin. Drug Metab. Toxicol. 2019, 15, 231–244. [Google Scholar] [CrossRef] [PubMed]
  74. Ye, Q.; Zou, B.; Yeo, Y.H.; Li, J.; Huang, D.Q.; Wu, Y.; Yang, H.; Liu, C.; Kam, L.Y.; Tan, X.X.E.; et al. Global prevalence, incidence, and outcomes of non-obese or lean non-alcoholic fatty liver disease: A systematic review and meta-analysis. Lancet Gastroenterol. Hepatol. 2020, 5, 739–752. [Google Scholar] [CrossRef] [PubMed]
  75. Idilman, I.S.; Ozdeniz, I.; Karcaaltincaba, M. Hepatic Steatosis: Etiology, Patterns, and Quantification. Semin. Ultrasound CT MR 2016, 37, 501–510. [Google Scholar] [CrossRef] [PubMed]
  76. Schuppan, D.; Surabattula, R.; Wang, X.Y. Determinants of fibrosis progression and regression in NASH. J. Hepatol. 2018, 68, 238–250. [Google Scholar] [CrossRef]
  77. Anstee, Q.M.; Reeves, H.L.; Kotsiliti, E.; Govaere, O.; Heikenwalder, M. From NASH to HCC: Current concepts and future challenges. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 411–428. [Google Scholar] [CrossRef]
  78. Neuschwander-Tetri, B.A.; Loomba, R.; Sanyal, A.J.; Lavine, J.E.; Van Natta, M.L.; Abdelmalek, M.F.; Chalasani, N.; Dasarathy, S.; Diehl, A.M.; Hameed, B.; et al. Farnesoid X nuclear receptor ligand obeticholic acid for non-cirrhotic, non-alcoholic steatohepatitis (FLINT): A multicentre, randomised, placebo-controlled trial. Lancet 2015, 385, 956–965. [Google Scholar] [CrossRef] [Green Version]
  79. Pouwels, S.; Sakran, N.; Graham, Y.; Leal, A.; Pintar, T.; Yang, W.; Kassir, R.; Singhal, R.; Mahawar, K.; Ramnarain, D. Non-alcoholic fatty liver disease (NAFLD): A review of pathophysiology, clinical management and effects of weight loss. BMC Endocr. Disord. 2022, 22, 63. [Google Scholar] [CrossRef]
  80. Li, M.; Xie, F.; Wang, L.; Zhu, G.; Qi, L.W.; Jiang, S. Celastrol: An Update on Its Hepatoprotective Properties and the Linked Molecular Mechanisms. Front. Pharmacol. 2022, 13, 857956. [Google Scholar] [CrossRef]
  81. Fan, N.; Zhao, J.; Zhao, W.; Zhang, X.; Song, Q.; Shen, Y.; Shum, H.C.; Wang, Y.; Rong, J. Celastrol-loaded lactosylated albumin nanoparticles attenuate hepatic steatosis in non-alcoholic fatty liver disease. J. Control. Release 2022, 347, 44–54. [Google Scholar] [CrossRef]
  82. Agrawal, N.; Sharma, M.; Singh, S.; Goyal, A. Recent Advances of α-Glucosidase Inhibitors: A Comprehensive Review. Curr. Top. Med. Chem. 2022, 22, 2069–2086. [Google Scholar] [CrossRef]
  83. Zhao, X.; Zhang, H.; Li, J.; Tian, M.; Yang, J.; Sun, S.; Hu, Q.; Yang, L.; Zhang, S. Orally administered saccharide-sequestering nanocomplex to manage carbohydrate metabolism disorders. Sci. Adv. 2021, 7, abf7311. [Google Scholar] [CrossRef] [PubMed]
  84. Lan, T.; Li, C.; Yang, G.; Sun, Y.; Zhuang, L.; Ou, Y.; Li, H.; Wang, G.; Kisseleva, T.; Brenner, D.; et al. Sphingosine kinase 1 promotes liver fibrosis by preventing miR-19b-3p-mediated inhibition of CCR2. Hepatology 2018, 68, 1070–1086. [Google Scholar] [CrossRef] [PubMed]
  85. Zanieri, F.; Levi, A.; Montefusco, D.; Longato, L.; De Chiara, F.; Frenguelli, L.; Omenetti, S.; Andreola, F.; Luong, T.V.; Massey, V.; et al. Exogenous Liposomal Ceramide-C6 Ameliorates Lipidomic Profile, Energy Homeostasis, and Anti-Oxidant Systems in NASH. Cells 2020, 9, 1237. [Google Scholar] [CrossRef]
  86. Ma, H.; Liu, X.; Zhang, M.; Niu, J. Liver sinusoidal endothelial cells are implicated in multiple fibrotic mechanisms. Mol. Biol. Rep. 2021, 48, 2803–2815. [Google Scholar] [CrossRef] [PubMed]
  87. Senoo, H.; Mezaki, Y.; Fujiwara, M. The stellate cell system (vitamin A-storing cell system). Anat. Sci. Int. 2017, 92, 387–455. [Google Scholar] [CrossRef]
  88. Duong, H.T.; Dong, Z.; Su, L.; Boyer, C.; George, J.; Davis, T.P.; Wang, J. The use of nanoparticles to deliver nitric oxide to hepatic stellate cells for treating liver fibrosis and portal hypertension. Small 2015, 11, 2291–2304. [Google Scholar] [CrossRef]
  89. Perri, R.E.; Langer, D.A.; Chatterjee, S.; Gibbons, S.J.; Gadgil, J.; Cao, S.; Farrugia, G.; Shah, V.H. Defects in cGMP-PKG pathway contribute to impaired NO-dependent responses in hepatic stellate cells upon activation. Am. J. Physiol. Gastrointest. Liver Physiol. 2006, 290, G535–G542. [Google Scholar] [CrossRef]
  90. Furuhashi, H.; Tomita, K.; Teratani, T.; Shimizu, M.; Nishikawa, M.; Higashiyama, M.; Takajo, T.; Shirakabe, K.; Maruta, K.; Okada, Y.; et al. Vitamin A-coupled liposome system targeting free cholesterol accumulation in hepatic stellate cells offers a beneficial therapeutic strategy for liver fibrosis. Hepatol. Res. 2018, 48, 397–407. [Google Scholar] [CrossRef]
  91. Dewidar, B.; Meyer, C.; Dooley, S.; Meindl-Beinker, A.N. TGF-β in Hepatic Stellate Cell Activation and Liver Fibrogenesis-Updated 2019. Cells 2019, 8, 1419. [Google Scholar] [CrossRef] [Green Version]
  92. Qiao, J.B.; Fan, Q.Q.; Xing, L.; Cui, P.F.; He, Y.J.; Zhu, J.C.; Wang, L.; Pang, T.; Oh, Y.K.; Zhang, C.; et al. Vitamin A-decorated biocompatible micelles for chemogene therapy of liver fibrosis. J. Control. Release 2018, 283, 113–125. [Google Scholar] [CrossRef]
  93. Ezhilarasan, D.; Karthikeyan, S.; Vivekanandan, P. Ameliorative effect of silibinin against N-nitrosodimethylamine-induced hepatic fibrosis in rats. Environ. Toxicol. Pharmacol. 2012, 34, 1004–1013. [Google Scholar] [CrossRef]
  94. Wang, Q.; Peng, Z.; Xiao, S.; Geng, S.; Yuan, J.; Li, Z. RNAi-mediated inhibition of COL1A1 and COL3A1 in human skin fibroblasts. Exp. Dermatol. 2007, 16, 611–617. [Google Scholar] [CrossRef] [PubMed]
  95. Kim, K.S.; Hur, W.; Park, S.J.; Hong, S.W.; Choi, J.E.; Goh, E.J.; Yoon, S.K.; Hahn, S.K. Bioimaging for targeted delivery of hyaluronic Acid derivatives to the livers in cirrhotic mice using quantum dots. ACS Nano 2010, 4, 3005–3014. [Google Scholar] [CrossRef] [PubMed]
  96. Zhang, L.F.; Wang, X.H.; Zhang, C.L.; Lee, J.; Duan, B.W.; Xing, L.; Li, L.; Oh, Y.K.; Jiang, H.L. Sequential Nano-Penetrators of Capillarized Liver Sinusoids and Extracellular Matrix Barriers for Liver Fibrosis Therapy. ACS Nano 2022, 16, 14029–14042. [Google Scholar] [CrossRef] [PubMed]
  97. Kaps, L.; Nuhn, L.; Aslam, M.; Brose, A.; Foerster, F.; Rosigkeit, S.; Renz, P.; Heck, R.; Kim, Y.O.; Lieberwirth, I.; et al. In Vivo Gene-Silencing in Fibrotic Liver by siRNA-Loaded Cationic Nanohydrogel Particles. Adv. Healthc. Mater. 2015, 4, 2809–2815. [Google Scholar] [CrossRef]
  98. Leber, N.; Kaps, L.; Aslam, M.; Schupp, J.; Brose, A.; Schäffel, D.; Fischer, K.; Diken, M.; Strand, D.; Koynov, K.; et al. SiRNA-mediated in vivo gene knockdown by acid-degradable cationic nanohydrogel particles. J. Control. Release 2017, 248, 10–23. [Google Scholar] [CrossRef]
  99. Yu, Z.; Guo, J.; Liu, Y.; Wang, M.; Liu, Z.; Gao, Y.; Huang, L. Nano delivery of simvastatin targets liver sinusoidal endothelial cells to remodel tumor microenvironment for hepatocellular carcinoma. J. Nanobiotechnol. 2022, 20, 9. [Google Scholar] [CrossRef]
  100. Bennett, R.G.; Heimann, D.G.; Singh, S.; Simpson, R.L.; Tuma, D.J. Relaxin decreases the severity of established hepatic fibrosis in mice. Liver Int. 2014, 34, 416–426. [Google Scholar] [CrossRef] [Green Version]
  101. Jaskiewicz, K.; Chasen, M.R. Differential expression of transforming growth factor alpha, adhesions molecules and integrins in primary, metastatic liver tumors and in liver cirrhosis. Anticancer Res. 1995, 15, 559–562. [Google Scholar]
  102. Kim, S.Y.; Lee, C.H.; Midura, B.V.; Yeung, C.; Mendoza, A.; Hong, S.H.; Ren, L.; Wong, D.; Korz, W.; Merzouk, A.; et al. Inhibition of the CXCR4/CXCL12 chemokine pathway reduces the development of murine pulmonary metastases. Clin. Exp. Metastasis 2008, 25, 201–211. [Google Scholar] [CrossRef] [Green Version]
  103. Zhou, R.; Zhang, M.; He, J.; Liu, J.; Sun, X.; Ni, P. Functional cRGD-Conjugated Polymer Prodrug for Targeted Drug Delivery to Liver Cancer Cells. ACS Omega 2022, 7, 21325–21336. [Google Scholar] [CrossRef]
  104. Gao, Y.; Zheng, Q.C.; Xu, S.; Yuan, Y.; Cheng, X.; Jiang, S.; Kenry; Yu, Q.; Song, Z.; Liu, B.; et al. Theranostic Nanodots with Aggregation-Induced Emission Characteristic for Targeted and Image-Guided Photodynamic Therapy of Hepatocellular Carcinoma. Theranostics 2019, 9, 1264–1279. [Google Scholar] [CrossRef] [PubMed]
  105. Schwarze, P.E.; Tolleshaug, H.; Seglen, P.O. Endocytosis of asialo-glycoproteins and attachment to asialo-glycoproteins of hepatocytes from carcinogen-treated rats. Toxicol. Pathol. 1987, 15, 88–92. [Google Scholar] [CrossRef] [PubMed]
  106. Duan, W.; Liu, Y. Targeted and synergistic therapy for hepatocellular carcinoma: Monosaccharide modified lipid nanoparticles for the co-delivery of doxorubicin and sorafenib. Drug Des. Dev. Ther. 2018, 12, 2149–2161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Li, X.; Wang, X.; Liu, N.; Wang, Q.; Hu, J. Inhibition of Metastatic Hepatocarcinoma by Combined Chemotherapy with Silencing VEGF/VEGFR2 Genes through a GalNAc-Modified Integrated Therapeutic System. Molecules 2022, 27, 2082. [Google Scholar] [CrossRef]
  108. Hu, J.; Hu, J.; Wu, W.; Qin, Y.; Fu, J.; Zhou, J.; Liu, C.; Yin, J. N-acetyl-galactosamine modified metal-organic frameworks to inhibit the growth and pulmonary metastasis of liver cancer stem cells through targeted chemotherapy and starvation therapy. Acta Biomater. 2022, 151, 588–599. [Google Scholar] [CrossRef]
  109. Asai, R.; Tsuchiya, H.; Amisaki, M.; Makimoto, K.; Takenaga, A.; Sakabe, T.; Hoi, S.; Koyama, S.; Shiota, G. CD44 standard isoform is involved in maintenance of cancer stem cells of a hepatocellular carcinoma cell line. Cancer Med. 2019, 8, 773–782. [Google Scholar] [CrossRef]
  110. Wang, L.; Su, W.; Liu, Z.; Zhou, M.; Chen, S.; Chen, Y.; Lu, D.; Liu, Y.; Fan, Y.; Zheng, Y.; et al. CD44 antibody-targeted liposomal nanoparticles for molecular imaging and therapy of hepatocellular carcinoma. Biomaterials 2012, 33, 5107–5114. [Google Scholar] [CrossRef]
  111. Liu, X.; Liu, H.; Wang, S.L.; Liu, J.W. Hyaluronic acid derivative-modified nano-structured lipid carrier for cancer targeting and therapy. J. Zhejiang Univ. Sci. B 2020, 21, 571–580. [Google Scholar] [CrossRef]
  112. Cannito, S.; Bincoletto, V.; Turato, C.; Pontisso, P.; Scupoli, M.T.; Ailuno, G.; Andreana, I.; Stella, B.; Arpicco, S.; Bocca, C. Hyaluronated and PEGylated Liposomes as a Potential Drug-Delivery Strategy to Specifically Target Liver Cancer and Inflammatory Cells. Molecules 2022, 27, 1062. [Google Scholar] [CrossRef]
  113. Li, W.; Gomez, E.; Zhang, Z. Immunohistochemical expression of stromal cell-derived factor-1 (SDF-1) and CXCR4 ligand receptor system in hepatocellular carcinoma. J. Exp. Clin. Cancer Res. CR 2007, 26, 527–533. [Google Scholar] [PubMed]
  114. Chen, Y.; Liu, Y.C.; Sung, Y.C.; Ramjiawan, R.R.; Lin, T.T.; Chang, C.C.; Jeng, K.S.; Chang, C.F.; Liu, C.H.; Gao, D.Y.; et al. Overcoming sorafenib evasion in hepatocellular carcinoma using CXCR4-targeted nanoparticles to co-deliver MEK-inhibitors. Sci. Rep. 2017, 7, 44123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Huang, D.; Rao, D.; Jin, Q.; Lai, M.; Zhang, J.; Lai, Z.; Shen, H.; Zhong, T. Role of CD147 in the development and diagnosis of hepatocellular carcinoma. Front. Immunol. 2023, 14, 1149931. [Google Scholar] [CrossRef]
  116. Tian, H.; Huang, Y.; He, J.; Zhang, M.; Ni, P. CD147 Monoclonal Antibody Targeted Reduction-Responsive Camptothecin Polyphosphoester Nanomedicine for Drug Delivery in Hepatocellular Carcinoma Cells. ACS Appl. Bio Mater. 2021, 4, 4422–4431. [Google Scholar] [CrossRef] [PubMed]
  117. Li, M.; Deng, L.; Li, J.; Yuan, W.; Gao, X.; Ni, J.; Jiang, H.; Zeng, J.; Ren, J.; Wang, P. Actively Targeted Magnetothermally Responsive Nanocarriers/Doxorubicin for Thermochemotherapy of Hepatoma. ACS Appl. Mater. Interfaces 2018, 10, 41107–41117. [Google Scholar] [CrossRef] [PubMed]
  118. Jin, C.; Bai, L.; Lin, L.; Wang, S.; Yin, X. Paclitaxel-loaded nanoparticles decorated with bivalent fragment HAb18 F(ab’)2 and cell penetrating peptide for improved therapeutic effect on hepatocellular carcinoma. Artif. Cells Nanomed. Biotechnol. 2018, 46, 1076–1084. [Google Scholar] [CrossRef] [Green Version]
  119. Komposch, K.; Sibilia, M. EGFR Signaling in Liver Diseases. Int. J. Mol. Sci. 2015, 17, 30. [Google Scholar] [CrossRef]
  120. Li, Z.; Zhao, R.; Wu, X.; Sun, Y.; Yao, M.; Li, J.; Xu, Y.; Gu, J. Identification and characterization of a novel peptide ligand of epidermal growth factor receptor for targeted delivery of therapeutics. FASEB J. 2005, 19, 1978–1985. [Google Scholar] [CrossRef]
  121. Abdulmalek, S.; Mostafa, N.; Gomaa, M.; El-Kersh, M.; Elkady, A.I.; Balbaa, M. Bee venom-loaded EGFR-targeting peptide-coupled chitosan nanoparticles for effective therapy of hepatocellular carcinoma by inhibiting EGFR-mediated MEK/ERK pathway. PLoS ONE 2022, 17, e0272776. [Google Scholar] [CrossRef]
  122. Jia, L.; Li, J.; Li, P.; Liu, D.; Li, J.; Shen, J.; Zhu, B.; Ma, C.; Zhao, T.; Lan, R.; et al. Site-specific glycoproteomic analysis revealing increased core-fucosylation on FOLR1 enhances folate uptake capacity of HCC cells to promote EMT. Theranostics 2021, 11, 6905–6921. [Google Scholar] [CrossRef]
  123. Ling, D.; Xia, H.; Park, W.; Hackett, M.J.; Song, C.; Na, K.; Hui, K.M.; Hyeon, T. pH-sensitive nanoformulated triptolide as a targeted therapeutic strategy for hepatocellular carcinoma. ACS Nano 2014, 8, 8027–8039. [Google Scholar] [CrossRef] [PubMed]
  124. Hu, B.G.; Liu, L.P.; Chen, G.G.; Ye, C.G.; Leung, K.K.; Ho, R.L.; Lin, M.C.; Lai, P.B. Therapeutic efficacy of improved α-fetoprotein promoter-mediated tBid delivered by folate-PEI600-cyclodextrin nanopolymer vector in hepatocellular carcinoma. Exp. Cell Res. 2014, 324, 183–191. [Google Scholar] [CrossRef] [PubMed]
  125. Cheng, M.; Dai, D. Inhibitory of active dual cancer targeting 5-Fluorouracil nanoparticles on liver cancer in vitro and in vivo. Front. Oncol. 2022, 12, 971475. [Google Scholar] [CrossRef]
  126. Cai, Y.; Xu, Y.; Chan, H.F.; Fang, X.; He, C.; Chen, M. Glycyrrhetinic Acid Mediated Drug Delivery Carriers for Hepatocellular Carcinoma Therapy. Mol. Pharm. 2016, 13, 699–709. [Google Scholar] [CrossRef] [PubMed]
  127. Wu, M.; Lian, B.; Deng, Y.; Feng, Z.; Zhong, C.; Wu, W.; Huang, Y.; Wang, L.; Zu, C.; Zhao, X. Resveratrol-loaded glycyrrhizic acid-conjugated human serum albumin nanoparticles wrapping resveratrol nanoparticles: Preparation, characterization, and targeting effect on liver tumors. J. Biomater. Appl. 2017, 32, 191–205. [Google Scholar] [CrossRef] [PubMed]
  128. Qu, Y.; Sun, F.; He, F.; Yu, C.; Lv, J.; Zhang, Q.; Liang, D.; Yu, C.; Wang, J.; Zhang, X.; et al. Glycyrrhetinic acid-modified graphene oxide mediated siRNA delivery for enhanced liver-cancer targeting therapy. Eur. J. Pharm. Sci. 2019, 139, 105036. [Google Scholar] [CrossRef]
  129. Li, Y.; Wu, J.; Lu, Q.; Liu, X.; Wen, J.; Qi, X.; Liu, J.; Lian, B.; Zhang, B.; Sun, H.; et al. GA&HA-Modified Liposomes for Co-Delivery of Aprepitant and Curcumin to Inhibit Drug-Resistance and Metastasis of Hepatocellular Carcinoma. Int. J. Nanomed. 2022, 17, 2559–2575. [Google Scholar] [CrossRef]
  130. Zhang, J.; Zhang, M.; Ma, H.; Song, X.; He, L.; Ye, X.; Li, X. Overexpression of glypican-3 is a predictor of poor prognosis in hepatocellular carcinoma: An updated meta-analysis. Medicine 2018, 97, e11130. [Google Scholar] [CrossRef]
  131. Ma, W.; Zhu, D.; Li, J.; Chen, X.; Xie, W.; Jiang, X.; Wu, L.; Wang, G.; Xiao, Y.; Liu, Z.; et al. Coating biomimetic nanoparticles with chimeric antigen receptor T cell-membrane provides high specificity for hepatocellular carcinoma photothermal therapy treatment. Theranostics 2020, 10, 1281–1295. [Google Scholar] [CrossRef]
  132. Su, D. The transcatheter arterial chemoembolization combined with targeted nanoparticle delivering sorafenib system for the treatment of microvascular invasion of hepatocellular carcinoma. Bioengineered 2021, 12, 11124–11135. [Google Scholar] [CrossRef]
  133. Chen, S.C.; Hu, T.H.; Huang, C.C.; Kung, M.L.; Chu, T.H.; Yi, L.N.; Huang, S.T.; Chan, H.H.; Chuang, J.H.; Liu, L.F.; et al. Hepatoma-derived growth factor/nucleolin axis as a novel oncogenic pathway in liver carcinogenesis. Oncotarget 2015, 6, 16253–16270. [Google Scholar] [CrossRef] [Green Version]
  134. Yazdian-Robati, R.; Bayat, P.; Oroojalian, F.; Zargari, M.; Ramezani, M.; Taghdisi, S.M.; Abnous, K. Therapeutic applications of AS1411 aptamer, an update review. Int. J. Biol. Macromol. 2020, 155, 1420–1431. [Google Scholar] [CrossRef]
  135. Zhang, X.; Gao, Q.; Zhuang, Q.; Zhang, L.; Wang, S.; Du, L.; Yuan, W.; Wang, C.; Tian, Q.; Yu, H.; et al. A dual-functional nanovehicle with fluorescent tracking and its targeted killing effects on hepatocellular carcinoma cells. RSC Adv. 2021, 11, 10986–10995. [Google Scholar] [CrossRef] [PubMed]
  136. Adachi, M.; Kai, K.; Yamaji, K.; Ide, T.; Noshiro, H.; Kawaguchi, A.; Aishima, S. Transferrin receptor 1 overexpression is associated with tumour de-differentiation and acts as a potential prognostic indicator of hepatocellular carcinoma. Histopathology 2019, 75, 63–73. [Google Scholar] [CrossRef] [PubMed]
  137. Jing, F.; Li, J.; Liu, D.; Wang, C.; Sui, Z. Dual ligands modified double targeted nano-system for liver targeted gene delivery. Pharm. Biol. 2013, 51, 643–649. [Google Scholar] [CrossRef] [Green Version]
  138. Meng, L.; Yang, L.; Zhao, X.; Zhang, L.; Zhu, H.; Liu, C.; Tan, W. Targeted delivery of chemotherapy agents using a liver cancer-specific aptamer. PLoS ONE 2012, 7, e33434. [Google Scholar] [CrossRef] [PubMed]
  139. Lo, A.; Lin, C.T.; Wu, H.C. Hepatocellular carcinoma cell-specific peptide ligand for targeted drug delivery. Mol. Cancer Ther. 2008, 7, 579–589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Ding, Z.; Wang, D.; Shi, W.; Yang, X.; Duan, S.; Mo, F.; Hou, X.; Liu, A.; Lu, X. In vivo Targeting of Liver Cancer with Tissue- and Nuclei-Specific Mesoporous Silica Nanoparticle-Based Nanocarriers in mice. Int. J. Nanomed. 2020, 15, 8383–8400. [Google Scholar] [CrossRef] [PubMed]
  141. Zhang, J.; Wang, X.; Cheng, L.; Yuan, J.; Zhong, Z. SP94 peptide mediating highly specific and efficacious delivery of polymersomal doxorubicin hydrochloride to hepatocellular carcinoma in vivo. Colloids Surf. B Biointerfaces 2021, 197, 111399. [Google Scholar] [CrossRef]
  142. De Bleser, P.J.; Jannes, P.; van Buul-Offers, S.C.; Hoogerbrugge, C.M.; van Schravendijk, C.F.; Niki, T.; Rogiers, V.; van den Brande, J.L.; Wisse, E.; Geerts, A. Insulinlike growth factor-II/mannose 6-phosphate receptor is expressed on CCl4-exposed rat fat-storing cells and facilitates activation of latent transforming growth factor-beta in cocultures with sinusoidal endothelial cells. Hepatology 1995, 21, 1429–1437. [Google Scholar] [CrossRef]
  143. Beljaars, L.; Molema, G.; Weert, B.; Bonnema, H.; Olinga, P.; Groothuis, G.M.; Meijer, D.K.; Poelstra, K. Albumin modified with mannose 6-phosphate: A potential carrier for selective delivery of antifibrotic drugs to rat and human hepatic stellate cells. Hepatology 1999, 29, 1486–1493. [Google Scholar] [CrossRef]
  144. Van Beuge, M.M.; Prakash, J.; Lacombe, M.; Gosens, R.; Post, E.; Reker-Smit, C.; Beljaars, L.; Poelstra, K. Reduction of fibrogenesis by selective delivery of a Rho kinase inhibitor to hepatic stellate cells in mice. J. Pharmacol. Exp. Ther. 2011, 337, 628–635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Van Beuge, M.M.; Prakash, J.; Lacombe, M.; Post, E.; Reker-Smit, C.; Beljaars, L.; Poelstra, K. Enhanced effectivity of an ALK5-inhibitor after cell-specific delivery to hepatic stellate cells in mice with liver injury. PLoS ONE 2013, 8, e56442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Kawaguchi, R.; Yu, J.; Honda, J.; Hu, J.; Whitelegge, J.; Ping, P.; Wiita, P.; Bok, D.; Sun, H. A membrane receptor for retinol binding protein mediates cellular uptake of vitamin A. Science 2007, 315, 820–825. [Google Scholar] [CrossRef]
  147. Senoo, H.; Yoshikawa, K.; Morii, M.; Miura, M.; Imai, K.; Mezaki, Y. Hepatic stellate cell (vitamin A-storing cell) and its relative--past, present and future. Cell Biol. Int. 2010, 34, 1247–1272. [Google Scholar] [CrossRef] [PubMed]
  148. Zhang, Z.; Wang, C.; Zha, Y.; Hu, W.; Gao, Z.; Zang, Y.; Chen, J.; Zhang, J.; Dong, L. Corona-directed nucleic acid delivery into hepatic stellate cells for liver fibrosis therapy. ACS Nano 2015, 9, 2405–2419. [Google Scholar] [CrossRef]
  149. Huang, J.; Huang, H.; Wang, Y.; Xu, B.; Lin, M.; Han, S.; Yuan, Y.; Wang, Y.; Shuai, X. Retinol-binding protein-hijacking nanopolyplex delivering siRNA to cytoplasm of hepatic stellate cell for liver fibrosis alleviation. Biomaterials 2023, 299, 122134. [Google Scholar] [CrossRef] [PubMed]
  150. Beljaars, L.; Molema, G.; Schuppan, D.; Geerts, A.; De Bleser, P.J.; Weert, B.; Meijer, D.K.; Poelstra, K. Successful targeting to rat hepatic stellate cells using albumin modified with cyclic peptides that recognize the collagen type VI receptor. J. Biol. Chem. 2000, 275, 12743–12751. [Google Scholar] [CrossRef] [Green Version]
  151. Hao, Y.; Song, K.; Tan, X.; Ren, L.; Guo, X.; Zhou, C.; Li, H.; Wen, J.; Meng, Y.; Lin, M.; et al. Reactive Oxygen Species-Responsive Polypeptide Drug Delivery System Targeted Activated Hepatic Stellate Cells to Ameliorate Liver Fibrosis. ACS Nano 2022, 16, 20739–20757. [Google Scholar] [CrossRef]
  152. Ashour, A.A.; El-Kamel, A.H.; Abdelmonsif, D.A.; Khalifa, H.M.; Ramadan, A.A. Modified Lipid Nanocapsules for Targeted Tanshinone IIA Delivery in Liver Fibrosis. Int. J. Nanomed. 2021, 16, 8013–8033. [Google Scholar] [CrossRef]
  153. Pinzani, M.; Milani, S.; Herbst, H.; DeFranco, R.; Grappone, C.; Gentilini, A.; Caligiuri, A.; Pellegrini, G.; Ngo, D.V.; Romanelli, R.G.; et al. Expression of platelet-derived growth factor and its receptors in normal human liver and during active hepatic fibrogenesis. Am. J. Pathol. 1996, 148, 785–800. [Google Scholar]
  154. Beljaars, L.; Weert, B.; Geerts, A.; Meijer, D.K.; Poelstra, K. The preferential homing of a platelet derived growth factor receptor-recognizing macromolecule to fibroblast-like cells in fibrotic tissue. Biochem. Pharmacol. 2003, 66, 1307–1317. [Google Scholar] [CrossRef] [PubMed]
  155. Bansal, R.; Prakash, J.; de Ruijter, M.; Beljaars, L.; Poelstra, K. Peptide-modified albumin carrier explored as a novel strategy for a cell-specific delivery of interferon gamma to treat liver fibrosis. Mol. Pharm. 2011, 8, 1899–1909. [Google Scholar] [CrossRef] [PubMed]
  156. Li, Q.; Ding, Y.; Guo, X.; Luo, S.; Zhuang, H.; Zhou, J.; Xu, N.; Yan, Z. Chemically modified liposomes carrying TRAIL target activated hepatic stellate cells and ameliorate hepatic fibrosis in vitro and in vivo. J. Cell. Mol. Med. 2019, 23, 1951–1962. [Google Scholar] [CrossRef]
  157. Bansal, R.; Poelstra, K. Hepatic Stellate Cell Targeting Using Peptide-Modified Biologicals. Methods Mol. Biol. 2023, 2669, 269–284. [Google Scholar] [CrossRef]
  158. Gantzel, R.H.; Kjær, M.B.; Laursen, T.L.; Kazankov, K.; George, J.; Møller, H.J.; Grønbæk, H. Macrophage Activation Markers, Soluble CD163 and Mannose Receptor, in Liver Fibrosis. Front. Med. 2020, 7, 615599. [Google Scholar] [CrossRef]
  159. Møller, L.N.; Knudsen, A.R.; Andersen, K.J.; Nyengaard, J.R.; Hamilton-Dutoit, S.; Okholm Møller, E.M.; Svendsen, P.; Møller, H.J.; Moestrup, S.K.; Graversen, J.H.; et al. Anti-CD163-dexamethasone protects against apoptosis after ischemia/reperfusion injuries in the rat liver. Ann. Med. Surg. 2015, 4, 331–337. [Google Scholar] [CrossRef] [Green Version]
  160. Svendsen, P.; Graversen, J.H.; Etzerodt, A.; Hager, H.; Røge, R.; Grønbæk, H.; Christensen, E.I.; Møller, H.J.; Vilstrup, H.; Moestrup, S.K. Antibody-Directed Glucocorticoid Targeting to CD163 in M2-type Macrophages Attenuates Fructose-Induced Liver Inflammatory Changes. Mol. Therapy. Methods Clin. Dev. 2017, 4, 50–61. [Google Scholar] [CrossRef] [Green Version]
  161. Uehara, K.; Harumoto, T.; Makino, A.; Koda, Y.; Iwano, J.; Suzuki, Y.; Tanigawa, M.; Iwai, H.; Asano, K.; Kurihara, K.; et al. Targeted delivery to macrophages and dendritic cells by chemically modified mannose ligand-conjugated siRNA. Nucleic Acids Res. 2022, 50, 4840–4859. [Google Scholar] [CrossRef] [PubMed]
  162. Wang, T.; Zhang, J.; Hou, T.; Yin, X.; Zhang, N. Selective targeting of tumor cells and tumor associated macrophages separately by twin-like core-shell nanoparticles for enhanced tumor-localized chemoimmunotherapy. Nanoscale 2019, 11, 13934–13946. [Google Scholar] [CrossRef]
  163. Liu, Y.; Liang, S.; Jiang, D.; Gao, T.; Fang, Y.; Fu, S.; Guan, L.; Zhang, Z.; Mu, W.; Chu, Q.; et al. Manipulation of TAMs functions to facilitate the immune therapy effects of immune checkpoint antibodies. J. Control. Release 2021, 336, 621–634. [Google Scholar] [CrossRef] [PubMed]
  164. Malovic, I.; Sørensen, K.K.; Elvevold, K.H.; Nedredal, G.I.; Paulsen, S.; Erofeev, A.V.; Smedsrød, B.H.; McCourt, P.A. The mannose receptor on murine liver sinusoidal endothelial cells is the main denatured collagen clearance receptor. Hepatology 2007, 45, 1454–1461. [Google Scholar] [CrossRef] [PubMed]
  165. Liu, Q.; Wang, X.; Liu, X.; Kumar, S.; Gochman, G.; Ji, Y.; Liao, Y.P.; Chang, C.H.; Situ, W.; Lu, J.; et al. Use of Polymeric Nanoparticle Platform Targeting the Liver To Induce Treg-Mediated Antigen-Specific Immune Tolerance in a Pulmonary Allergen Sensitization Model. ACS Nano 2019, 13, 4778–4794. [Google Scholar] [CrossRef]
  166. Xu, X.; Wang, X.; Liao, Y.P.; Luo, L.; Xia, T.; Nel, A.E. Use of a Liver-Targeting Immune-Tolerogenic mRNA Lipid Nanoparticle Platform to Treat Peanut-Induced Anaphylaxis by Single- and Multiple-Epitope Nucleotide Sequence Delivery. ACS Nano 2023, in press. [Google Scholar] [CrossRef] [PubMed]
  167. Li, R.; Oteiza, A.; Sørensen, K.K.; McCourt, P.; Olsen, R.; Smedsrød, B.; Svistounov, D. Role of liver sinusoidal endothelial cells and stabilins in elimination of oxidized low-density lipoproteins. Am. J. Physiol. Gastrointest. Liver Physiol. 2011, 300, G71–G81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Cabral, F.; Al-Rahem, M.; Skaggs, J.; Thomas, T.A.; Kumar, N.; Wu, Q.; Fadda, P.; Yu, L.; Robinson, J.M.; Kim, J.; et al. Stabilin receptors clear LPS and control systemic inflammation. iScience 2021, 24, 103337. [Google Scholar] [CrossRef]
  169. Pattipeiluhu, R.; Arias-Alpizar, G.; Basha, G.; Chan, K.Y.T.; Bussmann, J.; Sharp, T.H.; Moradi, M.A.; Sommerdijk, N.; Harris, E.N.; Cullis, P.R.; et al. Anionic Lipid Nanoparticles Preferentially Deliver mRNA to the Hepatic Reticuloendothelial System. Adv. Mater. 2022, 34, e2201095. [Google Scholar] [CrossRef]
  170. Paunovska, K.; Gil, C.J.; Lokugamage, M.P.; Sago, C.D.; Sato, M.; Lando, G.N.; Gamboa Castro, M.; Bryksin, A.V.; Dahlman, J.E. Analyzing 2000 in Vivo Drug Delivery Data Points Reveals Cholesterol Structure Impacts Nanoparticle Delivery. ACS Nano 2018, 12, 8341–8349. [Google Scholar] [CrossRef]
  171. Brindley, P.J.; Bachini, M.; Ilyas, S.I.; Khan, S.A.; Loukas, A.; Sirica, A.E.; Teh, B.T.; Wongkham, S.; Gores, G.J. Cholangiocarcinoma. Nat. Rev. Dis. Primers 2021, 7, 65. [Google Scholar] [CrossRef]
  172. Khemlina, G.; Ikeda, S.; Kurzrock, R. The biology of Hepatocellular carcinoma: Implications for genomic and immune therapies. Mol. Cancer 2017, 16, 149. [Google Scholar] [CrossRef]
  173. Tummala, K.S.; Brandt, M.; Teijeiro, A.; Graña, O.; Schwabe, R.F.; Perna, C.; Djouder, N. Hepatocellular Carcinomas Originate Predominantly from Hepatocytes and Benign Lesions from Hepatic Progenitor Cells. Cell Rep. 2017, 19, 584–600. [Google Scholar] [CrossRef] [Green Version]
  174. Huang, D.Q.; Mathurin, P.; Cortez-Pinto, H.; Loomba, R. Global epidemiology of alcohol-associated cirrhosis and HCC: Trends, projections and risk factors. Nat. Rev. Gastroenterol. Hepatol. 2023, 20, 37–49. [Google Scholar] [CrossRef] [PubMed]
  175. Ishiguro, S.; Inoue, M.; Kurahashi, N.; Iwasaki, M.; Sasazuki, S.; Tsugane, S. Risk factors of biliary tract cancer in a large-scale population-based cohort study in Japan (JPHC study); with special focus on cholelithiasis, body mass index, and their effect modification. Cancer Causes Control 2008, 19, 33–41. [Google Scholar] [CrossRef] [PubMed]
  176. Sano, T.; Shimada, K.; Sakamoto, Y.; Ojima, H.; Esaki, M.; Kosuge, T. Prognosis of perihilar cholangiocarcinoma: Hilar bile duct cancer versus intrahepatic cholangiocarcinoma involving the hepatic hilus. Ann. Surg. Oncol. 2008, 15, 590–599. [Google Scholar] [CrossRef]
  177. Yi, M.; Zheng, X.; Niu, M.; Zhu, S.; Ge, H.; Wu, K. Combination strategies with PD-1/PD-L1 blockade: Current advances and future directions. Mol. Cancer 2022, 21, 28. [Google Scholar] [CrossRef]
  178. Garcia, J.; Hurwitz, H.I.; Sandler, A.B.; Miles, D.; Coleman, R.L.; Deurloo, R.; Chinot, O.L. Bevacizumab (Avastin®) in cancer treatment: A review of 15 years of clinical experience and future outlook. Cancer Treat. Rev. 2020, 86, 102017. [Google Scholar] [CrossRef] [PubMed]
  179. Finn, R.S.; Qin, S.; Ikeda, M.; Galle, P.R.; Ducreux, M.; Kim, T.Y.; Kudo, M.; Breder, V.; Merle, P.; Kaseb, A.O.; et al. Atezolizumab plus Bevacizumab in Unresectable Hepatocellular Carcinoma. N. Engl. J. Med. 2020, 382, 1894–1905. [Google Scholar] [CrossRef]
  180. Oh, D.Y.; Lee, K.H.; Lee, D.W.; Yoon, J.; Kim, T.Y.; Bang, J.H.; Nam, A.R.; Oh, K.S.; Kim, J.M.; Lee, Y.; et al. Gemcitabine and cisplatin plus durvalumab with or without tremelimumab in chemotherapy-naive patients with advanced biliary tract cancer: An open-label, single-centre, phase 2 study. Lancet Gastroenterol. Hepatol. 2022, 7, 522–532. [Google Scholar] [CrossRef]
  181. Slesser, A.A.; Simillis, C.; Goldin, R.; Brown, G.; Mudan, S.; Tekkis, P.P. A meta-analysis comparing simultaneous versus delayed resections in patients with synchronous colorectal liver metastases. Surg. Oncol. 2013, 22, 36–47. [Google Scholar] [CrossRef]
  182. Brodt, P.; Fallavollita, L.; Bresalier, R.S.; Meterissian, S.; Norton, C.R.; Wolitzky, B.A. Liver endothelial E-selectin mediates carcinoma cell adhesion and promotes liver metastasis. Int. J. Cancer 1997, 71, 612–619. [Google Scholar] [CrossRef]
  183. Gout, S.; Tremblay, P.L.; Huot, J. Selectins and selectin ligands in extravasation of cancer cells and organ selectivity of metastasis. Clin. Exp. Metastasis 2008, 25, 335–344. [Google Scholar] [CrossRef]
  184. Yu, J.; Chen, G.G.; Lai, P.B.S. Targeting hepatocyte growth factor/c-mesenchymal-epithelial transition factor axis in hepatocellular carcinoma: Rationale and therapeutic strategies. Med. Res. Rev. 2021, 41, 507–524. [Google Scholar] [CrossRef]
  185. Yao, C.; Wu, S.; Kong, J.; Sun, Y.; Bai, Y.; Zhu, R.; Li, Z.; Sun, W.; Zheng, L. Angiogenesis in hepatocellular carcinoma: Mechanisms and anti-angiogenic therapies. Cancer Biol. Med. 2023, 20, 25–43. [Google Scholar] [CrossRef] [PubMed]
  186. Zhang, X.; Lu, M.; Xu, Y.; He, G.; Liu, Q.; Zhu, J.; Zhang, C.; Zhang, X. IL-10 promoter hypomethylation is associated with increased IL-10 expression and poor survival in hepatocellular carcinoma. Transl. Cancer Res. 2019, 8, 1466–1475. [Google Scholar] [CrossRef] [PubMed]
  187. Shen, Y.; Wei, Y.; Wang, Z.; Jing, Y.; He, H.; Yuan, J.; Li, R.; Zhao, Q.; Wei, L.; Yang, T.; et al. TGF-β regulates hepatocellular carcinoma progression by inducing Treg cell polarization. Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2015, 35, 1623–1632. [Google Scholar] [CrossRef] [PubMed]
  188. Huang, X.M.; Zhang, N.R.; Lin, X.T.; Zhu, C.Y.; Zou, Y.F.; Wu, X.J.; He, X.S.; He, X.W.; Wan, Y.L.; Lan, P. Antitumor immunity of low-dose cyclophosphamide: Changes in T cells and cytokines TGF-beta and IL-10 in mice with colon-cancer liver metastasis. Gastroenterol. Rep. 2020, 8, 56–65. [Google Scholar] [CrossRef]
  189. Lei, X.; Lei, Y.; Li, J.K.; Du, W.X.; Li, R.G.; Yang, J.; Li, J.; Li, F.; Tan, H.B. Immune cells within the tumor microenvironment: Biological functions and roles in cancer immunotherapy. Cancer Lett. 2020, 470, 126–133. [Google Scholar] [CrossRef]
  190. Donne, R.; Lujambio, A. The liver cancer immune microenvironment: Therapeutic implications for hepatocellular carcinoma. Hepatology 2022, 77, 1773–1796. [Google Scholar] [CrossRef]
  191. Biffi, G.; Tuveson, D.A. Diversity and Biology of Cancer-Associated Fibroblasts. Physiol. Rev. 2021, 101, 147–176. [Google Scholar] [CrossRef]
  192. Ezhilarasan, D. Hepatic stellate cells in the injured liver: Perspectives beyond hepatic fibrosis. J. Cell. Physiol. 2022, 237, 436–449. [Google Scholar] [CrossRef]
  193. Chandra Jena, B.; Sarkar, S.; Rout, L.; Mandal, M. The transformation of cancer-associated fibroblasts: Current perspectives on the role of TGF-β in CAF mediated tumor progression and therapeutic resistance. Cancer Lett. 2021, 520, 222–232. [Google Scholar] [CrossRef] [PubMed]
  194. Konstorum, A.; Lowengrub, J.S. Activation of the HGF/c-Met axis in the tumor microenvironment: A multispecies model. J. Theor. Biol. 2018, 439, 86–99. [Google Scholar] [CrossRef] [PubMed]
  195. Karakasheva, T.A.; Lin, E.W.; Tang, Q.; Qiao, E.; Waldron, T.J.; Soni, M.; Klein-Szanto, A.J.; Sahu, V.; Basu, D.; Ohashi, S.; et al. IL-6 Mediates Cross-Talk between Tumor Cells and Activated Fibroblasts in the Tumor Microenvironment. Cancer Res. 2018, 78, 4957–4970. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Kadomoto, S.; Izumi, K.; Mizokami, A. Roles of CCL2-CCR2 Axis in the Tumor Microenvironment. Int. J. Mol. Sci. 2021, 22, 8530. [Google Scholar] [CrossRef]
  197. Laviron, M.; Boissonnas, A. Ontogeny of Tumor-Associated Macrophages. Front. Immunol. 2019, 10, 1799. [Google Scholar] [CrossRef] [Green Version]
  198. Li, B.H.; Garstka, M.A.; Li, Z.F. Chemokines and their receptors promoting the recruitment of myeloid-derived suppressor cells into the tumor. Mol. Immunol. 2020, 117, 201–215. [Google Scholar] [CrossRef]
  199. Tengesdal, I.W.; Dinarello, A.; Powers, N.E.; Burchill, M.A.; Joosten, L.A.B.; Marchetti, C.; Dinarello, C.A. Tumor NLRP3-Derived IL-1β Drives the IL-6/STAT3 Axis Resulting in Sustained MDSC-Mediated Immunosuppression. Front. Immunol. 2021, 12, 661323. [Google Scholar] [CrossRef]
  200. Arvanitakis, K.; Mitroulis, I.; Germanidis, G. Tumor-Associated Neutrophils in Hepatocellular Carcinoma Pathogenesis, Prognosis, and Therapy. Cancers 2021, 13, 2899. [Google Scholar] [CrossRef]
  201. Wiedemann, G.M.; Röhrle, N.; Makeschin, M.C.; Fesseler, J.; Endres, S.; Mayr, D.; Anz, D. Peritumoural CCL1 and CCL22 expressing cells in hepatocellular carcinomas shape the tumour immune infiltrate. Pathology 2019, 51, 586–592. [Google Scholar] [CrossRef]
  202. Zhang, D.; Zhang, J.; Zeng, J.; Li, Z.; Zuo, H.; Huang, C.; Zhao, X. Nano-Gold Loaded with Resveratrol Enhance the Anti-Hepatoma Effect of Resveratrol In Vitro and In Vivo. J. Biomed. Nanotechnol. 2019, 15, 288–300. [Google Scholar] [CrossRef]
  203. Zhou, M.; Yi, Y.; Liu, L.; Lin, Y.; Li, J.; Ruan, J.; Zhong, Z. Polymeric micelles loading with ursolic acid enhancing anti-tumor effect on hepatocellular carcinoma. J. Cancer 2019, 10, 5820–5831. [Google Scholar] [CrossRef] [PubMed]
  204. Liu, L.; Dai, H.; Wu, Y.; Li, B.; Yi, J.; Xu, C.; Wu, X. In vitro and in vivo mechanism of hepatocellular carcinoma inhibition by β-TCP nanoparticles. Int. J. Nanomed. 2019, 14, 3491–3502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Vyas, D.; Patel, M.; Wairkar, S. Strategies for active tumor targeting-an update. Eur. J. Pharmacol. 2022, 915, 174512. [Google Scholar] [CrossRef]
  206. Cheng, T.M.; Chang, W.J.; Chu, H.Y.; De Luca, R.; Pedersen, J.Z.; Incerpi, S.; Li, Z.L.; Shih, Y.J.; Lin, H.Y.; Wang, K.; et al. Nano-Strategies Targeting the Integrin αvβ3 Network for Cancer Therapy. Cells 2021, 10, 1684. [Google Scholar] [CrossRef] [PubMed]
  207. Xu, J.; Xu, H.Y.; Zhang, Q.; Song, F.; Jiang, J.L.; Yang, X.M.; Mi, L.; Wen, N.; Tian, R.; Wang, L.; et al. HAb18G/CD147 functions in invasion and metastasis of hepatocellular carcinoma. Mol. Cancer Res. MCR 2007, 5, 605–614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Habault, J.; Poyet, J.L. Recent Advances in Cell Penetrating Peptide-Based Anticancer Therapies. Molecules 2019, 24, 927. [Google Scholar] [CrossRef] [Green Version]
  209. Zhang, Y.C.; Wu, C.G.; Li, A.M.; Liang, Y.; Ma, D.; Tang, X.L. Oxaliplatin and Gedatolisib (PKI-587) Co-Loaded Hollow Polydopamine Nano-Shells with Simultaneous Upstream and Downstream Action to Re-Sensitize Drugs-Resistant Hepatocellular Carcinoma to Chemotherapy. J. Biomed. Nanotechnol. 2021, 17, 18–36. [Google Scholar] [CrossRef]
  210. Wang, G.; Gao, X.; Gu, G.; Shao, Z.; Li, M.; Wang, P.; Yang, J.; Cai, X.; Li, Y. Polyethylene glycol-poly(ε-benzyloxycarbonyl-l-lysine)-conjugated VEGF siRNA for antiangiogenic gene therapy in hepatocellular carcinoma. Int. J. Nanomed. 2017, 12, 3591–3603. [Google Scholar] [CrossRef] [Green Version]
  211. Mo, Y.; He, L.; Lai, Z.; Wan, Z.; Chen, Q.; Pan, S.; Li, L.; Li, D.; Huang, J.; Xue, F.; et al. Gold nano-particles (AuNPs) carrying miR-326 targets PDK1/AKT/c-myc axis in hepatocellular carcinoma. Artif. Cells Nanomed. Biotechnol. 2019, 47, 2830–2837. [Google Scholar] [CrossRef]
  212. Balança, C.C.; Salvioni, A.; Scarlata, C.M.; Michelas, M.; Martinez-Gomez, C.; Gomez-Roca, C.; Sarradin, V.; Tosolini, M.; Valle, C.; Pont, F.; et al. PD-1 blockade restores helper activity of tumor-infiltrating, exhausted PD-1hiCD39+ CD4 T cells. JCI Insight 2021, 6, e142513. [Google Scholar] [CrossRef]
  213. Anwanwan, D.; Singh, S.K.; Singh, S.; Saikam, V.; Singh, R. Challenges in liver cancer and possible treatment approaches. Biochim. Biophys. Acta. Rev. Cancer 2020, 1873, 188314. [Google Scholar] [CrossRef]
  214. Nisar, S.; Hashem, S.; Macha, M.A.; Yadav, S.K.; Muralitharan, S.; Therachiyil, L.; Sageena, G.; Al-Naemi, H.; Haris, M.; Bhat, A.A. Exploring Dysregulated Signaling Pathways in Cancer. Curr. Pharm. Des. 2020, 26, 429–445. [Google Scholar] [CrossRef] [PubMed]
  215. Sun, Y.; Zhai, W.; Liu, X.; Song, X.; Gao, X.; Xu, K.; Tang, B. Homotypic cell membrane-cloaked biomimetic nanocarrier for the accurate photothermal-chemotherapy treatment of recurrent hepatocellular carcinoma. J. Nanobiotechnol. 2020, 18, 60. [Google Scholar] [CrossRef] [PubMed]
  216. Wang, H.; Wang, B.; Wang, S.; Chen, J.; Zhi, W.; Guan, Y.; Cai, B.; Zhu, Y.; Jia, Y.; Huang, S.; et al. Injectable in situ intelligent thermo-responsive hydrogel with glycyrrhetinic acid-conjugated nano graphene oxide for chemo-photothermal therapy of malignant hepatocellular tumor. J. Biomater. Appl. 2022, 37, 151–165. [Google Scholar] [CrossRef] [PubMed]
  217. He, Z.; Zhang, S. Tumor-Associated Macrophages and Their Functional Transformation in the Hypoxic Tumor Microenvironment. Front. Immunol. 2021, 12, 741305. [Google Scholar] [CrossRef]
  218. Li, Y.; Patel, S.P.; Roszik, J.; Qin, Y. Hypoxia-Driven Immunosuppressive Metabolites in the Tumor Microenvironment: New Approaches for Combinational Immunotherapy. Front. Immunol. 2018, 9, 1591. [Google Scholar] [CrossRef] [Green Version]
  219. Petrillo, M.; Patella, F.; Pesapane, F.; Suter, M.B.; Ierardi, A.M.; Angileri, S.A.; Floridi, C.; de Filippo, M.; Carrafiello, G. Hypoxia and tumor angiogenesis in the era of hepatocellular carcinoma transarterial loco-regional treatments. Future Oncol. 2018, 14, 2957–2967. [Google Scholar] [CrossRef]
  220. Chang, C.C.; Dinh, T.K.; Lee, Y.A.; Wang, F.N.; Sung, Y.C.; Yu, P.L.; Chiu, S.C.; Shih, Y.C.; Wu, C.Y.; Huang, Y.D.; et al. Nanoparticle Delivery of MnO2 and Antiangiogenic Therapy to Overcome Hypoxia-Driven Tumor Escape and Suppress Hepatocellular Carcinoma. ACS Appl. Mater. Interfaces 2020, 12, 44407–44419. [Google Scholar] [CrossRef]
  221. Wang, J.; Zheng, C.; Zhai, Y.; Cai, Y.; Lee, R.J.; Xing, J.; Wang, H.; Zhu, H.H.; Teng, L.; Li, Y.; et al. High-density lipoprotein modulates tumor-associated macrophage for chemoimmunotherapy of hepatocellular carcinoma. Nano Today 2021, 37, 101064. [Google Scholar] [CrossRef]
  222. Daei Farshchi Adli, A.; Jahanban-Esfahlan, R.; Seidi, K.; Samandari-Rad, S.; Zarghami, N. An overview on Vadimezan (DMXAA): The vascular disrupting agent. Chem. Biol. Drug Des. 2018, 91, 996–1006. [Google Scholar] [CrossRef]
  223. Abdel-Rahman, O.; Elsayed, Z.; Elhalawani, H. Gemcitabine-based chemotherapy for advanced biliary tract carcinomas. Cochrane Database Syst. Rev. 2018, 4, CD011746. [Google Scholar] [CrossRef] [PubMed]
  224. Guo, H.; Song, Y.; Li, F.; Fan, Y.; Li, Y.; Zhang, C.; Hou, H.; Shi, M.; Zhao, Z.; Chen, Z. ACT001 suppressing M1 polarization against inflammation via NF-κB and STAT1 signaling pathways alleviates acute lung injury in mice. Int. Immunopharmacol. 2022, 110, 108944. [Google Scholar] [CrossRef] [PubMed]
  225. Boutilier, A.J.; Elsawa, S.F. Macrophage Polarization States in the Tumor Microenvironment. Int. J. Mol. Sci. 2021, 22, 6995. [Google Scholar] [CrossRef]
  226. Li, T.; Liu, Z.; Fu, X.; Chen, Y.; Zhu, S.; Zhang, J. Co-delivery of Interleukin-12 and doxorubicin loaded Nano-delivery system for enhanced immunotherapy with polarization toward M1-type Macrophages. Eur. J. Pharm. Biopharm. 2022, 177, 175–183. [Google Scholar] [CrossRef] [PubMed]
  227. Park, D.J.; Sung, P.S.; Lee, G.W.; Cho, S.; Kim, S.M.; Kang, B.Y.; Hur, W.; Yang, H.; Lee, S.K.; Lee, S.H.; et al. Preferential Expression of Programmed Death Ligand 1 Protein in Tumor-Associated Macrophages and Its Potential Role in Immunotherapy for Hepatocellular Carcinoma. Int. J. Mol. Sci. 2021, 22, 4710. [Google Scholar] [CrossRef] [PubMed]
  228. Chamseddine, A.N.; Assi, T.; Mir, O.; Chouaib, S. Modulating tumor-associated macrophages to enhance the efficacy of immune checkpoint inhibitors: A TAM-pting approach. Pharmacol. Ther. 2022, 231, 107986. [Google Scholar] [CrossRef]
  229. Kuske, M.; Haist, M.; Jung, T.; Grabbe, S.; Bros, M. Immunomodulatory Properties of Immune Checkpoint Inhibitors-More than Boosting T-Cell Responses? Cancers 2022, 14, 1710. [Google Scholar] [CrossRef]
  230. Liu, X.; Zhou, J.; Wu, H.; Chen, S.; Zhang, L.; Tang, W.; Duan, L.; Wang, Y.; McCabe, E.; Hu, M.; et al. Fibrotic immune microenvironment remodeling mediates superior anti-tumor efficacy of a nano-PD-L1 trap in hepatocellular carcinoma. Mol. Ther. J. Am. Soc. Gene Ther. 2023, 31, 119–133. [Google Scholar] [CrossRef]
  231. Liu, Q.; Huang, W.; Liang, W.; Ye, Q. Current Strategies for Modulating Tumor-Associated Macrophages with Biomaterials in Hepatocellular Carcinoma. Molecules 2023, 28, 2211. [Google Scholar] [CrossRef]
  232. Marrone, G.; Maeso-Díaz, R.; García-Cardena, G.; Abraldes, J.G.; García-Pagán, J.C.; Bosch, J.; Gracia-Sancho, J. KLF2 exerts antifibrotic and vasoprotective effects in cirrhotic rat livers: Behind the molecular mechanisms of statins. Gut 2015, 64, 1434–1443. [Google Scholar] [CrossRef]
  233. Xie, G.; Wang, X.; Wang, L.; Wang, L.; Atkinson, R.D.; Kanel, G.C.; Gaarde, W.A.; Deleve, L.D. Role of differentiation of liver sinusoidal endothelial cells in progression and regression of hepatic fibrosis in rats. Gastroenterology 2012, 142, 918–927.e916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  234. Dasargyri, A.; Kümin, C.D.; Leroux, J.C. Targeting Nanocarriers with Anisamide: Fact or Artifact? Adv. Mater. 2017, 29, 1603451. [Google Scholar] [CrossRef]
  235. Hu, M.; Wang, Y.; Xu, L.; An, S.; Tang, Y.; Zhou, X.; Li, J.; Liu, R.; Huang, L. Relaxin gene delivery mitigates liver metastasis and synergizes with check point therapy. Nat. Commun. 2019, 10, 2993. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  236. Kaps, L.; Schuppan, D. Targeting Cancer Associated Fibroblasts in Liver Fibrosis and Liver Cancer Using Nanocarriers. Cells 2020, 9, 2027. [Google Scholar] [CrossRef] [PubMed]
  237. Thapa, P.; Farber, D.L. The Role of the Thymus in the Immune Response. Thorac. Surg. Clin. 2019, 29, 123–131. [Google Scholar] [CrossRef]
  238. Waisman, A.; Lukas, D.; Clausen, B.E.; Yogev, N. Dendritic cells as gatekeepers of tolerance. Semin. Immunopathol. 2017, 39, 153–163. [Google Scholar] [CrossRef]
  239. Hocking, A.M.; Buckner, J.H. Genetic basis of defects in immune tolerance underlying the development of autoimmunity. Front. Immunol. 2022, 13, 972121. [Google Scholar] [CrossRef]
  240. Liu, E.G.; Yin, X.; Swaminathan, A.; Eisenbarth, S.C. Antigen-Presenting Cells in Food Tolerance and Allergy. Front. Immunol. 2020, 11, 616020. [Google Scholar] [CrossRef]
  241. Drazdauskaitė, G.; Layhadi, J.A.; Shamji, M.H. Mechanisms of Allergen Immunotherapy in Allergic Rhinitis. Curr. Allergy Asthma Rep. 2020, 21, 2. [Google Scholar] [CrossRef]
  242. Richardson, N.; Wraith, D.C. Advancement of antigen-specific immunotherapy: Knowledge transfer between allergy and autoimmunity. Immunother. Adv. 2021, 1, ltab009. [Google Scholar] [CrossRef]
  243. Han, P.; Hanlon, D.; Sobolev, O.; Chaudhury, R.; Edelson, R.L. Ex vivo dendritic cell generation-A critical comparison of current approaches. Int. Rev. Cell Mol. Biol. 2019, 349, 251–307. [Google Scholar] [CrossRef] [PubMed]
  244. Mohammadi, B.; Saghafi, M.; Abdulsattar Faraj, T.; Kamal Kheder, R.; Sajid Abdulabbas, H.; Esmaeili, S.A. The role of tolerogenic dendritic cells in systematic lupus erythematosus progression and remission. Int. Immunopharmacol. 2023, 115, 109601. [Google Scholar] [CrossRef] [PubMed]
  245. Li, H.; Yang, Y.G.; Sun, T. Nanoparticle-Based Drug Delivery Systems for Induction of Tolerance and Treatment of Autoimmune Diseases. Front. Bioeng. Biotechnol. 2022, 10, 889291. [Google Scholar] [CrossRef]
  246. Tiegs, G.; Lohse, A.W. Immune tolerance: What is unique about the liver. J. Autoimmun. 2010, 34, 1–6. [Google Scholar] [CrossRef] [PubMed]
  247. Carambia, A.; Freund, B.; Schwinge, D.; Heine, M.; Laschtowitz, A.; Huber, S.; Wraith, D.C.; Korn, T.; Schramm, C.; Lohse, A.W.; et al. TGF-β-dependent induction of CD4+CD25+Foxp3+ Tregs by liver sinusoidal endothelial cells. J. Hepatol. 2014, 61, 594–599. [Google Scholar] [CrossRef]
  248. Carambia, A.; Frenzel, C.; Bruns, O.T.; Schwinge, D.; Reimer, R.; Hohenberg, H.; Huber, S.; Tiegs, G.; Schramm, C.; Lohse, A.W.; et al. Inhibition of inflammatory CD4 T cell activity by murine liver sinusoidal endothelial cells. J. Hepatol. 2013, 58, 112–118. [Google Scholar] [CrossRef]
  249. Shtykova, E.V.; Huang, X.; Gao, X.; Dyke, J.C.; Schmucker, A.L.; Dragnea, B.; Remmes, N.; Baxter, D.V.; Stein, B.; Konarev, P.V.; et al. Hydrophilic Monodisperse Magnetic Nanoparticles Protected by an Amphiphilic Alternating Copolymer. J. Phys. Chem. C Nanomater. Interfaces 2008, 112, 16809–16817. [Google Scholar] [CrossRef] [Green Version]
  250. Carambia, A.; Freund, B.; Schwinge, D.; Bruns, O.T.; Salmen, S.C.; Ittrich, H.; Reimer, R.; Heine, M.; Huber, S.; Waurisch, C.; et al. Nanoparticle-based autoantigen delivery to Treg-inducing liver sinusoidal endothelial cells enables control of autoimmunity in mice. J. Hepatol. 2015, 62, 1349–1356. [Google Scholar] [CrossRef]
  251. Elvevold, K.; Simon-Santamaria, J.; Hasvold, H.; McCourt, P.; Smedsrød, B.; Sørensen, K.K. Liver sinusoidal endothelial cells depend on mannose receptor-mediated recruitment of lysosomal enzymes for normal degradation capacity. Hepatology 2008, 48, 2007–2015. [Google Scholar] [CrossRef]
  252. Maldonado, R.A.; LaMothe, R.A.; Ferrari, J.D.; Zhang, A.H.; Rossi, R.J.; Kolte, P.N.; Griset, A.P.; O’Neil, C.; Altreuter, D.H.; Browning, E.; et al. Polymeric synthetic nanoparticles for the induction of antigen-specific immunological tolerance. Proc. Natl. Acad. Sci. USA 2015, 112, E156–E165. [Google Scholar] [CrossRef]
  253. Kishimoto, T.K.; Ferrari, J.D.; LaMothe, R.A.; Kolte, P.N.; Griset, A.P.; O’Neil, C.; Chan, V.; Browning, E.; Chalishazar, A.; Kuhlman, W.; et al. Improving the efficacy and safety of biologic drugs with tolerogenic nanoparticles. Nat. Nanotechnol. 2016, 11, 890–899. [Google Scholar] [CrossRef] [PubMed]
  254. Ilyinskii, P.O.; Roy, C.J.; LePrevost, J.; Rizzo, G.L.; Kishimoto, T.K. Enhancement of the Tolerogenic Phenotype in the Liver by ImmTOR Nanoparticles. Front. Immunol. 2021, 12, 637469. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kaps, L.; Limeres, M.J.; Schneider, P.; Svensson, M.; Zeyn, Y.; Fraude, S.; Cacicedo, M.L.; Galle, P.R.; Gehring, S.; Bros, M. Liver Cell Type-Specific Targeting by Nanoformulations for Therapeutic Applications. Int. J. Mol. Sci. 2023, 24, 11869. https://doi.org/10.3390/ijms241411869

AMA Style

Kaps L, Limeres MJ, Schneider P, Svensson M, Zeyn Y, Fraude S, Cacicedo ML, Galle PR, Gehring S, Bros M. Liver Cell Type-Specific Targeting by Nanoformulations for Therapeutic Applications. International Journal of Molecular Sciences. 2023; 24(14):11869. https://doi.org/10.3390/ijms241411869

Chicago/Turabian Style

Kaps, Leonard, María José Limeres, Paul Schneider, Malin Svensson, Yanira Zeyn, Silvia Fraude, Maximiliano L. Cacicedo, Peter R. Galle, Stephan Gehring, and Matthias Bros. 2023. "Liver Cell Type-Specific Targeting by Nanoformulations for Therapeutic Applications" International Journal of Molecular Sciences 24, no. 14: 11869. https://doi.org/10.3390/ijms241411869

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop