Keywords Claudins - monoclonal antibodies - therapeutic target - signaling - cell plasticity
Liver fibrosis and cancer are serious diseases with limited treatment options and
poor outcome. Recent research has revealed that proteins relevant in cell–cell contact
are also expressed on the surface of liver and cancer cells and can contribute to
the disease. For example, Claudin-1 and Occludin mediate infection of hepatocytes
by hepatitis C virus—a major cause of liver disease and cancer worldwide. Claudin-1
has been shown to play a function role in liver fibrosis and cancer development. Claudin-1
has been shown to be a target to treat liver fibrosis and cancer using monoclonal
antibodies. In liver cancers, Claudin-6 overexpression is associated with aggressive
behavior and treatment resistance. Clinical investigation is currently underway for
CLDN-6 overexpressing tumors using Claudin-6 targeting therapies. In conclusion, targeting
exposed Claudins offers a novel clinical opportunity for the treatment of liver fibrosis
and cancer.
Liver fibrosis and cancer are major global health issues with increasing incidence
due to prevalent risk factors including alcohol consumption, metabolic syndrome, and
steatotic liver disease (SLD).[1 ] The most prevalent chronic liver disease, metabolic dysfunction-associated steatotic
liver disease (MASLD), affects roughly 30% of the global population, and has shown
an increase of 50.4% between 1990 to 2006 and 2016 to 2019.[2 ] MASLD and its inflammatory and aggressive form, metabolic dysfunction-associated
steatohepatitis (MASH), significantly contribute to liver fibrosis, which is the most
significant risk factor for liver cirrhosis and decompensation. Research analyzing
the natural progression of MASH patients indicates that approximately 20% of patients
with F3 fibrosis develop cirrhosis or liver-related complications within 2 years.[3 ] Developing treatments for liver fibrosis has proven challenging. Clinical studies
of compounds in late-stage development, such as glucagon-like peptide-1 (GLP-1) and
thyroid hormone receptor-β agonists, have shown effects on metabolic and inflammatory
endpoints, with limited reduction in liver fibrosis, particularly in patients with
advanced fibrosis.[4 ]
[5 ]
[6 ]
Liver fibrosis is the key risk factor for liver cancer, which represent the third
leading cause of cancer-related deaths in the United States. The available treatment
options for hepatocellular carcinoma (HCC), which accounts for more than 80% of primary
liver cancers, are inadequate. Curative treatment is available for only a small number
of patients, and tumor recurrence often occurs after treatment. Moreover, no adjuvant
treatment has been authorized yet. Palliative systemic therapy can only be recommended
to advanced-stage patients with preserved liver function.[7 ] Therefore, innovative therapies targeting liver fibrosis and cancer remain an unmet
medical need.
Tight junction (TJ) proteins have emerged as targets for chronic diseases and cancers,
such as gastric cancer.[8 ] TJ proteins not only form intercellular junctions but are also exposed at the cell
surface outside the TJs in a nonjunctional form ([Fig. 1 ]). They have shown to play an important role in various liver diseases comprising
hepatitis C, liver fibrosis, and HCC and a large body of data show that nonjunctional
TJ proteins are therapeutic targets for these conditions.
Fig. 1 Tight junction (TJ) structure and biology. TJs are formed by transmembrane proteins
belonging to the claudin family (CLDNs), TJ-associated marvel proteins (TAMPs), blood
vessel epicardial substance (BVES) and junctional adhesion molecules (JAMs). The transmembrane
proteins are connected to the actin filaments and microtubules by the junctional plaque,
which contains several adaptor proteins, protein phosphates, kinases, GTP-binding
proteins, transcriptional, and posttranscriptional regulators such as ZO-1 associated
nucleic acid binding proteins (ZONAB). TJ transmembrane proteins are also localized
outside the junctions at the basolateral membrane. Nonjunctionally expressed proteins
not only have a major intracellular signaling role but are also used by pathogens
to enter in the cells. Nonjunctional proteins are accessible to drugs and are the
main therapeutic targets of TJ-targeting agents.
In this article, we review recent advances and perspectives on TJ proteins focusing
on Claudins as therapeutic targets for liver disease and HCC.
Functional Role of Tight Junction Proteins Expressed in and Outside the Junctions
in the Biology of the Liver
Functional Role of Tight Junction Proteins Expressed in and Outside the Junctions
in the Biology of the Liver
TJs are intercellular junctions responsible for regulating paracellular transport
and maintaining cell polarity. They are formed by transmembrane and cytosolic proteins.[9 ] Notably, transmembrane proteins forming TJs are also expressed outside of the junction
where they participate in signal transduction, intercellular communication,[10 ]
[11 ]
[12 ] and can influence cellular processes such as cell proliferation, migration, and
differentiation.[13 ]
[14 ] Nonjunctional TJ proteins comprise several proteins, including Claudins (CLDNs),
TJ-associated marvel proteins like occludin (OCLN), tricellulin, Marvel D3, junctional
adhesion molecules (JAMs), and angulins. Junctional and nonjunctional protein pools
are interconnected. Studies on the dynamic behavior of TJ have shown that TJ proteins
are highly mobile, constantly remodeling and exchanging between the junctional and
nonjunctional membrane and intracellular pools.[15 ] Junctional and nonjunctional TJs proteins have been shown to transmit signals through
various pathways to regulate cell differentiation and proliferation. These pathways
include the Hippo-pathway transcriptional coactivators YES-associated protein 1 (YAP1)
and transcriptional coactivator with PDZ-binding motif (TAZ),[16 ]
[17 ]
[18 ] AKT–mTOR[9 ]
[19 ] and JUN N-terminal kinase (JNK) mitogen-activated protein kinase (MAPK), and extracellular
signal–regulated kinase (ERK) signaling pathways.[20 ]
[21 ]
[22 ] It has been suggested that these pathways play important roles in controlling cellular
functions. Moreover, both junctional and nonjunctional TJ proteins engage in crosstalk
with focal adhesion, such as between integrins and claudins.[23 ]
[24 ]
[25 ]
[26 ] These interactions result in a complex interaction system with neighboring cells
and extracellular matrix, which further modifies cell proliferation and migration.[9 ]
In the liver, TJ proteins are predominantly expressed in epithelial cells, specifically
hepatocytes and cholangiocytes. CLDNs are the most prevalent and abundant TJ proteins.
In hepatocytes, junctional TJ proteins establish the blood–biliary barrier, which
separates the apical biliary pole from the basolateral membrane near the sinusoidal
space. In cholangiocytes, junctional TJ proteins secure the bile ducts and canals
and facilitate bile production and modification of composition.[27 ] In both types of cells, nonjunctional proteins are expressed at basolateral membrane
and play a significant role in signal transduction and cell–matrix interactions. Recent
studies have also shown that TJ proteins are also expressed in liver mesenchymal cells
in their nonjunctional form and are upregulated during inflammatory processes.[28 ]
[29 ] Exposed, nonjunctional, TJ proteins are accessible to pathogens (such as hepatitis
C virus) or drugs (such as antibodies) compared with their junctional counterparts.
Indeed, TJ proteins exposed outside the TJs at the cell surface play a significant
role in the pathogenesis of liver diseases, including viral infection, liver fibrosis,
and cancer.
HCV infection biology exemplifies the role of nonjunctional TJ proteins in liver disease
and their potential as therapeutic targets. Numerous studies have demonstrated that
nonjunctionally expressed TJ proteins, such as CLDN1 and OCLN are crucial in HCV cell
entry.[13 ]
[30 ] Together with CD81 and scavenger receptor BI, they are four of the primary host
entry factors for HCV necessary for hepatocyte infection.[13 ]
[31 ] Several studies have elegantly demonstrated that both nonjunctional forms of CLDN1
and OCLN mediate HCV entry.[32 ]
[33 ]
[34 ]
[35 ]
[36 ] It is important to note that HCV does not directly bind to CLDN1.[37 ] Instead, CLDN1 serves as a coreceptor implicated in viral entry steps following
viral binding of CD81.[32 ]
[38 ]
[39 ] Indeed, after HCV–CD81 binding, CLDN1 interacts with CD81 to facilitate viral internalization.[32 ]
[38 ]
[39 ] OCLN, as CLDN1, appears to not bind HCV and acts a cofactor necessary for late postbinding
events involved in HCV entry.[40 ]
[41 ]
[42 ] Additionally, OCLN and CLDN1 contribute to cell-to-cell transmission of HCV.[13 ]
[43 ]
[44 ]
Research in HCV biology has opened new insights into the role of nonjunction TJ proteins
in liver disease and their potential as therapeutic targets. CLDN1 was among the first
TJ proteins uncovered as an essential host factor allowing the virus to enter and
infect hepatocytes.[13 ] Monoclonal antibodies (mAbs) that recognize the conformation-dependent epitope within
ECL1 prevent and disrupt CD81–CLDN1 association at the basolateral membrane of cells,
thereby inhibiting viral internalization.[45 ]
[46 ] This mechanism confers to the CLDN1 mAbs a pan-genotypic effect enabling them to
prevent HCV infection and cure chronic hepatitis C.[44 ]
[45 ] In chimeric mice engrafted with human hepatocytes, CLDN1-specific mAbs were able
to prevent acute infection as well as cure of established chronic viral infection.[35 ] Moreover, these antibodies have an additional antiviral effect by modulating CLDN1
intracellular signaling, interfering with the MAPK pathway involved in maintaining
HCV infection.[13 ]
[35 ] Other mAbs targeting the extracellular loop-2 (ECL2) of CLDN1 have demonstrated
the ability to prevent HCV infection in chimeric mice with human liver.[13 ] Furthermore, CLDN1-derived peptides or recombinant proteins can compete with the
endogenous CLDN1 and prevent viral entry.[30 ]
Similarly, mAbs against ECL1 and 2 of OCLN have also been developed.[47 ]
[48 ] Interestingly, a mAb targeting ECL2 of OCLN prevented HCV infection in human hepatoma
Huh7.5.1 cells only when applied to the basolateral membrane instead of the apical
one, confirming the nonjunctional protein's entry role at the basolateral membrane.[48 ] In addition, the ECL2-directed mAb demonstrated more effective antiviral activity,
indicating that ECL2 is the primary site involved in HCV late entry steps.[13 ] Some of the OCLN antibodies have also been evaluated for safety and efficacy in
human liver chimeric mice with no signs of major toxicity.[47 ]
The mechanistic studies on nonjunctional CLDN1, OCLN, and HCV have provided crucial
insights into the biology of TJ proteins, their role in liver disease, and as therapeutic
targets. Studying the biology of HCV and targeting the intracellular signaling pathways
of TJ proteins has facilitated the development of precise mAbs for the treatment of
liver fibrosis and cancer, which are currently in clinical trials.[28 ]
[49 ]
Nonjunctional Tight Junction Proteins as Therapeutic Target for Liver Fibrosis
Nonjunctional Tight Junction Proteins as Therapeutic Target for Liver Fibrosis
Liver fibrosis is a common endpoint of chronic liver diseases, marked by excessive
extracellular matrix deposition, tissue remodeling, and liver regeneration. Advanced
liver fibrosis stages can result in end-stage liver disease and is associated with
increased risk of liver decompensation and HCC. The severity of liver fibrosis is
the most important prognostic factor in patients with MASLD[50 ] and is linked to the occurrence of liver-related events. Moreover, impaired liver
function in patients with HCC is a major barrier to cancer treatment. Treatment for
liver fibrosis is a major unmet medical need, but the development of such treatments
has been challenging, with no drug being approved so far. Inflammatory and metabolic
drugs for SLD have shown no major efficacy on liver fibrosis, a condition where collagen
remodeling and liver regeneration are impaired.
TJ proteins exposed at the basolateral membrane outside the TJs have been shown to
have a role in liver fibrosis pathogenesis and to influence mesenchymal cell activation
and liver cell regeneration. Among all the TJ proteins, CLDN1 has been the most extensively
studied. Multiple pieces of evidence support the role of nonjunctional CLDN1 as a
major contributing factor for the pathogenesis of liver fibrosis. Studies of CLDN1 expression have shown overexpression in cirrhotic livers.[51 ]
[52 ] CLDN1 levels, and in particular the nonjunctional form, increase alongside the fibrosis
stage.[28 ] Single-cell RNA-sequencing analysis revealed that CLDN1 is expressed in liver epithelial cells (hepatocytes and cholangiocytes), bipotent
progenitor cells, and stellate cells. Notably, individual cell expression appears
to be the highest in bipotent progenitor cells. TNFα, a cytokine implicated in liver
inflammation and regeneration, contributes to stellate cells activation into myofibroblast,
major players in collagen production. Additionally, it enhances CLDN1 expression levels in hepatocytes as well as myofibroblasts. Furthermore, in vivo
knockdown of CLDN1 considerably reduced liver fibrosis and tumor burden in a chimeric
NASH mouse model, providing a genetic validation for targeting CLDN1 for liver fibrosis
and HCC prevention.[28 ]
An mAb that targets nonjunctional CLDN1 has demonstrated a potent antifibrotic and
HCC prevention effect across several mouse and patient-derived models of liver fibrosis
progressing to HCC.[28 ] Mechanistic studies have shown that targeting CLDN1 suppresses profibrotic signal
transduction including a robust effect on the TNF-α–NFκB signaling pathway. CLDN1
interacts directly with other proteins, including epidermal growth factor receptor
(EGFR), EPCAM, the ECM receptor integrin α 5 (ITGA5), and the ECM component laminin
5 (LAMA5). By modulating this interaction, CLDN1 mAb inhibits EGFR and ERK phosphorylation
and suppresses SRC proto-oncogene and SRC signaling, a key downstream pathway of cell-ECM
mechanoreceptors.[53 ] These changes result in modulation of the plasticity of hepatocytes, progenitor
cells, and myofibroblasts, leading to a reversal of their pathological, immature,
and profibrotic profile to more differentiated and functionals, thereby reversing
fibrosis and reducing HCC development.[28 ]
[54 ]
[55 ]
[56 ] The function and intracellular signaling of CLDN1, along with the effects of the
nonjunctional CLDN1-specific mAb are presented in [Fig. 2 ]. In addition, the role of CLDN1 in fibrosis seems not to be limited to the liver.
CLDN1 has been found to be upregulated in both lung and kidney patients and models.[57 ]
[58 ]
[59 ] Moreover, treatment with CLDN1 mAbs efficiently reduced lung and kidney fibrosis
in several state-of-the-art mouse and patient models.[28 ]
[60 ]
Fig. 2 Nonjunctional CLDN1 as therapeutic target for liver fibrosis and cancer. Nonjunctional
CLDN1 (njCLDN1) interacts with other transmembrane proteins such as ITGA5 (Integrin
Subunit Alpha 5), EPCAM (epithelial cell adhesion molecule), EGFR (epidermal growth
factor receptor), and JAG1/2 (Jagged canonical Notch ligand) as described previously.[28 ] This interaction activates TNFα–NFκB signaling, MAPK/ERK, SRC, and Notch signaling
pathways, which control cell activation, differentiation, and plasticity as well as
proliferation. A CLDN1-specific mAb targeting exposed CLDN1 on hepatocytes or liver
cancer cells has demonstrated to robustly inhibit liver fibrosis and prevent and treat
HCC.[28 ]
[84 ] HCC, hepatocellular carcinoma; mAb, monoclonal antibody.
The clinical development of an mAb targeting nonjunctional CLDN1 in liver and other
organ fibrosis is currently ongoing. A fully humanized anti-human CLDN1 mAb has undergone
safely testing in cynomolgus monkeys[28 ] and healthy volunteers. Moreover, a phase Ib clinical trial is currently ongoing
for patients with advanced liver fibrosis or compensated cirrhosis (FEGATO-01, NCT05939947).
A phase II clinical trial is under way for patients with antineutrophil cytoplasmic
antibody-associated vasculitis with rapidly progressive glomerulonephritis, which
induces kidney fibrosis (RENAL-F02, NCT06047171).
Other TJ proteins have also been described in liver fibrosis and regeneration, although
their role as therapeutic target has been poorly investigated with no differentiation
between junctional and nonjunctional proteins. JAMs affect leukocyte recruitment and
migration, vascular permeability, and remodeling. In healthy livers, epithelial cells,
endothelial cells, and hepatic stellate cells express JAM-A, whereas JAM-B and -C
are restricted to endothelial cells. Complete and selective knockout of Jam-a in bone marrow-derived cells and endothelial cells exacerbates liver fibrosis in
CCl4 -induced model regulating nonsinusoidal vascular immune cell recruitment, liver sinusoid
capillarization, and hematopoietic stem cell quiescence.[61 ] In a mouse model of CCL4-induced fibrosis, JAM-B and -C expression increases in
endothelial cells and JAM-C expression is induced in myofibroblasts, thereby enhancing
their contractility capacity. These results suggest a potential role of these TJ proteins
in regulating liver immune cell recruitment and fibrosis-induced portal hypertension.[62 ] Tight-junction protein 2 (TJP2) is a TJ protein required for normal cortical distribution
of radixin, bile canalicular volume regulation, and biliary microvilli density. Tjp2 knockout deregulates CLDN1, and key bile acid transporters and detoxification enzymes
and it is associated with liver injury and fibrosis.[63 ] Experiments with mice fed a choline-deficient, ethionine-supplemented, or diethoxycarbonyl-1,4-dihydrocollidine
diet, models for chronic liver disease, biliary–blood barrier injury, and ductular
reaction, show reduced expression of several TJ proteins such as CLDN3, 5, and 7.[64 ] In the healthy liver, CLDN4 and CLDN7 are expressed only in hepatic progenitor cells
and cholangiocytes. However, in cirrhotic livers, CLDN4 and CLDN7 are detected in
hepatocytes at the edges of regeneration nodules, suggesting a role of these TJ proteins
in liver regeneration during extensive fibrosis.[65 ] Livers from patients with alcohol-related hepatitis, a condition usually associated
with extensive fibrosis, demonstrated upregulations of several CLDNs and gap junction
molecules linked to hepatocyte regeneration and hepatic stellate cell activation.
These include CLDN5, CLDN10, CLDN1 and connexin 26, 32, 43, 46.6 as well as gap junction
protein 12.[66 ] In the CCl4 fibrosis model, the expression of hepatic and small intestinal zonula occludens-1
(ZO-1) and OCLN significantly decreased, and these changes can be reverted by a Ginkgolide-A
treatment.[67 ] Finally, in rats treated with thioacetamide, a model of liver fibrosis with pronounced
ductular reaction, a deregulation of OCLN et CLDN2 driven by MAP-kinase, p38 MAP-kinase,
and PI3-kinase in response to IL-1β has been observed.[68 ] During liver regeneration after partial hepatectomy in rats, p38 MAP-kinase induces
downregulation of connexin 32 and upregulation of CLDN1. In vivo administration of
p38 MAP-kinase inhibitor SB203580 can effectively hinder connexin 32 downregulation
and promote CLDN-1 upregulation, suggesting a possible contribution of this compound
toward the restoration of hepatocytes following liver damage.[69 ]
[70 ]
In summary, CLDN1 is the first nonjunctional TJ protein that has undergone extensive
preclinical studies with completed proof of concept to treat liver fibrosis and prevent
liver cancer. A highly specific mAb targeting nonjunctional CLDN1 modulates the plasticity
of hepatocytes, progenitor cells, and myofibroblasts, restoring their differentiated
and functional cell phenotype that is disrupted by fibrosis and inflammation in chronic
liver disease and during the development of HCC.[28 ] The mAbs act mainly on CLDN1 expressed on hepatocytes and progenitor cells as well
as CLDN1 additionally expressed on stellate cells and myofibroblasts. The CLDN1-specific
mAbs exhibit a dual effect on liver fibrosis and HCC prevention and thus have potential
as a treatment for patients with active chronic liver disease and advanced fibrosis
or at a high risk of HCC development. Moreover, these CLDN1-specific mAbs may be employed
as HCC adjuvant treatment to reduce recurrence risk after surgery or locoregional
therapies or, once safety study have been completed, in patients with advanced cirrhosis
who are not eligible for liver transplantation. Compared with other compounds in clinical
development for MASH and fibrosis, the CLDN1 mAb displays a very robust antifibrotic
profile and has a shown a unique potential for preventing HCC in preclinical models.
Combining antimetabolic drugs such as GLP-1 and thyroid hormone receptor-β agonists
with the CLDN1 mAb will provide opportunities for treatment strategies that simultaneously
target MASH's metabolic and fibrotic hallmarks.
Targeting Tight Junction Proteins to Treat Hepatocellular Carcinoma
Targeting Tight Junction Proteins to Treat Hepatocellular Carcinoma
HCC represents the most frequent type of primary liver cancer and the fourth-leading
cause of cancer-related death worldwide.[71 ]
[72 ] A key characteristic of HCC carcinogenesis is its development in a chronically inflamed
and most frequently fibrotic liver microenvironment.[73 ] In this regard, Claudins as key regulators of cell–cell and cell–matrix interactions
have gained increasing interest as drivers of hepatocarcinogenesis. In fact, numerous
studies report dysregulated expression of different members of the TJ protein family
in HCC: while CLDN3 and 14 have been shown to be downregulated in HCC,[74 ]
[75 ] CLDN1, 5, and 10 are overexpressed in tumorous HCC liver tissue.[52 ]
[76 ]
[77 ]
[78 ] Indicating a functional impact of its differential abundance, CLDN1 overexpression
has been shown to be correlated with poor patients' survival.[79 ] Similarly, high levels of CLDN10 were found to be associated with worse patients'
outcome and tumor recurrence.[76 ]
[77 ]
[78 ] Only limited data exist on the expression of other TJ proteins, such as OCLN and
ZO in HCC. Ram et al reported decreased expression of ZO-1 in HCC tissue.[80 ] In line, Nagai et al described decreased ZO-1 levels in HCC as a predictive marker
of poor prognosis.[81 ] The data on the expression level of OCLN in HCC are controversial and both up- and
downregulation have been described, potentially reflecting a correlation of OCLN expression
with tumor cell differentiation[76 ]
[82 ] (for a review see[83 ]). Strikingly, the expression of Claudins has been associated with a specific differentiation
of the corresponding liver tumor. Hereby, not only the overall expression level, but
also particularly the subcellular localization seems to impact on tumor cell plasticity.
Thus, CLDN1 overexpression in HCC tumor cells was found to be accompanied by aberrant
nonjunctional delocalization in the cytoplasm and even nucleus.[84 ] These CLDN1 overexpressing liver tumors were further found to show a stem cell or
progenitor like phenotype.[84 ] CLDN4 and CLDN7 overexpressing liver tumors on the other hand show a ductular differentiation.[85 ] Similar observations were made in other solid cancer types: cytonuclear delocalization
of ZO-1 has been shown to be associated with epithelial–mesenchymal transition (EMT)
and highly invasive human lung cancer cells.[86 ] In breast cancer, ZO-1 is associated with a glandular differentiation.[87 ] Cytosolic OCLN expression was further associated with a well differentiation of
gastric tumor cells.[88 ]
Genetic and epigenetic alterations, as well as signaling pathways, can regulate the
overexpression and delocalization of TJ proteins, potentially through posttranslational
modifications. Recent research from our group has shown that Claudin-1 is upregulated
in AXIN1-mutated HCC, whereas downregulated in tumors with CTNNB1 mutations.[84 ] Additionally, claudin gene amplification has been described to affect expression
patterns.[89 ] Potential mechanisms of claudin upregulation also include epigenetic alterations.
In fact, specific histone modifications have been shown to contribute to CLDN14 overexpression
in HCC.[74 ] Signaling pathway-mediated posttranslational modifications are believed to have
the greatest impact on dynamic changes in TJ protein expression. A recent study found
that TNFα-NFκB signaling strongly upregulates Claudin-1 expression in liver parenchymal
and nonparenchymal cells in the context of inflammation.[28 ] Furthermore, our group identified hypoxia as a significant factor in the upregulation
of CLDN1 expression in HCC cancer cells.[84 ] Other studies suggest that oncogenic signaling pathways, including PI3K/AKT/mTOR
or MAPK/ERK signaling, can upregulate hepatic expression of different claudin family
members.[90 ]
[91 ] However, the molecular mechanisms underlying protein overexpression have not yet
been fully defined. The mechanism of TJ protein delocalization is also not yet understood.
Studies on colon cancer cell lines suggest that the cell adhesion protein EPCAM, which
is often overexpressed in cancer, stabilizes Claudin 1 and 7 at nonjunctional locations
and prevents their lysosomal degradation.[83 ]
[92 ]
Beyond its role as downstream targets of signaling cascades, Claudins and other members
of the TJ protein family also actively impact on key oncogenic cell processes by forming
bidirectional signaling hubs that connect the extracellular to the intracellular compartment
(for a review see[93 ]). As a key event during hepatocarcinogenesis, EMT has consistently been associated
with TJ protein alterations. However, given the role of TJ in maintaining cell polarity
as a key feature of epithelial cells, the questions arise whether alterations in the
expression reflect an active role of these proteins in the mechanism of EMT or rather
depict its consequence. While numerous reports in other solid cancers, including breast,
pancreatic, or colon cancer suggest a direct interaction of diverse TJ proteins with
EMT mediators (for a review see[94 ]
[93 ]), conclusive perturbation studies in HCC are mostly limited to Claudins. Suh et
al reported CLDN1 to induce EMT in HCC via c-Abl-ERK signaling induction and upregulation
of the transcription factors Slug and Zeb 1.[95 ] Moreover, Yoon et al revealed CLDN1 to promote EMT via c-Abl-OKC-δ-mediated upregulation
of MMP2.[96 ] Conversely, CLDN3 suppresses EMT in liver cancer by downregulation of Wnt-β-catenin
signaling.[75 ]
Beyond its impact on EMT, Claudins, and other TJ proteins have been associated with
cell–cell or cell–matrix interactions and key oncogenic cascades regulating cell growth.
Corroborating the role of CLDN1 in cell–cell communication, a recent study identified
nonjunctional CLDN1 to control Notch signaling upon cell–cell contact.[84 ] Zhang et al reported ZO-1 overexpression in liver cancer cells to inhibit cell proliferation
and migration in vitro.[97 ]
Given its active role in key oncogenic events, including tumor cell differentiation
and stemness, as well as EMT and invasion, TJ proteins have been studied extensively
as potential targets of cancer therapy, including HCC. Yet, treatment strategies for
patients with advanced HCC that are not eligible for surgical or locoregional therapies
are highly limited.[7 ] Currently, the combination therapy with atezolizumab and bevacizumab or durvalumab
and tremelimumab represent the preferred first line therapy for advanced HCC. However,
objective treatment response is below 30%,[98 ] and alternative treatment strategies such as receptor tyrosine kinases show only
limited efficacy in immunotherapy-resistant HCC's (for a review see[99 ]
[100 ]). Moreover, HCC recurrence after curative treatments such as resection or local
ablation is high, with rates exceeding 70% after 4 years. To date, there is no approved
adjuvant treatment to mitigate the risk of HCC recurrence. Thus, urgent development
of novel HCC therapies is imperative.
CLDN1 and CLDN6 are currently the targets with the most promising and advanced clinical
development. Targeting nonjunctional CLDN1 by a mAb has been shown to suppress tumor
initiation and progression in patient-derived ex vivo and in vivo models of HCC.[28 ]
[84 ] Mechanistically, tumor cell suppression was associated with broad inhibitory effects
on oncogenic signaling cascades, EMT, as well as cancer stemness.[84 ] These oncogenic pathways are also modified in other cancers such as head and neck
cancer.[101 ] Currently, a phase I/II clinical trial is investigating a CLDN1 mAb with or without
combination with pembrolizumab in patients with head and neck cancer (NCT06054477).
CLDN6 is highly expressed in embryonic stem cells and in HCC compared with normal
tissue. CLDN6 expression in HCC is associated with a biliary transdifferentation phenotype
and sorafenib resistance. CLDN6 overexpression activates the JAK2/STAT3 signaling
pathway, leading to increased BLC2 expression and inhibition of apoptosis in cancer
cells.[102 ] Additionally, CLDN6 competes with YAP1 for TJP2 binding, preventing YAP1 cytoplasmic
retention. This results in elevated levels of free YAP1, facilitating its nuclear
translocation and transcription of genes that promote cell proliferation and trigger
EMT[49 ]
[103 ] ([Fig. 3 ]). Anti-CLDN6 mAbs conjugated with cytotoxic agents further showed potent antitumor
efficiency as well as sorafenib synergy in mouse models of HCC.[49 ] Safety and efficacy of CLDN6 targeting chimeric antigen receptor (CAR)-T cells,
with or without a CAR-T Cell amplifying RNA vaccine, for any type of CLDN6+ cancers, including HCC, are currently being evaluated in a phase I/IIa trial (NCT04503278).
A nonprespecified interim analysis of 22 patients treated in this trial has been recently
published. Safety data show manageable toxicity with 46% of patients experiencing
cytokine release syndrome. The unconfirmed objective response was 33% with one complete
response. However, no enrolled patient had liver cancer.[104 ]
Fig. 3 Biology of CLDN6-overexpressing liver cancer. CLDN6 overexpression activates the
JAK2/STAT3 signaling pathways that upregulates BLC2 (B cell lymphoma 2) and reduces
cancer cell apoptosis. CLDN6 also competes with YAP1 (Yes-associated protein) for
binding to TJP2 (tight junction protein 2), preventing YAP1 translocation to the nucleus.
CLDN6 overexpression increases the levels of unbound YAP1 and favors YAP1 nuclear
translocation and transcription of genes that promote cell proliferation and induce
EMT. Anti-CLDN6 mAbs have been shown to have preclinical efficacy against HCC.[49 ] EMT, epithelial–mesenchymal transition; HCC, hepatocellular carcinoma.
While data on small molecule inhibitor mediated targeting of ZO protein is yet restricted
to gastrointestinal cancers other than HCC (for a review see[105 ]), reduction of proliferation and migration of liver cancer cells upon transgenic
ZO-1 overexpression may catalyze further research on ZO-targeting therapies in HCC.
In summary, targeting Claudins is a promising strategy for HCC treatment. CLDN1- and
CLDN6-based treatment approaches have shown strong efficacy in preclinical models
and clinical development has started for solid tumors outside the liver. Future studies
should prioritize the development of companion biomarkers and response predictors
as well as the investigation of combined therapies using immune checkpoint inhibitors
and/or antiangiogenic drugs to enhance treatment efficacy.
Conclusions
Studies on HCV biology have unveiled a key role of TJs proteins in liver pathophysiology
and development of fibrosis and cancer. Nonjunctionally exposed TJ proteins have been
reported to play a key role in oncogenic intracellular signaling and mediate cell
proliferation, differentiation, and function. As a treatment for liver fibrosis, CLDN1
is currently the most advanced target with ongoing clinical development. CLDN1 exists
in a junctional and nonjunctional form, and it is expressed not only in liver epithelial
cells but also in liver progenitors and activated myofibroblasts. Nonjunctional CLDN1
is has been uncovered as a driver for liver fibrosis and HCC via the activation of
multiple signaling pathways such as TNF-α–NFκB, EGFR, ERK, Notch-1, and SRC resulting
in modulation of plasticity and fate of fibrosis driver cells including hepatocytes
and myofibroblasts. Preclinical data demonstrate robust and significant antifibrotic
and HCC prevention efficacy, highlighting an opportunity for this mAb in patients
to treat advanced liver fibrosis (F3/4), those at high risk of HCC, and patients with
cirrhosis who are ineligible to liver transplantation.
Several studies investigated the role of TJ proteins such as CLDN1, CLDN4–7, OCLN,
ZO-1 in the biology of HCC. Preclinical studies have identified CLDN1 and 6 as therapeutic
targets. CLDN1- and CLDN6-based treatment approaches have shown robust efficacy in
preclinical models and clinical development has started for solid tumors outside the
liver. The antifibrotic activity of CLDN1 targeting therapies is a particular advantage
for HCC treatment, since the dual anticancer and antifibrotic effect may also improve
the underlying liver disease, which is often a key denominator of survival in HCC
patients. Further studies are needed for clinical proof of concept in HCC.