Metabolic Dysfunction-Associated Fatty Liver Disease (MAFLD) - An Update of The Recent Advances in Pharmacological Treatment
Metabolic Dysfunction-Associated Fatty Liver Disease (MAFLD) - An Update of The Recent Advances in Pharmacological Treatment
https://doi.org/10.1007/s13105-023-00954-4
REVIEW
Received: 24 November 2022 / Accepted: 28 February 2023 / Published online: 28 March 2023
© The Author(s) 2023
Abstract
Metabolic dysfunction–associated fatty liver disease (MAFLD) is nowadays considered the liver manifestation of metabolic
syndrome. Its prevalence is increasing worldwide in parallel to the epidemic of diabetes and obesity. MAFLD includes a
wide spectrum of liver injury including simple steatosis and non-alcoholic steatohepatitis (NASH) that may lead to serious
complications such as liver cirrhosis and liver cancer. The complexity of its pathophysiology and the intricate mechanisms
underlying disease progression explains the huge variety of molecules targeting diverse biological mechanisms that have
been tested in preclinical and clinical settings in the last two decades. Thanks to the large number of clinical trials of the
last few years, most of them still ongoing, the pharmacotherapy scenario of MAFLD is rapidly evolving. The three major
components of MAFLD, steatosis, inflammation, and fibrosis seem to be safely targeted with different agents at least in a
large proportion of patients. Likely, in the next few years more than one drug will be approved for the treatment of MAFLD
at different disease stages. The aim of this review is to synthesize the characteristics and the results of the most advanced
clinical trials for the treatment of NASH to evaluate the recent advances of pharmacotherapy in this disease.
                                                                          Introduction
Key Points • For the time being, there is no specific treatment
for NASH to avoid disease progression into hepatic fibrosis or to
                                                                          Definition and epidemiology
reduce established fibrosis.
    • As pathophysiology of NASH is very complex and not-                 Metabolic dysfunction–associated fatty liver disease
completely well understood, identification of biological drug             (MAFLD) has become one of the most relevant forms of
targets is challenging.
                                                                          chronic liver disease worldwide due to the progressively
    • Many clinical trials are under development to try to identify
drugs to diminish hepatic inflammation/fibrosis. A synthesis of           increased in obesity rates over the past 30–40 years [1].
the results of the most advanced clinical trials for the treatment of     Obesity is defined as a body mass index ≥ 30 kg/m2 and the
NASH is presented in this review.                                         World Health Organization estimates that over 13% of the
                                                                          world population (> 600 million people) suffer from it, with
Paloma Sangro, Manuel de la Torre Aláez Shared equal authorship.
                                                                          more prevalent rates among children and adolescents [2].
* Paloma Sangro                                                           The metabolic syndrome includes three out of the follow-
  [email protected]                                                         ing conditions: abdominal obesity, hypertriglyceridemia,
* Manuel de la Torre Aláez                                                low HDL cholesterol, arterial hypertension, and/or hyper-
  [email protected]                                                         glycemia. MAFLD is not only associated to those comor-
     Bruno Sangro                                                         bidities but also to insulin resistance. Indeed, the nomencla-
     [email protected]                                                      ture of non-alcoholic fatty liver disease (NAFLD) has been
     Delia D’Avola                                                        updated to MAFLD. This term describes better the liver dis-
     [email protected]                                                      ease associated with known metabolic dysfunction. In fact,
1                                                                         the new definition of MAFLD refers to hepatic steatosis in
     Liver Unit Clínica, Universidad de Navarra, Madrid, Spain
                                                                          addition to one of the following three criteria: overweight/
2
     Centro de Investigación Biomédica en Red de Enfermedades             obesity, presence of type 2 diabetes mellitus, or evidence of
     Hepáticas y Digestivas, Pamplona, Spain
                                                                                                                             13
                                                                                                                        Vol.:(0123456789)
870                                                                                                               P. Sangro et al.
metabolic dysregulation. The exclusion of other liver dis-      a significant association of a variant in “patatin-like phos-
eases including alcoholic, autoimmune, or viral hepatitis is    pholipase domain-containing 3” (PNPLA3) gene on chromo-
not a prerequisite for the diagnosis of MAFLD [3].              some 22 leading to modifications in retinol metabolism [12];
   Within the general population, the overall global preva-     or in “transmembrane 6 superfamily member 2” (TM6SF2)
lence of NAFLD (defined using imaging criteria) is esti-        gene on chromosome 19 leading to an impaired lipid trans-
mated to be 25% [4]. However, important variability was         porter, have been associated with fatty liver disease [13].
observed according to geographic regions, being up to 31.8%     Polymorphism in “ectoenzyme nucleotide pyrophosphate
in the Middle East, 30.4% in South America, and 13.5%           phosphodiesterase 1” (ENPP1 or PC1) and in “insulin recep-
in Africa [4]. NAFLD incidence has rarely been measured.        tor substrate-1” (IRS1) genes, which are related to insulin
   Given the common risk factors between NAFLD and              resistance, have also been described in NAFLD patients
cardiovascular risk factors, cardiac-related death is one of    [14]. Even, a polymorphism in “membrane bound O-acyl-
the leading causes of death for NAFLD patients [5]. Other       transferase domain-containing 7” (MBOAT7-TMC4) gene,
causes include liver-related death, malignancy, or other        involved in oxidative stress, increases the risk of fibrosis in
causes such as infections, type 2 diabetes mellitus, or pul-    patients with NAFLD [15]. Genetic variants in “glucokinase
monary embolism.                                                regulatory protein” (GCKR) gene [16], “solute carrier family
   NAFLD is defined as the presence of steatosis in > 5%        2-member 1” (SLC2A1) gene [17], and “17-beta hydroxys-
of hepatocytes in the absence of other competing chronic        teroid dehydrogenase 13” (HSD17B13) gene [18], have also
liver diseases and without significant alcohol consumption      been identified in patients with fatty liver disease.
(< 20 g/day in women and < 30 g/day in men). NAFLD                 NASH is the result of multiple intracellular signals
includes non-alcoholic fatty liver (NAFL) and non-alcoholic     derived either from proinflammatory molecules and con-
steatohepatitis (NASH), the latter being more severe as it      tact with immune cells and/or from external stimulation
includes hepatocyte damage with the risk of subsequent          through visceral adipose tissue and gut microbiome, the
development of significant fibrosis, cirrhosis, and/or hepa-    last two conditioned by diet. The outcome of these interac-
tocellular carcinoma [6]. In this context, NASH has become      tions on hepatocytes includes modified insulin signaling,
one of the leading causes of cirrhosis in adults in the USA     lipogenesis, mitochondrial dysfunction, and activation or
and NASH-related cirrhosis is currently the second indica-      abnormal functioning of nuclear receptors such as bile acid
tion of liver transplantation in the USA [7].                   receptors farnesoid X receptor (FXR) [19], liver X receptor
   Whereas the definitive diagnosis of both MAFLD and           (LXR) [20], pregnane X receptor (PXR) [21], and vitamin
NASH requires a liver biopsy, imagine techniques such as        D receptor (VDR) [22] leading to hepatic inflammation by
magnetic resonance imaging (MRI) (MRI proton density            different downstream effects. Hepatic stellate cells are acti-
fat fraction [MRI-PDFF] or magnetic resonance spectros-         vated by these signals may trigger a fibrogenic response with
copy), computed tomography (CT), ultrasound (US) with           extracellular matrix production [23].
controlled attenuation parameter (CAP), and/or elastogra-          Diet with excess carbohydrates and saturated fat lead to
phy may provide a valuable assessment of fat deposition         an energy metabolism imbalance with lipid deposition in
and significant fibrosis [6]. Although no specific treatments   skeletal muscle. Subsequent muscle insulin resistance due
other than the control of the associated metabolic disorders    to increased intramyocellular lipid content impedes the stor-
is available, follow-up to control comorbidities is recom-      age of ingested glucose as muscle glycogen [24]. Glucose is
mended to detect disease progression.                           then rerouted to the liver where insulin resistance stimulates
                                                                sterol regulatory element–binding protein 1c (SREBP1c)
Genetics and pathogenesis                                       [25] which increases the expression of hepatic enzymes
                                                                that regulate de novo lipogenesis with higher VLDL produc-
Most individuals with MAFLD have no symptoms, and the           tion and hypertriglyceridemia. Other transcription factors
disease may remain silent until it has progressed to cirrho-    such as carbohydrate-responsive element–binding protein
sis [8]. Recent data suggest that the transition from NAFL      (ChREBP) [26], peroxisome proliferator–activated recep-
to NASH is quite dynamic whereas fibrosis development is        tor gamma coactivator 1-beta (PPARg coactivator 1-b) [27],
significantly slower in NAFLD than in NASH, needing even        and LXR [28] also foster liver lipogenesis.
up to 14 years [9]. Up to 20% of patients with NASH are            Although visceral adipose tissue has been related to the
considered as “rapid progressors” where, although predic-       pathogenesis of NASH, visceral adipose tissue may just be
tors are largely unknown, genetic susceptibility might play     an additional site for lipid accumulation when subcutane-
a role [10]. The relevance of the genetic determinants is       ous adipose tissue capacity is surpassed [29]. Circulating
just beginning to be elucidated [11], but they may involve      triglyceride-enriched lipoproteins are cleared by peripheral
intrahepatic lipolysis, triglyceride export, hepatic mito-      lipoprotein lipase (Lpl) [30]. However, an increase in endog-
chondrial oxidation, or glucokinase activity. For instance,     enous Lpl inhibitors, such as apolipoprotein C3 (ApoC3)
13
Metabolic dysfunction–associated fatty liver disease (MAFLD): an update of the recent advances…                                       871
[31], angiopoietin-like proteins 3/8 (ANGPTL3/8) complex,                     In the last 10 years, more than 20 molecules have been
and ANGPTL4 [32], impedes clearance of these circulating                   tested for the treatment of NASH. Most drugs were dis-
triglyceride-enriched lipoproteins and therefore, induces an               carded after unsuccessful clinical trials, others are still in
increased hepatic triglyceride uptake. Increases in endog-                 early stage of clinical development (phase 1 and 2 clinical
enous Lpl inhibitors have been described in NASH patients.                 trials) and a few have reached phase 3 trials. So far, drugs
   All in all, NASH is a heterogeneous disease with a com-                 at most advanced clinical development include antidiabetic
plex pathophysiology that makes finding a single effective                 drugs, FXR agonists, PPAR agonists, and thyroid hormone
treatment a major challenge.                                               receptor (THR) agonists (Fig. 1). Table 1 includes a synthe-
                                                                           sis of the clinical trials (phase II/III) in treatment of NASH.
It is well-known that the first approach to treat NAFLD                    As insulin resistance and type 2 diabetes mellitus are closely
patients is the nutritional intervention with a change in life-            associated with NASH, the efficacy of several antidiabetic
styles. However, the complexity of NASH pathophysiology                    agents has been studied in NASH. Pioglitazone showed his-
and the interplay of multiple genetic and environmental fac-               tological benefits (improved NAFLD activity score, also
tors in disease progression explains why the pharmacother-                 called NAS or improvement in single histological com-
apy of this clinical condition includes a variety of molecules             ponents of NASH) in three randomized trials in diabetic
with different biological targets, from drugs used to treat                and prediabetic patients with NASH. However, the effect
of the diseases that contribute to NASH development and                    on liver fibrosis was not significant, although worsening of
progression (i.e., antidiabetic agents) to drugs targeting liver           liver fibrosis was not observed [33–35].
inflammation and fibrosis. Its long natural history and wide                  Newer antidiabetic agents including SGLT1/2 (sodium-
spectrum of severity, from mild inflammation to end-stage                  glucose co- transporter-1/2) inhibitors and GLP-1R (gluca-
cirrhosis, are challenges in the evaluation of pharmacologi-               gon-like peptide 1 receptor) agonists are being tested in
cal interventions and requires specific interventions for each             NASH. Dapagliflozin and semaglutide have reached late-
disease stage.                                                             stage clinical development.
                                                                                                                               13
     Table 1  Main clinical trials in treatment of NASH/NAFLD (phase II/III)
                                                                                                                                                                                                       872
     Molecule and main trial        Mechanism of action             Primary objective                   Patients Grade of fibrosis Main results                    Most frequent adverse events
                                                                                                        recruited
13
                                                                                                        (n)
                                                                                               Well tolerated
                                                                                                                                                               the placebo or control group in patients with NASH (image-
                                                                                                                                                               based diagnosis) significantly lowered ALT (weighted mean
                                                                                                                                                               difference (WMD): − 6.62U/L; 95%CI: − 12.66, − 0.58;
                                                                                                                                                               p = 0.03) and AST levels (WMD: − 4.20U/L; 95%CI: − 7.92,-
                                                                                                 worsening of NASH on liver                                    0.47; p = 0.03). Gamma-glutamyl transferase (GGT) levels
                                                                                                 MRS. Improvement in liver
1–3
                                                                                                                               123
                       (n)
by MRI
Inhibitor of mitochondrial
CoA synthetase 4
                                                                                                                               (NCT04321343) phase 2
                       Molecule and main trial
PXL065
                                                                                                                                                                                                                    13
874                                                                                                                 P. Sangro et al.
from baseline were also observed in the treated groups             placebo in histological resolution of NASH (49% and 39%,
without any acute pancreatitis episode. A phase 3 clinical         respectively, vs. 22%), histological improvement of fibrosis
trial (ESSENCE, NCT04822181) is recruiting non-cirrhotic           (48% and 34%, respectively, vs. 29%), or both (35% and
NASH patients to evaluate the potential resolution of steato-      25%, respectively, vs. 9%) were observed [43]. Nausea, diar-
hepatitis and the improvement in fibrosis with this GLP-1R         rhea, peripheral edema, anemia, and weight gain occurred
agonist.                                                           more frequently with lanifibranor than with placebo.
    The LEAN trial [38], a phase 2 trial evaluating the effect        The PPARα agonist pemafibrate has been tested in
of liraglutide (1.8 mg daily) compared to placebo in patients      NASH patients (MRI diagnosis with ALT elevation) [44].
with biopsy-confirmed NASH, showed histological resolu-            The primary endpoint of percentage change in liver fat
tion of NASH (39% with liraglutide vs. 9% with placebo,            content measured by MRI at week 24 was not met (− 5.3%
p = 0.019). Fibrosis progression was observed in 36% of            vs − 4.2%; treatment difference − 1.0%, p = 0.85). However,
patients receiving placebo vs in 9% of liraglutide-treated         liver stiffness measured by MRI decreased at week 48 (treat-
patients (p = 0.04). Adverse events reported were mainly           ment difference − 5.7%, p = 0.036), and was maintained at
gastrointestinal disorders or administration site reactions.       week 72 (treatment difference − 6.2%, p = 0.024).
    Others GLP1R such as cotadutide (NCT04019561), tirze-
patide (NCT04166773), or efinopegdutide (NCT04944992)              FXR agonists
have been tested or are still being tested in phase 2 trials and
the results have not been published yet.                           FXR modulation may have multiple implications in the treat-
                                                                   ment of NASH since, besides bile acid (BA) synthesis, this
PPAR modulators                                                    receptor is involved in glucose and lipid metabolism, and in
                                                                   the regulation of inflammation [45]. FXR activation down-
The peroxisome proliferator-activator receptor (PPAR)              regulates bile acid synthesis through the upregulation of
family is formed by PPARα, PPARγ, and PPARδ which                  fibroblast growth factor (FGF)-19 expression and by reduc-
are mainly located in liver, brown adipose tissue, and mac-        ing the expression of CYP7A1, the rate-limiting enzyme of
rophage. They activate fatty acid oxidation, lower synthesis       the BA synthesis. This results in a protective effect against
of triglycerides, and increase insulin sensitivity. Saroglitazar   the toxic accumulation of BAs through increased conjuga-
is a dual PPARα and PPARγ agonist, that was first approved         tion in the liver and secretion into the bile canaliculi. FXR
in India for the treatment of type 2 diabetes mellitus patients    activation improves glucose tolerance by reducing hepatic
with hypertriglyceridemia [39]. Since 2020, it is widely used      gluconeogenesis and increasing glycogen synthesis [46].
as treatment for NASH patients in India. Among patients            Additionally, FXR activation reduces fat accumulation in
with NAFLD (stages 0–3) diagnosed by imaging (ultra-               the liver by targeting SHP expression and CYP7A1 activ-
sound, CT scan, or MRI) or liver biopsy showing NASH or            ity [47]. Obeticholic acid (OCA) is a semi-synthetic bile
simple steatosis, and alanine aminotransferase (ALT) > 1.5         acid analog of chenodesoxicholic acid with a potent ago-
upper limit of normal recruited into a phase 3 trial (EVI-         nist activity on the FXR in the liver and in the intestine.
DENCES II), saroglitazar reduced ALT levels after 16 weeks         OCA, as the other members of FXR agonists family, may
of therapy [40]. Patients receiving saroglitazar 4 mg reduced      exert multiple therapeutic effects on NASH by improving
the most their levels of ALT (− 45.8% vs. 3.4%, p < 0.001).        hepatic lipid and glucose metabolism and through its anti-
Histological improvement of NASH after 16 weeks of treat-          inflammatory and anti-fibrotic activity. OCA lowers plasma
ment was observed with 4 mg of saroglitazar (reduction in          triglycerides by downregulating the expression of SREBP1c
liver fat content − 19.7% vs. 4.1% with placebo (p = 0.004)).      expression (48) and increases hepatic fatty acid oxidation
Safety and tolerability of saroglitazar were further assessed      through upregulation of pyruvate dehydrogenase kinase 4
in a phase 2 study (EVIDENCES IV), and the most frequent           (PDK4) and modulating glucose-dependent lipogenic genes
adverse effects were diarrhea and cough [41].                      [49]. It has been shown that OCA reduces insulin resist-
    After a successful phase 2 b trial, elafibranor, a dual        ance and improves glucose homeostasis in diabetic patients
PPAR-alpha/delta agonist, was tested in a phase 3 clinical         with NASH [50]. OCA may also contribute to reduce portal
trial in patients with non-cirrhotic NASH. In this trial, elafi-   pressure by increasing nitric oxide synthases (iNOS) and
branor did not meet histological endpoints (NASH resolution        decreasing inflammation mediators.
without worsening of fibrosis) nor the key secondary end-             After a phase 2b clinical trial in patients with biopsy-
point (fibrosis improvement at least one stage) [42].              proven NASH without cirrhosis (FLINT trial) in which OCA
    Lanifibranor, the only pan-PPAR agonist, was assessed          displayed a beneficial impact on fibrosis without resolution
in non-cirrhotic biopsy-confirmed highly active NASH               of advanced fibrosis and at least of 2 points improvement
(stages 0–3) patients in a phase 2b trial (NATIVE study)           in NASH compared to placebo [51], this drug is now being
were 1200 mg showed better results than 800 mg dose or             tested in a large phase 3 randomized, placebo-controlled trial
13
Metabolic dysfunction–associated fatty liver disease (MAFLD): an update of the recent advances…                                       875
(REGENERATE trial, NCT2548351) to evaluate the long-                       inflammation and fibrosis measured by using a non-invasive
term effects on NASH and fibrosis in patients with stage 1–3               composite marker assessed by MRI (cT1) and MRI-PDFF
fibrosis. The results of an interim analysis showed a signifi-             [57].
cant improvement of fibrosis in approximately 20% of patients
treated with OCA vs. 12% of placebo [52]. The NASH resolu-
tion endpoint was not met in this preliminary analysis. The                FGF analogs
most significant side effect was a dose-dependent pruritus,
that in some cases (< 10%) required treatment discontinua-                 FGF19 and FGF21 are members of FGF superfamily that
tion. Increased cholesterol levels were frequently observed,               exert similar but not identical beneficial effects on lipid
and statins were newly prescribed in almost 50% of patients                and glucose metabolism. Indeed, the administration of
receiving OCA [52].                                                        both FGF19 and FGF21 in animal models improve insulin
    At the same time, the antifibrogenic effect of OCA                     sensitivity, improve body weight and fat mass, lipid levels,
is being tested in a phase 3 trial including patients with                 and liver steatosis. This latter effect probably depends on
compensated cirrhosis due to NASH (REVERSE trial,                          the inhibition of SREBP1 and reduced expression of genes
NCT03439254).                                                              involved in triglyceride synthesis [58].
    MET642 and MET 409 are other structurally optimized                       In the last years, different FGF21 analogs and one FGF19
synthetic FXR agonists. Preliminary reports suggest that                   analog have been tested in phase 1 and 2 trials for the treat-
these compounds produce less pruritus than OCA. MET642                     ment of NASH.
is now being tested in a phase 2 clinical trial in patients with              In a phase 2b trial, the FGF19 analog aldafermin failed
NASH (NCT0477396). After the results of a phase 1b trial                   to improve liver fibrosis among patients with NASH-related
in which MET409 showed its ability to reduce the liver fat                 stage 2 or 3 fibrosis. Other FGF21 analogs are pegbelfermin
content in monotherapy, this drug is now being tested in a                 and efruxifermin. In a phase 2 trial in patients with stage
phase 2b trial in combination with empagliflozin.                          1–3 liver fibrosis, efruxifermin reduced liver fat content,
    Tropifexor, another highly potent non-bile acid FXR                    improved liver function tests, and fibrosis and inflammation
agonist, is being tested in a phase 2 clinical trial (FLIGHT               markers (including NAS score). Fibrosis stage improvement
FXR, NCT02855164) in patients with stage 1–3 fibrosis                      of at least 1 point was observed in almost half of the patients
due to NASH. In a preliminary analysis, treatment with                     and NASH resolution in almost one third. Safety profile was
tropifexor lead to a significant reduction of hepatic fat, liver           favorable [59].
transaminases, and body weight compared to placebo with
a favorable safety profile. Complete results are expected to
be released in the next few months [53].                                   THR beta agonists
    Cilofexor is another nonsteroidal FXR agonist that
showed efficacy in decreasing liver steatosis a phase 2                    THR-β is involved in the regulation of lipid metabolism,
trial [54]. The preliminary results of a phase 2 trial testing             plays a role in insulin sensitivity, and promotes liver
cilofexor in combination with firsocostat (an acetyl-CoA car-              regeneration and reduces hepatocyte apoptosis in the liver.
boxylase inhibitor) and semaglutide suggest a synergistic                  In a phase 2 trial in patients with NASH and fibrosis, the
activity of this combination in improving liver steatosis and              THR-β agonist resmetirom decreased liver fat content and
liver biochemistry [55].                                                   improved lipid metabolism parameters and liver function
    EDP305 is another FXR agonist. A phase 2 randomized,                   tests, with a positive impact on lipid profile and fibrosis
double-blind, placebo-controlled, dose-ranging trial                       markers, with no significant effect on body weight [60]. A
(ARGON-1) has shown that when administered to non-cir-                     phase 3 study on patients with non-invasive diagnosis of
rhotic biopsy-proven NASH patients, a 12-week course of                    NAFLD showed that 100 mg of resmetiron administered
EDP305 reduced liver fat content and decreased ALT [56].                   daily for 52 weeks improved liver fat content in approxi-
The most frequent adverse event was pruritus and changes                   mately 50% of treated patients vs 8% of placebo-treated
in lipid parameters were milder than with first-in-clasee                  patients; liver fibrosis in approximately 20% of treated
FXR agonists. ARGON-2 is a phase 2b study testing the                      patients vs. 10% of placebo-treated patients (both meas-
efficacy of EDP305 in NASH patients with stage 2–3 fibrosis                ured by a non-invasive test). Lipid metabolism param-
(NCT04378010).                                                             eters, liver enzymes, and inflammatory biomarkers also
    Finally, TERN-101 is a nonsteroidal FXR agonist with                   improved compared to placebo [61]. There were no major
enhanced liver distribution. In a preliminary analysis of a                safety concerns. Main adverse events were gastrointestinal
phase 2 trial (LIFT study) in patients with stage 1–3 liver                (diarrhea, nausea). A phase 3 study in NASH patients with
fibrosis, a 12-week course of TERN-101 rapidly decreased                   fibrosis is still ongoing and data will be available soon
ALT and GGT and seemed to have positive effect on                          (MAESTRO-NASH, NCT03900429).
                                                                                                                               13
876                                                                                                             P. Sangro et al.
   A liver-directed THR-β agonist, VK2809, improved            in patients with stage 3 fibrosis; and STELLAR-4, in
liver fat content compared to placebo among patients with      patients with compensated cirrhosis) [66].
NAFLD treated with two different doses in a preliminary
analysis of a phase 2 trial [62]. No serious adverse events    Other metabolic treatments
were reported.
                                                               Molecules involved in other metabolic pathways are at dif-
                                                               ferent stage of clinical development for the treatment of
Anti‑fibrotic and anti‑inflammatory agents                     NASH.
                                                                   Stearoyl-coenzyme A desaturase-1 (SCD-1) is consid-
Currently, many agents with anti-fibrotic and anti-inflam-     ered a mediator of liver steatosis and fibrosis because of its
matory effects are being tested in NASH. Most of them are      role in fatty acid biosynthesis [67] [68]. Aramchol is an oral
still in early stage of clinical development: in phase 1 (as   SCD1 modulator which showed an ability to reduce liver
for example GB1211 targeting galectin 3, DFV890 target-        fat and improve NASH and fibrosis with favorable tolerance
ing NLPR-3, or nimacimab targeting CB1) and phase 2 (as        among patients with overweight or obesity and prediabetes
for example tipelukast, a leucotrien, or nitazoxanide, an      in a phase 2b trial (ARREST) [69]. An ongoing phase 3 trial
antiparasitic agent). The description of all of them exceeds   (ARMOR study, NCT04104321) in patients with advanced
the objective of this review.                                  fibrosis and NASH aims to evaluate the efficacy of aramchol
    The available information from molecules in later          vs. placebo on NASH resolution, fibrosis improvement, and
stages of clinical development is summarized below.            clinical outcomes related with NASH progression.
    Cenicriviroc (CVC) is an orally administered, small            Icosabutate is a synthetic omega 3 fatty acid (a structur-
molecule antagonist that blocks chemokine 2 and 5 recep-       ally engineered eicosapentaenoic acid that resist oxidation
tors, both with well-known roles in liver inflammation         and does not accumulate in hepatocytes) that could confer
and fibrosis. In a phase 2b trial (CENTAUR), the primary       beneficial effects on hepatic oxidative stress, inflammation,
objective of histological improvement in NASH was not          and fibrosis. A phase 2b study is evaluating the efficacy of
met although CVC improved measurable liver fibrosis            different doses of icosabutate on the resolution of NASH
without worsening NASH [59]. A phase 3 trial (AURORA,          without worsening of fibrosis. The primary endpoint is
NCT03028740) was prematurely stopped when an interim           to evaluate the percentage of patients with resolution of
analysis casted doubts on the efficacy of the drug. Prelimi-   NASH defined as disappearance of ballooning with lobular
nary results of a phase 2b trial (TANDEM, NCT03517540)         inflammation without worsening in fibrosis (ICONA study,
showed that the combination of tropifexor with CVC was         NCT04052516).
safe and able to reduce body weight and ALT in patients            MSDC-0602 K is a thiazolidine-dione designed to
with biopsy-proven NASH with fibrosis. However, the            modulate the mitochondrial pyruvate carrier (MPC), a pro-
combination was not superior to either drug in monother-       tein complex that regulates the entry of pyruvate into the
apy in terms of histological endpoints [63].                   mitochondria. A phase 2b placebo-controlled randomized
    Belapectin is a complex carbohydrate that targets          trial (EMMINENCE) failed to show improved histological
galectin-3, a B-galactoside-binding lectin that plays a        outcomes (≥ 2-point NAS improvement without worsening
role in inflammatory response and fibrosis [64]. A phase       fibrosis, NASH resolution, and fibrosis reduction) among
2b clinical trial (NASH-CX, NCT02462967) showed that           patients with biopsy-proven NASH and stage 1–3 fibrosis
belapectin has not any significant effect on inflammation      [70]. A dose-dependent improvement in the glycemic control
and fibrosis compared to placebo. However, it produced         and liver enzymes were nevertheless observed.
a significant decrease of the hepatic venous pressure gra-         A novel, deuterium-stabilized R-pioglitazone, PXL065,
dient in patients with NASH-related cirrhosis without          that lacks PPAR-gamma activity but exerts its non-genomic
esophageal varices [65]. A phase 2b/3 trial (NAVIGATE,         target activities (mitochondrial pyruvate carrier and acyl-
NCT04365868) is assessing the proportion of NASH               CoA synthetase 4 inhibition) has been tested in a phase 2
patients with compensated cirrhosis who develop new            trial in non-cirrhotic patients with NASH (NCT04321343).
esophageal varices after an 18-month course of belapectin      According to the preliminary results reported at AASLD 2022
compared to placebo, as well as the incidence of long-term     meeting, this drug can reduce the liver fat content in 40% of
clinically significant cirrhosis-related events.               patients and improve at least 1 fibrosis stage in 30–50% of
    Selonsertib is an oral inhibitor of the apoptosis sig-     treated patients. In this trial, up to 30% of patients showed
nal–regulating kinase-1 (ASK1). Although well tolerated,       NASH resolution after 36 weeks of treatment with a good
it failed to show improvement in fibrosis without worsen-      safety profile since it lacks the PPAR-gamma side effects of
ing of NASH in two different phase 3 trials (STELLAR-3,        glitazones [71]. The phase 3 trial has not started yet.
13
Metabolic dysfunction–associated fatty liver disease (MAFLD): an update of the recent advances…                                                                877
                                                                                                                                                      13
878                                                                                                                                P. Sangro et al.
17. Vazquez-Chantada M, Gonzalez-Lahera A, Martinez-Arranz                36. Sun L, Deng C, Gu Y, He Y, Yang L, Shi J (2022) Effects of
    I, Garcia-Monzon C, Regueiro MM, Garcia-Rodriguez JL et al                dapagliflozin in patients with nonalcoholic fatty liver disease: a
    (2013) Solute carrier family 2 member 1 is involved in the                systematic review and meta-analysis of randomized controlled
    development of nonalcoholic fatty liver disease. Hepatology               trials. Clin Res Hepatol Gastroenterol 46(4):101876
    57(2):505–514                                                         37. Newsome PN, Buchholtz K, Cusi K, Linder M, Okanoue T,
18. Abul-Husn NS, Cheng X, Li AH, Xin Y, Schurmann C, Stevis P                Ratziu V et al (2021) A placebo-controlled trial of subcutane-
    et al (2018) A protein-truncating HSD17B13 variant and protec-            ous semaglutide in nonalcoholic steatohepatitis. N Engl J Med
    tion from chronic liver disease. N Engl J Med 378(12):1096–1106           384(12):1113–1124
19. Chávez-Talavera O, Tailleux A, Lefebvre P, Staels B (2017) Bile       38. Armstrong MJ, Gaunt P, Aithal GP, Barton D, Hull D, Parker
    acid control of metabolism and inflammation in obesity, type 2            R et al (2016) Liraglutide safety and efficacy in patients with
    diabetes, dyslipidemia, and nonalcoholic fatty liver disease. Gas-        non-alcoholic steatohepatitis (LEAN): a multicentre, double-
    troenterology 152(7):1679-1694.e3                                         blind, randomised, placebo-controlled phase 2 study. The Lancet
20. Beaven SW, Matveyenko A, Wroblewski K, Chao L, Wilpitz D,                 387(10019):679–690
    Hsu TW et al (2013) Reciprocal regulation of hepatic and adipose      39. Jani RH, Pai V, Jha P, Jariwala G, Mukhopadhyay S, Bhansali
    lipogenesis by liver X receptors in obesity and insulin resistance.       A et al (2014) A multicenter, prospective, randomized, double-
    Cell Metab 18(1):106–117                                                  blind study to evaluate the safety and efficacy of saroglitazar
21 Cave MC, Clair HB, Hardesty JE, Falkner KC, Feng W, Clark BJ et al         2 and 4 mg compared with placebo in type 2 diabetes mellitus
    (1859) 2016 Nuclear receptors and nonalcoholic fatty liver disease.       patients having hypertriglyceridemia not controlled with ator-
    Biochimica et Biophysica Acta (BBA) - Gene Reg Mech 9:1083–99             vastatin therapy (PRES. Diabetes Technol Ther 16(2):63–71
22. Cao Y, Shu X-B, Yao Z, Ji G, Zhang L (2020) Is vitamin D recep-       40. Gawrieh S, Noureddin M, Loo N, Mohseni R, Awasty V, Cusi
    tor a druggable target for non-alcoholic steatohepatitis? World J         K et al (2021) Saroglitazar, a PPAR-α/γ Agonist, for treatment
    Gastroenterol 26(38):5812–5821                                            of NAFLD: a randomized controlled double-blind phase 2 trial.
23. Watanabe A, Sohail MA, Gomes DA, Hashmi A, Nagata J, Sut-                 Hepatology 74(4):1809–1824
    terwala FS et al (2009) Inflammasome-mediated regulation of           41. Siddiqui MS, Idowu MO, Parmar D, Borg BB, Denham D, Loo
    hepatic stellate cells. Am J Physiol-Gastrointestinal Liver Physiol       NM et al (2021) A phase 2 double blinded, randomized con-
    296(6):G1248-57                                                           trolled trial of saroglitazar in patients with nonalcoholic stea-
24. Flannery C, Dufour S, Rabøl R, Shulman GI, Petersen KF (2012)             tohepatitis. Clin Gastroenterol Hepatol 19(12):2670–2672
    Skeletal muscle insulin resistance promotes increased hepatic         42. Stephen A Harrison, Vlad Ratziu, Pierre Bedossa, Jean-Fran-
    de novo lipogenesis, hyperlipidemia, and hepatic steatosis in the         cois Dufour, Frederik Kruger, Jörn M Schattenberg, Sven M.
    elderly. Diabetes 61(11):2711–2717                                        Francque, Marco Arrese, Jacob George, Elisabetta Bugianesi,
25. Brown MS, Goldstein JL (2008) Selective versus total insulin              Helena Cortez-Pinto, Quentin M. Anstee, Adrian C. Gadano,
    resistance: a pathogenic paradox. Cell Metab 7(2):95–96                   Manuel Romero-Gomez, DWH and AJS. RESOLVE-IT phase
26. Uyeda K, Repa JJ (2006) Carbohydrate response element binding             3 trial of Elafibranor in NASH: final results of the week 72
    protein, ChREBP, a transcription factor coupling hepatic glucose          interim surrogate efficacy analysis. 2022.
    utilization and lipid synthesis. Cell Metab 4(2):107–110              43. Francque SM, Bedossa P, Ratziu V, Anstee QM, Bugianesi
27. Nagai Y, Yonemitsu S, Erion DM, Iwasaki T, Stark R, Weismann              E, Sanyal AJ et al (2021) A randomized, controlled trial of
    D et al (2009) The role of peroxisome proliferator-activated recep-       the pan-PPAR agonist lanifibranor in NASH. N Engl J Med
    tor γ coactivator-1 β in the pathogenesis of fructose-induced insu-       385(17):1547–1558
    lin resistance. Cell Metab 9(3):252–264                               44. Nakajima A, Eguchi Y, Yoneda M, Imajo K, Tamaki N, Suganami
28. Bindesbøll C, Fan Q, Nørgaard RC, MacPherson L, Ruan H-B,                 H et al (2021) Randomised clinical trial: pemafibrate, a novel
    Wu J et al (2015) Liver X receptor regulates hepatic nuclear              selective peroxisome proliferator-activated receptor α modulator
    O-GlcNAc signaling and carbohydrate responsive element-bind-              (SPPARMα), versus placebo in patients with non-alcoholic fatty
    ing protein activity. J Lipid Res 56(4):771–785                           liver disease. Aliment Pharmacol Ther 54(10):1263–1277
29. Cuthbertson DJ, Steele T, Wilding JP, Halford JC, Harrold JA,         45. Caligiuri A, Gentilini A, Marra F (2016) Molecular pathogenesis
    Hamer M et al (2017) What have human experimental overfeeding             of NASH. Int J Mol Sci 17(9):1575
    studies taught us about adipose tissue expansion and susceptibility   46. Jiao Y, Lu Y, Li X (2015) Farnesoid X receptor: a master regulator
    to obesity and metabolic complications? Int J Obes 41(6):853–865          of hepatic triglyceride and glucose homeostasis. Acta Pharmacol
30. Loomba R, Friedman SL, Shulman GI (2021) Mechanisms and                   Sin 36(1):44–50
    disease consequences of nonalcoholic fatty liver disease. Cell        47 Panzitt K (1867) Wagner M 2021 FXR in liver physiology: multi-
    184(10):2537–2564                                                         ple faces to regulate liver metabolism. Biochimica et Biophysica
31. Richart C, Auguet T, Terra X (2010) Apolipoprotein C3 gene                Acta (BBA) - Mol Basis Disease 7:166133
    variants in nonalcoholic fatty liver disease. N Engl J Med            48. Watanabe M, Houten SM, Wang L, Moschetta A, Mangelsdorf DJ,
    363(2):193–195                                                            Heyman RA et al (2004) Bile acids lower triglyceride levels via
32. Petersen MC, Shulman GI (2018) Mechanisms of insulin action               a pathway involving FXR, SHP, and SREBP-1c. J Clin Investig
    and insulin resistance. Physiol Rev 98(4):2133–2223                       113(10):1408–1418
33. Belfort R, Harrison SA, Brown K, Darland C, Finch J, Har-             49. Savkur RS, Bramlett KS, Michael LF, Burris TP (2005) Regula-
    dies J et al (2006) A placebo-controlled trial of pioglitazone            tion of pyruvate dehydrogenase kinase expression by the farnesoid
    in subjects with nonalcoholic steatohepatitis. N Engl J Med               X receptor. Biochem Biophys Res Commun 329(1):391–396
    355(22):2297–2307                                                     50. Mudaliar S, Henry RR, Sanyal AJ, Morrow L, Marschall H-U, Kip-
34. Cusi K, Orsak B, Bril F, Lomonaco R, Hecht J, Ortiz-Lopez C               nes M et al (2013) Efficacy and safety of the farnesoid X receptor
    et al (2016) Long-term pioglitazone treatment for patients with           agonist obeticholic acid in patients with type 2 diabetes and nonal-
    nonalcoholic steatohepatitis and prediabetes or type 2 diabetes           coholic fatty liver disease. Gastroenterology 145(3):574-582.e1
    mellitus. Ann Intern Med 165(5):305                                   51. Neuschwander-Tetri BA, Loomba R, Sanyal AJ, Lavine JE, Van
35. Sanyal AJ, Chalasani N, Kowdley KV, McCullough A, Diehl AM,               Natta ML, Abdelmalek MF et al (2015) Farnesoid X nuclear
    Bass NM et al (2010) Pioglitazone, vitamin E, or placebo for              receptor ligand obeticholic acid for non-cirrhotic, non-alcoholic
    nonalcoholic steatohepatitis. N Engl J Med 362(18):1675–1685
13
Metabolic dysfunction–associated fatty liver disease (MAFLD): an update of the recent advances…                                                                 879
      steatohepatitis (FLINT): a multicentre, randomised, placebo-                63. Pedrosa M, Seyedkazemi S, Francque S, Sanyal A, Rinella M,
      controlled trial. The Lancet 385(9972):956–965                                  Charlton M et al (2020) A randomized, double-blind, multicenter,
52.   Younossi ZM, Ratziu V, Loomba R, Rinella M, Anstee QM,                          phase 2b study to evaluate the safety and efficacy of a combina-
      Goodman Z et al (2019) Obeticholic acid for the treatment of                    tion of tropifexor and cenicriviroc in patients with nonalcoholic
      non-alcoholic steatohepatitis: interim analysis from a multicen-                steatohepatitis and liver fibrosis: study design of the TANDEM
      tre, randomised, placebo-controlled phase 3 trial. The Lancet                   trial. Contemp Clin Trials 88:105889
      394(10215):2184–2196                                                        64. Di Lella S, Sundblad V, Cerliani JP, Guardia CM, Estrin DA,
53.   Quentin M. Anstee, Kathryn Jean Lucas, Sven M. Francque,                        Vasta GR et al (2011) When galectins recognize glycans: from
      Manal F. Abdelmalek, Arun J Sanyal, Vlad Ratziu, Adrian C.                      biochemistry to physiology and back again. Biochemistry
      Gadano, Mary E. Rinella, Michael R. Charlton, Rohit Loomba,                     50(37):7842–7857
      Edward Mena, George Boon Bee Goh, Jossy Kochuparampil Li                    65. Chalasani N, Abdelmalek MF, Garcia-Tsao G, Vuppalanchi R,
      Chen, Gerardo Rodrigue SL and MCP. Safety and efficacy of                       Alkhouri N, Rinella M et al (2020) Effects of belapectin, an
      tropifexor plus cenicriviroc combination therapy in adult patients              inhibitor of galectin-3, in patients with nonalcoholic steatohepa-
      with fibrotic NASH: 48 week results from the phase 2b tandem                    titis with cirrhosis and portal hypertension. Gastroenterology
      study. Hepatology. 2020 Nov 1;72(S1):1A-130A                                    158(5):1334-1345.e5
54.   Patel K, Harrison SA, Elkhashab M, Trotter JF, Herring R, Rojter            66 Harrison SA, Wong VW-S, Okanoue T, Bzowej N, Vuppa-
      SE et al (2020) Cilofexor, a Nonsteroidal FXR agonist, in patients              lanchi R, Younes Z et al (2020) Selonsertib for patients with
      with noncirrhotic NASH: a phase 2 randomized controlled trial.                  bridging fibrosis or compensated cirrhosis due to NASH:
      Hepatology 72(1):58–71                                                          results from randomized phase III STELLAR trials. J Hepatol
55.   Alkhouri N, Herring R, Kabler H, Kayali Z, Hassanein T, Kohli A et al           73(1):26–39
      (2022) Safety and efficacy of combination therapy with semaglutide,         67. Miyazaki M, Flowers MT, Sampath H, Chu K, Otzelberger C,
      cilofexor and firsocostat in patients with non-alcoholic steatohepatitis:       Liu X et al (2007) Hepatic stearoyl-CoA desaturase-1 deficiency
      a randomised, open-label phase II trial. J Hepatol 77(3):607–618                protects mice from carbohydrate-induced adiposity and hepatic
56.   Ratziu V, Rinella ME, Neuschwander-Tetri BA, Lawitz E, Denham                   steatosis. Cell Metab 6(6):484–496
      D, Kayali Z et al (2022) EDP-305 in patients with NASH: a phase             68. Iruarrizaga-Lejarreta M, Varela-Rey M, Fernández-Ramos D,
      II double-blind placebo-controlled dose-ranging study. J Hepatol                Martínez-Arranz I, Delgado TC, Simon J et al (2017) Role of
      76(3):506–517                                                                   aramchol in steatohepatitis and fibrosis in mice. Hepatol Commun
57.   Rohit Loomba, Kris V Kowdley, Raj Vuppalanchi, Tarek Has-                       1(9):911–927
      sanein, Sergio E. Rojter, Muhammad Y Sheikh, Sam Moussa,                    69. Ratziu V, de Guevara L, Safadi R, Poordad F, Fuster F, Flores-
      Diana Chung, Clarence Eng, Tonya Marmon, Mona Qureshi, Erin                     Figueroa J et al (2021) Aramchol in patients with nonalcoholic
      Quirk DC and EJL. Liver distributed FXR agonist TERN-101                        steatohepatitis: a randomized, double-blind, placebo-controlled
      demonstrates favorable safety and efficacy profile in NASH phase                phase 2b trial. Nat Med 27(10):1825–1835
      2a LIFT study. Hepatology. 2021 Oct 14;74(S1):1–156                         70. Harrison SA, Alkhouri N, Davison BA, Sanyal A, Edwards C,
58.   Talukdar S, Kharitonenkov A (2021) FGF19 and FGF21. In                          Colca JR et al (2020) Insulin sensitizer MSDC-0602K in non-
      NASH we trust Mol Metab 46:101152                                               alcoholic steatohepatitis: a randomized, double-blind, placebo-
59.   Harrison SA, Ruane PJ, Freilich BL, Neff G, Patil R, Behling                    controlled phase IIb study. J Hepatol 72(4):613–626
      CA et al (2021) Efruxifermin in non-alcoholic steatohepatitis: a            71. Harrison SA, Thang C, Bozec S, Bolze S, DeWitt S, Moller
      randomized, double-blind, placebo-controlled, phase 2a trial. Nat               DE PF (2022) PXL065 (deuterium-stabilized R-enantiomer
      Med 27(7):1262–1271                                                             of pioglitazone) reduces liver fat content and improves liver
60.   Harrison SA, Bashir MR, Guy CD, Zhou R, Moylan CA, Frias                        histology without PPARG -mediated side effects in patients
      JP et al (2019) Resmetirom (MGL-3196) for the treatment                         with NASH: analysis of a 36 week placebo-controlled phase
      of non-alcoholic steatohepatitis: a multicentre, randomised,                    2 trial (DESTINY1). [Internet]. Available from: https://www.
      double-blind, placebo-controlled, phase 2 trial. The Lancet                     aasld.o rg/t he-l iver-m eetin g/p xl065-d euter ium-s tabil ized-r-
      394(10213):2012–2024                                                            enantiomer-pioglitazone-reduces-liver-fat-content. Accessed
61.   Harrison S, Taub R, Neff G, Moussa S, Alkhouri N, Bashir M                      24 Mar 2023
      (2022) Primary data analyses of MAESTRO-NAFLD-1 a 52 week                   72. Dufour J-F, Caussy C, Loomba R (2020) Combination therapy for
      double-blind placebo-controlled phase 3 clinical trial of resmeti-              non-alcoholic steatohepatitis: rationale, opportunities and chal-
      rom in patients with NAFLD. J Hepatol 1(77):S14                                 lenges. Gut 69(10):1877–1884
62.   Lian B, Loomba R, Neutel J, Margaritescu C, Homer K, Luk A
      et al (2020) VK2809, a novel liver-directed thyroid receptor ago-           Publisher's note Springer Nature remains neutral with regard to
      nist, produces durable reductions in liver fat in patients with non-        jurisdictional claims in published maps and institutional affiliations.
      alcoholic fatty liver disease: results of 4-week follow-up assess-
      ment from a 12-week phase 2 randomized, placebo-controlled
      trial. J Hepatol 73:S53
13