ISSN 0006-2979, Biochemistry (Moscow), 2025, Vol. 90, No. 1, pp. 1-18 © Pleiades Publishing, Ltd., 2025.
Russian Text © The Author(s), 2025, published in Biokhimiya, 2025, Vol. 90, No. 1, pp. 3-21.
1
REVIEW
4-Methylumbelliferone, an Inhibitor
of Hyaluronan Synthase, Prevents the Development
of Oncological, Inflammatory, Degenerative,
and Autoimmune Diseases
Viktoriya V. Fedorova
1
, Alexandra Tsitrina
2
, Noreen Halimani
1
,
and Yuri V. Kotelevtsev
1,a
*
1
Skolkovo Institute of Science and Technology, 121205 Moscow, Russia
2
Ben-Gurion University of the Negev, 8410501 Be’er Sheva, Israel
a
e-mail: y.kotelevtsev@skoltech.ru
Received September 24, 2024
Revised November 24, 2024
Accepted December 8, 2024
AbstractHyaluronic acid (HA) is the main structure-forming polymer of the extracellular matrix. HA metab-
olism plays an important role in intercellular interaction in healthy organism and in various pathologies.
HA is synthesized by hyaluronan synthase (HAS); mammals have three highly homologous isoforms of this
enzyme: HAS1, HAS2, and HAS3. No highly specific competitive inhibitors of HASs have been described so
far. 4-Methylumbelliferone (4-MU), a natural coumarin compound, is commonly used to inhibit HA synthesis
in vivo and in cell cultures. The review is focused on the molecular mechanisms underlying the therapeutic
effects of 4-MU and discusses results of 4-MU application in tissue cultures and animal disease models, as well
as in first clinical trials of this compound. It was found that along with receptors and transcription factors,
one of the pharmacological targets of 4-MU is HAS2, which is most common isoform of HAS. Moreover, it is
inhibition of HA synthesis that underlies the pharmacological effects of 4-MU in oncological, autoimmune,
degenerative, and hypercompensated regenerative processes (fibrosis, scar formation). New clinical drugs
based on specific HAS2 inhibitors will be the first-in-class compounds to treat a wide range of diseases.
DOI: 10.1134/S0006297924603459
Keywords: 4-methylumbelliferone, Odeston, Hymecromone, hyaluronan synthase inhibition, hyaluronan synthase
Abbreviations: HA, hyaluronic acid; ECM, extracellular matrix; 4-MU, 4-methylumbelliferone; 4-MUG, 4-methylumbellif-
erone beta-D-glucuronide; HAS, hyaluronan synthase.
* To whom correspondence should be addressed.
“The most fruitful basis of the discovery of a new drug is to start with an old one”
Sir James Black, Nobel Prize Laureate 1988
INTRODUCTION
Development of new drugs in the post-genomic
era is based on detailed knowledge of signaling path-
ways and key effectors or pharmacological targets
(enzymes, receptors, and transcription factors). At the
same time, physiologically active substances still play
an important role in the identification and validation
of pharmacological targets. Numerous experimental
data have confirmed the therapeutic effect of the
natural coumarin compound 4-methylumbelliferone
(4-MU) in animal models of oncological, autoimmune,
degenerative, and hyperproliferative diseases. This
review is focused on the studies on the validation of
hyaluronan synthase (HAS) as the main 4-MU phar-
macological target, which is an essential step in the
development first-in-class drugs, namely, inhibitors of
hyaluronic acid (HA) synthesis.
FEDOROVA et al.2
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
THE ROLE OF EXTRACELLULAR MATRIX
IN NORMAL AND PATHOLOGICAL STATES
Extracellular matrix (ECM), which forms the basis
of connective tissue, is a highly organized interstitial
structure that ensures mechanical integrity and cell–
cell interaction.
ECM consists of polymeric carbohydrates glycos-
aminoglycans (GAGs), proteins (mainly, fibrillar), and
proteoglycans (PGs). ECM is a barrier and, at the same
time, a depot for peptide hormones and cytokines.
It also directly generates chemical and mechanical
signals essential for the maintenance of tissue homeo-
stasis. Pathological processes typical of many systemic
diseases lead to the ECM rearrangement and chang-
es in its structure, which eventually contributes to
changes in the tissue architecture and results in the
development of diseases, such as fibrosis, osteoarthri-
tis, and cancer [1, 2].
HA is a linear polymer consisting of D-glucuronic
acid and D-N-acetylglucosamine residues connected by
alternating β-1,4- and β-1,3-glycosidic bonds; it is the
main ECM component by weight. HA homeostasis is
maintained through the synthetic activity of HAS en-
zymes, decomposition by hyaluronidases, and chemical
degradation mainly via the action of reactive oxygen
species. There are three HAS isoforms. HAS1 is ac-
tive during embryogenesis. HAS2 is the main isoform
both in embryogenesis and in most tissues during the
postnatal period; it synthesizes high-molecular-weight
(HMW) HA fraction with a weight of 1000-6000 kDa;
HAS3 synthesizes low-molecular-weight (LMW) forms
of HA weighing less than 250  kDa. HMW HA usually
has the anti-inflammatory, antiangiogenic, and an-
ti-cancer properties. On the contrary, LMW fractions
of HA exhibit proinflammatory and proangiogenic ef-
fects and promote cell adhesion. Although these prop-
erties of HA have been well established, the mech-
anisms underlying them are poorly understood and
need further exploration [3].
Human body also has various types of hyaluro-
nidases (HYALs) that cleave HA. The most thoroughly
characterized of them are HYAL1 and HYAL2. HYAL2
degrades HA into the fragments approximately 50
monomers in length (~20kDa), while HYAL1 hydrolyz-
es HA into tetrasaccharides (~1600Da), which undergo
further degradation in the lysosomes [4]. Pathological
processes, such as disruption of HA metabolism, can-
cer, tissue damage, and inflammation, can change this
balance, thus increasing the concentration of LMW
HA. There is a large body of evidence indicating HA
involvement in the chronic inflammation character-
istic of type  2 diabetes, liver cirrhosis, asthma, and
cancer progression and metastasis. Thus, HA pro-
motes adhesion and motility of metastatic melanoma
cells  [5], enhances motility of pancreatic [6] and pros-
tate cancer cells [7], hinders drug delivery to tumors
[8-10], promotes drug resistance  [11], stimulates cell
division [12], and acts as an immune regulatory fac-
tor  [13]. Upregulated HA synthesis in the tumor stro-
ma is a negative prognostic factor [14-18].
The blood level of HA is a marker of liver fibro-
sis. In a fibrotic liver, HA is synthesized by fibroblasts
originated from activated stellate cells. Normally, stel-
late cells do not express HAS2 (the main enzyme that
produces HA in adult tissues) and do not synthesize
HA. Liver damage leads to the production of TGF-β,
which triggers transdifferentiation of stellate cells
into myofibroblasts and dramatically increases HAS2
expression in them [19]. HA accumulation in the pa-
renchyma results in the activation of Notch1 signaling
pathway in stellate cells, leading to their activation,
increased synthesis of the ECM, and development of
fibrosis [20]. Therefore, HAS2 and HAS3 are important
pharmacological targets in the treatment of diseases
associated with pathological activation of HA synthe-
sis, in particular, liver fibrosis.
Elucidation of molecular mechanisms of HA syn-
thesis by mammalian HASs has become important in
the context of the targeted search for their specific in-
hibitors that can be used as drugs. These mechanisms
have been discussed in most detail in the review by
DeAngelis and Zimmer [21]. Within a few years af-
ter the discovery of bacterial HAS in Streptococcus
pyogenes (SpHAS), three isoforms of vertebrate en-
zyme (HAS1-3) and viral HAS (CvHAS of Paramecium
bursaria Chlorella virus-1, PBCV-1) have been identi-
fied. CvHAS is similar to the vertebrate enzymes in
the overall architecture of the cytoplasmic domain
containing the active site and transmembrane (TM)
domains, with two TM helices at the N-terminus and
four at the C-terminus. All these enzymes belong to
class I glycosyltransferases, but vertebrate HASs and
viral CvHAS add sugars to the nonreducing end of the
growing HA chain (Fig.1), whereas SpHAS adds sugars
to the reducing end.
Vertebrate HASs, CvHAS, and SpHAS have the
glycosyltransferase domain of the second type (GT-2),
which allows to incorporate both uridine 5′-diphos-
phoglucuronic acid (UDP-GlcA) and uridine 5′-disphos-
phate N-acetylglucosamine (UDP-GlcNAc). The three-di-
mensional structure of CvHAS has been determined by
electron cryomicroscopy [22]. 4-MU, which significant-
ly reduces expression of HAS2/HAS3 [1], is a widely
used and the only well-characterized inhibitor of HA
synthesis that is known under commercial names of
Hymecromone and Odeston. It has been approved for
the clinical application in Europe and Asia and is rou-
tinely used as a hepatoprotector, antispasmodic, and
choleretic in biliary dyskinesia. In Italy, this drug has
been approved by the Italian Medicines Agency (AIC
no.02130002) and is sold under the name Cantabiline.
THERAPEUTIC PROPERTIES OF 4-METHYLUMBELLIFERONE 3
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
Fig.  1. HA synthesis by HAS enzymes. UDP-GlcNAc, uridine diphosphate N-acetylglucosamine; GlcA, glucuronic acid.
Fig.  2. Proposed mechanism of 4-MU effect on HA synthesis. a) Normal pathway of HA synthesis. b) UDP substitution by
4-MU resulting in the suppression of HA synthesis by HAS (from Nagyetal. [26]).
THE MECHANISM OF 4-MU ACTION
ON HYALURONIC ACID SYNTHESIS
There is no evidence of competitive inhibition or
even direct interaction of 4-MU with HAS. 4-MU does
not affect the enzymatic activity of the solubilized
HAS [23]. The most common hypothesis is that 4-MU
acts a competitive substrate for uridine 5′-diphos-
phate-glucuronosyltransferase (UGT), thus depleting
the cellular pool of uridine 5′-UDP-GlcA utilized in HA
synthesis [24-26] (Fig. 2).
However, this hypothesis has not been con-
firmed experimentally. As an evidence against it, it
was shown that 4-MU does not affect the synthesis
of other glycosaminoglycans, which utilizes the same
monomers as the synthesis of HA. In addition, couma-
rins with alkylated 7-hydroxy group, which cannot be
the substrates for UGT, were still found to inhibit HA
synthesis with a high efficiency in  vitro [27]. 4-MU has
been shown to reduce the expression level of HAS2
mRNA  [25,  28,  29] and simultaneously upregulate ex-
pression of Hyal1 gene [30]. It also reduced the levels
of phosphorylase and uridine 5′-diphosphate glucose
dehydrogenase [31]. It still remains unknown how HA
synthesis is regulated at the transcriptional level and
Fig.  3. Mechanisms of HA synthesis inhibition by 4-MU:
competition with HA precursor UDP-GlcA; inhibition of HAS2
gene expression; indirect inhibition of HASs (from Vitale
et al. [1]).
FEDOROVA et al.4
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
whether involved mechanisms are selective for these
particular mRNAs (Fig. 3).
As has been shown by our group and other re-
searchers [32,  33], 4-MU has multiple targets not di-
rectly related to HA metabolism. It is possible that a
decreased HA accumulation observed in  vitro studies
is the cumulative effect of several parallel processes,
including possible substrate depletion, downregula-
tion of HAS2 expression (as experimentally demon-
strated), and activation of Hyal1 expression [30]. HAS2
expression is also known to be regulated by nuclear
receptors, in particular, glucocorticoid receptor. Thus,
expression of HAS2 was almost completely suppressed
by dexamethasone [34]. Cells exposed to 4-MU demon-
strated alterations in the cell cycle and p53 signaling
cascade [35, 36].
EFFECT OF 4-MU ON VARIOUS TYPES
OF CANCER AND AUTOIMMUNE
AND INFLAMMATORY PROCESSES
4-MU multiple processes associated with tumor
progression, such as migration, proliferation, and in-
vasion of cancer cell and angiogenesis, as well as in-
fluences cells of tumor microenvironment (fibroblasts,
endothelial and immune cells). Cancer development
involves rapid changes in the structure and composi-
tion of the ECM (whose main component is HA), which
requires creation of new drugs capable of changing
the properties of the ECM. This approach is promis-
ing in the treatment of various types of cancer, and
4-MU has already been approved for the use in this
new capacity.
Using a mouse model of carbon tetrachloride-in-
duced liver fibrosis, we demonstrated that formation
of collagen fibers is preceded by HA synthesis along
the boundaries of liver lobules. 4-MU inhibited HA
synthesis and significantly decreased formation of col-
lagen fibers around hepatic lobules [30]. In our recent
study, siRNA-mediated knockdown of the HAS2 gene
reproduced the effect of 4-MU on several signaling
pathways and transcription of some key genes, result-
ing in suppression of liver fibrosis [37].
The use 4-MU in the treatment of brain cancer is
especially interesting. The ECM of malignant gliomas
and glioblastomas is characterized by an increased HA
content; HA stimulates adhesive and invasive process-
es of tumor cells  [38]. 4-MU is a small molecule ca-
pable of passing through the blood-brain barrier and
inhibiting the synthesis of HA, which has promoted
studies on its possible application in the treatment
for gliomas and glioblastomas. Thus, in mouse mod-
els, high doses of 4-MU reduced HA synthesis, reduced
proliferation and migration of glioblastoma cells, and
stimulated their apoptosis [39-41]. As shown in in  vitro
and in vivo experiments, 4-MU reduced prolifera-
tion of glioma cells by regulating autophagy [42].
Chistyakov et al. [43] demonstrated that 4-MU inhibited
the inflammatory response of astrocytes. Oral adminis-
tration of 4-MU in mice caused a significant decrease
in the HA content in the spinal cord and brain, re-
duction in synaptic stability, and reactivation of neu-
roplasticity, which resulted in improved memory [44].
The data on preclinical studies on 4-MU applica-
tion for the treatment of various diseases are given
in Table 1.
THE PROSPECTS OF COMBINED THERAPY
WITH 4-MU. THE EFFECT OF 4-MU
ON THE TUMOR PHYSICAL BARRIER.
THE USE OF 4-MU AS A HYALURONIC ACID
SYNTHESIS INHIBITOR
HA-rich ECM forms a biological barrier of the
tumor microenvironment. This barrier regulates the
activity of immune effectors  [13,  98], prevents drug
diffusion  [99], hinders the adsorption of transgenic
vectors in gene therapy  [100], and plays an import-
ant role in the acquisition of resistance to anticancer
drugs [1,  11,  101].
The possibility of changing the properties of tu-
mor microenvironment in order to improve the result
of antitumor therapy has been actively investigated.
The pathological tumor microenvironment is charac-
terized by hypoxia and high interstitial fluid pressure,
leading to tumor progression and resistance to treat-
ment [102]. Increased interstitial pressure is consid-
ered to be the most important barrier for efficient
drug distribution within the tumor. The reasons for
the increased interstitial pressure in the tumor are
numerous and include extensive intratumor vascular
network, insufficient development of lymphatic ves-
sels, changes in the ECM components, and pressure
created by constantly dividing tumor cells [103, 104].
An increased HA content in tissues surrounding the
tumor contributes to the increase in the ECM volume
and, as a result, increases in the pressure inside the
tumor [105, 106]. Such high HA content in the tumor
microenvironment forms a physical barrier that re-
stricts the access of monoclonal antibodies and immune
cells to the tumor tissue, which is one of the mech-
anisms of tumor resistance to immunotherapy [107].
Due to the ability to inhibit the synthesis of
HA, 4-MU was suggested for the adjuvant therapy in
combination with the primary anticancer therapy.
Using various models, it has been shown that the use
of 4-MU in a combined therapy for various types of
cancer increased the treatment efficacy, reduced the
toxicity of antitumor drugs, and helped to overcome
emerging chemoresistance (Table 2).
THERAPEUTIC PROPERTIES OF 4-METHYLUMBELLIFERONE 5
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
Table  1. Preclinical studies on the application of 4-MU in the treatment of various diseases
Organ/system Studied disease Year Type of investigation Reference
Inflammation
acute respiratory distress syndrome (ARDS)
2013 in vitro [45]
2015 in vitro [46]
allergic inflammation 2022 in vitro [47]
allergic rhinitis 2022 in vitro/in vivo [48]
inflammation 2022 in vitro [49]
Head and neck oral squamous cell carcinomas 2022 in vitro [50]
Bile ducts
biliary dyskinesia 1984 in vivo [51]
biliary colic 1995 in vivo [52]
Immune response
Graves’ orbitopathy 2020 in vitro [53]
transplant rejection 2021 in vitro/in vivo [54]
autoimmune response to transplanted
islets of Langerhans
2020 in vitro/in vivo [55]
acute lung allograft rejection 2021 in vitro/in vivo [56]
Bone marrow chronic myeloid leukemia
2013 in vitro [57]
2016 in vitro [58]
2017 in vitro [59]
Lungs
pleural mesothelioma 2017 in vitro/in vivo [60]
pulmonary fibrosis,
pulmonary hypertension
2017 in vivo [61]
Mammary glands breast cancer
2019 in vitro [62]
2022 in vitro [63]
Bladder bladder cancer 2017 in vitro/in vivo [64]
Peripheral
nervous system
malignant peripheral nerve sheath tumor 2017 in vitro/in vivo [65]
Liver
hepatocellular carcinoma
2012 in vitro/in vivo [66]
2015 in vitro/in vivo [67]
2019 in vitro/in vivo [29]
2021 in vitro/in vivo [68]
2022 in vitro/in vivo [69]
liver metastasis of malignant melanoma 2005 in vitro/in vivo [70]
liver fibrosis 2019 in vivo [30]
steatohepatitis 2021 in vitro/in vivo [71]
FEDOROVA et al.6
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
Table 1 (cont.)
Organ/system Studied disease Year Type of investigation Reference
Pancreas
pancreatic cancer
2006 in vitro/in vivo [72]
2016 in vitro/in vivo [73]
2017 in vitro/in vivo [74]
2018 in vitro/in vivo [75, 76]
pancreatic ductal adenocarcinoma 2019 in vitro [77, 78]
Kidneys
renal cell carcinoma 2013 in vitro [79]
kidney ischemia-reperfusion injury 2013 in vivo [80]
metastatic renal cell carcinoma 2020 in vitro [81]
diabetic nephropathy 2021 in vivo [82]
advanced renal cell carcinoma 2022 in vitro/in vivo [83]
Prostate prostate cancer
2010 in vitro [84]
2015 in vitro/in vivo [85]
Connective tissue fibrosarcoma
2017 in vitro [86]
2019 in vitro [87]
2020 in vitro [88]
2021 in vitro [89]
Large intestine colorectal carcinoma 2015 in vitro/in vivo [90]
Central nervous
system
glioblastoma
2021 in vitro [39, 40]
2022 in vitro/in vivo [91]
Endometrium endometriosis
2016 in vitro/in vivo [92]
2020 in vitro [93]
2023 in vivo [94]
Ovaries ovarian cancer
2014 in vitro [95]
2019 in vitro/
in vivo [96]
2020 in vitro [97]
Table 2. Preclinical studies on the treatment of various types of cancer using combinations of anticancer drugs
and 4-MU
Type of cancer Main treatment Type of study Year Reference
Hepatocellular
carcinoma
immunotherapy:
IL-12-encoding adenovirus (AdIL-12)
in vitro 2018 [111]
Glioblastoma temozolomide in vitro 2023 [41]
THERAPEUTIC PROPERTIES OF 4-METHYLUMBELLIFERONE 7
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
Table 2 (cont.)
Type of cancer Main treatment Type of study Year Reference
Malignant pleural
mesothelioma
trametinib in vitro/in vivo 2017 [60]
Colorectal
carcinoma
cyclophosphamide
with immunotherapy (AdIL-12)
in vitro/in vivo 2015 [90]
Melanoma vemurafenib in vitro 2021 [109]
Esophageal squa-
mous cell carcinoma
dichloroacetic acid in vitro/in vivo 2019 [108]
Oral squamous cell
carcinoma
radiotherapy in vitro 2022 [50]
Renal cell
carcinoma
sorafenib in vitro 2013 [79]
Advanced renal cell
carcinoma
sorafenib in vitro/in vivo 2022 [83]
Pancreatic cancer 5-fluorouracil in vitro/in vivo 2018 [76]
Pancreatic cancer gemcitabine in vitro/in vivo 2006 [72]
Ovarian cancer carboplatin in vitro/in vivo 2019 [96]
Bladder urothelial
carcinoma
cisplatin or doxorubicin in vivo 2019 [110]
Fibrosarcoma radiotherapy
in vitro 2021 [89]
in vitro 2019 [87]
in vitro 2017 [86]
Chronic myeloid
leukemia
imatinib in vitro 2017 [59]
Chronic myeloid
leukemia
doxorubicin in vitro 2016 [58]
According to the data on the use of 4-MU as an
addition to the main therapy, 4-MU enhanced the ra-
diosensitivity of radiation-resistant cells in oral squa-
mous cell carcinoma [50] and fibrosarcoma [86-89].
Inrenal cell carcinoma, sorafenib in combination with
4-MU inhibited more efficiently proliferation and in-
vasion of cancer cells, suppressed capillary formation,
and induces apoptosis of tumor and endothelial cells
[79, 83]. 4-MU increased the efficacy of 5-fluorouracil
[68] and gemcitabine [72] against pancreatic cancer,
inhibited cell proliferation, and decreased the size of
primary tumors and metastases, as well as promot-
ed survival of affected animals. 4-MU increased the
sensitivity of glioblastoma cells to temozolomide by
enhancing the cytotoxic effect of the drug [41]. 4-MU
exacerbated the cytotoxic effect of carboplatin on che-
moresistant ovarian cancer cells [96]. A combined use
of dichloroacetate and 4-MU in a model of esopha-
geal squamous cell carcinoma promoted apoptosis of
cancer cells and inhibited tumor growth [108]. 4-MU
increased the sensitivity of myeloid leukemia cells to
doxorubicin  [58] and promoted their senescence  [59].
A combination of vemurafenib with 4-MU reduced the
survival of melanoma cells more efficiently compared
to vemurafenib monotherapy [109]. 4-MU enhanced
the chemosensitivity of bladder urothelial carcinoma
cells to doxorubicin and cisplatin  [110]. 4-MU signifi-
cantly reduced the interstitial tumor pressure and im-
proved perfusion, thus ensuring more efficient expres-
sion of the adenovirus transgene in the IL-12 (AdIL-12)
immunotherapy of colorectal cancer [90]. In a liver
cancer model, a combination of 4-MU with AdIL-12
FEDOROVA et al.8
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
led to a more pronounced inhibition of tumor growth
and increased survival of mice compared to AdIL-12
monotherapy [111].
THE USE OF 4-MU AS A HEPATOPROTECTOR
AND CHOLESTATIC AGENT TO REDUCE
THE HEPATOTOXICITY OF PRIMARY THERAPY
Immune checkpoint inhibitors, cytokines, and
antibodies against these proteins are used as immu-
nomodulators to enhance the body immune response
to tumors and chronic inflammation foci in rheuma-
toid, autoimmune, and inflammatory diseases [112,
113]. These drugs have successfully passed clinical
trials and have been approved for the use in clinical
practice by the European and American drug agen-
cies [114]. However, up to 17% patients receiving such
immunotherapy suffer from complications associated
with the damage of liver and bile duct [115-119].
Depending on the severity of complications, the
treatment for hepatotoxicity might include cessation
of therapy with immune checkpoint inhibitors. Cor-
ticosteroids and immunosuppression (in more se-
vere cases) can be recommended as well [120-123].
Ursodeoxycholic acid (UDCA) is used to improve the
liver function in the case of cholestatic hepatotoxicity,
when corticosteroids are ineffective [124-127]. UDCA
has the hepatoprotective and choleretic effects and
is considered as the treatment standard for choles-
tatic liver diseases with the autoimmune component
(primary biliary cholangitis, primary sclerosing chol-
angitis) [128-130]. To our knowledge, there are no
reports on the effect of 4-MU on the risk of hepato-
toxicity development in response to immunotherapy.
However, its established cholestatic and hepatoprotec-
tive properties make 4-MU a promising agent for such
studies.
The data accumulated strongly suggest the need
for the clinical trials of 4-MU as an agent for the adju-
vant/additional antitumor therapy that would reduce
the HA content, modify the ECM and tumor micro-
environment, decrease interstitial pressure, improve
tumor perfusion, facilitate drug access, and produce
hepatoprotective and cholestatic effects, thus decreas-
ing the risks of hepatotoxicity during immunotherapy.
TOPICAL APPLICATION OF 4-MU
TO PREVENT FORMATION OF STRETCH MARKS,
SCARS, KELOID SCARS, SUNBURNS,
AND HYPOPIGMENTATION FOCI
Topical application of 4-MU leads to efficient in-
hibition of HA synthesis in the skin [131]. 4-MU has
been shown to prevent keratinocyte activation and to
reduce epidermal hyperproliferation [132] and migra-
tion rate of keloid keratinocytes, thus decreasing the
likelihood of keloid scar formation [133].
4-MU enhances the processes of melanogenesis,
which makes it a promising agent in the treatment
of skin conditions associated with hypopigmenta-
tion, as well as a cosmetic product to provide natural
tan [134].
METABOLISM OF 4-MU.
TOXICITY AND SAFETY FOR HUMANS
Like all coumarins, 4-MU is poorly soluble in wa-
ter. It is a nonpolar molecule and therefore, can easily
pass through the lipid barrier in the intestine. It is
almost completely absorbed upon oral administration
and is excreted in urine and bile [26]. The methyl
group at position 4 ensures low toxicity of 4-MU by
preventing its metabolism to coumarin 3,4-epoxide
by cytochrome P450, as well as weak anticoagulation
properties compared to other coumarins, such as
dicoumarol and warfarin [135].
When ingested, 4-MU is very rapidly and almost
completely metabolized to 4-methylumbelliferone be-
ta-D-glucuronide (4-MUG) in the liver and small intes-
tine, which until recently, has limited its use in the
treatment of bile ducts only [1,  136-138]. Less than 3%
of orally administered 4-MU remains unchanged at
the systemic level, while intravenous administration
of 4-MU provides 10-30 times higher concentration of
this compound in the blood [26,  139]. The half-life of
orally administered 4-MU is only 28 min for humans
and 3 min for mice [140,  141]. At the same time, the
median concentration of 4-MUG in the plasma is more
than 3000 times higher than the concentration of
4-MU [26,  141], i.e., most of 4-MU is present as 4-MUG
in a body. However, despite its low bioavailability and
short half-life, orally taken 4-MU efficiently inhibits
HA synthesis. 4-MUG was proven to be as efficient as
4-MU in inhibiting HA synthesis; moreover, it is hydro-
lyzed back to 4-MU inside the cells [137]. Therefore,
to evaluate the pharmacodynamics of 4-MU, it is nec-
essary to take into account the effect of its metabo-
lite 4-MUG. These data suggest that 4-MU can be used
for the treatment of diseases beyond the biliary tract.
For example, as a small nonpolar molecule, 4-MU is
able to cross the blood-brain barrier and inhibit pro-
liferation of glioma cell [42].
A typical regimen of 4-MU administration for an
adult is 900-2400 mg/day [26]. No mutagenic or geno-
toxic effects of 4-MU have been found [1,  142,  143].
Clinical trials in patients with chronic hepatitis  B
and C (NCT00225537), healthy individuals, and pa-
tients with respiratory diseases (NCT02780752) [144]
have proven the safety of 4-MU (see Table 3).
THERAPEUTIC PROPERTIES OF 4-METHYLUMBELLIFERONE 9
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
Table 3. Clinical trials on the use of 4-MU in the treatment of various diseases
Disease (study) Status Year
Reference/
Identifier clinicaltrials.gov
Interstitial lung diseases
(SOLID Study)
phase II;
recruitment of participants
has not started
2024 NCT06325696
Primary sclerosing cholangitis
phase II;
participants are being recruited
2022 NCT05295680
COVID-19 no information available 2022 NCT05386420
Pulmonary hypertension,
including interstitial lung diseases
(SATURN Study)
phase II; completed 2021 NCT05128929
Healthy participants; study
of 4-MU effect on HA synthesis
phase I; completed 2016 [144]/ NCT02780752
Biliary sludge stage 2 no information available 2016 [145]
Chronic hepatitis C virus
and hepatitis B virus
no information available 2005 NCT00225537
Biliary dyskinesia no information available 2005 [146]
Biliary dyskinesia no information available 2001 [147]
Biliary dyskinesia no information available 1995 [52]
Study of 4-MU bioavailability no information available 1993 [141]
Symptoms after bile duct surgery no information available 1988 [148]
Biliary dyskinesia after
cholecystectomy
no information available 1984 [51]
CONCLUSION
Despite numerous experimental studies demon-
strating the efficacy of 4-MU in various animal mod-
els of oncological, immune, and degenerative diseases,
the molecular mechanisms of its action remain hypo-
thetical. It was demonstrated (at least in the model of
liver fibrosis) that the knockdown of the gene encod-
ing HAS2 led not only to the suppression of fibrosis,
but also to changes in the transcriptome that were
similar to those observed upon oral administration of
4-MU [37]. At the same time, it cannot be excluded
that some of effects of 4-MU may be independent of
the HA synthesis inhibition [62,  149]. 4-MU may also
act through different mechanisms depending on the
type of cancer. However, taken together, the data on
the effectiveness of 4-MU prove the need for a detailed
study of its pharmacokinetic and pharmacodynamic
properties to develop the treatment regimen (admin-
istration route, doses affecting 4-MU bioavailability, in-
tervals between doses, and administration schedule).
The first toxicology studies have already been con-
ducted in phase I clinical trials [144], which allowed
to proceed to clinical studies of the drug effectiveness
(phaseIIa). This is a worldwide trend. Thus, it is cur-
rently planned to conduct clinical trials on the use
of 4-MU in the treatment of interstitial lung diseases
and cholangitis (see Table3).
The clinical trials of 4-MU include selection of
appropriate doses for particular pathologies and in-
vestigation of metabolite excretion rates and drug bio-
availability. Another important factor is development
of new dosage forms (for example, 4-MU-containing
nanoparticles) that will not only increase 4-MU bio-
availability, but will also lead to the patent protection
of a new drug.
Finally, if HAS is indeed the main pharmacologi-
cal target of 4-MU, development of new chemical com-
pounds using 3D models of HAS2/HAS3 and docking
of potential ligands with the help of artificial intelli-
gence will inevitably result in the creation of original,
first-in-class targeted drugs based on HAS inhibitors.
FEDOROVA et al.10
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
Contributions. Yu.V.K. participated in the devel-
opment of review concept and selection of articles and
supervised experimental work of the authors present-
ed in this review; A.C. and N.Kh. provided key con-
clusions from their experimental articles, edited the
manuscript, and participated in its discussion; V.V.F.
conducted literature search and analysis and wrote
the original version of the manuscript.
Ethics approval and consent to participate.
This work does not contain any studies involving hu-
man and animal subjects.
Conflict of interest. The authors of this work de-
clare that they have no conflicts of interest.
REFERENCES
1. Vitale, D. L., Icardi, A., Rosales, P., Spinelli, F. M.,
Sevic,I., and Alaniz, L.D. (2021) Targeting the tumor
extracellular matrix by the natural molecule 4-meth-
ylumbelliferone: a complementary and alternative
cancer therapeutic strategy, Front. Oncol., 11, 710061,
https://doi.org/10.3389/fonc.2021.710061.
2. Rosales,P., Vitale,D., Icardi,A., Sevic,I., and Alaniz,L.
(2024) Role of Hyaluronic acid and its chemical de-
rivatives in immunity during homeostasis, cancer
and tissue regeneration, Semin. Immunopathol., 46,
15, https://doi.org/10.1007/s00281-024-01024-7.
3. Tavianatou, A.G., Caon,I., Franchi,M., Piperigkou,Z.,
Galesso,D., and Karamanos, N.K. (2019) Hyaluronan:
molecular size-dependent signaling and biological
functions in inflammation and cancer, FEBS J., 286,
2883-2908, https://doi.org/10.1111/febs.14777.
4. Jung,H. (2020) Hyaluronidase: an overview of its prop-
erties, applications, and side effects, Arch. Plast. Surg.,
47, 297-300, https://doi.org/10.5999/aps.2020.00752.
5. Kudo,D., Kon,A., Yoshihara,S., Kakizaki,I., Sasaki,M.,
Endo,M., and Takagaki,K. (2004) Effect of a hyaluro-
nan synthase suppressor, 4-methylumbelliferone, on
B16F-10 melanoma cell adhesion and locomotion,
Biochem. Biophys. Res. Commun., 321, 783-787, https://
doi.org/10.1016/j.bbrc.2004.07.041.
6. Cheng, X.B., Kohi,S., Koga,A., Hirata,K., and Sato,N.
(2016) Hyaluronan stimulates pancreatic cancer
cell motility, Oncotarget, 7, 4829-4840, https://doi.
org/10.18632/oncotarget.6617.
7. Ricciardelli, C., Russell, D.L., Ween, M.P., Mayne,K.,
Suwiwat,S., Byers,S., Marshall, V.R., Tilley, W.D., and
Horsfall, D. J. (2007) Formation of hyaluronan- and
versican-rich pericellular matrix by prostate cancer
cells promotes cell motility, J.Biol. Chem., 282, 10814-
10825, https://doi.org/10.1074/jbc.M606991200.
8. Jacobetz, M. A., Chan, D.S., Neesse,A., Bapiro, T.E.,
Cook, N., Frese, K. K., Feig, C., Nakagawa, T., Cald-
well, M. E., Zecchini, H. I., Lolkema, M. P., Jiang, P.,
Kultti,A., Thompson, C.B., Maneval, D.C., Jodrell,D.I.,
Frost,G.I., Shepard, H.M., Skepper, J.N., and Tuveson,
D. A. (2013) Hyaluronan impairs vascular function
and drug delivery in a mouse model of pancreat-
ic cancer, Gut, 62, 112-120, https://doi.org/10.1136/
gutjnl-2012-302529.
9. Misra,S., Ghatak,S., Zoltan-Jones,A., and Toole, B.P.
(2003) Regulation of multidrug resistance in cancer
cells by hyaluronan, J.Biol. Chem., 278, 25285-25288,
https://doi.org/10.1074/jbc.C300173200.
10. Toole, B.P., and Slomiany, M.G. (2008) Hyaluronan:
aconstitutive regulator of chemoresistance and ma-
lignancy in cancer cells, Semin. Cancer Biol., 18, 244-
250, https://doi.org/10.1016/j.semcancer.2008.03.009.
11. Vitale, D. L., Spinelli, F. M., Del Dago, D., Icardi, A.,
Demarchi,G., Caon, I., García,M., Bolontrade, M.F.,
Passi, A., Cristina, C., and Alaniz, L. (2018) Co-treat-
ment of tumor cells with hyaluronan plus doxorubi-
cin affects endothelial cell behavior independently of
VEGF expression, Oncotarget, 9, 36585-36602, https://
doi.org/10.18632/oncotarget.26379.
12. Kultti, A., Zhao, C., Singha, N. C., Zimmerman, S.,
Osgood, R.J., Symons,R., Jiang,P., Li,X., Thompson,
C. B., Infante, J. R., Jacobetz, M. A., Tuveson, D. A.,
Frost, G.I., Shepard, H.M., and Huang,Z. (2014) Accu-
mulation of extracellular hyaluronan by hyaluronan
synthase3 promotes tumor growth and modulates the
pancreatic cancer microenvironment, Biomed. Res.
Int., 2014, 817613, https://doi.org/10.1155/2014/817613.
13. McBride, W. H., and Bard, J. B. (1979) Hyaluroni-
dase-sensitive halos around adherent cells. Their
role in blocking lymphocyte-mediated cytolysis,
J. Exp. Med., 149, 507-515, https://doi.org/10.1084/
jem.149.2.507.
14. Lipponen, P., Aaltomaa, S., Tammi, R., Tammi, M.,
Agren, U., and Kosma, V. M. (2001) High stromal
hyaluronan level is associated with poor differ-
entiation and metastasis in prostate cancer, Eur. J.
Cancer., 37, 849-856, https://doi.org/10.1016/s0959-
8049(00)00448-2.
15. Setälä, L. P., Tammi, M. I., Tammi, R. H., Eskelinen,
M.J., Lipponen, P.K., Agren, U.M., Parkkinen,J., Alha-
va, E.M., and Kosma, V.M. (1999) Hyaluronan expres-
sion in gastric cancer cells is associated with local and
nodal spread and reduced survival rate, Br.J. Cancer.,
79, 1133-1138, https://doi.org/10.1038/sj.bjc.6690180.
16. Anttila, M. A., Tammi, R. H., Tammi, MI., Syrjänen,
K.J., Saarikoski, S.V., and Kosma, V.M. (2000) High
levels of stromal hyaluronan predict poor disease
outcome in epithelial ovarian cancer, Cancer Res., 60,
150-155.
17. Bharadwaj, A.G., Kovar, J.L., Loughman,E., Elowsky,C.,
Oakley, G. G., and Simpson, M. A. (2009) Sponta-
neous metastasis of prostate cancer is promoted by
excess hyaluronan synthesis and processing, Am. J.
Pathol., 174, 1027-1036, https://doi.org/10.2353/
ajpath.2009.080501.
THERAPEUTIC PROPERTIES OF 4-METHYLUMBELLIFERONE 11
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
18. Sironen, R.K., Tammi,M., Tammi, R., Auvinen, P. K.,
Anttila, M., and Kosma, V. M. (2011) Hyaluronan in
human malignancies, Exp. Cell Res., 317, 383-391,
https://doi.org/10.1016/j.yexcr.2010.11.017.
19. Kim,J., and Seki,E. (2023) Hyaluronan in liver fibro-
sis: basic mechanisms, clinical implications, and ther-
apeutic targets, Hepatol. Commun., 7, e0083, https://
doi.org/10.1097/HC9.0000000000000083.
20. Yang, Y. M., Noureddin, M., Liu, C., Ohashi, K., Kim,
S. Y., Ramnath, D., Powell, E. E., Sweet, M. J., Roh,
Y.S., Hsin, I. F., Deng, N., Liu,Z., Liang, J., Mena, E.,
Shouhed,D., Schwabe, R.F., Jiang,D., Lu, S.C., Noble,
P.W., and Seki,E. (2019) Hyaluronan synthase 2-medi-
ated hyaluronan production mediates Notch1 activa-
tion and liver fibrosis, Sci. Transl. Med., 11, eaat9284,
https://doi.org/10.1126/scitranslmed.aat9284.
21. DeAngelis, P. L., and Zimmer, J. (2023) Hyaluronan
synthases; mechanisms, myths, and mysteries of
three types of unique bifunctional glycosyltransferas-
es, Glycobiology, 33, 1117-1127, https://doi.org/10.1093/
glycob/cwad075.
22. Maloney, F.P., Kuklewicz,J., Corey, R.A., Bi,Y., Ho,R.,
Mateusiak,L., Pardon,E., Steyaert, J., Stansfeld, P.J.,
and Zimmer,J. (2022) Structure, substrate recognition
and initiation of hyaluronan synthase, Nature, 604,
195-201, https://doi.org/10.1038/s41586-022-04534-2.
23. Nakamura,T., Funahashi, M., Takagaki,K., Munaka-
ta,H., Tanaka,K., Saito,Y., and Endo,M. (1997) Effect
of 4-methylumbelliferone on cell-free synthesis of
hyaluronic acid, Biochem. Mol. Biol. Int., 43, 263-268,
https://doi.org/10.1080/15216549700204041.
24. Kakizaki, I., Kojima, K., Takagaki, K., Endo, M.,
Kannagi, R., Ito, M., Maruo, Y., Sato, H., Yasuda, T.,
Mita, S., Kimata, K., and Itano, N. (2004) A novel
mechanism for the inhibition of hyaluronan biosyn-
thesis by 4-methylumbelliferone, J. Biol. Chem., 279,
33281-33289, https://doi.org/10.1074/jbc.M405918200.
25. Kultti, A., Pasonen-Seppänen, S., Jauhiainen, M.,
Rilla, KJ., Kärnä, R., Pyöriä, E., Tammi, R. H., and
Tammi, M. I. (2009) 4-Methylumbelliferone inhib-
its hyaluronan synthesis by depletion of cellular
UDP-glucuronic acid and downregulation of hyaluro-
nan synthase2 and3, Exp. Cell Res., 315, 1914-1923,
https://doi.org/10.1016/j.yexcr.2009.03.002.
26. Nagy,N., Kuipers, H.F., Frymoyer, A.R., Ishak, H. D.,
Bollyky, J. B., Wight, T. N., and Bollyky, P. L. (2015)
4-methylumbelliferone treatment and hyaluronan
inhibition as a therapeutic strategy in inflammation,
autoimmunity, and cancer, Front. Immunol., 6, 123,
https://doi.org/10.3389/fimmu.2015.00123.
27. Tsitrina, A. A., Krasylov, I. V., Maltsev, D. I.,
Andreichenko, I.N., Moskvina, V. S., Ivankov, D.N.,
Bulgakova, E.V., Nesterchuk, M., Shashkovskaya, V.,
Dashenkova, N. O., Khilya, V. P., Mikaelyan, A., and
Kotelevtsev,Y. (2021) Inhibition of hyaluronan secre-
tion by novel coumarin compounds and chitin syn-
thesis inhibitors, Glycobiology, 31, 959-974, https://
doi.org/10.1093/glycob/cwab038.
28. Saito,T., Dai,T., and Asano,R. (2013) Thehyaluronan
synthesis inhibitor 4-methylumbelliferone exhibits
antitumor effects against mesenchymal-like canine
mammary tumor cells, Oncol. Lett., 5, 1068-1074,
https://doi.org/10.3892/ol.2013.1124.
29. Sukowati, C. H. C., Anfuso, B., Fiore, E., Ie, S. I.,
Raseni, A., Vascotto, F., Avellini, C., Mazzolini, G.,
and Tiribelli, C. (2019) Hyaluronic acid inhibition
by 4-methylumbelliferone reduces the expression
of cancer stem cells markers during hepatocarcino-
genesis, Sci. Rep., 9, 4026, https://doi.org/10.1038/
s41598-019-40436-6.
30. Andreichenko, I. N., Tsitrina, A. A., Fokin, A. V.,
Gabdulkhakova, A.I., Maltsev, D.I., Perelman, G. S.,
Bulgakova, E.V., Kulikov, A.M., Mikaelyan, A.S., and
Kotelevtsev, Y. V. (2019) 4-methylumbelliferone pre-
vents liver fibrosis by affecting hyaluronan deposi-
tion, FSTL1 expression and cell localization, Int.J. Mol.
Sci., 20, 6301, https://doi.org/10.3390/ijms20246301.
31. Vigetti, D., Rizzi, M., Viola, M., Karousou, E.,
Genasetti, A., Clerici, M., Bartolini, B., Hascall, V. C.,
De Luca,G., and Passi,A. (2009) Theeffects of 4-meth-
ylumbelliferone on hyaluronan synthesis, MMP2
activity, proliferation, and motility of human aortic
smooth muscle cells, Glycobiology, 19, 537-546, https://
doi.org/10.1093/glycob/cwp022.
32. Tsitrina, A. A., Halimani, N., Andreichenko, I. N.,
Sabirov, M., Nesterchuk, M., Dashenkova, N. O., Ro-
manov, R., Bulgakova, E. V., Mikaelyan, A., and Ko-
televtsev, Y. (2023) 4-methylumbelliferone targets
revealed by public data analysis and liver transcrip-
tome sequencing, Int. J. Mol. Sci., 24, 2129, https://
doi.org/10.3390/ijms24032129.
33. Díaz,M., Pibuel,M., Paglilla,N., Poodts,D., Álvarez,E.,
Papademetrio, D. L., Hajos, S. E., and Lompardía,
S. L. (2021) 4-Methylumbelliferone induces antitu-
mor effects independently of hyaluronan synthe-
sis inhibition in human acute leukemia cell lines,
Life Sci., 287, 120065, https://doi.org/10.1016/j.lfs.
2021.120065.
34. Zhang, W., Watson, C. E., Liu,C., Williams, K.J., and
Werth, V.P. (2000) Glucocorticoids induce a near-total
suppression of hyaluronan synthase mRNA in der-
mal fibroblasts and in osteoblasts: a molecular mech-
anism contributing to organ atrophy, Biochem. J.,
349, 91-97, https://doi.org/10.1042/0264-6021:3490091.
35. Saito, T., Tamura, D., Nakamura, T., Makita, Y.,
Ariyama,H., Komiyama,K., Yoshihara,T., and Asano,R.
(2013) 4-methylumbelliferone leads to growth ar-
rest and apoptosis in canine mammary tumor cells,
Oncol. Rep., 29, 335-342, https://doi.org/10.3892/
or.2012.2100.
36. Ban,H., Uchakina,O., and McKallip, R.J. (2015) Hyal-
uronic acid inhibitor 4-methylumbelliferone activates
FEDOROVA et al.12
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
the intrinsic apoptosis pathway in K562 chronic my-
elogenous leukemia cells, Anticancer Res., 35, 5231-
5240.
37. Halimani, N., Nesterchuk, M., Tsitrina, A. A.,
Sabirov, M., Andreichenko, I. N., Dashenkova, N.O.,
Petrova,E., Kulikov, A.M., Zatsepin, T. S., Romanov,
R. A., Mikaelyan, A. S., and Kotelevtsev, Y. V. (2024)
Knockdown of Hyaluronan synthase 2 suppresses
liver fibrosis in mice via induction of transcriptomic
changes similar to 4MU treatment, Sci. Rep., 14, 2797,
https://doi.org/10.1038/s41598-024-53089-x.
38. Kim,Y., and Kumar,S. (2014) CD44-mediated adhesion
to hyaluronic acid contributes to mechanosensing
and invasive motility, Mol. Cancer Res., 12, 1416-1429,
https://doi.org/10.1158/1541-7786.MCR-13-0629.
39. Pibuel, M. A., Díaz, M., Molinari, Y., Poodts, D.,
Silvestroff,L., Lompardía, S.L., Franco,P., and Hajos,
S. E. (2021) 4-Methylumbelliferone as a potent and
selective antitumor drug on a glioblastoma mod-
el, Glycobiology, 31, 29-43, https://doi.org/10.1093/
glycob/cwaa046.
40. Pibuel, M. A., Poodts, D., Díaz, M., Molinari, Y. A.,
Franco, P.G., Hajos, S.E., and Lompardía, S.L. (2021)
Antitumor effect of 4MU on glioblastoma cells is me-
diated by senescence induction and CD44, RHAMM
and p-ERK modulation, Cell Death Discov., 7, 280,
https://doi.org/10.1038/s41420-021-00672-0.
41. Pibuel, M.A., Poodts,D., Sias, S.A., Byrne,A., Hajos,
S.E., Franco, P.G., and Lompardía, S.L. (2023) 4-Meth-
ylumbelliferone enhances the effects of chemother-
apy on both temozolomide-sensitive and resistant
glioblastoma cells, Sci. Rep., 13, 9356, https://doi.org/
10.1038/s41598-023-35045-3.
42. Yan,T., Chen,X., Zhan,H., Yao,P., Wang,N., Yang,H.,
Zhang, C., Wang, K., Hu, H., Li, J., Sun, J., Dong, Y.,
Lu,E., Zheng,Z., Zhang,R., Wang,X., Ma,J., Gao,M.,
Ye,J., Wang,X., Teng,L., Liu, H., and Zhao,S. (2021)
Interfering with hyaluronic acid metabolism sup-
presses glioma cell proliferation by regulating auto-
phagy, Cell Death Dis., 12, 486, https://doi.org/10.1038/
s41419-021-03747-z.
43. Chistyakov, D. V., Nikolskaya, A. I., Goriainov, S. V.,
Astakhova, A.A., and Sergeeva, M.G. (2020) Inhibitor
of hyaluronic acid synthesis 4-methylumbelliferone
as an anti-inflammatory modulator of LPS-mediated
astrocyte responses, Int. J. Mol. Sci., 21, 8203, https://
doi.org/10.3390/ijms21218203.
44. Dubisova, J., Burianova, J. S., Svobodova, L.,
Makovicky,P., Martinez-Varea, N., Cimpean,A., Faw-
cett, J.W., Kwok, J.C.F., and Kubinova,S. (2022) Oral
treatment of 4-methylumbelliferone reduced peri-
neuronal nets and improved recognition memory in
mice, Brain Res. Bull., 181, 144-156, https://doi.org/
10.1016/j.brainresbull.2022.01.011.
45. McKallip, R. J., Hagele, H. F., and Uchakina, O. N.
(2013) Treatment with the hyaluronic acid synthesis
inhibitor 4-methylumbelliferone suppresses SEB-in-
duced lung inflammation, Toxins (Basel), 5, 1814-1826,
https://doi.org/10.3390/toxins5101814.
46. McKallip, R. J., Ban, H., and Uchakina, O. N. (2015)
Treatment with the hyaluronic Acid synthesis inhib-
itor 4-methylumbelliferone suppresses LPS-induced
lung inflammation, Inflammation, 38, 1250-1259,
https://doi.org/10.1007/s10753-014-0092-y.
47. Wang, H.N., Xiang, Q.A., Lin, H.H., Chen, J.N., Guo,
W. J., Guo, W. M., Yue, X. N., Zhao, Z. F., Ji, K., and
Chen, J.J. (2022) Plant-derived molecule 4-methylum-
belliferone suppresses FcεRI-mediated mast cell acti-
vation and allergic inflammation, Molecules, 27, 1577,
https://doi.org/10.3390/molecules27051577.
48. Lee, S.N., Yoon, S.A., Song, J.M., Kim, H.C., Cho, H.J.,
Choi, A.M.K., and Yoon, J.H. (2022) Cell-type-specific
expression of hyaluronan synthases HAS2 and HAS3
promotes goblet cell hyperplasia in allergic airway in-
flammation, Am.J. Respir. Cell Mol. Biol., 67, 360-374,
https://doi.org/10.1165/rcmb.2021-0527OC.
49. Galkina, S. I., Fedorova, N. V., Ksenofontov, A. L.,
Golenkina, E. A., Serebryakova, M. V., Stadnichuk,
V.I., Baratova, L.A., and Sud’ina, G.F. (2022) Inhibitor
of hyaluronic acid synthesis 4-methylumbelliferone
suppresses the secretory processes that ensure the in-
vasion of neutrophils into tissues and induce inflam-
mation, Biomedicines, 10, 314, https://doi.org/10.3390/
biomedicines10020314.
50. Hasegawa, K., Saga, R., Ohuchi, K., Kuwahara, Y.,
Tomita, K., Okumura, K., Sato, T., Fukumoto, M.,
Tsuruga, E., and Hosokawa, Y. (2022) 4-Methylume-
belliferone enhances radiosensitizing effects of ra-
dioresistant oral squamous cell carcinoma cells via
hyaluronan synthase 3 suppression, Cells, 11, 3780,
https://doi.org/10.3390/cells11233780.
51. Quaranta,S., Rossetti,S., and Camarri,E. (1984) Dou-
ble-blind clinical study on hymecromone and placebo
in motor disorders of the bile ducts after cholecystec-
tomy, Clin. Ter., 108, 513-517.
52. Krawzak, H.W., Heistermann, H. P., Andrejewski,K.,
and Hohlbach, G. (1995) Postprandial bile-duct ki-
netics under the influence of 4-methylumbellifer-
one (hymecromone), Int. J. Clin. Pharmacol. Ther.,
33, 569-572.
53. Yoon, Y., Chae, M. K., Lee, E.J., and Yoon, J. S. (2020)
4-Methylumbelliferone suppresses hyaluronan and
adipogenesis in primary cultured orbital fibroblasts
from Graves’ orbitopathy, Graefes Arch. Clin. Exp.
Ophthalmol., 258, 1095-1102, https://doi.org/10.1007/
s00417-019-04528-3.
54. Marshall, P. L., Nagy, N., Kaber, G., Barlow, G. L.,
Ramesh, A., Xie, B. J., Linde, M. H., Haddock, N. L.,
Lester, C. A., Tran, Q. L., de Vries, C. R., Hargil, A.,
Malkovskiy, A. V., Gurevich, I., Martinez, H. A.,
Kuipers, H. F., Yadava, K., Zhang, X., Evanko, S. P.,
Gebe, J. A., Wang, X., Vernon, R. B., de la Motte, C.,
THERAPEUTIC PROPERTIES OF 4-METHYLUMBELLIFERONE 13
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
Wight, T. N., Engleman, E. G., Krams, S. M., Meyer,
E. H., and Bollyky, P. L. (2021) Hyaluronan synthe-
sis inhibition impairs antigen presentation and de-
lays transplantation rejection, Matrix Biol., 96, 69-86,
https://doi.org/10.1016/j.matbio.2020.12.001.
55. Gebe, J. A., Gooden, M. D., Workman, G., Nagy, N.,
Bollyky, P. L., Wight, T. N., and Vernon, R. B. (2020)
Modulation of hyaluronan synthases and involve-
ment of T cell-derived hyaluronan in autoimmune
responses to transplanted islets, Matrix Biol. Plus, 9,
100052, https://doi.org/10.1016/j.mbplus.2020.100052.
56. Imani,J., Liu,K., Cui,Y., Assaker, J.P., Han,J., Ghosh,
A. J., Ng, J., Shrestha, S., Lamattina, A. M., Louis,
P.H., Hentschel,A., Esposito, A.J., Rosas, I.O., Liu,X.,
Perrella, M.A., Azzi,J., Visner,G., and El-Chemaly,S.
(2021) Blocking hyaluronan synthesis alleviates acute
lung allograft rejection, JCI Insight., 6, e142217, https://
doi.org/10.1172/jci.insight.142217.
57. Uchakina, O. N., Ban, H., and McKallip, R. J. (2013)
Targeting hyaluronic acid production for the treat-
ment of leukemia: treatment with 4-methylumbel-
liferone leads to induction of MAPK-mediated apop-
tosis in K562 leukemia, Leuk. Res., 37, 1294-1301,
https://doi.org/10.1016/j.leukres.2013.07.009.
58. Uchakina, O.N., Ban,H., Hostetler, B.J., and McKallip,
R. J. (2016) Inhibition of hyaluronic acid formation
sensitizes chronic myelogenous leukemia to treat-
ment with doxorubicin, Glycobiology, 26, 1171-1179,
https://doi.org/10.1093/glycob/cww064.
59. Lompardía, S. L., Díaz, M., Papademetrio, D. L.,
Pibuel,M., Álvarez,É., and Hajos, S.E. (2017) 4-meth-
ylumbelliferone and imatinib combination enhances
senescence induction in chronic myeloid leukemia
cell lines, Invest. New Drugs, 35, 1-10, https://doi.org/
10.1007/s10637-016-0397-9.
60. Cho,H., Matsumoto,S., Fujita,Y., Kuroda,A., Menju,T.,
Sonobe,M., Kondo,N., Torii,I., Nakano,T., Lara, P.N.,
Gandara, D. R., Date, H., and Hasegawa, S. (2017)
Trametinib plus 4-methylumbelliferone exhibits anti-
tumor effects by ERK blockade and CD44 downregula-
tion and affects PD-1 and PD-L1 in malignant pleural
mesothelioma, J.Thorac. Oncol., 12, 477-490, https://
doi.org/10.1016/j.jtho.2016.10.023.
61. Collum, S. D., Chen, N. Y., Hernandez, A. M.,
Hanmandlu,A., Sweeney,H., Mertens, T.C.J., Weng,T.,
Luo,F., Molina, J.G., Davies,J., Horan, I.P., Morrell,
N. W., Amione-Guerra, J., Al-Jabbari, O., Youker, K.,
Sun, W., Rajadas, J., Bollyky, P. L., Akkanti, B. H.,
Jyothula, S., Sinha, N., Guha, A., and Karmouty-
Quintana,H. (2017) Inhibition of hyaluronan synthe-
sis attenuates pulmonary hypertension associated
with lung fibrosis, Br. J. Pharmacol., 174, 3284-3301,
https://doi.org/10.1111/bph.13947.
62. Karalis, T. T., Heldin, P., Vynios, D. H., Neill, T.,
Buraschi, S., Iozzo, R. V., Karamanos, N. K., and
Skandalis, S.S. (2019) Tumor-suppressive functions of
4-MU on breast cancer cells of different ER status:
regulation of hyaluronan/HAS2/CD44 and specific
matrix effectors, Matrix Biol., 78-79, 118-138, https://
doi.org/10.1016/j.matbio.2018.04.007.
63. Choi, B.H., Ryoo,I., Sim, K. H., Ahn, H.J., Lee, Y. J.,
and Kwak, M.K. (2022) High levels of hyaluronic acid
synthase-2 mediate NRF2-driven chemoresistance in
breast cancer cells, Biomol. Ther. (Seoul), 30, 368-379,
https://doi.org/10.4062/biomolther.2022.074.
64. Morera, D.S., Hennig, M.S., Talukder,A., Lokeshwar,
S.D., Wang, J., Garcia-Roig,M., Ortiz, N., Yates, T. J.,
Lopez, L.E., Kallifatidis,G., Kramer, M.W., Jordan, A.R.,
Merseburger, A. S., Manoharan, M., Soloway, M. S.,
Terris, M.K., and Lokeshwar, V. B. (2017) Hyaluronic
acid family in bladder cancer: potential prognostic
biomarkers and therapeutic targets, Br. J. Cancer, 117,
1507-1517, https://doi.org/10.1038/bjc.2017.318.
65. Ikuta, K., Ota, T., Zhuo, L., Urakawa,H., Kozawa, E.,
Hamada,S., Kimata,K., Ishiguro, N., and Nishida,Y.
(2017) Antitumor effects of 4-methylumbelliferone, a
hyaluronan synthesis inhibitor, on malignant periph-
eral nerve sheath tumor, Int. J. Cancer, 140, 469-479,
https://doi.org/10.1002/ijc.30460.
66. Piccioni,F., Malvicini,M., Garcia, MG., Rodriguez,A.,
Atorrasagasti, C., Kippes, N., Piedra Buena, I. T.,
Rizzo, M.M., Bayo,J., Aquino,J., Viola,M., Passi,A.,
Alaniz, L., and Mazzolini, G. (2012) Antitumor ef-
fects of hyaluronic acid inhibitor 4-methylumbel-
liferone in an orthotopic hepatocellular carcinoma
model in mice, Glycobiology, 22, 400-410, https://
doi.org/10.1093/glycob/cwr158.
67. Piccioni, F., Fiore, E., Bayo, J., Atorrasagasti, C.,
Peixoto,E., Rizzo,M., Malvicini,M., Tirado-González,I.,
García, M. G., Alaniz, L., and Mazzolini, G. (2015)
4-methylumbelliferone inhibits hepatocellular car-
cinoma growth by decreasing IL-6 production and
angiogenesis, Glycobiology, 25, 825-835, https://
doi.org/10.1093/glycob/cwv023.
68. Rodríguez, M.M., Onorato,A., Cantero, M.J., Domín-
guez,L., Bayo,J., Fiore,E., García,M., Atorrasagasti,C.,
Canbay,A., Malvicini,M., and Mazzolini, G.D. (2021)
4-methylumbelliferone-mediated polarization of
M1 macrophages correlate with decreased hepato-
cellular carcinoma aggressiveness in mice, Sci. Rep.,
11, 6310, https://doi.org/10.1038/s41598-021-85491-0.
69. Weiz, G., Molejon, MI., Malvicini, M., Sukowati,
C.H.C., Tiribelli, C., Mazzolini,G., and Breccia, J.D.
(2022) Glycosylated 4-methylumbelliferone as a tar-
geted therapy for hepatocellular carcinoma, Liver Int.,
42, 444-457, https://doi.org/10.1111/liv.15084.
70. Yoshihara, S., Kon, A., Kudo, D., Nakazawa, H.,
Kakizaki, I., Sasaki, M., Endo, M., and Takagaki, K.
(2005) A hyaluronan synthase suppressor, 4-meth-
ylumbelliferone, inhibits liver metastasis of mel-
anoma cells, FEBS Lett., 579, 2722-2726, https://
doi.org/10.1016/j.febslet.2005.03.079.
FEDOROVA et al.14
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
71. Yang, Y. M., Wang, Z., Matsuda, M., and Seki, E.
(2021) Inhibition of hyaluronan synthesis by
4-methylumbelliferone ameliorates non-alcoholic
steatohepatitis in choline-deficient L-amino acid-
defined diet-induced murine model, Arch. Pharm.
Res., 44, 230-240, https://doi.org/10.1007/s12272-
021-01309-7.
72. Nakazawa,H., Yoshihara,S., Kudo,D., Morohashi,H.,
Kakizaki, I., Kon, A., Takagaki, K., and Sasaki, M.
(2006) 4-methylumbelliferone, a hyaluronan syn-
thase suppressor, enhances the anticancer activ-
ity of gemcitabine in human pancreatic cancer
cells, Cancer Chemother. Pharmacol., 57, 165-170,
https://doi.org/10.1007/s00280-005-0016-5.
73. Yoshida, E., Kudo, D., Nagase, H., Shimoda, H.,
Suto,S., Negishi,M., Kakizaki,I., Endo,M., and Haka-
mada, K. (2016) Antitumor effects of the hyaluro-
nan inhibitor 4-methylumbelliferone on pancreatic
cancer, Oncol. Lett., 12, 2337-2344, https://doi.org/
10.3892/ol.2016.4930.
74. Nagase, H., Kudo, D., Suto, A., Yoshida, E., Suto, S.,
Negishi, M., Kakizaki, I., and Hakamada, K. (2017)
4-methylumbelliferone suppresses hyaluronan
synthesis and tumor progression in SCID mice in-
tra-abdominally inoculated with pancreatic cancer
cells, Pancreas, 46, 190-197, https://doi.org/10.1097/
MPA.0000000000000741.
75. Cheng, X.B., Sato,N., Kohi,S., Koga,A., and Hirata,K.
(2018) 4-Methylumbelliferone inhibits enhanced
hyaluronan synthesis and cell migration in pan-
creatic cancer cells in response to tumor-stromal
interactions, Oncol. Lett., 15, 6297-6301, https://
doi.org/10.3892/ol.2018.8147.
76. Yoshida,E., Kudo,D., Nagase,H., Suto,A., Shimoda,H.,
Suto, S., Kakizaki, I., Endo, M., and Hakamada, K.
(2018) 4-methylumbelliferone decreases the hyaluro-
nan-rich extracellular matrix and increases the effec-
tiveness of 5-fluorouracil, Anticancer Res., 38, 5799-
5804, https://doi.org/10.21873/anticanres.12919.
77. Kudo,Y., Kohi,S., Hirata,K., Goggins,M., and Sato,N.
(2019) Hyaluronan activated-metabolism pheno-
type (HAMP) in pancreatic ductal adenocarcinoma,
Oncotarget, 10, 5592-5604, https://doi.org/10.18632/
oncotarget.27172.
78. Suto, A., Kudo, D., Yoshida, E., Nagase, H., Suto, S.,
Mimura,J., Itoh,K., and Hakamada,K. (2019) Increase
of tumor infiltrating γδ T-cells in pancreatic ductal
adenocarcinoma through remodeling of the extracel-
lular matrix by a hyaluronan synthesis suppressor,
4-methylumbelliferone, Pancreas, 48, 292-298, https://
doi.org/10.1097/MPA.0000000000001211.
79. Benitez,A., Yates, T.J., Shamaldevi,N., Bowen,T., and
Lokeshwar, V. B. (2013) Dietary supplement hymec-
romone and sorafenib: a novel combination for the
control of renal cell carcinoma, J. Urol., 190, 285-290,
https://doi.org/10.1016/j.juro.2012.12.011.
80. Colombaro, V., Declèves, A. E., Jadot, I., Voisin, V.,
Giordano, L., Habsch, I., Nonclercq, D., Flamion, B.,
and Caron, N. (2013) Inhibition of hyaluronan is
protective against renal ischaemia-reperfusion inju-
ry, Nephrol. Dial. Transplant., 28, 2484-2493, https://
doi.org/10.1093/ndt/gft314.
81. Jordan, A. R., Wang, J., Yates, T. J., Hasanali, S. L.,
Lokeshwar, S. D., Morera, D. S., Shamaladevi, N.,
Li, C. S., Klaassen, Z., Terris, M. K., Thangaraju, M.,
Singh, A. B., Soloway, M. S., and Lokeshwar, V. B.
(2020) Molecular targeting of renal cell carcinoma
by an oral combination, Oncogenesis, 9, 52, https://
doi.org/10.1038/s41389-020-0233-0.
82. Selman, G., Martinez, L., Lightle, A., Aguilar, A.,
Woltmann, D., Xiao, Y., Vazquez-Padron, R. I., and
Salman, L.H. (2021) Ahyaluronan synthesis inhibitor
delays the progression of diabetic kidney disease in
a mouse experimental model, Kidney360, 2, 809-818,
https://doi.org/10.34067/KID.0004642020.
83. Wang,J., Jordan, A.R., Zhu,H., Hasanali, S.L., Thom-
as,E., Lokeshwar, S.D., Morera, D. S., Alexander, S.,
McDaniels, J., Sharma, A., Aguilar, K., Sarcan, S.,
Zhu,T., Soloway, M.S., Terris, M.K., Thangaraju,M.,
Lopez, L. E., and Lokeshwar, V. B. (2022) Targeting
hyaluronic acid synthase-3 (HAS3) for the treatment
of advanced renal cell carcinoma, Cancer Cell Int.,
22, 421, https://doi.org/10.1186/s12935-022-02818-1.
84. Lokeshwar, V. B., Lopez, L. E., Munoz, D., Chi, A.,
Shirodkar, S. P., Lokeshwar, S. D., Escudero, D. O.,
Dhir, N., and Altman, N. (2010) Antitumor activi-
ty of hyaluronic acid synthesis inhibitor 4-methy-
lumbelliferone in prostate cancer cells, Cancer Res.,
70, 2613-2623, https://doi.org/10.1158/0008-5472.
CAN-09-3185.
85. Yates, T. J., Lopez, L. E., Lokeshwar, S. D., Ortiz, N.,
Kallifatidis,G., Jordan, A., Hoye,K., Altman, N., and
Lokeshwar, V. B. (2015) Dietary supplement 4-meth-
ylumbelliferone: an effective chemopreventive and
therapeutic agent for prostate cancer, J. Natl. Cancer
Inst., 107, djv085, https://doi.org/10.1093/jnci/djv085.
86. Saga, R., Monzen, S., Chiba, M., Yoshino, H.,
Nakamura, T., and Hosokawa, Y. (2017) Anti-tumor
and anti-invasion effects of a combination of 4-meth-
ylumbelliferone and ionizing radiation in human
fibrosarcoma cells, Oncol. Lett., 13, 410-416, https://
doi.org/10.3892/ol.2016.5385.
87. Saga, R., Hasegawa, K., Murata, K., Chiba, M.,
Nakamura,T., Okumura,K., Tsuruga,E., and Hosoka-
wa,Y. (2019) Regulation of radiosensitivity by 4-meth-
ylumbelliferone via the suppression of interleukin-1
in fibrosarcoma cells, Oncol. Lett., 17, 3555-3561,
https://doi.org/10.3892/ol.2019.9990.
88. Hasegawa, K., Saga, R., Takahashi, R., Fukui, R.,
Chiba, M., Okumura, K., Tsuruga, E., and Hosoka-
wa, Y. (2020) 4-methylumbelliferone inhibits clono-
genic potency by suppressing high molecular weight-
THERAPEUTIC PROPERTIES OF 4-METHYLUMBELLIFERONE 15
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
hyaluronan in fibrosarcoma cells, Oncol. Lett., 19,
2801-2808, https://doi.org/10.3892/ol.2020.11370.
89. Saga, R., Matsuya, Y., Takahashi, R., Hasegawa, K.,
Date,H., and Hosokawa, Y. (2021) 4-Methylumbellif-
erone administration enhances radiosensitivity of
human fibrosarcoma by intercellular communica-
tion, Sci. Rep., 11, 8258, https://doi.org/10.1038/s41598-
021-87850-3.
90. Malvicini,M., Fiore, E., Ghiaccio,V., Piccioni,F., Riz-
zo,M., Olmedo Bonadeo,L., García,M., Rodríguez,M.,
Bayo, J., Peixoto, E., Atorrasagasti, C., Alaniz, L.,
Aquino,J., Matar,P., and Mazzolini,G. (2015) Tumor
microenvironment remodeling by 4-methylumbellif-
erone boosts the antitumor effect of combined immu-
notherapy in murine colorectal carcinoma, Mol. Ther.,
23, 1444-1455, https://doi.org/10.1038/mt.2015.112.
91. Zhan,D., Yalcin,F., Ma,D., Fu,Y., Wei,S., Lal,B., Li,Y.,
Dzaye,O., Laterra,J., Ying,M., Lopez-Bertoni,H., and
Xia, S. (2021) Targeting UDP-α-d-glucose 6-dehydro-
genase alters the CNS tumor immune microenviron-
ment and inhibits glioblastoma growth, Genes Dis., 9,
717-730, https://doi.org/10.1016/j.gendis.2021.08.008.
92. Olivares, C.N., Alaniz, L. D., Menger, M.D., Barañao,
R.I., Laschke, M.W., and Meresman, G. F. (2016) In-
hibition of hyaluronic acid synthesis suppresses an-
giogenesis in developing endometriotic lesions, PLoS
One, 11, e0152302, https://doi.org/10.1371/journal.
pone.0152302.
93. McLaughlin, J. E., Santos, M. T., Binkley, P. A.,
Sultana, M., Tekmal, R. R., Schenken, R. S., and
Knudtson, J. F. (2020) Inhibition of hyaluronic acid
synthesis decreases endometrial cell attachment,
migration, and invasion, Reprod. Sci., 27, 1058-1063,
https://doi.org/10.1007/s43032-019-00100-w.
94. Olivares, C.N., Ricci, A.G., Bilotas, M. A., Alaniz,L.,
Barañao, R.I., and Meresman, G.F. (2023) Effects of
pharmacological inhibition of hyaluronic acid synthe-
sis on experimental endometriosis, Eur.J. Clin. Invest.,
53, e13899, https://doi.org/10.1111/eci.13899.
95. Tamura, R., Yokoyama, Y., Yoshida, H., Imaizumi, T.,
and Mizunuma, H. (2014) 4-Methylumbelliferone
inhibits ovarian cancer growth by suppressing thy-
midine phosphorylase expression, J. Ovarian Res.,
7, 94, https://doi.org/10.1186/s13048-014-0094-2.
96. Lokman, N.A., Price, Z.K., Hawkins, E.K., Macpherson,
A.M., Oehler, M.K., and Ricciardelli,C. (2019) 4-Meth-
ylumbelliferone inhibits cancer stem cell activation
and overcomes chemoresistance in ovarian can-
cer, Cancers (Basel), 11, 1187, https://doi.org/10.3390/
cancers11081187.
97. An,G., Park, S., Lee, M., Lim,W., and Song,G. (2020)
Antiproliferative effect of 4-methylumbelliferone in
epithelial ovarian cancer cells is mediated by dis-
ruption of intracellular homeostasis and regulation
of PI3K/AKT and MAPK signaling, Pharmaceutics, 12,
640, https://doi.org/10.3390/pharmaceutics12070640.
98. Spinelli, F. M., Vitale, D. L., Icardi, A., Caon, I.,
Brandone, A., Giannoni, P., Saturno, V., Passi, A.,
García,M., Sevic,I., and Alaniz,L. (2019) Hyaluronan
preconditioning of monocytes/macrophages affects
their angiogenic behavior and regulation of TSG-6
expression in a tumor type-specific manner, FEBSJ.,
286, 3433-3449, https://doi.org/10.1111/febs.14871.
99. Höbarth,K., Maier,U., and Marberger,M. (1992) Top-
ical chemoprophylaxis of superficial bladder cancer
with mitomycinC and adjuvant hyaluronidase, Eur.
Urol., 21, 206-210, https://doi.org/10.1159/000474839.
100. Ruponen,M., Honkakoski,P., Tammi,M., and Urtti,A.
(2004) Cell-surface glycosaminoglycans inhibit cat-
ion-mediated gene transfer, J. Gene Med., 6, 405-414,
https://doi.org/10.1002/jgm.522.
101. Miyamoto,H., Murakami,T., Tsuchida,K., Sugino,H.,
Miyake,H., and Tashiro, S. (2004) Tumor-stroma in-
teraction of human pancreatic cancer: acquired re-
sistance to anticancer drugs and proliferation reg-
ulation is dependent on extracellular matrix pro-
teins, Pancreas, 28, 38-44, https://doi.org/10.1097/
00006676-200401000-00006.
102. Fang, H., and Declerck, Y. A. (2013) Targeting the
tumor microenvironment: from understanding path-
ways to effective clinical trials, Cancer Res., 73, 4965-
4977, https://doi.org/10.1158/0008-5472.CAN-13-0661.
103. Heldin, C. H., Rubin, K., Pietras, K., and Ostman, A.
(2004) High interstitial fluid pressure– an obstacle in
cancer therapy, Nat. Rev. Cancer, 4, 806-813, https://
doi.org/10.1038/nrc1456.
104. Padera, T.P., Stoll, B.R., Tooredman, J.B., Capen, D.,
di Tomaso,E., and Jain, R.K. (2004) Pathology: cancer
cells compress intratumour vessels, Nature, 427, 695,
https://doi.org/10.1038/427695a.
105. Toole, B. P. (2004) Hyaluronan: from extracellular
glue to pericellular cue, Nat. Rev. Cancer, 4, 528-539,
https://doi.org/10.1038/nrc1391.
106. Friman, T., Gustafsson, R., Stuhr, LB., Chidiac, J.,
Heldin, N.E., Reed, R.K., Oldberg, A., and Rubin,K.
(2012) Increased fibrosis and interstitial fluid pres-
sure in two different types of syngeneic murine car-
cinoma grown in integrin β3-subunit deficient mice,
PLoS One, 7, e34082, https://doi.org/10.1371/journal.
pone.0034082.
107. Singha, N. C., Nekoroski, T., Zhao, C., Symons, R.,
Jiang, P., Frost, G. I., Huang, Z., and Shepard, H. M.
(2015) Tumor-associated hyaluronan limits effica-
cy of monoclonal antibody therapy, Mol. Cancer
Ther., 14, 523-532, https://doi.org/10.1158/1535-7163.
MCT-14-0580.
108. Twarock, S., Reichert, C., Bach, K., Reiners, O.,
Kretschmer,I., Gorski, D.J., Gorges,K., Grandoch,M.,
and Fischer, J.W. (2019) Inhibition of the hyaluronan
matrix enhances metabolic anticancer therapy by
dichloroacetate invitro and invivo, Br. J. Pharmacol.,
176, 4474-4490, https://doi.org/10.1111/bph.14808.
FEDOROVA et al.16
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
109. Abildgaard,C., Rizza,S., Christiansen,H., Schmidt,S.,
Dahl, C., Abdul-Al, A., Christensen, A., Filomeni, G.,
and Guldberg, P. (2021) Screening of metabolic
modulators identifies new strategies to target met-
abolic reprogramming in melanoma, Sci. Rep., 11,
4390, https://doi.org/10.1038/s41598-021-83796-8.
110. Anand, V., Khandelwal, M., Appunni, S., Gupta, N.,
Seth,A., Singh,P., Mathur,S., and Sharma,A. (2019)
CD44 splice variant (CD44v3) promotes progression
of urothelial carcinoma of bladder through Akt/
ERK/STAT3 pathways: novel therapeutic approach,
J. Cancer Res. Clin. Oncol., 145, 2649-2661, https://
doi.org/10.1007/s00432-019-03024-9.
111. Rodríguez, M.M., Fiore,E., Bayo,J., Atorrasagasti,C.,
García,M., Onorato,A., Domínguez,L., Malvicini,M.,
and Mazzolini,G. (2018) 4Mu decreases CD47 expres-
sion on hepatic cancer stem cells and primes a potent
antitumor Tcell response induced by interleukin-12,
Mol. Ther., 26, 2738-2750, https://doi.org/10.1016/
j.ymthe.2018.09.012.
112. Hountondji, L., Ferreira De Matos, C., Lebossé, F.,
Quantin,X., Lesage,C., Palassin,P., Rivet,V., Faure,S.,
Pageaux, G. P., Assenat, É., Alric, L., Zahhaf, A.,
Larrey, D., Witkowski Durand Viel, P., Riviere, B.,
Janick, S., Dalle, S., Maria, A. T. J., Comont, T., and
Meunier, L. (2023) Clinical pattern of checkpoint
inhibitor-induced liver injury in a multicentre co-
hort, JHEP Rep., 5, 100719, https://doi.org/10.1016/
j.jhepr.2023.100719.
113. Tawbi, H.A., Schadendorf,D., Lipson, E.J., Ascierto,
P.A., Matamala,L., Castillo Gutiérrez,E., Rutkowski,P.,
Gogas, H. J., Lao, C. D., De Menezes, J. J., Dalle, S.,
Arance,A., Grob, J.-J., Srivastava,S., Abaskharoun,M.,
Hamilton, M., Keidel, S., Simonsen, K. L., Sobiesk,
A.M., Li,B., Hodi, F.S., and Long, G. V. (2022) Relat-
limab and nivolumab versus nivolumab in untreat-
ed advanced melanoma, N.Engl. J. Med., 386, 24-34,
https://doi.org/10.1056/NEJMoa2109970.
114. Malnick, S. D. H., Abdullah, A., and Neuman, M. G.
(2021) Checkpoint Inhibitors and Hepatotoxici-
ty, Biomedicines, 9, 101, https://doi.org/10.3390/
biomedicines9020101.
115. De Martin,E., Michot,JM., Rosmorduc,O., Guettier,C.,
and Samuel, D. (2020) Liver toxicity as a limiting
factor to the increasing use of immune checkpoint
inhibitors, JHEP Rep., 2, 100170, https://doi.org/
10.1016/j.jhepr.2020.100170.
116. Hernandez, N., and Bessone, F. (2022) Hepatotox-
icity induced by biological agents: clinical features
and current controversies, J. Clin. Transl. Hepa-
tol., 10, 486-495, https://doi.org/10.14218/JCTH.
2021.00243.
117. Shah,P., Sundaram,V., and Björnsson, E. (2020) Bio-
logic and checkpoint inhibitor-induced liver injury:
a systematic literature review, Hepatol. Commun.,
4, 172-184, https://doi.org/10.1002/hep4.1465.
118. Peeraphatdit, T.B., Wang,J., Odenwald, M.A., Hu,S.,
Hart, J., and Charlton, M. R. (2020) Hepatotoxicity
from immune checkpoint inhibitors: a systematic re-
view and management recommendation, Hepatology,
72, 315-329, https://doi.org/10.1002/hep.31227.
119. Delire,B., De Martin, E., Meunier,L., Larrey,D., and
Horsmans,Y. (2022) Immunotherapy and gene ther-
apy: new challenges in the diagnosis and manage-
ment of drug-induced liver injury, Front. Pharmacol.,
12, 786174, https://doi.org/10.3389/fphar.2021.786174.
120. Haanen,J., Obeid,M., Spain,L., Carbonnel,F., Wang,Y.,
Robert,C., Lyon, A.R., Wick,W., Kostine,M., Peters,S.,
Jordan,K., and Larkin,J. (2022) Management of tox-
icities from immunotherapy: ESMO clinical practice
guideline for diagnosis, treatment and follow-up,
Ann. Oncol., 33, 1217-1238, https://doi.org/10.1016/
j.annonc.2022.10.001.
121. Puzanov,I., Diab,A., Abdallah,K., Bingham, C.O.,3rd,
Brogdon,C., Dadu,R., Hamad, L., Kim,S., Lacouture,
M.E., LeBoeuf, N. R., Lenihan,D., Onofrei,C., Shan-
non,V., Sharma, R., Silk, A. W., Skondra,D., Suarez-
Almazor, M. E., Wang, Y., Wiley, K., Kaufman, H. L.,
and Ernstoff, M. S. (2017) Managing toxicities asso-
ciated with immune checkpoint inhibitors: consen-
sus recommendations from the Society for Immu-
notherapy of Cancer (SITC) Toxicity Management
Working Group, J. Immunother. Cancer, 5, 95, https://
doi.org/10.1186/s40425-017-0300-z.
122. Brahmer, J. R., Lacchetti, C., Schneider, B. J., Atkins,
M.B., Brassil, K.J., Caterino, J.M., Chau,I., Ernstoff,
M. S., Gardner, J. M., Ginex, P., Hallmeyer, S., Holter
Chakrabarty, J., Leighl, N. B., Mammen, J. S.,
McDermott, D.F., Naing,A., Nastoupil, L.J., Phillips,T.,
Porter, L.D., Puzanov,I., Reichner, C.A., Santomasso,
B. D., Seigel, C., Spira, A., Suarez-Almazor, M. E.,
Wang,Y., Weber, J.S., Wolchok, J.D., and Thompson,
J.A. (2018) Management of immune-related adverse
events in patients treated with immune checkpoint
inhibitor therapy: American society of clinical on-
cology clinical practice guideline, J. Clin. Oncol., 36,
1714-1768, https://doi.org/10.1200/JCO.2017.77.6385.
123. Dougan,M., Wang,Y., Rubio-Tapia,A., and Lim, J.K.
(2021) AGA Clinical practice update on diagnosis
and management of immune checkpoint inhibitor
colitis and hepatitis: expert review, Gastroenterol-
ogy, 160, 1384-1393, https://doi.org/10.1053/j.gastro.
2020.08.063.
124. Doherty, G.J., Duckworth, A.M., Davies, S. E., Mells,
G. F., Brais, R., Harden, S. V., Parkinson, C. A., and
Corrie, P.G. (2017) Severe steroid-resistant anti-PD1
T-cell checkpoint inhibitor-induced hepatotoxici-
ty driven by biliary injury, ESMO Open, 2, e000268,
https://doi.org/10.1136/esmoopen-2017-000268.
125. Onishi, S., Tajika, M., Bando, H., Matsubara, Y.,
Hosoda,W., Muro,K., and Niwa,Y. (2020) Ursodeoxy-
cholic acid and bezafibrate were useful for steroid-
THERAPEUTIC PROPERTIES OF 4-METHYLUMBELLIFERONE 17
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
refractory, immune-related hepatitis: a case report,
J. Med. Case Rep., 14, 230, https://doi.org/10.1186/
s13256-020-02541-3.
126. Sato,K., Hayashi,M., Abe,K., Fujita,M., Takahashi,A.,
and Ohira, H. (2020) Pembrolizumab-induced scle-
rosing cholangitis in a lung adenocarcinoma patient
with a remarkable response to chemotherapy: a case
report, Clin. J. Gastroenterol., 13, 1310-1314, https://
doi.org/10.1007/s12328-020-01178-5.
127. Robles-Díaz, M., Nezic, L., Vujic-Aleksic, V., and
Björnsson, E. S. (2021) Role of ursodeoxycholic acid
in treating and preventing idiosyncratic drug-induced
liver injury. A systematic review, Front. Pharmacol.,
12, 744488, https://doi.org/10.3389/fphar.2021.744488.
128. Cullen, S. N., Rust, C., Fleming, K., Edwards, C.,
Beuers,U., and Chapman, R.W. (2008) High dose ur-
sodeoxycholic acid for the treatment of primary scle-
rosing cholangitis is safe and effective, J. Hepatol.,
48, 792-800, https://doi.org/10.1016/j.jhep.2007.12.023.
129. Ma,H., Zeng,M., Han,Y., Yan,H., Tang,H., Sheng,J.,
Hu, H., Cheng, L., Xie, Q., Zhu, Y., Chen, G., Gao, Z.,
Xie, W., Wang, J., Wu, S., Wang, G., Miao, X., Fu, X.,
Duan, L., Xu, J., Wei, L., Shi, G., Chen, C., Chen, M.,
Ning, Q., Yao, C., and Jia, J. (2016) A multicenter,
randomized, double-blind trial comparing the ef-
ficacy and safety of TUDCA and UDCA in Chinese
patients with primary biliary cholangitis, Medi-
cine (Baltimore), 95, e5391, https://doi.org/10.1097/
MD.0000000000005391.
130. Xiang, X., Yang, X., Shen, M., Huang, C., Liu, Y.,
Fan, X., and Yang, L. (2021) Ursodeoxycholic acid at
18-22mg/kg/d showed a promising capacity for treat-
ing refractory primary biliary cholangitis, Can. J.
Gastroenterol. Hepatol., 2021, 6691425, https://doi.org/
10.1155/2021/6691425.
131. Steen, E.H., Short, W.D., Li,H., Parikh, U.M., Blum,A.,
Templeman,N., Nagy,N., Bollyky, P.L., Keswani, S.G.,
and Balaji, S. (2021) Skin-specific knockdown of hy-
aluronan in mice by an optimized topical 4-methy-
lumbelliferone formulation, Drug Deliv., 28, 422-432,
https://doi.org/10.1080/10717544.2021.1886376.
132. Rilla,K., Pasonen-Seppänen,S., Rieppo,J., Tammi,M.,
and Tammi,R. (2004) Thehyaluronan synthesis inhib-
itor 4-methylumbelliferone prevents keratinocyte ac-
tivation and epidermal hyperproliferation induced by
epidermal growth factor, J.Invest. Dermatol., 123, 708-
714, https://doi.org/10.1111/j.0022-202X.2004.23409.x.
133. Supp, D.M., Hahn, J. M., McFarland, K.L., and Gla-
ser, K. (2014) Inhibition of hyaluronan synthase 2
reduces the abnormal migration rate of keloid ke-
ratinocytes, J. Burn Care Res., 35, 84-92, https://
doi.org/10.1097/BCR.0b013e3182a2a9dd.
134. Kim, T., Kim, K. B., and Hyun, C. G. (2023) A 7-hy-
droxy 4-methylcoumarin enhances melanogene-
sis in B16-F10 melanoma cells, Molecules, 28, 3039,
https://doi.org/10.3390/molecules28073039.
135. Vassallo, J. D., Hicks, S. M., Born, S. L., and Daston,
G. P. (2004) Roles for epoxidation and detoxifica-
tion of coumarin in determining species differences
in Clara cell toxicity, Toxicol. Sci., 82, 26-33, https://
doi.org/10.1093/toxsci/kfh237.
136. Egan,D., O’Kennedy,R., Moran,E., Cox,D., Prosser,E.,
and Thornes, R.D. (1990) Thepharmacology, metab-
olism, analysis, and applications of coumarin and
coumarin-related compounds, Drug Metab. Rev., 22,
503-529, https://doi.org/10.3109/03602539008991449.
137. Nagy, N., Gurevich, I., Kuipers, H.F., Ruppert, S. M.,
Marshall, P. L., Xie, B. J., Sun, W., Malkovskiy, A. V.,
Rajadas, J., Grandoch, M., Fischer, J. W., Frymoyer,
A. R., Kaber, G., and Bollyky, P. L. (2019) 4-Methy-
lumbelliferyl glucuronide contributes to hyaluronan
synthesis inhibition, J. Biol Chem., 294, 7864-7877,
https://doi.org/10.1074/jbc.RA118.006166.
138. Okhlobystin, A. V., and Ufimtseva, A. K. (2020) The
use of hymecromone in diseases of the biliary tract:
opportunities and prospects [in Russian], Vopr.
Detsk. Dietol., 18, 66-74, https://doi.org/10.20953/1727-
5784-2020-5-66-74.
139. Garrett, E. R., and Venitz, J. (1994) Comparisons of
detections, stabilities, and kinetics of degradation of
hymecromone and its glucuronide and sulfate me-
tabolites, J. Pharm. Sci., 83, 115-116, https://doi.org/
10.1002/jps.2600830128.
140. Takeda, S., and Aburada, M. (1981) The choleretic
mechanism of coumarin compounds and phenolic
compounds, J. Pharmacobiodyn., 4, 724-734, https://
doi.org/10.1248/bpb1978.4.724.
141. Garrett, E.R., Venitz,J., Eberst,K., and Cerda, J.J. (1993)
Pharmacokinetics and bioavailabilities of hymecro-
mone in human volunteers, Biopharm. Drug Dispos.,
14, 13-39, https://doi.org/10.1002/bdd.2510140103.
142. Marshall, M.E., Mohler, J.L., Edmonds,K., Williams,B.,
Butler,K., Ryles,M., Weiss,L., Urban,D., Bueschen,A.,
and Markiewicz,M. (1994) Anupdated review of the
clinical development of coumarin (1,2-benzopyrone)
and 7-hydroxycoumarin, J. Cancer Res. Clin. Oncol.,
120, S39-S42, https://doi.org/10.1007/BF01377124.
143. Lake, B. G. (1999) Coumarin metabolism., toxicity
and carcinogenicity: relevance for human risk as-
sessment, Food Chem. Toxicol., 37, 423-453, https://
doi.org/10.1016/s0278-6915(99)00010-1.
144. Rosser, J.I., Nagy,N., Goel,R., Kaber,G., Demirdjian,S.,
Saxena, J., Bollyky, J. B., Frymoyer, A. R., Pacheco-
Navarro, A.E., Burgener, E.B., Rajadas,J., Wang,Z.,
Arbach, O., Dunn, C. E., Kalinowski, A., Milla, C. E.,
and Bollyky, P.L. (2022) Oral hymecromone decreases
hyaluronan in human study participants, J. Clin. In-
vest., 132, e157983, https://doi.org/10.1172/JCI157983.
145. Selezneva, E. Ya., Mechetina, T. A., Orlova, Yu. N.,
Koricheva, E. S., Voynovan, I. N., Bezaeva, I. V.,
Dubtsova, E. A., and Bordin, D. S. (2016) Compara-
tive study of the UDCA monotherapy effectiveness
FEDOROVA et al.18
BIOCHEMISTRY (Moscow) Vol. 90 No. 1 2025
and UDCA- hymecromone combination in patients
with stage 2 biliary sludge [in Russian], Eksp. Klin.
Gastroenterol., 10, 94-98.
146. Hoffmann, R.M., Schwarz,G., Pohl,C., Ziegenhagen,
D.J., and Kruis,W. (2005) Bile acid-independent effect
of hymecromone on bile secretion and common bile
duct motility, Dtsch. Med. Wochenschr., 130, 1938-1943,
https://doi.org/10.1055/s-2005-872606.
147. Abate, A., Dimartino, V., Spina, P., Costa, P. L.,
Lombardo, C., Santini, A., Del Piano, M., and
Alimonti, P. (2001) Hymecromone in the treatment
of motor disorders of the bile ducts: a multicenter.,
double-blind, placebo-controlled clinical study,
Drugs Exp. Clin. Res., 27, 223-231.
148. Camarri,E., and Marchettini,G. (1988) Hymecromone
in the treatment of symptoms following surgery
ofthe bile ducts, Recenti Prog. Med., 79, 198-202.
149. Ishizuka,S., Askew, E.B., Ishizuka,N., Knudson, C.B.,
and Knudson, W. (2016) 4-methylumbelliferone di-
minishes catabolically activated articular chondro-
cytes and cartilage explants via a mechanism inde-
pendent of hyaluronan inhibition, J.Biol. Chem., 291,
12087-12104, https://doi.org/10.1074/jbc.M115.709683.
Publishers Note. Pleiades Publishing remains
neutral with regard to jurisdictional claims in published
maps and institutional affiliations. AI tools may have
been used in the translation or editing of this article.