10.1007@s00280 020 04046 8
10.1007@s00280 020 04046 8
10.1007@s00280 020 04046 8
https://doi.org/10.1007/s00280-020-04046-8
REVIEW ARTICLE
Abstract
Drug resistance is a well-known phenomenon leading to a reduction in the effectiveness of pharmaceutical treatments. Resist-
ance to chemotherapeutic agents can involve various intrinsic cellular processes including drug efflux, increased resistance
to apoptosis, increased DNA damage repair capabilities in response to platinum salts or other DNA-damaging drugs, drug
inactivation, drug target alteration, epithelial–mesenchymal transition (EMT), inherent cell heterogeneity, epigenetic effects,
or any combination of these mechanisms. Deubiquitinating enzymes (DUBs) reverse ubiquitination of target proteins, main-
taining a balance between ubiquitination and deubiquitination of proteins to maintain cell homeostasis. Increasing evidence
supports an association of altered DUB activity with development of several cancers. Thus, DUBs are promising candidates
for targeted drug development. In this review, we outline the involvement of DUBs, particularly ubiquitin-specific proteases,
and their roles in drug resistance in different types of cancer. We also review potential small molecule DUB inhibitors that
can be used as drugs for cancer treatment.
Keywords DUB inhibitors · Cancer progression · Drug resistance · Cancer cell proliferation · Ubiquitin-specific proteases
13
Vol.:(0123456789)
Cancer Chemotherapy and Pharmacology
BCR B-cell receptor downstream signaling. Various reports have shown that
MYD88 Myeloid differentiation primary inhibiting proteosomal cysteine deubiquitinating enzymes
response 88 such as USP14 and UCHL5 has cytotoxic effects on cancer
PBMC Peripheral blood mononuclear cell cells and causes increased concentrations of ubiquitinated
MDR Multidrug resistance proteins.
HCC Hepatocellular carcinoma The isopeptide bond at the C-terminus of the ubiquitin
CSCs Cancer stem cells molecule is broken selectively by DUBs [9]. DUB functions
SIRT1 Silent mating type information regula- include removing ubiquitin from substrates and activation,
tion 2 homolog 1 deactivation, and localization of regulatory proteins. DUBs
MRP1 Multidrug resistance-associated pro- are involved in various cellular processes such as regula-
tein 1 tion of gene expression, apoptosis, and cytokine signaling,
NAFLD Non-alcoholic fatty liver disease and alterations in the activities of DUBs are associated with
TAK1 Transforming growth factor beta- pathological disorders including cancer and multiple neu-
activated kinase 1 rological disorders [10–13]. This suggests that DUBs may
TLS Translesion synthesis repair pathway be explored as potential drug targets. Based on recent data,
DUBs are being explored rigorously as a drug target in can-
cer since they have an important role in maintenance and
Introduction treatment response of multiple cancer types [14–16].
Drug resistance associated with various diseases is of
Cellular homeostasis is an essential process for cellular major concern and can be specific to a particular disease.
health and is maintained through regulation of protein syn- Various data suggest drug resistance as an emerging concern
thesis and degradation. Thirty percent of mammalian pro- in cancer that is thought to be mediated by mechanisms of
teins have a very short half-life and then become degraded drug efflux, epigenetics, or alterations in target molecules.
[1]. This mechanism of protein degradation is very well Other mechanisms of drug resistance include changes in the
regulated. The ubiquitin–proteasome system (UPS) mainly metabolome and mutations.
controls protein function and stabilization. This is a non-lys- This review gives a detailed description of the role of
osomal intracellular protein degradation pathway mediated DUBs, specifically ubiquitin-specific proteases (USPs), in
by proteasome holoenzymes, ubiquitin ligases, and deubiq- the context of drug resistance in cancer.
uitinating enzymes (DUBs) [2]. This UPS has a pivotal role
in various cellular processes [3–6].
Studies have shown that DUBs are differentially Ubiquitination and deubiquitination
expressed in various forms of cancer including non-small
cell lung cancer, breast cancer, lung cancer, and pancreatic A 76-amino acid polypeptide, ubiquitin, when covalently
ductal adenocarcinoma (Table 1) [7, 8]. These enzymes are bound with its target substrate causes multicatalytic 26S pro-
responsible for catalyzing the re-editing of polyubiquitin teasome complex-mediated degradation of protein [6, 17].
chains and/or removing ubiquitin from its target substrates. This process of ubiquitination is important in both degrada-
This causes alterations in the stability of proteins and/or tion of proteins and regulation of their functions [18]. Two
13
Cancer Chemotherapy and Pharmacology
families of enzymes with contrast activity determine the lysine group of the target protein and the carboxyl terminal
degree of ubiquitination of proteins; ubiquitin ligases cause glycine group of ubiquitin. E3 enzymes serve as substrate
addition of ubiquitin moieties to a protein, while deubiquit- recognition modules for the systems and can interact with
inating enzymes remove these moieties [19]. Ubiquitination both E2 and substrate. After formation of the ubiquitin, E2,
is a well-known phenomenon that has a significant role in E3, and substrate complex, the substrate is bound to ubiqui-
cellular proteins at post-translational levels [20, 21]. tin; this complete process is facilitated by E3 ligase. Later,
Ubiquitination is a process with multiple steps and com- E2 and E3 are removed, and the protein either degrades or
prises three types: (i) mono-ubiquitination, wherein only a is removed from ubiquitin (Fig. 1).
single ubiquitin molecule is attached to the target protein; A very important role of the ubiquitin proteosomal path-
(ii) multi-ubiquitination or poly-mono-ubiquitination, in way (UPP) has been established in the pathogenesis of vari-
which various molecules of ubiquitin are attached to the ous diseases [5]. It has been well established that deregu-
target protein; and (iii) poly-ubiquitination, in which a poly- lation of this UPP pathway has a strong co-relation with
ubiquitin chain is attached to the protein [16, 22, 23]. The various human disease pathologies [17, 24]. Studies suggest
enzymes involved in ubiquitination are ubiquitin-activating that inhibiting this pathway could be an innovative strat-
enzymes (E1), ubiquitin-conjugating enzymes (E2), and egy for therapeutic purposes and can involve inhibition at
ubiquitin ligases or E3 ubiquitin ligases (E3). two levels: the proteosomal level and that of ubiquitinating
Ubiquitination involves activation of ubiquitin by E1 in an E3 ligases. Much research has been performed to develop
ATP-dependent mechanism, followed by thioester bond for- small molecule inhibitors against various other components
mation between cysteine on the active site of enzyme E1 and of the UPP, including design of inhibitors for E1 conjugat-
the glycine residue of ubiquitin on the carboxyl side. Next, ing enzymes or E3 ubiquitin ligases. Studies report that E3
the ubiquitin molecule transfers from E1 to the cysteine ubiquitin ligases are an efficient target for therapeutic impli-
residue of E2. E3 ubiquitin ligases catalyze the final step of cations considering their role in numerous human diseases
ubiquitination by creating an isopeptide bond between the [25].
Fig. 1 The ubiquitin proteasome pathway. Ubiquitination occurs mediate ligation of ubiquitin to target protein substrates. DUBs
through the actions of E1 (ubiquitin-activating enzyme), E2 (ubiq- reverse E3 ligase-mediated ubiquitination and are involved in recy-
uitin-conjugating enzyme), and E3 enzymes (ubiquitin ligase) that cling of ubiquitin molecules
13
Cancer Chemotherapy and Pharmacology
DUBs are known to reverse the ubiquitination process modify the ubiquitination status of a protein. Such a pos-
by precisely cleaving the C-terminus isopeptide bond of sibility should be considered while designing novel DUB
ubiquitin [26]. The main function of proteases is to cleave inhibitors and targeted therapeutic strategies [32].
ubiquitin from proteins and other molecules [27, 28]. Pro- Substantial scientific evidence has shown the importance
teases are vital constituents of the ubiquitin–proteasome of UPS in regulating the proliferation and survival of cancer
system and have three different modes by which they act: cells. This system includes a small molecule ubiquitin along
through de novo ubiquitin synthesis by generating or releas- with a proteasome as a multi-subunit proteolytic complex
ing free ubiquitin; by cleaving polyubiquitinated chains; and that leads to breakdown of undesirable cellular proteins into
by removing the ubiquitin sequence from the ubiquitinated peptides that are used for metabolic processes [4, 33]. Vari-
proteins. ous inhibitors of these proteases have been designed, and
Studies have shown that DUBs are highly selective for their activity has been evaluated in in vitro systems as well
their substrates. They also are very specific to a protein as in vivo animal models [17, 34].
and can be even specific for branching pattern or shape of Despite their commendable potential, the available pro-
polyubiquitin chain. The key domain of DUBs [9] is the teasome inhibitors face the issues of drug resistance, toxic-
ubiquitin-binding domain (UBD), which is comprised of a ity, and a small therapeutic index, indicating the need for
ubiquitin-specific zinc finger protease domain (ZnF-UBP careful patient dosing [35]. This might be due to the inter-
domain), a ubiquitin-interacting motif (UIM), and a ubiqui- connection of proteasomes with many pathways within the
tin-associated domain (UBA domain) [29]. Various groups cell. To combat this issue, upstream components of the UPS
have shown that it is the ZnF-UBP domain that defines the have been targeted. Studies performed to date suggest a need
specific target proteins for DUBs [9]. to explore UPS further to identify novel drug targets.
The human genome encodes nearly 100 DUBs [19, 30].
These DUBs can be further classified into five sub-families,
four that are cysteine proteases and one comprised of metal-
loproteases. These sub-families are individually classified Role of ubiquitin‑specific proteases in cancer
as ubiquitin C-terminal hydrolases (UCHs), ubiquitin-spe- development
cific proteases (USPs or UBPs), ovarian tumor proteases
(OTUs), Machado–Joseph disease proteases (MJDs), and Ubiquitin-specific proteases are present in most forms of
JAB1/MPN/Mov34 metalloenzymes (JAMMs) (Table 2) cancer and are involved in regulation of various pathways
[31]. In addition, another class of DUB-like proteases that that have a role in cancer progression (Table 3) [36–39].
do not belong to the same gene family has been identified These proteases may serve as an effective target for thera-
and named M48USP [9]. This indicates that there might be peutic intervention. It has been observed that various dis-
other unexplored gene families that act in the UPS and can eases, including diseases related to the nervous system,
USP JAMM
USP1, USP2, USP3, USP4, USP5, USP6, USP7 COPS6, EIF3F, PSMD7
USP8, USP9X, USP9Y, USP10, USP11, USP12, AMSH-LP, AMSH,
USP13, USP14, USP15, USP16, USP17, USP18 PRPF8, BRCC36
USP19, USP20, USP21, USP22, USP23, USP24 MPND, MYSM1
USP25, USP26, USP27, USP28, USP28, USP30 EIF3H, COPS5
USP31, USP32, USP33, USP44, USP45, USP46 PSMD14
USP47, USP48, USP49, USP50, USP51, USP52
USP53, USP54, CYLD
OTU
OTUD1, YOD1, OTUB2, OTUB1, TRABID, OTUD7A
OTUD7B, A2O, HIN1L, OTUD4, OTUD6A, OTUD6B
OTUD3, OTUD5, VCPIP1
MJD
ATXN3, ATXN3L, JOSD1, JOSD2
UCH
UCH L1, UCH L3, UCH L5, BAP 1
13
Cancer Chemotherapy and Pharmacology
USP1 Downregulated Plays a vital role in DNA damage, DNA replication, and Fanconi anemia by co- ordinating [141, 147]
DNA repair by promoting the FA pathway and inhibiting the TLS pathway
USP2 Upregulated Deubiquitinates polyubiquitinated target proteins such as fatty acid synthase, murine double [110]
minute 2 (MDM2), MDM4/MDMX, and cyclin D1
USP3 Upregulated Plays a vital role in cell proliferation and spreading by regulating cell cycle control; stabilizes [148, 149]
KLF5 protein
USP4 Upregulated Positive regulator of NF-κB, TGF-β, Wnt/β-catenin, p53, and spliceosome pathways [150]
USP5 Upregulated Efficient repair of DNA double-strand breaks; stabilizes β-catenin protein [122, 151]
USP6 Upregulated Regulates the Wnt signaling pathway [152]
USP7 Upregulated Mediates stabilization of DNMT1 protein and DNA methylation and regulates MDM2/p53 [153]
USP8 Upregulated Promotes trafficking and degradation of chemokine receptor 4 (CXCR4); regulates epidermal [154]
growth factor receptor ubiquitination and trafficking
USP9 Upregulated Regulation of ERα action [84]
USP10 Upregulated Plays a vital role in stabilization of FLT3-ITD oncoprotein [40]
USP11 Upregulated Controls VGLL4 protein stability by promoting its deubiquitination [155]
USP14 Downregulated Downregulated in the apoptotic model and upregulated in the adhesive model of multiple [156]
myeloma in the Bcl-xl apoptotic pathway
USP18 Downregulated Regulatory role in non-alcoholic fatty liver disease [109]
USP20 Upregulated Alters claspin protein level [157]
USP22 Upregulated Interacts with SIRT1 protein and regulates its expression [105]
USP28 Upregulated Stabilizes oncogenic function of LIN28a protein and regulates c-myc protein [158]
USP39 and USP44 Upregulated Maintains mitotic spindle checkpoint and integrity [134, 159]
USP48 Downregulated Promotes mdm2 stability and enhances p53-mediated deubiquitination [160]
UCHL5 Upregulated Plays a vital role in regulation of TGF-β signaling [161]
cancer, and various viral infections, are related to abnor- and fewer side effects. Although nearly 60 USPs have been
malities in processes mediated through ubiquitin. found in humans, the number of active inhibitors is far
Data reported to date suggest that various USPs includ- smaller.
ing USP1-USP8, USP9X, and many more have significance Genetic mutations and abnormal regulation of growth and
in development and progression of cancer. Two therapeutic survival pathways in cancer lead to its diversity and resist-
strategies could be helpful in treating cancer: inhibiting the ance to existing therapies [41–43]. The FDA has approved
DUBs that act as oncoproteins and activating the DUBs that bortezomib as an inhibitor of 20S proteosome for treating
cause tumor suppression [25]. multiple myeloma and mantle cell lymphoma. This was
Weisberg et al. reported that DUBs help stabilize proteins the first inhibitor targeting UPS to be validated for cancer
responsible for disease, specifically in cancer. FLT3-ITD, an therapy [44–47]. This molecule was also associated with
oncoprotein, is stabilized by USP10, which removes the deg- the problems of toxicity and resistance, which limited its
radative ubiquitin tag in cases of acute myeloid leukemia. usage and efficacy [45]. The reason for this resistance is still
Inhibiting USP10 pharmacologically induced degradation unknown [48, 49]. Ongoing studies have suggested muta-
of FLT3-ITD, but did not degrade FLT3. Oncogenic FLT- tions in the catalytic domain as one reason for resistance to
3-expressing acute myeloid leukemia cells were selectively bortezomib. Another mechanism behind resistance to this
inhibited both in vitro and in vivo under these conditions molecule could be over-expression of anti-apoptotic proteins
[40]. such as BCl2 and BCL-XL [50, 51].
The US FDA has also approved second-generation pro-
teasome inhibitors, such as carfilzomib and ixazomib, for
Drug design for DUBs and the effects treatment of multiple myeloma. Ixazomib is an oral protea-
of DUBs in drug resistance some inhibitor that binds reversibly and inhibits the 20S pro-
teasome with a dissociation rate less than half that of borte-
Studies have shown that USPs serve as promising drug zomib (18 min for ixazomib versus 110 min for bortezomib)
targets for design of small molecule inhibitors as they are and has a higher PK/PD profile [52–56]. Ixazomib showed
potential-specific targets, possibly leading to better activity synergistic anti-multiple myeloma activity in combination
13
Cancer Chemotherapy and Pharmacology
with dexamethasone and lenelidomide, providing an addi- Cisplatin-resistant cells did not proliferate after synergistic
tional mechanism to overcome resistance to bortezomib. treatment with inhibitors for USP1/UAF1. Therefore, this
Calfilzomib, another proteasome inhibitor, binds irrevers- complex may serve as a prospective combination target for
ibly to the β5 proteasome subunit with greater affinity than overcoming cisplatin resistance in tumor cells.
bortezomib. This proteasome inhibitor is an isolated agent USP7, also known as HAUSP, serves as a deubiquitinat-
used specifically for treatment of relapsed or refractory mul- ing enzyme for various tumor suppressors [61]. Silencing
tiple myeloma patients who have undergone more than two of USP7 reduces CCDC6 level in HCT116 cells to increase
previous therapies including one with bortezomib. Carfil- the sensitivity of these cells to olaparib. Combining cisplatin
zomib has shown an excellent anti-MM effect in combina- with olaparib produced a synergistic effect on the cells.
tion with lenalidomide and dexamethasone [52–56]. Higher expression of USP7 increases the sensitivity of
Marizomib and oprozomib are other notable inhibitors cancer cell lines to tamoxifen (TAM) [62, 63]. Cell growth
that are currently in the first phase of clinical trial. Mari- inhibition by USP7 might be mediated by increased expres-
zomib, an oral irreversible protease inhibitor, binds to a sion of phosphatase and tensin homolog (PTEN). USP7
catalytic subunit of the 20S proteasome isolated from Salin- has also been established as a deubiquitinating enzyme for
ispora tropica. Phase 1 studies of Marizomib confirmed coiled-coil domain-containing protein 6 (CCDC6), a tumor
comparatively low toxicity with no symptoms of thrombo- suppressor. Reports suggest that NSCLC cells that have low
cytopenia. Marizomib showed great synergy in vitro with expression of CCDC6 protein are more susceptible to the
several other anti-myeloma agents, such as HDACs. Opro- PARP inhibitor olaparib. However, H1975 cells, with high
zomib, an oral irreversible protease inhibitor, is highly selec- expression of CCDC6, were resistant to olaparib. Knocking
tive for the β5 subunit of the 20S proteasome that exhibited down CCDC6 reverted the sensitivity of these cells [64].
similar potency to that of carfilzomib in toxicity assays. The data suggest that inhibitors for USP7 could increase the
Oprozomib has established an acceptable safety profile with response to olaparib and help to enhance response in other
little occurrence of neuropathy in two-phase 1b/2 studies therapeutic agents by downregulating CCDC6 level [65,
[52, 54–59]. 66]. MDM2, a murine oncogene (HDM2, human ortholog),
Cisplatin is a well-known drug used to treat many types acts as the substrate for USP7 and negatively regulates p53
of solid tumors and non-small cell lung cancer. However, protein by ubiquitination and subsequent proteasome-medi-
the response associated with this drug is of a short duration, ated degradation [67–69]. USP7 helps stabilize the level
after which the tumor no longer responds. This resistance of MDM2 during this process. It has been demonstrated
to cisplatin is dependent on various mechanisms that have that siRNA or knockout of USP7 halts deubiquitination of
not been completely elucidated. To dissect the mechanism MDM2 and stabilizes p53 [70, 71]. In addition, p53 mutates
behind this resistance, the effects of cisplatin on transla- in multiple myeloma, and its activation could be a thera-
tion of mRNA in a matched cisplatin-sensitive NSCLC peutic strategy [72]. USP7 is involved in deubiquitination
cell line were compared with those in a resistant cell line. of other onco-targets such as PTEN, FOXO4, and orclaspin
The researchers found that the mRNAs of a few genes were [66].
translated differentially. Among these was the gene encod- A chemical inhibitor of USP7, P5091, causes apoptosis
ing ubiquitin-specific peptidase 1 (USP1), which has signifi- in multiple myeloma cells with resistance to bortezomib
cance in DNA repair mechanisms; its mRNA translation was therapy. These preclinical studies emphasize the need for
inhibited by cisplatin in cisplatin-sensitive cells, but resistant USP7 inhibitors alone or in combination therapy for multi-
cells were unaffected [60]. ple myeloma [66]. A cyano-indenopyrazine derivative found
Sourisseau et al. found that siRNA or ML323 inhibi- by Colland et al. inhibits USP7 at a sub-micromolar IC50.
tion of USP1 expression reversed the resistance of resistant Further studies confirmed that p53 ubiquitination was inhib-
cells, which were surprisingly re-sensitized to cisplatin. The ited by this compound both in vitro and in vivo. The P5091
data suggest that a deficit in downregulation of USP1 at the inhibitor induced dose-dependent activity against various
translational level is the major mechanism for resistance to multiple myeloma cell lines, including cells resistant to
cisplatin in NSCLC, and that this could serve as a marker available therapies such as dexamethasone, doxorubicin, or
for resistance and as a potential drug target in cases with cis- melphalan [73].
platin resistance. This approach of analyzing mRNA at the FT671 is another potent, selective USP7 inhibitor, with an
translational level could aid in identification of new targets IC50 of 52 nM and that binds to the USP7 catalytic domain.
for overcoming drug resistance. FT671 destabilizes USP7 substrates such as MDM2, increas-
USP1 has a role in DNA repair as a deubiquitinat- ing the level of p53 protein and inhibiting tumor growth by
ing enzyme for monoubiquitinated FANCD2 [60]. USP1 promoting transcription of p53 target genes and tumor sup-
possesses a low level of deubiquitinating activity but pressor p21. In vitro studies showed that FT671 enhances
shows greater activity in a complex with UAF1 [61]. the p53 protein level in bone osteosarcoma (U2OS) cell
13
Cancer Chemotherapy and Pharmacology
lines and HCT116. FT-6717 is well tolerated at high doses patients with ERα-positive breast cancer [84], and drug
(100 mg/kg and 200 mg/kg) in mice and was administered in resistance to tamoxifen is a major issue, the mechanism of
a dose that significantly correlated with inhibition of tumor which has yet to be elucidated. Studies were performed to
growth [74, 75]. dissect the resistance mechanism, wherein identification
GNE-6640, another novel selective and non-covalent of resistant genes was performed using large-scale loss
inhibitor of USP7, has IC50 values of 0.75 μM (full length of function genetic screening in ZR-75–1 luminal breast
USP7), 0.43 μM (USP7 catalytic domain), and 20.3 μM (full cancer cells. This revealed that functional loss of USP9X
length USP43). Ubiquitin-specific protease-7 (USP7) con- inhibited the activity of tamoxifen but not the activity of
trols the stability of the p53 tumor suppressor and various another endocrine agent, fulvestrant, which is an ER down
other proteins critical for tumor cell survival. GNE-6640 regulator. Knockdown of USP9X not only stabilized the
facilitates endogenous MDM2 ubiquitination and targets cel- interaction of ERα and chromatin, but also induced tamox-
lular USP7 and p53 signaling pathways, inducing tumor cell ifen-mediated activation of ERα. In addition, proliferation
death, enhancing cytotoxicity, and decreasing the viability of cells responsive to ERα was stimulated in the presence
of around 108 cell lines with IC50 ≤ 10 μM with chemothera- of tamoxifen.
peutic agents such as PIM kinase inhibitors [76]. Various reports suggest that a peptidase specific for
USP8 has a number of substrates including epidermal USP9X regulates various cellular behaviors, including
growth factor receptor (EGFR), binding to which leads tumor growth, invasion, and resistance to available therapies.
to degradation [77]. Inhibition of USP8, either by knock- WP1130 inhibitor exhibits anti-tumor activity by targeting
down or using a synthetic small molecule, led to a decrease a number of DUBs [27]. Increased cisplatin cytotoxicity
in range of receptor tyrosine kinase (RTK) activities and was observed in ER-negative tumor cells, but ER-positive
inhibited cell proliferation in both gefitinib-resistant and cells remained unaffected. Western blotting confirmed that
gefitinib-sensitive non-small cell lung cancer cell lines [77]. expression of USP9X was significantly lower in ER-positive
The BAG3 gene encodes a protein in humans known as BAG cells compared to ER-negative cells. Moreover, expression
family molecular chaperone regulator 3 (BAG3). This pro- of USP9X and Mcl-1 was reduced post-treatment with
tein causes stabilization of Mcl-1, which is a differentiation WP1130 in ER-negative cells but not in ER-positive cells.
protein in stimulated myeloid leukemia cells, leading to drug In breast cancer cells, knockdown of USP9X reduced the
resistance. BAG3 and USP9X interact to stabilize the Mcl-1 chemosensitization activity of WP1130 in the presence
protein; when this interaction was inhibited by WP1130, it of cisplatin. These data indicate that combination therapy
reduced the Mcl-1 level and simultaneously blocked Bcr- of cisplatin with WP1130 increases treatment sensitivity
Abl kinase signaling. This initiated the process of apoptosis, dependent on USP9X in ER-negative breast cancer [85].
overcoming the resistance of cancer cells. Knockdown studies using specific siRNAs of two DUBs,
Another major issue is resistance to tamoxifen, an estro- USP14, and UCHL5, which are highly expressed in multiple
gen receptor (ER) antagonist, in breast cancer. Breast cancer myeloma cells compared to normal plasma cells, resulted
accounts for most of the cancer-related deaths in females in reduced cell viability in multiple myeloma. Both of these
throughout the world [78]. Estrogen helps in growth and DUBs have reversible binding efficacy with 19S regulatory
differentiation of mammary glands [79]. A large percentage proteins and have a role in cancer. Inhibiting these DUBs
of breast cancer patients are ER positive; hence, blocking may lead to reduced uptake of protein substrates to be
this receptor is the ideal therapy for such patients [80]. In degraded [86]. Based on these findings, various other DUB
patients not positive for ER, this therapy fails [81]. This inhibitors were designed. One such inhibitor is b-AP15,
receptor has a very important role in normal growth of the which selectively blocks USP14 and UCHL5 deubiquitinase
breast as well as cancer cell growth in that region [82]. ER activity without affecting 20S core particle activity [16]. A
serves as a prominent biomarker for disease-free survival in potential USP14 inhibitor showed an IC50 dose of 4–5 µM
patients who have had breast cancer. Signaling mediated by and accelerated degradation of protein due to blocking of the
ER has intricate interaction with many other growth signal- proteasome-linked USP14 [87].
ing pathways such as HER2 in breast cancer, and it has a role Another notable mechanism of drug resistance in multiple
in drug resistance that involves various pathways [83]. One myeloma is cell adhesion-mediated drug resistance (CAM-
strategy to combat drug resistance in cancer is to understand DR). This form of cancer occurs due to accumulation of
the methods for drug target alteration. clonal neoplastic cells in the bone marrow and causes bone
Recent studies conducted on USP9X in the context of lesions [88, 89]. Multiple myeloma is also associated with
ERα-positive breast cancer revealed a very interesting drug resistance despite many available treatments [90, 91].
role, wherein loss of USP9X leads to stimulation of tamox- Myeloma cells adhere like glue in the stromal cells of bone
ifen despite resistance due to inhibition of ERα activity. marrow or in the extracellular matrix as the major cause
This drug has been widely used for endocrine therapy for of resistance [92, 93]. This adherence of myeloma cells or
13
Cancer Chemotherapy and Pharmacology
a fibronectin-coated surface protects myeloma cells from potential drug targets that can combat this issue. A promi-
apoptosis, leading to drug resistance [94]. nent role of USP22 has been found in MDR of HCC cells
USP14 is over-expressed in the glue model of multiple (Fig. 2). Knockdown of USP22 led to cell cycle arrest and
myeloma, while it is downregulated in the apoptotic model. hampered cell growth in HepG2, an HCC cell line. The role
It also contributes to CAM-DR in multiple myeloma by of cancer stem cells, which have the properties of self-repair
interconnecting the BCL-xl and Wnt pathways. Studies and differentiation, has been reported in the context of MDR
report that USP14 is involved in various types of cancer in HCC [105].
including colorectal cancer, intra-hepatic cholangiocarci- There are reports showing that Sirtuin 1 (SIRT1) serves
noma, ovarian serous cystadenocarcinoma, and lung car- as a functional negotiator of USP22, promoting HCC cell
cinoma [95–98]. In leukemia cells, USP14 is upregulated. resistance to chemotherapy. Overexpression of SIRT1 in
Treatment with USP14 and UCHL5 inhibitors can induce HCC leads to HCC cell proliferative capacity and causes
apoptosis in multiple myeloma cells and overcome resist- resistance to existing chemotherapies [106]. Protein expres-
ance to bortezomib [86]. This suggests a clear role of USP14 sion of SIRT1 is positively regulated when it interacts with
in CAM-DR. USP22. The expression of SIRT1 and USP22 has a sig-
Walderstrom macroglobulinemia (WM) is a lymphoma of nificant effect on the AKT pathway and MRP1 expression,
B cells, wherein toll-like receptors along with B-cell recep- both of which are inhibited by LY294002. Both USP22 and
tors and MYD88 aid in tumor survival [99]. Higher expres- MRP1 were expressed in more than 150 clinical samples of
sion levels of USP14 and UCHL5 were found in WM tumor HCC according to multiple myeloma histochemical stain-
cells, which were resistant to drug treatment compared with ing. This demonstrates that USP22 promotes MDR in HCC
normal human peripheral blood monocytes or bone mar- by affecting SIRT1/AKT/MRP1 pathways and suggest that
row cells. An inhibitor of USP14/UCHL5 was used to target could serve as a drug target for reversal of multidrug resist-
their co-disruption, and this caused apoptotic death in WM ance in HCC [105].
cells under both ex vivo and in vivo conditions. This further DUBs are engaged in regulation of cell-signaling path-
showed the importance of USP14/UCHL5 as a potential ways, which are usually distorted in cancers. DUBs also
target for overcoming drug-resistant WM and has a trans- have a role in regulation of p53, which is a well-studied
lational implication [100]. A novel proteosome inhibitory tumor suppressor protein mutated in various types of can-
molecule, b-AP15, selectively inhibited USP14 and UCHL5 cer cells [107]. DUBs such as USP7 and USP15 are mainly
without affecting non-proteosomal DUBs [31]. This mol- involved in stabilization of both P53 and MDM2, whereas
ecule was found to be effective against cancer resistance to USP15, USP2, USP4, USP5, USP10, and USP29 play a role
bortezomib. in regulation of p53 alone [16, 70, 108].
The mode of action of b-AP15 is different from that of DUBs have a wide array of roles in various metabolic
bortezomib [101]. It acts by reducing the cell viability in disorders such as non-alcoholic fatty liver disease (NAFLD),
multiple myeloma cells, even in the presence of bone mar- characterized by hepatic steatosis, impaired insulin sensitiv-
row stromal cells [86, 102]. The b-AP15 acted by arrest- ity, and chronic low-grade inflammation. USP18 protects
ing the cell growth of multiple myeloma by downregulating against these symptoms through its deubiquitinating activity.
CDC2, CDC25C, and cyclin B1, leading to caspase-depend-
ent apoptosis. Tumor growth inhibition and prolonged sur-
vival were observed in a xenograft model of human multi-
ple myeloma treated with b-AP15. This efficacy of b-AP15
against multiple myeloma supports the potential of DUBs as
therapeutic targets. Further clinical studies using inhibitors
of USP14/UCHL5 aim to provide clinical intervention for
patients opposed to proteosomal inhibitors [103].
Since DUBs belong to different classes, small molecule
inhibition of associated enzymes would be possible only in
terms of broad-spectrum DUB inhibition. A number of small
molecules have been designed to act against proteosomal
DUBs. One such molecule is RA-9, which is capable of
inhibiting proteosomal deubiquitinase activity and thereby
causing stress and cell death [31, 104].
The emerging issue of multidrug resistance is of great
concern for hepatocellular carcinoma (HCC). Dissecting
the mechanism for drug resistance will help in identifying Fig. 2 USP22-mediated multidrug resistance in HCC cells
13
Cancer Chemotherapy and Pharmacology
Due to limited information on its mechanism of patho- 5. Ciechanover A, Orian A, Schwartz AL (2000) Ubiquitin-medi-
genicity, there are very few possible treatment regimens for ated proteolysis: biological regulation via destruction. BioEssays
22:442–451
NAFLD. Recently, the role of USP18 has been elucidated in 6. Ciechanover A (2005) Proteolysis: from the lysosome to ubiqui-
progression of NAFLD, with USP18 deubiquitinizing TAK1 tin and the proteasome. Nat Rev Mol Cell Biol 6:79–86
and negatively regulating the NF-KB and JNK downstream 7. Zhang L, Xu B, Qiang Y et al (2015) Overexpression of deubiq-
signaling pathways. Exploring the role of USP18 could help uitinating enzyme USP28 promoted non-small cell lung cancer
growth. J Cell Mol Med 19:799–805
in understanding the pathogenesis of NAFLD and develop- 8. Cox JL, Wilder PJ, Wuebben EL et al (2014) Context-dependent
ing new therapies against this disorder [109]. function of the deubiquitinating enzyme USP9X in pancreatic
ductal adenocarcinoma. Cancer Biol Ther 15:1042–1052
9. Komander D, Clague MJ, Urbé S (2009) Breaking the chains:
structure and function of the deubiquitinases. Nat Rev Mol Cell
Biol 10:550–563
Future perspectives 10. Pfoh R, Lacdao IK, Saridakis V (2015) Deubiquitinases and the
new therapeutic opportunities offered to cancer. Endocr Relat
The data presented above emphasize the importance of Cancer 22:T35–54
DUBs as a promising therapeutic target for various forms 11. Wilkinson KD (2009) DUBs at a glance. J Cell Sci
122:2325–2329
of cancer and other diseases; however, the area needs to 12. Ramakrishna S, Suresh B, Baek KH (2011) The role of deubiq-
be explored further. Designing small molecule inhibitors uitinating enzymes in apoptosis. Cell Mol Life Sci 68:15–26
against DUBs could provide treatment for various diseases. 13. Farshi P, Deshmukh RR, Nwankwo JO et al (2015) Deubiquit-
Drug resistance in cancer is very complex, and current stud- inases (DUBs) and DUB inhibitors: a patent review. Expert Opin
Ther Pat 25(10):1191–1208
ies suggest that combination therapy could be the best option 14. Xie X (2019) Deubiquitinase as potential targets for cancer
for overcoming this problem. Despite identification of many immunotherapy. J Cell Immunol 1:1–3
inhibitors of DUBs, most are still in the preclinical stage or 15. Yuan T, Yan F, Ying M et al (2018) Inhibition of ubiquitin-spe-
in the lab for research. These inhibitors should be explored cific proteases as a novel anticancer therapeutic strategy. Front
Pharmacol 9:1–10
further, and rigorous attempts should be made to help them 16. D’Arcy P, Wang X, Linder S (2015) Deubiquitinase inhibition as
reach clinics. The small molecule inhibitors available for a cancer therapeutic strategy. Pharmacol Ther 147:32–54
DUBs act by interfering with DUB interactions with sub- 17. Adams J (2004) The proteasome: a suitable antineoplastic target.
strates, or they inhibit their catalytic activity. The available Nat Rev Cancer 4:349–360
18. Song MS, Salmena L, Carracedo A et al (2008) The deubiquit-
inhibitors are mostly non-specific, targeting multiple DUBs. inylation and localization of PTEN are regulated by a HAUSP-
However, the clinical use of molecules that target a specific PML network. Nature 455:813–817
DUB is yet to be proved, and there is a strong possibility of 19. Nijman SMB, Luna-Vargas MPA, Velds A et al (2005) A
resistance against specific and selective inhibitors. genomic and functional inventory of deubiquitinating enzymes.
Cell 123:773–786
20. Zhou MJ, Chen FZ, Chen HC (2014) Ubiquitination involved
Acknowledgements This research was supported by the Basic Sci-
enzymes and cancer. Med Oncol 31:93
ence Research Program through the National Research Founda-
21. Voutsadakis IA (2012) Ubiquitination and the ubiquitin-protea-
tion of Korea (NRF), which is funded by the Ministry of Education
some system as regulators of transcription and transcription fac-
(2017M3A9C6061361, 2018M3A9H3022412, 2017R1A2B2008727
tors in epithelial mesenchymal transition of cancer. Tumour Biol
and 2017M3A9B3061830).
33:897–910
22. Lander GC, Estrin E, Matyskiela ME et al (2012) Complete sub-
Compliance with ethical standards unit architecture of the proteasome regulatory particle. Nature
482:186–191
Conflict of interest The authors have no potential conflicts of interest 23. Haq S, Suresh B, Ramakrishna S (2018) Deubiquitylating
to disclose. enzymes as cancer stem cell therapeutics. Biochim Biophys Acta
Rev Cancer 1869:1–10
24. Hoeller D, Hecker CM, Dikic I (2006) Ubiquitin and ubiquitin-
like proteins in cancer pathogenesis. Nat Rev Cancer 6:776–788
25. Nicholson B, Marblestone JG, Butt TR, Mattern MR (2007) Deu-
References biquitinating enzymes as novel anticancer targets. Futur Oncol
3:191–199
1. Yewdell JW, Schubert U, Antón LC et al (2000) Rapid degrada- 26. Nicholson B, Leach CA, Goldenberg SJ et al (2008) Charac-
tion of a large fraction of newly synthesized proteins by protea- terization of ubiquitin and ubiquitin-like-protein isopeptidase
somes. Nature 404:770–774 activities. Protein Sci 17:1035–1043
2. Hershko A (2005) The ubiquitin system for protein degradation 27. Kapuria V, Peterson LF, Fang D et al (2010) Deubiquitinase inhi-
and some of its roles in the control of the cell division cycle. Cell bition by small-molecule WP1130 triggers aggresome formation
Death Differ 12:1191–1197 and tumor cell apoptosis. Cancer Res 70:9265–9276
3. Coux O, Tanaka K, Goldberg AL (1996) Structure and functions 28. McClurg UL, Robson CN (2015) Deubiquitinating enzymes as
of the 20S and 26S proteasomes. Annu Rev Biochem 65:801–847 oncotargets. Oncotarget 6:9657–9668
4. Hershko A, Ciechanover A, Varshavsky A (2000) The ubiquitin 29. Hicke L, Schubert HL, Hill CP (2005) Ubiquitin-binding
system. Nat Med 6:1073–1081 domains. Nat Rev Mol Cell Biol 6:610–621
13
Cancer Chemotherapy and Pharmacology
30. Fraile JM, Quesada V, Rodríguez D et al (2012) Deubiquitinases pro-apoptotic PMAIP1/noxa in neuroblastoma, thereby deter-
in cancer: new functions and therapeutic options. Oncogene mining bortezomib sensitivity independent of prosurvival
31:2373–2388 MCL1 expression. J Biol Chem 285:6904–6912
31. Brnjic S, Mazurkiewicz M, Fryknäs M et al (2014) Induction 52. Chhabra S (2017) Novel proteasome inhibitors and histone dea-
of tumor cell apoptosis by a proteasome deubiquitinase inhibi- cetylase inhibitors: progress in myeloma therapeutics. Pharma-
tor is associated with oxidative stress. Antioxid Redox Signal ceuticals 10:40
21:2271–2285 53. Hungria VTM, Crusoe EQ, Bittencourt RI et al (2019) New
32. Sgorbissa A, Potu H, Brancolini C (2010) Isopeptidases in proteasome inhibitors in the treatment of multiple myeloma.
anticancer therapy: looking for inhibitors. Am J Transl Res Hematol Transfus Cell Ther 41:76–83
2:235–247 54. Manasanch EE, Orlowski RZ (2017) Proteasome inhibitors in
33. Goldberg AL (2007) Functions of the proteasome: from protein cancer therapy. Nat Rev Clin Oncol 14:417–433
degradation and immune surveillance to cancer therapy. Biochem 55. Naymagon L, Abdul-Hay M (2016) Novel agents in the treat-
Soc Trans 35:12–17 ment of multiple myeloma: a review about the future. J Hema-
34. Kisselev AF, Goldberg AL (2001) Proteasome inhibitors: from tol Oncol 9:52
research tools to drug candidates. Chem Biol 8:739–758 56. Allegra A, Alonci A, Gerace D et al (2014) New orally active
35. Guédat P, Colland F (2007) Patented small molecule inhibitors in proteasome inhibitors in multiple myeloma. Leuk Res 38:1–9
the ubiquitin proteasome system. BMC Biochem 8(Suppl):S14 57. Moreau P, de Wit E (2017) Recent progress in relapsed multi-
36. Li T, Yan B, Ma Y et al (2018) Ubiquitin-specific protease 4 ple myeloma therapy: implications for treatment decisions. Br
promotes hepatocellular carcinoma progression via cyclophilin J Haematol 179:198–218
A stabilization and deubiquitination. Cell Death Dis 9:148 58. Szalat R, Munshi NC (2019) Novel agents in multiple mye-
37. Pal A, Donato NJ (2014) Ubiquitin-specific proteases as thera- loma. Cancer J 25:45–53
peutic targets for the treatment of breast cancer. Breast Cancer 59. Kubiczkova L, Pour L, Sedlarikova L et al (2014) Proteasome
Res 16:5–11 inhibitors—molecular basis and current perspectives in multi-
38. Young MJ, Hsu KC, Lin TE et al (2019) The role of ubiquitin- ple myeloma. J Cell Mol Med 18:947–961
specific peptidases in cancer progression. J Biomed Sci 26:1–14 60. Sourisseau T, Helissey C, Lefebvre C et al (2016) Translational
39. Melo-Cardenas J, Zhang Y, Zhang DD, Fang D (2016) Ubiquitin- regulation of the mRNA encoding the ubiquitin peptidase
specific peptidase 22 functions and its involvement in disease. USP1 involved in the DNA damage response as a determinant
Oncotarget 7:44848–44856 of Cisplatin resistance. Cell Cycle 15:295–302
40. Weisberg EL, Schauer NJ, Yang J et al (2017) Inhibition of 61. Reyes-Turcu FE, Ventii KH, Wilkinson KD (2009) Regula-
USP10 induces degradation of oncogenic FLT3. Nat Chem Biol tion and cellular roles of ubiquitin-specific deubiquitinating
13:1207–1215 enzymes. Annu Rev Biochem 78:363–397
41. Tyagi N, Tyagi M, Pachauri M, Ghosh PC (2015) Potential thera- 62. Morra F, Merolla F, Criscuolo D et al (2019) CCDC6 and
peutic applications of plant toxin-ricin in cancer: challenges and USP7 expression levels suggest novel treatment options in
advances. Tumor Biol 36:8239–8246 high-grade urothelial bladder cancer. J Exp Clin Cancer Res
42. Srivastava SK, Bhardwaj A, Arora S et al (2015) MicroRNA-345 38:1–14
induces apoptosis in pancreatic cancer cells through potentiation 63. Malapelle U, Morra F, Ilardi G et al (2017) USP7 inhibitors,
of caspase-dependent and -independent pathways. Br J Cancer downregulating CCDC6, sensitize lung neuroendocrine cancer
113:660–668 cells to PARP-inhibitor drugs. Lung Cancer 107:41–49
43. Arora S, Tyagi N, Bhardwaj A et al (2015) Silver nanoparticles 64. Morra F, Luise C, Merolla F et al (2015) FBXW7 and USP7
protect human keratinocytes against UVB radiation-induced regulate CCDC6 turnover during the cell cycle and affect cancer
DNA damage and apoptosis: potential for prevention of skin drugs susceptibility in NSCLC. Oncotarget 6:12697–12709
carcinogenesis. Nanomedicine 11:1265–1275 65. Nicholson B, Suresh Kumar KG (2011) The multifaceted roles
44. Dou Q, Zonder J (2014) Overview of proteasome inhibitor-based of USP7: new therapeutic opportunities. Cell Biochem Biophys
anti-cancer therapies: perspective on bortezomib and second gen- 60:61–68
eration proteasome inhibitors versus future generation inhibi- 66. Chauhan D, Tian Z, Nicholson B et al (2012) A small molecule
tors of ubiquitin-proteasome system. Curr Cancer Drug Targets inhibitor of ubiquitin-specific protease-7 induces apoptosis in
14:517–536 multiple myeloma cells and overcomes bortezomib resistance.
45. Chauhan D, Catley L, Li G et al (2005) A novel orally active Cancer Cell 22:345–358
proteasome inhibitor induces apoptosis in multiple myeloma 67. Cummins JM, Rago C, Kohli M et al (2004) Tumour suppression:
cells with mechanisms distinct from Bortezomib. Cancer Cell disruption of HAUSP gene stabilizes p53. Nature 416:648–653
8:407–419 68. Fang S, Jensen JP, Ludwig RL et al (2000) Mdm2 is a RING
46. Crosas B (2014) Deubiquitinating enzyme inhibitors and finger-dependent ubiquitin protein ligase for itself and p53. J Biol
their potential in cancer therapy. Curr Cancer Drug Targets Chem 275:8945–8951
14:506–516 69. Itahana K, Mao H, Jin A et al (2007) Targeted inactivation of
47. Lim K-H, Baek K-H (2013) Deubiquitinating enzymes as thera- Mdm2 RING finger E3 ubiquitin ligase activity in the mouse
peutic targets in cancer. Curr Pharm Des 19:4039–4052 reveals mechanistic insights into p53 regulation. Cancer Cell
48. Kale AJ, Moore BS (2012) Molecular mechanisms of acquired 12:355–366
proteasome inhibitor resistance. J Med Chem 55:10317–10327 70. Kon N, Kobayashi Y, Li M et al (2010) Inactivation of HAUSP
49. Niewerth D, Jansen G, Assaraf YG et al (2015) Molecular basis in vivo modulates p53 function. Oncogene 29:1270–1279
of resistance to proteasome inhibitors in hematological malignan- 71. Meulmeester E, Maurice MM, Boutell C et al (2005) Loss of
cies. Drug Resist Updat 18:18–35 HAUSP-mediated deubiquitination contributes to DNA dam-
50. Neslund-dudas C, Mitra B, Dou QP (2013) From bortezomib to age-induced destabilization of Hdmx and Hdm2. Mol Cell
other inhibitors of the proteasome and beyond. Curr Pharm Des 18:565–576
19:4025–4038 72. Anderson KC (2007) Targeted therapy of multiple myeloma
51. Hagenbuchner J, Ausserlechner MJ, Porto V et al (2010) based upon tumor-microenvironmental interactions. Exp Hema-
The anti-apoptotic protein BCL2L1/Bcl-xL is neutralized by tol 35:155–162
13
Cancer Chemotherapy and Pharmacology
73. Tian X, Isamiddinova NS, Peroutka RJ et al (2011) Characteriza- 94. Hazlehurst LA, Argilagos RF, Emmons M et al (2006) Cell
tion of selective ubiquitin and ubiquitin-like protease inhibitors adhesion to fibronectin (CAM-DR) influences acquired mitox-
using a fluorescence-based multiplex assay format. Assay Drug antrone resistance in U937 cells. Cancer Res 66:2338–2345
Dev Technol 9:165–173 95. Shinji S, Naito Z, Ishiwata S et al (2006) Ubiquitin-specific
74. Turnbull AP, Ioannidis S, Krajewski WW et al (2017) Molecular protease 14 expression in colorectal cancer is associated with
basis of USP7 inhibition by selective small molecule inhibitors. liver and lymph node metastases. Oncol Rep 15:539–543
Nature 550:481–486 96. Chuensumran U, Saelee P, Punyarit P et al (2011) Ubiquitin-
75. Wang Z, Kang W, You Y et al (2019) USP7: novel drug target in specific protease 14 expression associated with intrahepatic
cancer therapy. Front Pharmacol 10:1–15 cholangiocarcinoma cell differentiation. Asian Pac J Cancer
76. Kategaya L, Di Lello P, Rougé L et al (2017) USP7 small- Prev 12:775–779
molecule inhibitors interfere with ubiquitin binding. Nature 97. Yang Y, Hou JQ, Qu LY et al (2007) Differential expression
550:534–538 of USP2, USP14 and UBE4A between ovarian serous cystad-
77. Byun S, Lee SY, Lee J et al (2013) USP8 is a novel target for enocarcinoma and adjacent normal tissues. Xi Bao Yu Fen Zi
overcoming gefitinib resistance in lung cancer. Clin Cancer Res Mian Yi Xue Za Zhi 23:504–506
19:3894–3904 98. Wu N, Liu C, Bai C et al (2013) Over-expression of deubiq-
78. Bleyer A, Welch HG (2012) Effect of three decades of screen- uitinating enzyme USP14 in lung adenocarcinoma promotes
ing mammography on breast-cancer incidence. N Engl J Med proliferation through the accumulation of β-catenin. Int J Mol
367:1998–2005 Sci 14:10749–10760
79. Vesuna F, Lisok A, Kimble B et al (2012) Twist contributes to 99. Treon SP, Tripsas CK, Meid K et al (2015) Ibrutinib in previ-
hormone resistance in breast cancer by downregulating estrogen ously treated Waldenström’s macroglobulinemia. N Engl J Med
receptor-α. Oncogene 31:3223–3234 372:1430–1440
80. Schwarz LJ, Fox EM, Balko JM et al (2014) LYN-activating 100. Paulus A, Akhtar S, Caulfield TR et al (2016) Coinhibition
mutations mediate antiestrogen resistance in estrogen receptor- of the deubiquitinating enzymes, USP14 and UCHL5, with
positive breast cancer. J Clin Invest 124:5490–5502 VLX1570 is lethal to ibrutinib- or bortezomib-resistant Wal-
81. Santos-Martínez N, Díaz L, Ordaz-Rosado D et al (2014) Cal- denstrom macroglobulinemia tumor cells. Blood Cancer J
citriol restores antiestrogen responsiveness in estrogen recep- 6:e492
tor negative breast cancer cells: A potential new therapeutic 101. Aftab O, Engskog MK, Haglof J et al (2014) NMR spectros-
approach. BMC Cancer 14:230 copy-based metabolic profiling of drug-induced changes in vitro
82. Fishman J, Osborne MP, Telang NT (1995) The role of estrogen can discriminate between pharmacological classes. J Chem Inf
in mammary carcinogenesis. Ann N Y Acad Sci 768:91–100 Model 54:3251–3258
83. Massarweh S, Osborne CK, Creighton CJ et al (2008) Tamoxifen 102. Schmidt M, Altdörfer V, Schnitte S et al (2019) The deubiquit-
resistance in breast tumors is driven by growth factor receptor inase inhibitor b-AP15 and its effect on phenotype and function
signaling with repression of classic estrogen receptor genomic of monocyte-derived dendritic cells. Neoplasia 21:653–664
function. Cancer Res 68:826–833 103. Selvaraju K, Mazurkiewicz M, Wang X et al (2015) Inhibition
84. Oosterkamp HM, Hijmans EM, Brummelkamp TR et al (2014) of proteasome deubiquitinase activity: a strategy to overcome
USP9X downregulation renders breast cancer cells resistant to resistance to conventional proteasome inhibitors? Drug Resist
tamoxifen. Cancer Res 74:3810–3820 Updat 21–22:20–29
85. Fu P, Du F, Liu Y et al (2017) WP1130 increases cisplatin sensi- 104. Coughlin K, Anchoori R, Iizuka Y et al (2014) Small-molecule
tivity through inhibition of usp9x in estrogen receptor-negative RA-9 inhibits proteasome-associated DUBs and ovarian cancer
breast cancer cells. Am J Transl Res 9:1783–1791 in vitro and in vivo via exacerbating unfolded protein responses.
86. Tian Z, D’Arcy P, Wang X et al (2014) A novel small molecule Clin Cancer Res 20:3174–3186
inhibitor of deubiquitylating enzyme USP14 and UCHL5 induces 105. Ling S, Li J, Shan Q et al (2017) USP22 mediates the multid-
apoptosis in multiple myeloma and overcomes bortezomib resist- rug resistance of hepatocellular carcinoma via the SIRT1/AKT/
ance. Blood 123:706–716 MRP1 signaling pathway. Mol Oncol 11:682–695
87. Lee BH, Lee MJ, Park S et al (2010) Enhancement of protea- 106. Chen HC, Jeng YM, Yuan RH et al (2012) SIRT1 promotes
some activity by a small-molecule inhibitor of USP14. Nature tumorigenesis and resistance to chemotherapy in hepatocellular
467:179–184 carcinoma and its expression predicts poor prognosis. Ann Surg
88. Reeder SB, Hu HH, Sirlin CB et al (1999) Cell adhesion medi- Oncol 19:2011–2019
ated drug resistance (CAM-DR): role of integrins and resistance 107. Harris SL, Levine AJ (2005) The p53 pathway: positive and
to apoptosis in human myeloma cell lines. Blood 93:1658–1667 negative feedback loops. Oncogene 24:2899–2908
89. Furukawa Y, Kikuchi J (2015) Molecular pathogenesis of multi- 108. Zou Q, Jin J, Hu H et al (2014) USP15 stabilizes MDM2 to medi-
ple myeloma. Int J Clin Oncol 20:413–422 ate cancer-cell survival and inhibit antitumor T cell responses.
90. Raninga PV, Di Trapani G, Vuckovic S et al (2015) Inhibition Nat Immunol 15:562–570
of thioredoxin 1 leads to apoptosis in drug-resistant multiple 109. An S, Zhao LP, Shen LJ et al (2017) USP18 protects against
myeloma. Oncotarget 6:15410–15424 hepatic steatosis and insulin resistance through its deubiquitinat-
91. Tsubaki M, Takeda T, Ogawa N et al (2015) Overexpression ing activity. Hepatology 66:1866–1884
of survivin via activation of ERK1/2, Akt, and NF-κB plays a 110. Davis MI, Pragani R, Fox JT et al (2016) Small molecule inhibi-
central role in vincristine resistance in multiple myeloma cells. tion of the ubiquitin-specific protease USP2 accelerates cyclin D1
Leuk Res 39:445–452 degradation and leads to cell cycle arrest in colorectal cancer and
92. Hazlehurst LA, Valkov N, Wisner L et al (2001) Reduction in mantle cell lymphoma models. J Biol Chem 291:24628–24640
drug-induced DNA double-strand breaks associated with β1 111. Xu X, Huang A, Cui X et al (2019) Ubiquitin specific peptidase 5
integrin-mediated adhesion correlates with drug resistance in regulates colorectal cancer cell growth by stabilizing Tu transla-
U937 cells. Blood 98:1897–1903 tion elongation factor. Theranostics 9:4208–4220
93. Taylor ST, Hickman JA, Dive C (2000) Epigenetic determinants 112. Zhi Y, Shoujun H, Yuanzhou S et al (2012) STAT3 repressed
of resistance to etoposide regulation of Bcl- x(L) and Bax by USP7 expression is crucial for colon cancer development. FEBS
tumor microenvironmental factors. J Natl Cancer Inst 92:18–23 Lett 586:3013–3017
13
Cancer Chemotherapy and Pharmacology
113. Khan OM, Carvalho J, Spencer-Dene B et al (2018) The deu- glioblastoma: possible link to acquired resistance to anti-VEGF
biquitinase USP9X regulates FBW7 stability and suppresses therapy. Oncotarget 5:6353–6364
colorectal cancer. J Clin Invest 128:1326–1337 133. Wang X, Zhang Q, Wang Y et al (2018) Clinical significance of
114. Li W, Cui K, Prochownik EV, Li Y (2018) The deubiquitinase ubiquitin specific protease 7 (USP7) in predicting prognosis of
USP21 stabilizes MEK2 to promote tumor growth. Cell Death hepatocellular carcinoma and its functional mechanisms. Med
Dis 9:482 Sci Monit 24:1742–1750
115. Kosinsky RL, Zerche M, Saul D et al (2019) USP22 exerts 134. Zhang Y, vanDeursen J, Galardy PJ (2011) Overexpression of
tumor-suppressive functions in colorectalS cancer by decreas- ubiquitin specific protease 44 (USP44) induces chromosomal
ing mTOR activity. Cell Death Differ. https://doi.org/10.1038/ instability and is frequently observed in human T-Cell leukemia.
s41418-019-0420-8 PLoS ONE 6(8):e23389
116. Burkhart RA, Peng Y, Norris ZA et al (2013) Mitoxantrone 135. Yasunaga J, Lin FC, Lu X, Jeang K-T (2011) Ubiquitin-spe-
targets human ubiquitin-specific peptidase 11 (USP11) and is a cific peptidase 20 targets TRAF6 and human T cell leukemia
potent inhibitor of pancreatic cancer cell survival. Mol Cancer virus type 1 tax to negatively regulate NF-B signaling. J Virol
Res 11:901–911 85:6212–6219
117. Cai J, Liu T, Huang P et al (2017) USP39, a direct target of 136. Liu Y, Xu X, Lin P et al (2019) Inhibition of the deubiquitinase
microRNA-133a, promotes progression of pancreatic can- USP9x induces pre-B cell homeobox 1 (PBX1) degradation and
cer via the AKT pathway. Biochem Biophys Res Commun thereby stimulates prostate cancer cell apoptosis. J Biol Chem
486:184–190 294:4572–4582
118. Hou K, Zhu Z, Wang Y et al (2016) Overexpression and biologi- 137. Shan H, Li X, Xiao X et al (2018) USP7 deubiquitinates and sta-
cal function of ubiquitin-spSecific protease 42 in gastric cancer. bilizes NOTCH1 in T-cell acute lymphoblastic leukemia. Signal
PLoS ONE 11:1–11 Transduct Target Ther 3:1–10
119. Nishimura S, Oki E, Ando K et al (2017) High ubiquitin-specific 138. Chae YC, Jung H, Kim JY et al (2019) Ubiquitin-specific pepti-
protease 44 expression induces DNA aneuploidy and provides dase 3 induces TPA-mediated leukemia cell differentiation via
independent prognostic information in gastric cancer. Cancer regulating H2AK119ub. Anim Cells Syst 23:311–317
Med 6:1453–1464 139. Mistry H, Hsieh G, Buhrlage SJ et al (2013) Small-molecule
120. Gu YY, Yang M, Zhao M et al (2015) The de-ubiquitinase inhibitors of USP1 target ID1 degradation in leukemic cells. Mol
UCHL1 promotes gastric cancer metastasis via the Akt and Cancer Ther 12:2651–2662
Erk1/2 pathways. Tumor Biol 36:8379–8387 140. Hahn M, Bürckert JP, Luttenberger CA et al (2018) Aberrant
121. Zhong M, Jiang Q, Jin R (2018) USP4 expression independently splicing of the tumor suppressor CYLD promotes the develop-
predicts favorable survival in lung adenocarcinoma. IUBMB Life ment of chronic lymphocytic leukemia via sustained NF-κB sign-
70:670–677 aling. Leukemia 32:72–82
122. Ma X, Qi W, Pan H et al (2018) Overexpression of USP5 con- 141. Nijman SMB, Huang TT, Dirac AMG et al (2005) The deubiq-
tributes to tumorigenesis in non-small cell lung cancer via the uitinating enzyme USP1 regulates the fanconi anemia pathway.
stabilization of β-catenin protein. Am J Cancer Res 8:2284–2295 Mol Cell 17:331–339
123. Cui S, Fan L, Li Y, et al (2019) Targeting the USP1 dependent 142. Sonego M, Pellarin I, Costa A et al (2019) USP1 links platinum
KDM4A protein stability as a potential prostate cancer therapy. resistance to cancer cell dissemination by regulating snail stabil-
(In Press). https://doi.org/10.1111/cas.14375 ity. Sci Adv 5:eaav3235
124. Guo F, Zhang C, Wang F et al (2019) Deubiquitinating enzyme 143. Kim SY, Kwon SK, Lee SY, Baek KH (2018) Ubiquitin-specific
USP33 restrains docetaxel-induced apoptosis via stabilising the peptidase 5 and ovarian tumor deubiquitinase 6A are differen-
phosphatase DUSP1 in prostate cancer. Cell Death Differ. https tially expressed in p53+/+ and p53−/− HCT116 cells. Int J
://doi.org/10.1038/s41418-019-0473-8 Oncol 52:1705–1714
125. Lee JK, Chang N, Yoon Y et al (2016) USP1 targeting impedes 144. Ma M, Yu N (2016) Ubiquitin-specific protease 7 expression is a
GBM growth by inhibiting stem cell maintenance and radiore- prognostic factor in epithelial ovarian cancer and correlates with
sistance. Neuro Oncol 18:37–47 lymph node metastasis. Onco Targets Ther 9:1559–1569
126. Pal A, Young MA, Donato NJ (2014) Emerging potential of 145. Yan C, Yuan J, Xu J et al (2019) Ubiquitin-specific peptidase 39
therapeutic targeting of ubiquitin-specific proteases in the treat- regulates the process of proliferation and migration of human
ment of cancer. Cancer Res 74:4955–4966 ovarian cancer via p53/p21 pathway and EMT. Med Oncol
127. Ding K, Ji J, Zhang X et al (2019) RNA splicing factor USP39 36:1–13
promotes glioma progression by inducing TAZ mRNA matura- 146. Jin C, Yu W, Lou X et al (2013) UCHL1 is a putative tumor
tion. Oncogene 38:6414–6428 suppressor in ovarian cancer cells and contributes to cisplatin
128. Qiu GZ, Mao XY, Ma Y et al (2018) Ubiquitin-specific protease resistance. J Cancer 4:662–670
22 acts as an oncoprotein to maintain glioma malignancy through 147. Jacq X, Kemp M, Martin NMB, Jackson SP (2013) Deubiquity-
deubiquitinating B cell-specific Moloney murine leukemia virus lating enzymes and DNA damage response pathways. Cell Bio-
integration site 1 for stabilization. Cancer Sci 109:2199 chem Biophys 67:25–43
129. Oikonomaki M, Bady P, Hegi ME (2017) Ubiquitin Specific 148. Fang CL, Lin CC, Chen HK et al (2018) Ubiquitin-specific pro-
Peptidase 15 (USP15) suppresses glioblastoma cell growth via tease 3 overexpression promotes gastric carcinogenesis and is
stabilization of HECTD1 E3 ligase attenuating WNT pathway predictive of poor patient prognosis. Cancer Sci 109:3438–3449
activity. Oncotarget 8:110490–110502 149. Wu Y, Qin Q, Li F et al (2019) USP3 promotes breast can-
130. Hwang SJ, Lee HW, Kim HR et al (2016) Ubiquitin-specific cer cell proliferation by deubiquitinating KLF5. J Biol Chem
protease 4 controls metastatic potential through β-catenin stabi- 294:17837–17847
lization in brain metastatic lung adenocarcinoma. Sci Rep 6:1–13 150. Il YS, Kim HH, Yoon JH et al (2015) Ubiquitin specific protease
131. Hussain S, Zhang Y, Galardy PJ (2009) DUBs and cancer: the 4 positively regulates the WNT/β-catenin signaling in colorectal
role of deubiquitinating enzymes as oncogenes, non-oncogenes cancer. Mol Oncol 9:1834–1851
and tumor suppressors. Cell Cycle 8:1688–1697 151. Nakajima S, Lan L, Wei L et al (2014) Ubiquitin-specific pro-
132. Guo J, Shinriki S, Su Y et al (2014) Hypoxia suppresses cylin- tease 5 is required for the efficient repair of DNA double-strand
dromatosis (CYLD) expression to promote inflammation in breaks. PLoS ONE 9(1):e84899
13
Cancer Chemotherapy and Pharmacology
152. Madana B, Walkerb MP, Young R et al (2016) USP6 oncogene 158. Haq S, Das S, Kim DH et al (2019) The stability and oncogenic
promotes Wnt signaling by deubiquitylating Frizzleds. Proc Natl function of LIN28A are regulated by USP28. Biochim Biophys
Acad Sci USA 113:E2945–E2954 Acta Mol Basis Dis 1865:599–610
153. Cheng J, Yang H, Fang J et al (2015) Molecular mechanism for 159. Van Leuken RJ, Luna-Vargas MP, Sixma TK et al (2008) Usp39
USP7-mediated DNMT1 stabilization by acetylation. Nat Com- is essential for mitotic spindle checkpoint integrity and controls
mun 6:1–11 mRNA-levels of aurora B. Cell Cycle 7:2710–2719
154. Berlin I, Higginbotham KM, Dise RS et al (2010) The deubiqui- 160. Cetkovská K, Šustová H, Uldrijan S (2017) Ubiquitin-specific
tinating enzyme USP8 promotes trafficking and degradation of peptidase 48 regulates Mdm2 protein levels independent of its
the chemokine receptor 4 at the sorting endosome. J Biol Chem deubiquitinase activity. Sci Rep 7:1–9
285:37895–37908 161. Zhou Z, Yao X, Pang S et al (2018) The deubiquitinase UCHL5/
155. Zhang E, Shen B, Mu X et al (2016) Ubiquitin-specific protease UCH37 positively regulates Hedgehog signaling by deubiquit-
11 (USP11) functions as a tumor suppressor through deubiq- inating Smoothened. J Mol Cell Biol 10:243–257
uitinating and stabilizing VGLL4 protein. Am J Cancer Res
6:2901–2909 Publisher’s Note Springer Nature remains neutral with regard to
156. Xu X, Liu J, Shen C et al (2017) The role of ubiquitin-specific jurisdictional claims in published maps and institutional affiliations.
protease 14 (USP14) in cell adhesion-mediated drug resistance
(CAM-DR) of multiple myeloma cells. Eur J Haematol 98:4–12
157. Wang C, Yang C, Ji J et al (2017) Deubiquitinating enzyme
USP20 is a positive regulator of claspin and suppresses the
malignant characteristics of gastric cancer cells. Int J Oncol
50:1136–1146
13