+ All Categories
Home > Documents > Recent advances in proteasome inhibitor discovery

Recent advances in proteasome inhibitor discovery

Date post: 09-Dec-2016
Category:
Upload: kenyon
View: 216 times
Download: 1 times
Share this document with a friend
32
1. Introduction 2. Currently approved proteasome inhibitors 3. Compound screening 4. Rational design 5. Expert opinion Review Recent advances in proteasome inhibitor discovery Yuri Pevzner, Rainer Metcalf, Melanie Kantor, Desiree Sagaro & Kenyon Daniel Moffitt Cancer Center, Screening and Modeling Unit, Chemical Biology Core, Tampa, FL, USA Introduction: Proteasome inhibition is a quickly advancing subject of research and has a significant potential to become a potent therapeutic modality for many diseases and disorders. The aim of this review is to present the reader with the variety of approaches to the proteasome inhibitor discovery as well as highlight the diversity of scaffolds being considered for this task. Areas covered: This review focuses on current developments in proteasome inhibitor discovery, including an account of research efforts covered in the liter- ature from the years 2009 -- 2012, although some of the earlier work is also men- tioned. Specifically, presented are the type of experiments performed, the compounds and compound families investigated along with their activities and assessment for potential therapeutic value. In particular, authors highlight differ- ent paths to discovery of the proteasome inhibitors such as screening of large libraries, repurposing of existing therapeutics, development of compounds with known proteasome inhibitory activities as well as utilizing novel scaffolds. Expert opinion: Discovery of therapeutically successful proteasome inhibitors depends on a number of factors and demands a multipronged approach. Screening protocols, choice of assays, desired mode of action, selection of a binding pocket, targeting and delivery strategy, all require careful consideration when attempting to target the proteasome. Keywords: cancer, drug discovery, inhibition, modeling, proteolysis, rational design, screening Expert Opin. Drug Discov. (2013) 8(5):537-568 1. Introduction The maintenance of proteins is determined by the rate of expression antagonized by rate of degradation. This turnover process is essential for proper regulation of the amount and function of proteins. The proteasome is vital in this process and is a nearly ubiquitous cellular component [1,2]. The pathway for selective proteolysis is a complex process involving discretionary ubiquitination of target proteins by ubiquitin ligases. Ubiqitin is a 76-residue, highly conserved protein that is first activated by the E1 enzyme, complexed with E2, and finally conjugated with its target protein by E3 [3]. This results in a polyubiquitin chain marking the desired protein for destruction [4]. These ubiquitin chains can then bind to the proteasome via specific recognition sites on the regulatory particle (RP), or PA700 cap, of the proteasome [5]. The 26S mammalian proteasome is a massive 2,400 kDa molecule composed of a 20S core particle (CP) and one or two 19S 18-subunit RP (Figure 1A). The CP is a 700 kDa barrel-shaped particle formed by four axially stacked heptameric rings [6]. The b subunits of the inner rings include six proteases whose active sites face an inte- rior lumen where proteolysis occurs (Figure 1B) [7]. These active sites are further anat- omized into a catalytic region (S1) and recognition region (S2) [8]. The S1 pockets are all very similar and incorporate a catalytic THR1 residue [7]. The S2 pockets of the active sites display preferential binding to certain residue sequences (Figure 1C) [9]. These sites are known as the b1/PRE3 peptidylglutamyl peptide 10.1517/17460441.2013.780020 © 2013 Informa UK, Ltd. ISSN 1746-0441, e-ISSN 1746-045X 537 All rights reserved: reproduction in whole or in part not permitted Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Regina on 05/01/13 For personal use only.
Transcript
Page 1: Recent advances in proteasome inhibitor discovery

1. Introduction

2. Currently approved

proteasome inhibitors

3. Compound screening

4. Rational design

5. Expert opinion

Review

Recent advances in proteasomeinhibitor discoveryYuri Pevzner, Rainer Metcalf, Melanie Kantor, Desiree Sagaro &Kenyon Daniel††Moffitt Cancer Center, Screening and Modeling Unit, Chemical Biology Core, Tampa, FL, USA

Introduction: Proteasome inhibition is a quickly advancing subject of research

and has a significant potential to become a potent therapeutic modality for

many diseases and disorders. The aim of this review is to present the reader

with the variety of approaches to the proteasome inhibitor discovery as well

as highlight the diversity of scaffolds being considered for this task.

Areas covered: This review focuses on current developments in proteasome

inhibitor discovery, including an account of research efforts covered in the liter-

ature from the years 2009 -- 2012, although some of the earlier work is also men-

tioned. Specifically, presented are the type of experiments performed, the

compounds and compound families investigated along with their activities and

assessment for potential therapeutic value. In particular, authors highlight differ-

ent paths to discovery of the proteasome inhibitors such as screening of large

libraries, repurposing of existing therapeutics, development of compounds

with known proteasome inhibitory activities as well as utilizing novel scaffolds.

Expert opinion: Discovery of therapeutically successful proteasome inhibitors

depends on a number of factors and demands a multipronged approach.

Screening protocols, choice of assays, desired mode of action, selection of a

binding pocket, targeting and delivery strategy, all require careful consideration

when attempting to target the proteasome.

Keywords: cancer, drug discovery, inhibition, modeling, proteolysis, rational design, screening

Expert Opin. Drug Discov. (2013) 8(5):537-568

1. Introduction

The maintenance of proteins is determined by the rate of expression antagonized byrate of degradation. This turnover process is essential for proper regulation of theamount and function of proteins. The proteasome is vital in this process and is anearly ubiquitous cellular component [1,2].

The pathway for selective proteolysis is a complex process involving discretionaryubiquitination of target proteins by ubiquitin ligases. Ubiqitin is a 76-residue, highlyconserved protein that is first activated by the E1 enzyme, complexed with E2, andfinally conjugated with its target protein by E3 [3]. This results in a polyubiquitinchain marking the desired protein for destruction [4]. These ubiquitin chains canthen bind to the proteasome via specific recognition sites on the regulatory particle(RP), or PA700 cap, of the proteasome [5].

The 26S mammalian proteasome is a massive 2,400 kDa molecule composed of a20S core particle (CP) and one or two 19S 18-subunit RP (Figure 1A). The CP is a700 kDa barrel-shaped particle formed by four axially stacked heptameric rings [6].The b subunits of the inner rings include six proteases whose active sites face an inte-rior lumen where proteolysis occurs (Figure 1B) [7]. These active sites are further anat-omized into a catalytic region (S1) and recognition region (S2) [8]. The S1 pocketsare all very similar and incorporate a catalytic THR1 residue [7]. The S2 pocketsof the active sites display preferential binding to certain residue sequences(Figure 1C) [9]. These sites are known as the b1/PRE3 peptidylglutamyl peptide

10.1517/17460441.2013.780020 © 2013 Informa UK, Ltd. ISSN 1746-0441, e-ISSN 1746-045X 537All rights reserved: reproduction in whole or in part not permitted

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 2: Recent advances in proteasome inhibitor discovery

caspase-like recognition (PGPH), the b2/PUP1 trypsin-like recognition (T-L) and the b5/PRE2 chymotrypsin-like recognition (CT-L) [10]. Other structural variants of theproteasome have been discovered and exhibit slightly alteredfunctions of the beta subunits and regulatory caps. The immu-noproteasome consists of smaller 11S RPs and a modified 20SCP containing ib1, ib2 and ib5 subunits; structural isoformsof the constitutive b1, b2 and b5 subunits, respectively [11].The thymoproteasome is another proteasomal polymorphismmore recently elucidated which incorporates the ib1 andib2 immunoproteasome subunits but integrates a constitu-tively differing tb5 subunit [12].Inhibition of the proteasome has been of recent interest, due

to its effect on cancer cells [13]. Disruption of the ubiquitin-proteasome pathway (UPP) can lead to significant build-up of cytotoxic proteins and activation of apoptotic pathways,particularly in rapidly proliferating cells [14]. Further, termi-nally differentiated cells appear to be protected by proteasomeinhibition, leading to the idea that oncogenic cells are espe-cially susceptible to proteasome inhibition while quiescent cellsmay be preserved to some extent [15]. Proteasome inhibitionmay also be useful in understanding and possibly treating neu-rodegenerative disorders [16], cardiac disease [17,18] and organtransplant rejection [19].Proteasome inhibitors can be classified into many different

groups based on structure and reaction mechanism. Peptidemimics are an emerging class of compounds designed totake advantage of the specificity of proteins as well as theabsorption and stability of small molecules [20]. These typeof compounds and other small molecules contain common

functional groups, such as aldehyde, vinyl sulfone, boronate,epoxyketone, and b-lactone. The typical pharmacophore forthese molecules is the targeting of the CT-L recognition siteand reacting with the catalytic threonine in the activesite [21]. In this review, we survey recent developments inproteasome inhibitor families, screening, and rational design.

2. Currently approved proteasome inhibitors

The peptide boronate, Bortezomib, was the first proteasomeinhibitor approved by the Food and Drug Administration(FDA) for treatment of patients with multiple myeloma [22]

and mantle cell lymphoma [23]. Bortezomib carries a boronicacid “warhead” that forms a covalent bond with the catalyticTHR1 residue of the b5 subunit of the proteasome [24].Although it is considered a reversible inhibitor, Bortezomibhas a slow dissociation rate rendering it virtually irreversible [10].This may contribute to reported side effects which includefatigue, nausea, sensory neuropathy and adverse cardiovasculareffects [25,26]. In clinical use, Bortezomib has been reported tohave up to an 88% response rate [24]. However, despite suc-cesses, about 60% of the patients treated with Bortezomibdevelop resistance to it within an average of 1 year from thebeginning of the treatment [24]. In 2012, the FDA gave anaccelerated approval for an epoxyketone Carfilzomib for treat-ment of patients with multiple myeloma [27]. Carfilzomib, a“second-generation” proteasome inhibitor, was developed toaddress Bortezomib’s toxicity and drug resistance while achiev-ing comparable potency [28]. Carfilzomib is a true irreversibleinhibitor of the proteasome. However, it has an increased selec-tivity for the CT-L activity as compared to Bortezomib and canbe administered on a more aggressive schedule [29]. In clinic,Carfilzomib demonstrated a 22.9% overall response rate andshowed reduced incidence of peripheral neuropathies com-pared to Bortezomib [10,27]. Still, Carfilzomib shows side effectsincluding pneumonia, acute renal failure, pyrexia, congestiveheart failure, cytopenia, fatigue, nausea and dyspnea [27,28].

The fast-tracked approval and successes of Carfilzomibgives validation to the search for follow-on drugs. Intravenousdelivery, the need for aggressive dosing, and side effects, allshow that there is still plenty of room for improvementwhen it comes to proteasome inhibition. An increase in siteselectivity, more effective tissue targeting, improved pharma-codynamic, and pharmacokinetic properties can create betterproteasome inhibitors that are both convenient for patientsand more effective pharmaceutical agents. An expansion ofinhibitor armamentarium would allow for selective targetingof different proteasomal activities through a wide range ofmechanisms, each more fitted to a specific goal.

3. Compound screening

The practice of screening libraries of compounds for potentiallead molecules is common for a number of targets including,and especially, the proteasome. Following the identification

Article highlights.

. The proteasome is an important cellular componentresponsible for the regulation of protein turnover,therefore its targeting provides an avenue to controlcrucial cellular processes.

. Proteasome inhibition has been considered a promisingstrategy to combating a number of diseasesincluding cancer.

. Compound library screening allows to probe a diversevariety of scaffolds that may yield promising candidates.

. Development of the classes of compounds that havepreviously been shown to inhibit proteasome allows toimprove on existing therapeutics and address concernsthat accompany them.

. Metal complexing is a viable strategy for selectivelytargeting the proteasome, a component essential forboth healthy and diseased cells.

. Drug repurposing or adaptation of existing therapeuticswith no previous proteasome inhibitory indications canprovide a viable strategy for getting promisingcompounds to the clinic.

. Development of novel scaffolds can enrich the pool ofinhibitor candidates and provide new avenues for finetuning the regulation of proteasomal activity.

This box summarizes key points contained in the article.

Y. Pevzner et al.

538 Expert Opin. Drug Discov. (2013) 8(5)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 3: Recent advances in proteasome inhibitor discovery

of potential leads from a screening campaign, compoundscan be further characterized and developed into promisingcandidates. Screening can be performed using large librariescontaining hundreds of thousands of compounds or smaller“diversity” subsets that are representative of the chemicaldiversity of a larger library. A variety of biochemical and cel-lular assays can be used in such screenings as long as they canbe adapted to a high-throughput approach. Alternatively, aninitial computational screening can be performed to discarda substantial portion of compounds as nonbinders prior tobiochemical screening. The advantage of such a techniqueis that a larger library, numbering in millions of com-pounds, can be screened computationally with significantlyless time and cost than a biochemical screen [30]. However,the disadvantage of this is the approximate nature of scoringfunctions that can result in false positives as well as falsenegatives [31,32].

Daniel and colleagues analyzed 1,990 drug-like compoundsof the National Cancer Institute (NCI) Diversity Set Library [33]

in a combination of biochemical and cellular assays and foundthat compound NCI-109268 (Table 1, 1a) inhibited the CT-Lactivity of the proteasome in Jurcat T cells and 20S rabbitproteasome isolates with an IC50 » 6 µM [34]. Similarly, theNCI Diversity Set [33] was screened by Lavelin and colleaguesusing a novel live cell image screening method based on thetranslocation of a fluorescent reporter, the nuclear localizationsignal-deficient p53 derivative, from the cytoplasm to thenucleus [35]. The most active of the several resulting hits inhib-ited the 26S proteasome at EC50 = 100 µM (Table 1, 1b,bromocopper; (dipyridin-2-ylmethylideneamino)-[methylsulfanyl

(sulfoniumylidene)methyl]milacicazanide). Lawrence et al. alsoscreened the NCI Diversity Set [33] specifically targeting theCT-L activity of the 20S proteasome. Following the initialscreen, a smaller focused library of naphthoquinone analogs ofthe lead compound (Table 1, 1c, 4-[(3-chloro-1,4-dioxonaph-thalen-2-yl)amino]-N-pyridin-2-ylbenzenesulfonamide) wassynthesized and evaluated in subsequent assays in order todetermine the structure--activity relationship (SAR) of itsCT-L inhibition [36]. Ge and colleagues performed a libraryscreening of 20,000 compounds in a biochemical assay thatidentified a hydronaphthoquinone containing compound tosynthesize and evaluate a focused library. This approach haslead to an identification of a compound (Table 1, 1d, N-(3-(1H-Tetrazol-5-ylthio)-4-hydroxynaphthalen-1-yl)-4-(tri-fluoromethyl)biphenyl-4-sulfonamide) with an inhibition ofthe CT-L activity in the range of IC50 » 0.44 to 1.01 µM inintact breast cancer cells [37]. Another set of naphthoquinonederivatives based on the hit identified by Lawrence et al. wasdesigned by Xu and colleagues [38]. Biochemical assay andtumor cell line evaluation of these compounds identified acompound (Table 1, 1e) with activity improved or comparableto that of the original compound found by Lawrence et al. [38].

A secondary metabolite secreted by the fungus Aspergillusterreus, terrein (Table 1, 1f) displayed proteasome inhibitionwhen Demasi et al. performed a screening of fungi extractsagainst a purified 20S horse proteasome [39]. The initial screenwas followed by assays to determine the site specificity ofterrein and its effect on proteolytic activity of the 20S isolatesas well as its performance in cells [39]. Terrein inhibitedCT-L and T-L activities at IC50 = 300 µM and promoted

β Ring subunits

β1

β2

β5

20S CP

Caspase-like

(PGPH)

Trypsin-like

Chymotrypsin-like

(T-L)

(CT-L)

A. B. C.

19SCap

19SCap

Lid

Base

α Subunits

α Subunits

β Subunits

Proteasome

Figure 1. Structural diagram of the constitutive proteasome. A. 19S RPs presented in grey (lid) and red (base) and 20S CP in

blue (a) and turquoise (b). B. 20S CP shown with b1, b2 and b5 subunits localized in central lumen. C. b1, b2 and b5 subunits

displayed as an electrostatic surface potential map with red representing negative charge, white representing neutral and

blue representing positive. The proteolytic threonine 1 (green) is highlighted in each.

Recent advances in proteasome inhibitor discovery

Expert Opin. Drug Discov. (2013) 8(5) 539

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 4: Recent advances in proteasome inhibitor discovery

Table 1. Survey of screening compounds.

Compound reference number 2-D Compound structure Description Inhibition activity* Refs.

Compound screening1a A,B,C 20S (CT-L) 6 µM [34]

1b A 26S (CT-L) 100 µM EC50 [35]

1c A,C,F (CT-L) 1.0 ± 0.63 µM [36]

1d A,C (CT-L) 0.44 -- 1.01 µM [37]

1e A,C (CT-L) 3.65 µM [38]

1f A,C,D (CT-L) (T-L) 300 µM [39]

*Inhibition activity reported as IC50 unless otherwise noted.

A: Biochemical assay; B: Cellular assay; C: CT-L activity assayed; D: T-L activity assayed; E: PGPH activity assayed; F: Covalent inhibitor; G: Noncovalent inhibitor;

H: Peptide; I: Peptidomimetic; J: Structure determined, no data reported.

Y. Pevzner et al.

540 Expert Opin. Drug Discov. (2013) 8(5)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 5: Recent advances in proteasome inhibitor discovery

80% cell death in fibroblasts at 150 and 250 µM after only4 h [39]. Blackburn et al. employed a multistep strategy con-sisting of: library screening, X-ray crystallography, andstructure-based design to develop a series of highly potentand selective inhibitors of the CT-L activity of the 20S consti-tutive and immunoproteasome [40]. The inhibitors were devel-oped using structure-guided design based on the crystalstructure of a hit compound that was identified by a high-

throughput cell-based screening of a library of 350,000 com-pounds [40]. After a series of focused library screens, the mostpotent compound (Table 1, 1g, N-(2,2-dimethylpropyl)-N˜2˜-[1H-indol-3-yl(oxo)acetyl]-L-asparaginyl-N-(2-methyl-benzyl)-3-pyridin-4-yl-L-alaninamide) was identified withinhibitory activity IC50 = 0.0012 µM with a proposed nonco-valent mode of inhibition [40]. Long and colleagues developedan assay utilizing human colon cell lines 4Ub-Luc DLD-1 [41]

Table 1. Survey of screening compounds (continued).

Compound reference number 2-D Compound structure Description Inhibition activity* Refs.

1g A,C,D,E,G (CT-L) 1.2 � 10-3 µM [40]

1h J - [42]

1i J - [42]

1j A,C,D,E,G (CT-L) 1.9 µM [43]

1k A,C,G (CT-L) 0.34 ± 0.04 µM [44]

*Inhibition activity reported as IC50 unless otherwise noted.

A: Biochemical assay; B: Cellular assay; C: CT-L activity assayed; D: T-L activity assayed; E: PGPH activity assayed; F: Covalent inhibitor; G: Noncovalent inhibitor;

H: Peptide; I: Peptidomimetic; J: Structure determined, no data reported.

Recent advances in proteasome inhibitor discovery

Expert Opin. Drug Discov. (2013) 8(5) 541

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 6: Recent advances in proteasome inhibitor discovery

that express 4-ubiquitin luciferase reporter protein to screen12,000 plant extracts and 62,000 pure compounds [42]. Thisassay led to identification and characterization of severalcycloartane derivatives from the leaves of Neobutonia melleri,some of which (Table 1, 1h -- Neoboutomellerone and1i -- 22-de-O-acetylneoboutomellerone) showed activity ina cellular proteasome assay [42]. A combination of compu-tational and biochemical screening was employed byBasse et al. [43]. Following the initial docking of a 300,000compound library, 200 were evaluated experimentally andseveral compounds were validated showing low micromolarinhibition of one or more proteolytic activities of 20S protea-some. The most active compound (Table 1, 1j) inhibited theCT-L activity at IC50 = 1.9 µM [43]. Similar to the approachtaken by Blackburn et al. [40], an investigation combininglibrary screening, crystallographic structure determinationand structure-guided SAR was conducted by Gallastegui andcolleagues [44]. The SAR study was performed on a series ofnoncovalent hydroxyurea derivatives which emerged from aninitial fluorogenic tetrapeptide substrate library screen [44].The most active compound (Table 1, 1k, 1-hydroxy-1-[(2R)-4-3-[(3S,5S,7S)-tricyclo[3.3.1.1˜3,7˜]dec-1-yloxy]phenyl-but-3-yn-2-yl]urea) exhibited inhibition of the CT-L activity ofthe proteasome at IC50 = 0.34 µM [44].Compound library screening is an important and widely

used approach that helps to search for novel inhibitors byallowing exploration of broad and diverse body of com-pounds. As this review demonstrates, the diversity of com-pounds exhibiting proteasome inhibitory activity hints at theimportance of venturing into the vastness of chemical space,a task that compound screening helps to achieve.

4. Rational design

In addition to screening compound libraries, there are a num-ber of compound families that have received significant atten-tion for targeting the proteasome in recent years. These haveemerged from earlier studies, current clinically approved drugs,and/or promising leads. Important structural features that thesecompounds possess are often used in the development of novelscaffolds. Substitution of these moieties onto a lead compoundmay increase its inhibitory activity or specificity toward one ormore of the proteasome’s catalytic sites. These scaffolds ofteninclude peptides, peptidomimetics, and natural products.

4.1 Boron-containing compoundsThe development of Bortezomib as a proteasome inhibitorapproved by the FDA for the treatment of multiple myelomahas accelerated active efforts to improve on its capability anddiminish some of its drawbacks for the next generation ofboron-containing proteasome inhibitors. Although Bortezo-mib is a potent and effective proteasome inhibitor, it hasbeen found to have multiple and often serious side effectsincluding: fatigue, nausea, sensory neuropathy and adversecardiovascular effects [25,26]. As a part of the effort to develop

second-generation proteasome inhibitors with high potencyand reduced side effects, the ability of Bortezomib to selec-tively bind to the b5 site of the 20S proteasome via its boronicacid moiety is a frequent design strategy.

Zhu et al. reported a SAR study on a series of tripeptidealdehyde and boronate-based compounds that demonstratelow nanomolar inhibitory activity [45], the most potent(Table 2, 2a, N-(2-pyrazinecarbonyl)-L-leucine-L-(2-naph-thyl)-alanine-L-leucine boronic acid) exhibiting inhibition ofthe CT-L activity of the isolated mouse 20S proteasome atIC50 = 0.00079 µM [45]. This study was followed by a com-prehensive SAR of a large number of dipeptidyl boronateinhibitors [26]. The best performing compounds (Table 2, 2b,[(1R)-1-[[(2S)-3-phenyl-2-[((S)-(-)-1,2,3,4-tetrahedro-naph-thoic-1-carbonyl)amino]-1-oxopropyl]amino]-3-methylbutyl]boronic acid and 2c, [(1R)-1-[[(2S)-3-phenyl-2-[(5,6,7,8-tet-rahedro-naphthoic-1-carbonyl)amino]-1-oxopropyl]amino]-3-methylbutyl] boronic acid) in this study were assayed in10 tumor cell lines that included multiple myeloma, large celllung cancer, human non-small cell lung cancer, colon carcinoma,and other cell lines, and showed an improvement over Bortezo-mib in seven out of ten cell lines with IC50 values ranging from0.0098 to 0.07 µM [26]. To further develop dipeptidyl boronicacids as proteasome inhibitors, in 2010, Zhu and colleagues per-formed another SAR study on this class of compounds [46].However, this time, in an effort to increase the activity and sta-bility, the peptides were composed of b-amino acids [46]. Thestudy also included toxicity assessments and docking simulationsfor binding mode characterization [47]. The best performingcompound (Table 2, 2d, [(1R)-1-[[(3R)-3-(4-methoxyphenyl)-3-[(phenylcarbonyl)amino]-1-oxopropyl]amino]-3-methylbutyl]boronic acid), although reduced in potency compared to Borte-zomib (IC50 = 0.0096 µM vs 0.00248 µM), was less toxic inzebrafish embryo and Sprague--Dawley rats and possessed a bet-ter pharmacokinetic profile [46]. Following this study, a morecomprehensive SAR evaluation of b-amino acid dipeptidylboronic-acid inhibitors was performed using a similar approachthat included a biochemical 20S proteasome screen followedby cell line cytotoxicity studies as well as pharmacokinetic andtoxicity evaluation in rodent models [47]. The most potent com-pound (Table 2, 2e, (R)-1-[3-(3-fluorophenyl)-3-((S)-1,2,3,4-tet-rahydronaphthalene-4-carboxamido)propanamido]-3-methyl-butyl boronic acid) performed comparably to Bortezomib in a20S proteasome inhibitory assay (IC50 = 0.00102 µM vs0.00154 µM) and showed potent cytotoxicity while maintainingimproved toxicity and pharmacokinetic profiles [47].

The recently discovered proteasome inhibitor tyropep-tin [48,49] was chosen by Watanabe et al. as the scaffoldfor development of tyropeptin boronic acid derivatives [50].A resulting lead compound (Table 2, 2f, (1R)-1-[N-(1-naph-thylacetyl)-L-O-methyltyrosyl-L-valyl]amino-3-methyl-1-butylboronic acid) inhibited the CT-L activity of the purifiedhuman 20S proteasome with IC50 = 0.0026 µM [50]. To fur-ther characterize tyropeptin boronic acid derivatives, a SARstudy was expanded to a total of 53 derivatives and tested for

Y. Pevzner et al.

542 Expert Opin. Drug Discov. (2013) 8(5)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 7: Recent advances in proteasome inhibitor discovery

Table 2. Survey of boron-containing compounds.

Compound reference

number

2-D Compound structure Description Inhibition activity* Refs.

Boron-containing compounds2a A,C,D,E,H (CT-L) (7.9 ± 1.1) � 10-5 µM [45]

2b A,C,F,H (CT-L) (1.2 ± 0.1) � 10-3 µM [26]

2c A,C,F,H (CT-L) (1.6 ± 0.2) � 10-3 µM [26]

2d A,C,F,H (CT-L) (9.6 ± 1.3) � 10-3 µM [46]

2e A,C,H (CT-L) 1.02 � 103 µM [47]

2f A,C,D,E,F,H (CT-L) 2.6 � 10-3 µM [50]

*Inhibition activity reported as IC50 unless otherwise noted.

A: Biochemical assay; B: Cellular assay; C: CT-L activity assayed; D: T-L activity assayed; E: PGPH activity assayed; F: Covalent inhibitor; G: Noncovalent inhibitor;

H: Peptide; I: Peptidomimetic; J: Structure determined, no data reported.

Recent advances in proteasome inhibitor discovery

Expert Opin. Drug Discov. (2013) 8(5) 543

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 8: Recent advances in proteasome inhibitor discovery

Table 2. Survey of boron-containing compounds (continued).

Compound reference

number

2-D Compound structure Description Inhibition activity* Refs.

2g A,C,D,E,F,H (CT-L) 4.1 � 10-3 µM [51]

2h A,C,D,E,F,H (Cytotoxicity) 4.9 � 10-3 µM [51]

2i A,C,F,H (CT-L) 0.28 ± 0.04 µM [54]

2j A,C,F,H (CT-L) 0.54 ± 0.14 µM [54]

2k A,C,F,H (CT-L) 2.5 � 10-4 µM [55]

2l A,C,I (CT-L) (3.5 ± 0.5) � 10-3 µM [56]

*Inhibition activity reported as IC50 unless otherwise noted.

A: Biochemical assay; B: Cellular assay; C: CT-L activity assayed; D: T-L activity assayed; E: PGPH activity assayed; F: Covalent inhibitor; G: Noncovalent inhibitor;

H: Peptide; I: Peptidomimetic; J: Structure determined, no data reported.

Y. Pevzner et al.

544 Expert Opin. Drug Discov. (2013) 8(5)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 9: Recent advances in proteasome inhibitor discovery

both purified 20S proteasome inhibition and cytotoxicity inmultiple myeloma-derived cells [51]. The compound exhibitingthe most potency in a biochemical assay (Table 2, 2g, 3-phe-noxyphenylacetamide) showed inhibition of the CT-L activitywith an IC50 = 0.0041 µM, while the most cytotoxic com-pound (Table 2, 2h, 3-fluoro picolinamide) possessed an IC50

of 0.0049 µM [51]. Nakamura et al., in addition to exploitingthe boron peptide moiety, have turned to structural featuresfrom the natural product Belactosin C -- a compound isolatedfrom Streptomyces sp. and shown to inhibit rabbit 20S protea-some [52,53]. The result was a series of low micromolar com-pounds with both cell growth inhibition and 20S proteasomeinhibition capacity, representatives of which are shownin Table 2 (2i and 2j) [54]. A computationally generated three-dimensional (3D) pharmacophore model was used as a startingpoint to elucidate a series of boron-containing dipeptide inhib-itors by Lei et al. [55]. The most active compound (Table 2, 2k,[(1R)-1-[[(2S)-3-(1-Naphthyl)-2-[(1-naphthyl-2-carbonyl)-amino]-1-oxopropyl]amino]-3-methylbutyl]boronic acid)inhibited the CT-L activity of the 20S proteasome with anIC50 of 0.00025 µM [55]. The assays were followed by adocking study of the most active compound to elucidate itsbinding mode within the b5 subunit of the 20S particle [55].As part of their search for novel proteasome inhibitors,Iqbal et al. have explored chiral boronate-derived peptidomi-metic compounds [56]. The most active of those (Table 2, 2l,2-[[(1R)-1-[[(2S-5-[[Imino(nitroamino)methyl]-2-[((R,S)-10-cyano-2-cyclopentyldecanoyl)amino-1-oxopentyl]amino]-3-methylbutyl]]-(3aS,4S,6S,7aR)-hexahydro-3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborole and 2m, 2-[[(1R)-1-[[(2S-5-[[Imino(nitroamino)methyl]-2-[((R,S)-10-N-phtha-limido-2-cyclopentyldecanoyl)amino-1-oxopentyl]amino]-

3-methylbutyl]]-(3aS,4S,6S,7aR)-hexahydro3a,5,5-trimethyl-4,6-methano-1,3,2-benzodioxaborole) inhibited CT-L activityof 20S proteasome at IC50 = 0.0035 µM and showed activ-ity in a number of human and rodent cancer cell lines [56].Furthermore, these compounds served as a starting pointfor a study of a series of (2S,3R)-2-amino-3-hydroxybutyricacid derivatives that led to discovery of a potent inhibitor(Table 2, 2n, [(1R)-1-[[(2S,3R)-3-hydroxy-2-[(6-phenylpy-ridine-2-carbonyl)amino]-1-oxobutyl]amino]-3-methylbu-tyl]boronic acid) that demonstrated efficacy in a cellular assayalong with a favorable toxicity profile and was selected for pre-clinical profiling for multiple myeloma therapy [57].

Boron-containing compounds have proven themselves aspotent proteasome inhibitors, albeit not without caveats.Thus, it is important to build on this progress and improveon past efforts to take boronates from “the lesser of two evils”to an optimal choice of treatment.

4.2 Fluorine-containing compoundsFluorine-containing compounds have been playing anincreasingly important role in drug design over the past sev-eral years [58-61]. The popularity of fluorine is largely due tothe characteristics of carbon-bonded fluorine such as smallsize, ability to withdraw electrons, and hydrophobicity of flu-orocarbons that facilitates tighter binding to their targets.Additionally, it has been proposed that addition of fluoro sub-stituents may help enhance metabolic stability of anticancercompounds [62,63]. In particular, the trifluoromethyl groupwith its unique properties and suitable geometry is oftenused to improve the stability and activity of compounds [64].Due to this, many investigators have turned to this class ofcompounds in the search for potent proteasome inhibitors.

Table 2. Survey of boron-containing compounds (continued).

Compound reference

number

2-D Compound structure Description Inhibition activity* Refs.

2m A,C,I (CT-L) (3.5 ± 1.3) � 10-3 µM [56]

2n A,C,I (CT-L) (3.8 ± 1) � 10-3 µM [57]

*Inhibition activity reported as IC50 unless otherwise noted.

A: Biochemical assay; B: Cellular assay; C: CT-L activity assayed; D: T-L activity assayed; E: PGPH activity assayed; F: Covalent inhibitor; G: Noncovalent inhibitor;

H: Peptide; I: Peptidomimetic; J: Structure determined, no data reported.

Recent advances in proteasome inhibitor discovery

Expert Opin. Drug Discov. (2013) 8(5) 545

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 10: Recent advances in proteasome inhibitor discovery

Table 3. Survey of fluorine- and aldehyde-containing compounds.

Compound reference

number

2-D Compound structure Description Inhibition activity* Refs.

Flourine-containing compounds3a A,C,D,E,G,I (CT-L) 1.6 ± 0.1 µM

(PGPH) 2.7 ± 0.1 µM(T-L) 8.4 ± 1.3 µM

[65]

3b A,C,D,E,G,I (CT-L) 32 ± 2 µM(PGPH) 6 ± 0.5 µM(T-L) 30% inhibition

[65]

3c A,C,D,E,G,I (CT-L) 5.9 ± 0.5 µM(PGPH) 2.7 ± 0.1 µM(T-L) 4.4 ± 1.2 µM

[65]

3d A,C (CT-L) 4.44 µM [62]

3e A,C,D,E,G,H 26S (CT-L) 2.0 � 10-3 µM [70]

Aldehyde-containing compounds4a A,C,F,H (CT-L) 0.028 ± 0.006 µM [81]

*Inhibition activity reported as IC50 unless otherwise noted.

A: Biochemical assay; B: Cellular assay; C: CT-L activity assayed; D: T-L activity assayed; E: PGPH activity assayed; F: Covalent inhibitor; G: Noncovalent inhibitor;

H: Peptide; I: Peptidomimetic; J: Structure determined, no data reported.

Y. Pevzner et al.

546 Expert Opin. Drug Discov. (2013) 8(5)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 11: Recent advances in proteasome inhibitor discovery

Formicola et al. developed a series of pseudopeptides basedon the tryfluoromethyl-b-hydrazino acid scaffold the mostactive of which (Table 3, 3a, 2-[3-(N-6-Amino-2-[2-(3-phe-noxyphenyl)acetylamino]hexanoylhyd)hydrazino)-4,4,4-tri-fluoro-butyrylamino]-3-phenyl-propionic acid methyl ester,trifluoro-acetic acid salt, 3b, 2-3-[N-(6-Amino-2-tert-butoxy-carbonylaminohexanoyl)hydrazino]-4,4,4-trifluorobutyryla-mino-3-phenylpropionic acid methyl ester, citric acid saltand 3c, 2-3-[N-(2-Amino-6-benzyloxycarbonylaminohexa-noyl)hydrazino]-4,4,4-trifluoro-butyrylamino-3-phenylpro-pionic acid methyl ester) have shown differential inhibitorycapabilities toward the CT-L, T-L and PGPH activities of20S proteasome at micromolar concentrations [65]. It haspreviously been shown that an active phenolic natural productextracted from rhizome of the tropical Southeast Asian plantCurcuma longai curcumin [66-68], inhibited the proteasomeactivity in human colon cancer cells [69]. Padhye et al. devel-oped curcumin’s fluoro Knoevenagel condensates, Schiff bases,and copper complexes [62]. The study resulted in a potentinhibitor (Table 3, 3d, 4-Salicylidene-1,7-bis(4-hydroxy,3-methoxyphenyl)-1,6-heptadiene-3,5-dione) of both 20S pro-teasome CT-L activity and colon cancer cell growth [62]. Further-more, over the course of developing a series of fluorinatedinhibitors, the most potent one of which (Table 3, 3e (S)-2-azido-N-((S)-1-((S)-1-((S)-4-methyl-1-((R)-2-methyloxiran-2-yl)-1-oxopentan-2-ylamino)-1-oxo-3-(perfluorophenyl)propan-2-ylamino)-1-oxo-3-phenylpropan-2-yl)-3-phenylpropanamide,N3PhePhePhe(F5)Leu-EK) inhibited the CT-L activity ofpurified 26S proteasome at IC50 = 0.002 µM Geurink et al.demonstrated that the presence of fluorinated phenylalaninederivatives in peptide-based compounds improved their CT-Linhibitory potency and selectivity [70].

With a good track record, fluorine-containing pharmaceu-tical agents have proven themselves both within and outsideof the proteasome inhibition arena. Further inquiry into andthe development of these compounds have the potential toresult in effective clinical candidates.

4.3 Aldehyde-containing compoundsAldehydes, and specifically peptide aldehydes, have proventhemselves to be promising compounds when it comes to pro-teasome inhibition [7,71-77]. In addition, a wealth of experi-mental data has been accumulated over the years, which hasallowed investigators to draw important conclusions andbuild on the existing body of work. In a recent study Maand colleagues, guided by a crystal structure of a peptide alde-hyde inhibitor MG101 [78] in complex with 20S proteasome,synthesized and evaluated the activities of a new series ofpeptide aldehyde derivatives in a combination of computa-tional and biochemical experiments. The best performingcompound (Table 3, 4a, Cbz-Glu(OtBu)-Phe-Leucinal) inan isolated 20S proteasome assay exhibited activity atIC50 = 0.028 µM, outperforming a known potent MG101derivative peptide aldehyde MG132 [79,80] by an order ofmagnitude [81]. These results suggest that aldehyde-containing

compounds are a promising avenue to explore in the searchfor effective proteasome inhibitors.

4.4 Vinyl sulfone and vinyl ester-containing

compoundsVinyl sulfones are another example of a pharmacophoric groupoften used in developing proteasome inhibitors [82-86]. Theybelong to a class of inhibitors that contain Michael acceptorsand have been shown to inhibit the proteasome via formationof a covalent bond with the catalytic THR1 residues of theproteasomal active sites [21,87,88].

Ettari and coworkers developed a series of conformationallyconstrained tripeptidyl vinyl sulfones, the best of which(Table 4, 5a, Benzyl-1-((S)-1-((S,E)-1-(ethylsulfonyl)-5-meth-ylhex-1-en-3-ylamino)-1-oxo-3-phenylpropan-2-yl)-2-oxo-1,2-dihydropyridin-3-ylcarbamate, 5b, Allyl-1-((S)-1-((S,E)-1-(ethylsulfonyl)-5-methylhex-1-en-3-ylamino)-4-methyl-1-oxo-pentan-2-yl)-2-oxo-1,2-dihydropyridin-3-ylcarbamate and 5c,Allyl-1-((S)-1-((S,E)-1-(ethylsulfonyl)-5-methylhex-1-en-3-ylamino)-1-oxo-3-phenylpropan-2-yl)-2-oxo-1,2-dihydropyridin-3-ylcarbamate) showed good selectivity and inhibition of CT-L activity of 20S proteasome in the submicromolar range [82].As a strategy to enhance proteasome inhibition and cellularuptake, Vivier et al. incorporated vinyl sulfone groups into aseries of inhibitors [83]. The most potent of these compounds(Table 4, 5d, N1-(2-(Dipropylamino)ethyl)-N4-((S)-4-methyl-1-((S)-4-methyl-1-((S)-4-methyl-1-oxopentan-2-ylamino)-1-oxopentan-2-ylamino)-1-oxopentan-2-yl)terephthalimide,hydrochloride salt and 5e, N1-((Diethylamino)ethyl)-N4-(4-methyl-1-oxo-1-(1-oxo-3-phenylpropan-2-ylamino)pen-tan-2-yl)terephthalamide), while possessing decreasedcytotoxicity in comparison with their aldehyde-containinganalogs, provided potential low micromolar candidatesfor further assessment [83]. Similarly, Screen and colleaguesinvestigated the effects of replacing epoxyketone withvinyl sulfone moieties in proteasome inhibitors [84]. Thesubstitution resulted in an increased specificity toward pro-teasomal CT-L activity, which was proposed to be the rea-son for decreased overall cytotoxicity of vinyl sulfoneinhibitors [84].

Vinyl ester groups were proposed to inhibit the proteasomeby interacting with the catalytic threonine residue in a mannersimilar to that of vinyl sulfones [85]. Baldisserotto et al. exam-ined a series of cyclic peptides containing the moiety andshowed selective and potent inhibition of the PGPH activityin a biochemical assay at IC50 = 0.065 µM in 20S purifiedproteasome and in a cellular assay at IC50 = 0.091 µM bythe most active compound (Table 4, 5f c[Ser-Leu-Leu-Glu(Leu-VE)]) [85]. In a later study, however Baldisserotto andcolleagues saw a marked reduction in the inhibitory activityof a series of N-allyl vinyl ester-based peptides [89]. The mostpotent N-allylic vinyl ester derivative (Table 4, 5g HMB-Leu-Leu-N-AllylLeu-VE) exhibited its strongest inhibition atIC50 = 0.29 µM comparing to IC50 = 0.059 µM of the refer-ence vinyl ester peptide toward the T-L activity of the 20S

Recent advances in proteasome inhibitor discovery

Expert Opin. Drug Discov. (2013) 8(5) 547

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 12: Recent advances in proteasome inhibitor discovery

Table 4. Survey of vinyl sulfone, vinyl ester, and epoxyketone-containing compounds.

Compound reference

number

2-D Compound structure Description Inhibition

activity*

Refs.

Vinyl sulfone-containing compounds5a A,C,D,E,I (CT-L) 0.33 µM Ki [82]

5b A,C,D,E,I (CT-L) 0.61 µM Ki [82]

5c A,C,D,E,I (CT-L) 0.37 µM Ki [82]

5d B,G,H 0.71 ± 0.34 µM [83]

5e B,G,H 0.64 µM [83]

5f A,B,C,D,E,F,H (PGPH)0.065 µM (A)(PGPH)0.091 µM (B)

[85]

5g A,C,D,E,H (T-L)0.29 ± 0.33 µM

[89]

*Inhibition activity reported as IC50 unless otherwise noted.

A: Biochemical assay; B: Cellular assay; C: CT-L activity assayed; D: T-L activity assayed; E: PGPH activity assayed; F: Covalent inhibitor; G: Noncovalent inhibitor;

H: Peptide; I: Peptidomimetic; J: Structure determined, no data reported.

Y. Pevzner et al.

548 Expert Opin. Drug Discov. (2013) 8(5)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 13: Recent advances in proteasome inhibitor discovery

proteasome [89]. In another study, Chen and colleagues evalu-ated a set of vinyl ester-containing peptides that had alsoshown mixed results [86]. Finally, based on the 26S protea-some and cell extract assays, Screen et al. concluded that con-trary to the earlier studies vinyl ester-containing peptidesshow no inhibition of any of the proteolytic activities [84].This result shows the importance of assaying the inhibitoryactivities of compounds in 26S proteasome assays in additionto 20S and cell-based assays in order to confirm the inhibitionof the entire proteasome particle and rule out other waysthrough which an inhibitor may be exhibiting activity.

High specificity toward the CT-L activity of the protea-some, tight covalent binding to catalytic threonine and rela-tively low overall reactivity [83] make vinyl sulfones anattractive alternative to some of the other families of protea-some inhibitors. In addition to these hallmarks of a potentproteasome inhibitor, vinyl sulfones exhibit a potential foralleviating some of the side effects that other alternatives suf-fer. Therefore, further development of these compounds isneeded to ensure that this potential is realized in the clinic.

4.5 Epoxyketone-containing compoundsAs the success of Bortezomib has been fueling efforts todevelop boron-containing compounds, Carfilzomib [90,91]

has paved the way for the development for epoxyketone-containing compounds. Although a highly selective andpotent inhibitor of the CT-L activity of the proteasome [90-96],Carfilzomib shares some of the patient inconveniences of Bor-tezomib such as frequent intravenous dosing that often has tobe carried out for months [91]. As a result, efforts have beenundertaken to develop inhibitors that provide the samepotency and selectivity, yet are more patient friendly.

Zhou et al. performed a systematic SAR analysis in orderto develop orally bioavailable Carfilzomib analogs [97]. Thestudy resulted in a orally bioavailable and selective candidate(Table 4, 6a, (2S)-3-Methoxy-2-[(2S)-3-methoxy-2-[(2-methyl-1,3-thiazol-5-yl)formamido]propanamido]-N-[(2S)-1-[(2R)-

2-methyloxiran-2-yl]-1-oxo-3-phenylpropan-2-yl]propanamide)with favorable pharmacological properties [97].

In a proteasome inhibitor pharmacophore study, Screenand colleagues concluded that epoxyketones, althoughexhibiting preference toward the CT-L activity, also inhibitboth T-L and PGPH activities and result in overallincreased cytotoxicity in comparison with the vinyl sulfoneanalogs [84].

Approval of Carfilzomib, undoubtedly, gave epoxyketonesa necessary credibility in the arena of proteasome inhibition.However, the success of epoxyketones in the clinic is still lim-ited by many common patient inconveniences and sideeffects. A continued development of this family of com-pounds should, over time, address many of these concernsand improve the success rate of epoxyketones making thema more safe and effective therapeutic.

4.6 SyrbactinsTwo representatives of this class of natural products: syringo-lins, isolated from Pseudomonas syringae pv syringae [98] andstructurally related glidobactins isolated from a species of Bur-kholderiales [99] had received increased attention as candidatesfor proteasome inhibition [100-103]. This is largely because arepresentative of this class Syringolin A (SylA) has beenobserved to induce apoptosis in human neuroblastoma andovarian cancer cells [104] and subsequent evidence that itinhibited eukaryotic proteasomes via a novel mechanism inwhich the catalytic threonine of the proteasome performsMichael-type addition to the double bond of C4 on the12-membered ring system of the inhibitor [105].

Building on these initial findings Clerc et al., guided by crystalstructure, rationally designed a successful SylA-based lipophilicderivative (Table 5, 7a) that showed a 100-fold improvement inpotency over the original SylA toward the CT-L activity [100].In a later study, the same group developed a series of pegylatedSylA derivatives, the most potent of which (Table 5, 7b, SylA-L-L) inhibited CT-L activity of the purified human 20S

Table 4. Survey of vinyl sulfone, vinyl ester, and epoxyketone-containing compounds (continued).

Compound reference

number

2-D Compound structure Description Inhibition

activity*

Refs.

Epoxyketone-containing compounds6a A,C,H (CT-L)

0.055 ± 0.019 µM[97]

*Inhibition activity reported as IC50 unless otherwise noted.

A: Biochemical assay; B: Cellular assay; C: CT-L activity assayed; D: T-L activity assayed; E: PGPH activity assayed; F: Covalent inhibitor; G: Noncovalent inhibitor;

H: Peptide; I: Peptidomimetic; J: Structure determined, no data reported.

Recent advances in proteasome inhibitor discovery

Expert Opin. Drug Discov. (2013) 8(5) 549

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 14: Recent advances in proteasome inhibitor discovery

proteasome at Ki = 0.401 µM [101]. In order to get further insightinto the activities and modes of action of syrbactins, Clerc et al.developed a Syringolin A--Glidobactin A hybrid (Table 5, 7c) inaddition to GlbA. Both turned out to be potent proteasomeinhibitors, but have displayed different specificity toward consti-tutive proteasome, immunoproteasome, and thymoproteasomecatalytic subunits [106]. Archer and colleagues followed up onthe study of the SylA--GlbA hybrid with its crystal structureand additional assays including biochemical, cellular, and mul-tiple tumor cell lines. The study revealed that the SylA--GlbAhybrid is a promising proteasome inhibitor with a biochemically

assayed CT-L activity inhibition at Ki = 0.0125 µM andcell culture proteasome inhibition at IC50 = 2.87 µM(Table 5, 7c) [102]. Van der Linden and associates employed astrategy of combining structural features of syringolins andother moieties known to improve proteasome inhibition suchas epoxyketones and vinyl sulfones. The study yielded selectiveCT-L inhibitors (Table 5, 7d, tBuO-Val-urea-Val-Leu-EK2and 7e, Benzylamide-Val-urea-Val-Leu2-EK) with activityexceeding that of syringolin A (IC50 = 0.016 µM and 0.008 µMvs 1.3 µM) as well as a potent (IC50 = 0.46 µM) PGPH activityinhibitor (Table 5, 7f, Benzylamide-Val-urea-Val-Leu2-EK2) [103].

Table 5. Survey of syrbactins.

Compound

reference number

2-D Compound structure Description Inhibition activity* Refs.

Syrbactins7a A,C,D,E,F,H (CT-L)

(8.65 ± 1.33) � 10-3µM Ki

[100]

7b A,C,F,H (CT-L) 0.401 ± 0.037 µM Ki [101]

7c A,C,D,E,F,H (CT-L) 0.0125 µM KI (A)(CT-L) 2.87 ± 1.32 µM Ki (B)

[102]

7d A,C,D,E,H 26S (CT-L) 0.016 µM [103]

7e A,C,D,E,H 26S (CT-L) 0.008 µM [103]

7f A,C,D,E,H 26S (PGPH) 0.46 µM [103]

*Inhibition activity reported as IC50 unless otherwise noted.

A: Biochemical assay; B: Cellular assay; C: CT-L activity assayed; D: T-L activity assayed; E: PGPH activity assayed; F: Covalent inhibitor; G: Noncovalent inhibitor;

H: Peptide; I: Peptidomimetic; J: Structure determined, no data reported.

Y. Pevzner et al.

550 Expert Opin. Drug Discov. (2013) 8(5)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 15: Recent advances in proteasome inhibitor discovery

A relative newcomers to the field of proteasomeinhibition, Syrbactins have established themselves as apowerful tool in the hands of medicinal chemists. Contin-ued development of these compounds will result in potentand specific proteasome inhibitors that may see success inthe clinic.

4.7 FlavonoidsThis large class of compounds, frequently found in edibleplants, has received considerable attention due to the evidencesuggesting it has marked antitumor, anti-inflammatory andantioxidant activities [107]. Moreover, flavonoid activities havebeen linked with proteasome inhibition [108,109]. In recent yearsinquiry has continued into the inhibitory effects this widevariety of compounds has on the proteasome.

Chang investigated 26 flavonoids for their inhibitorypotential of 26S isolates across all proteolytic activities [110].The most potent compound (Table 6, 8a, 5,6,3,4-tetrahy-droxy-7-methoxyflavone) exhibited broad micromolar inhibi-tory activity with a slight selectivity toward CT-L [110]. Shimperformed biochemical screening of several flavonoids isolatedfrom the stem of Spatholobus suberectus against the CT-L activity of the 20S proteasome and identified severalcompounds (Table 6, 8b Isoliquiritigenin, 8c, Genisteinand 8d, a.7-Hydroxyflavanone) with inhibitory activity atlow micromolar concentrations [111].

One representative of this class of compounds, epigallocate-chin 3-gallate or EGCG, has been extensively studied as a pro-teasome inhibitor due to evidence linking this compound withthe reduction or growth inhibition of several types ofcancer [112-119] as well as evidence of EGCG’s targeting of theproteasome specifically [120]. In the effort to improve bioavail-ability of EGCG analogs, Huo and colleagues screened severalcompounds for their activity against both 20S and 26S protea-somes using biochemical and cellular assays in addition to per-forming an assay measuring the resistance of each compoundto methylation, which decreases bioavailability, in additionto elucidating the binding mode of EGCG and some of itsanalogs [121-124]. The experiment revealed the most cytotoxiccompound (Table 6, 8e) that is a prodrug of the mostmethylation-resistant inhibitor whose binding mode was thenmodeled computationally to elucidate its mode of actionwithin the b5 site of the proteasome [125]. A complimentarystudy by Bonfili et al. employed a combination of computa-tional studies and experimental assays to evaluate the stabilityof EGCG under neutral-alkaline and cell physiological condi-tions as well as to identify biologically active EGCG oxidativering-fission product (Table 6, 8g) [126]. The metabolite showedboth comparable inhibition of isolated 20S (IC50 = 173 µMfor CT-L activity) across all proteolytic activities as well as similarto EGCG activity in human cervical carcinoma cells [126].

Flavonoids do not exhibit the same level of potency seen inother compounds, which largely renders them unfit for treat-ment purposes. Their low toxicity, however, in combinationwith observed proteasome inhibition and antioxidant

activities make flavonoids a low risk agent that can beincorporated in a daily diet as part of a preventative strategy.

4.8 SalinosporamidesSalinosporamides have received considerable attention dueto recent success of a marine natural product-derived com-pound Salinosporamide A (salino A) that has been showingpromising signs and may become an option for cancertreatment [127-129].

Nguyen et al. have synthesized and enzymatically assayeda series of (-)-salinosporamide A (salino A) derivatives andidentified compounds that exhibit activities comparableto those of the original salino A and better than the activi-ties of Bortezomib with the most potent compound(Table 6, 9a (-)-homosalino) inhibiting CT-L activity atIC50 = 0.0007 µM and 0.0023 µM in 20S and 26S isolatesas compared to salino A at IC50 = 0.0008 µM and0.0025 µM and Bortezomib at IC50 = 0.0026 µM and0.0074 µM [130]. In a different approach, Rachid and col-leagues identified the genes of and utilized the enzymes ofStreptomyces sp. responsible for the biosynthesis of cinna-baramides, compounds closely related to salinosporamides,in order to generate novel compounds with inhibitorypotential toward the proteasome [131]. The activity of theresulting compounds was then tested in 20S isolates andin several tumor cell lines. The most potent (Table 6, 9b,17-chlorocinnabaramide A) of the compounds showedactivity (IC50 = 0.0088 µM against CT-L activity), butfailed to outperform salino A (IC50 = 0.0031 µM) [131].

Marine natural product-derived compounds have servedas a source for promising pharmaceutical agents in thepast [132,133], therefore it is not surprising to see a representa-tive of this class of compounds showing potential when itcomes to inhibition of the proteasome. However, it is a longway from the ocean to the clinic, and in order to surmountthe obstacles faced by a promising lead, an ongoing researcheffort is essential. Salinosporamide has shown the potentialand will, undoubtedly, continue to be of interest to researcherslooking for an effective proteasome inhibitor.

4.9 TMC-95ATMC-95A is a cyclic tripeptide from ascomycete fungusApiospora montagnei. It is one of the few potent noncovalentinhibitors of the proteasome that inhibits all three proteolyticactivities [134]. Its synthesis, however, is a difficult task [135-138]

and in the past few years there have been numerous attemptsto develop its mimics and derivatives [139-141].

Groll and colleagues developed a series of linear TMC-95Amimics, the most potent of which (Table 6, 10a) exhibited anincreased specificity toward CT-L activity of the proteasomein a biochemical assay superior to TMC-95A [142]. A seriesof pegylated monomers and dimers of a known linearTMC-95A mimic [143] were synthesized and evaluated in abiochemical assay by Marechal et al. and found that the pegy-lated dimers are much more potent toward the CT-L activity

Recent advances in proteasome inhibitor discovery

Expert Opin. Drug Discov. (2013) 8(5) 551

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 16: Recent advances in proteasome inhibitor discovery

Table 6. Survey of flavonoids, salinosporamides, and TMC-95A compounds.

Compound

reference number

2-D compound structure Description Inhibition activity* Refs.

Flavonoids8a A,C,D,E,H 26S (CT-L) 14.0 µM [110]

8b A,C (CT-L) 4.88 ± 1.5 µM [111]

8c A,C (CT-L) 9.26 ± 1.2 µM [111]

8d A,C (CT-L) 5.21 ± 1.5 µM [111]

8e A,C (CT-L) 29 µM [123,125]

8f A,C,D,E (CT-L) 173 ± 26 µM Ki [126]

*Inhibition activity reported as IC50 unless otherwise noted.

A: Biochemical assay; B: Cellular assay; C: CT-L activity assayed; D: T-L activity assayed; E: PGPH activity assayed; F: Covalent inhibitor; G: Noncovalent inhibitor;

H: Peptide; I: Peptidomimetic; J: Structure determined, no data reported.

Y. Pevzner et al.

552 Expert Opin. Drug Discov. (2013) 8(5)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 17: Recent advances in proteasome inhibitor discovery

Table 6. Survey of flavonoids, salinosporamides, and TMC-95A compounds (continued).

Compound

reference number

2-D compound structure Description Inhibition activity* Refs.

9a A,C,D,E 20S (CT-L)(7.0 ± 0.4) � 10-3 µM26S (CT-L)(2.3 ± 1.1) � 10-3 µM

[130]

9b A,C,D,E (CT-L)(8.8 ± 5.5) � 10-3 µM

[131]

TMC-95A10a A,C,G,I (CT-L) 1.5 ± 0.1 µM [142]

10b A,C,G,I (CT-L) 0.18 ± 0.01 µM [144]

*Inhibition activity reported as IC50 unless otherwise noted.

A: Biochemical assay; B: Cellular assay; C: CT-L activity assayed; D: T-L activity assayed; E: PGPH activity assayed; F: Covalent inhibitor; G: Noncovalent inhibitor;

H: Peptide; I: Peptidomimetic; J: Structure determined, no data reported.

Recent advances in proteasome inhibitor discovery

Expert Opin. Drug Discov. (2013) 8(5) 553

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 18: Recent advances in proteasome inhibitor discovery

of the proteasome 20S proteasome with the most active(Table 6, 10b) exhibiting activity at IC50 = 0.18 µM as comparedto IC50 = 141 µM of the original linear mimic [144].The noncovalent binding mode of TMC-95A makes this

compound an attractive alternative to its more potent covalentcounterparts. If developed further this compound can exhibitselective, potent and yet reversible, thus potentially less toxic,inhibition of the proteasome.

4.10 Metal complexesOver the years, a number of metal complexes have been inves-tigated for their potential to inhibit activity of the proteasome.Among those receiving attention are gold [145,146], nickel [147,148],zinc [147,148] and to a greater degree copper [34,149-152].Gold complexes initially showed promise when bis-

chelated gold(I) phosphine complexes had induced apoptosisin breast cancer cells [153]. Specific targeting of the proteasomeby gold complexes was performed by Milacic and colleagueswho showed that gold(III) complex had inhibited all threeactivities of 20S proteasome with a preference for CT-Lactivity [145]. More recently, Frezza et al. have tested Nickel(II) and Zinc(II) complexes for their inhibitory potentialagainst 26S and 20S proteasome [148]. While Nickel(II)-con-taining complex did not exhibit any marked activity againstthe target, Zinc(II) complex (Table 7, 11a, [Zn(LIA)2]) hadshown promise by inhibiting CT-L activity in both particlesat low micromolar concentrations [148].Special attention has been given to copper complexes as

potential proteasome inhibitors, in particular as a promisingstrategy in cancer therapy [34,149-152]. It has been shown thatmany types of cancers accumulate copper in their cells at levelsgreater than those of healthy cells, providing avenue for tar-geted treatment of tumors [154-157]. In one of the first studiesthat exploited the copper accumulation in cancer cells, itwas reported that certain copper organic compounds werepotent and selective inhibitors of the CT-L and speculatedthat these otherwise innocuous compounds can becomepotent in the copper-rich environment of cancer cells [34].This hypothesis was supported when the 8-hydroxyquinolineanalog clioquinol and a known copper binding compoundpyrrolidine dithiocarbamate inhibited the CT-L activity ofthe proteasome, blocked proliferation, and induced apoptosisin breast cancer cells while remaining nontoxic to noncancer-ous breast cells [149].In 2009, Hindo et al. characterized and evaluated a series of

Copper (II) complexes for 20S proteasome inhibition. Two ofthe complexes showed inhibition of CT-L activity of the pro-teasome and induction of prostate cancer cell death with themost potent (Table 7, 11b, [Cu(LI)Cl]) inducing cell deathat IC50 = 3.82 µM [151]. Milacic and colleagues performedextract-based, whole-cell, and proliferation assays to screen aseries of 8-hydroxyquinoline analogs [150]. The result showedthat otherwise nontoxic compounds (see Table 7, 11c) exhibittumor-suppressing and proteasome-inhibitory activity whenmixed with copper [150]. Zuo et al. have used a combination

of biological assays and computational methods to profileactivities and modes of action of several amino acid Schiffbase--copper (II) complexes. The investigation resulted in twocomplexes (Table 7, 11d [Cu(L1)(Phen)]·9H2O and 11e, [Cu(L2)(Phen)]·3H2O) that inhibit CT-L activity of both purified20S and cellular 26S proteasomes as well as induce growth inhi-bition and apoptosis in human breast and prostate cancercells [152].

Metal complexing compounds described above provideresearchers with an important avenue of specifically target-ing diseased cells. In particular, the characteristic accumula-tion of copper in cancerous cells provides an additional levelof precision to the inhibitors targeting the proteasome, aprotein equally important to both cancerous and healthycells. Moreover, a wide range of copper binding moietiesmeans additional flexibility in inhibitor design. Theseunique characteristics, in combination with the deeperinsight into the mechanisms of proteasome inhibition willundoubtedly lead up to some intriguing developments inthe field.

4.11 Known therapeutics as proteasome inhibitorsIn addition to discovering novel proteasome inhibitors and thedevelopment of existing compounds to improve their perfor-mance, another approach is evaluating known therapeutic com-pounds for their potential to inhibit the proteasome [158,158-161].The mechanism of action of such compounds in terms of pro-teasome inhibition is usually not known. However, their widerange of activities which often include antiproliferative andanticancer effects makes them good candidates for furtherinquiry regarding their potential as proteasome inhibitors. Inaddition, having been clinically evaluated, such compoundshave well understood toxicity profiles.

Schoof and colleagues synthesized and profiled a series ofderivatives of thiostrepton, a potent antibiotic and an anti-malarial agent [162-164]. Among other goals, the study wasaimed at finding a link between the antiplasmodial activityand the inhibition of the proteasome [158]. As a result, a seriesof new compounds (see Table 7, 13a for a representative com-pound) showed inhibition of CT-L and PGPH activities ofproteasome that correlated with their antiplasmodialactivity [158].

Over the years, a number of anti-HIV drugs including rito-navir [165] and bevirimat [159,166] have been shown to inhibitproteasome. Bevirimat, a known anti-HIV-1 agent is derivedfrom betulinic acid (BA), a compound with a number ofknown biological activities including antibacterial, antimalar-ial, anti-inflammatory, anticancer, antioxidant and anthelmicactivities [167]. Qian and colleagues biochemically assayedinhibition of CT-L activity of the proteasome by a series ofBA derivatives and identified several low micromolar inhibi-tors, the most potent of which (Table 7, 13b) exhibited activityat IC50 = 1.42 µM [167].

Fenbendazole (FZ) (methyl N-(6-phenylsulfanyl-1H-ben-zimidazol-2-yl) carbamate) is a known anthelmintic drug

Y. Pevzner et al.

554 Expert Opin. Drug Discov. (2013) 8(5)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 19: Recent advances in proteasome inhibitor discovery

Table 7. Survey of metal complexes and known therapeutic classes of compounds.

Compound reference

number

2-D Compound structure Description Inhibition

activity*

Refs.

Metal complexes11a A,C,F 26S (CT-L) 5.7 µM

20S (CT-L) 16.6 µM[148]

11b A,C (CT-L) 3.82 ± 0.01 µM [151]

11c A,C (CT-L) 26% inhib.at 5 µM(CT-L) 52% inhib.at 10 µM

[150]

11d A,C (CT-L) 0.0133 µM [152]

*Inhibition activity reported as IC50 unless otherwise noted.

A: Biochemical assay; B: Cellular assay; C: CT-L activity assayed; D: T-L activity assayed; E: PGPH activity assayed; F: Covalent inhibitor; G: Noncovalent inhibitor;

H: Peptide; I: Peptidomimetic; J: Structure determined, no data reported.

Recent advances in proteasome inhibitor discovery

Expert Opin. Drug Discov. (2013) 8(5) 555

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 20: Recent advances in proteasome inhibitor discovery

Table 7. Survey of metal complexes and known therapeutic classes of compounds (continued).

Compound reference

number

2-D Compound structure Description Inhibition

activity*

Refs.

11e A,C (CT-L) 0.0113 µM [152]

Known therapeutics classes of compounds13a See reference for structure A,C,D,E (CT-L) 0.32 µM

(PGPH) 0.1 µM[158]

13b A,C (CT-L) 1.42 µM [167]

13c A,C (CT-L) 1.2 -- 1.6 µM [160]

13d A,C,D,E (CT-L) 0.75 µM(T-L) 0.53 µM(PGPH) 0.46 µM

[161]

*Inhibition activity reported as IC50 unless otherwise noted.

A: Biochemical assay; B: Cellular assay; C: CT-L activity assayed; D: T-L activity assayed; E: PGPH activity assayed; F: Covalent inhibitor; G: Noncovalent inhibitor;

H: Peptide; I: Peptidomimetic; J: Structure determined, no data reported.

Y. Pevzner et al.

556 Expert Opin. Drug Discov. (2013) 8(5)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 21: Recent advances in proteasome inhibitor discovery

used to treat pinworm in animals [160]. Its two benzimidazolederivatives mebendazole(methyl 5-benzoyl-2-benzimidazole-carbamate) and albendazole (methyl (5-propylthio)-1H-ben-zimidazol-2-yl carbamate methylester) have also been usedto treat pulmonary helminitic infection and alveolar echino-coccosis in humans [168] with low incidence of severe sideeffects despite long-term high dosage treatment [169,170]. Dograand Mukhopadhyay performed a comprehensive inquiry intothe effects that FZ had on tumor cells and proteasomalactivity specifically. The study demonstrated, among otherfindings, that FZ inhibits growth and induces apoptosisof human non-small cell lung cancer cells and inhibitsproteasome function at low micromolar concentrations(Table 7, 13c) [160].

Santoro and colleagues assessed the ability of porphyrins,molecules that among a multitude of other uses serve as pho-tosensitizers in photodynamic cancer therapy [171,172], to actas proteasome inhibitors. Inhibitory capacity of several vari-ous types of porphyrins toward 20S and 26S proteasomeswere assessed using cell lysates and protein extracts [161].The experiment demonstrated that tetracationic porphyrinsand some of their metallo derivatives may inhibit all threeactivities of the proteasome, with the most active compound(Table 7, 13d, meso-tetrakis(4-N-methylpyridyl)-porphyrin)exhibiting inhibition in the submicromolar range [161].

Lastly, the anti-alcoholism drug disulfiram, that has alsobeen shown to bind copper [173,174] has been closely investi-gated as a potential anticancer agent and a proteasome inhibitorin particular [174,175]. Chen et al. demonstrated an induction ofapoptosis in breast cancer cells and xenografts treated withdisulfiram and showed an inhibition of CT-L activity of puri-fied 20S proteasome by disulfiram (Table 7, 13e) in presenceof copper at IC50 » 7.5 µM [174]. Conticello and colleaguessimilarly saw cytotoxic effect that disulfiram--copper complexexhibited in hematological malignant cells, however an inquiryinto the effects on the 20S proteasome showed no direct evi-dence of activity of either disulfiram--copper or disulfiramalone [175].

Drug repurposing has, of late, been considered a viablestrategy for getting drugs through the regulatory hurdles andinto clinic [176]. Existing therapeutics have known toxicity

profiles and although their primary clinical purpose is differ-ent from the one sought, their effects may provide importantinitial clues toward their potential off-target activity. A closerlook at the multitude of known therapeutics is bound to resultin a number good candidates for development of proteasomeinhibitors.

4.12 Newly emerging classes of compoundsIn addition to some of the classes of compounds that havealready established themselves as proteasome inhibitors,others are just emerging onto the scene. As investigatorsexplore alternatives to some of the existing inhibition strate-gies and continue to look for potent compounds with goodbioavailability and fewer side effects, new families of com-pounds have been emerging and receiving more attention inrecent years. One of such compound is a cyclic heptapeptideisolated from a myxobacterium argyrin. Initially, it was shownthat inhibition of proteasome by argyrin A has led to the sta-bilization of the tumor suppressor protein p27 [177]. In a seriesof subsequent studies, argyrin A was characterized and theproteasome inhibitory capacities of its derivatives wereelucidated (Table 8, 12a, argyrin F) [178,179].

Building on their previous work with vinyl esters [85],Baldisserotto and colleagues synthesized and evaluated a seriesof novel N-acylpyrrole-containing peptide-based compoundswith the hypothesis that the new pharmacophore will beable to mimic the functionality of vinyl ester moiety [180].These were evaluated in purified proteasome and cellularassays where the most potent compounds (Table 8, 12b,Z-NH-(CH2)7-Val-Ser-Leu-VAP) exhibited activities atIC50 = 0.036 µM and IC50 = 0.058 µM toward PGPHactivity in biochemical and cellular assays respectively [180].

The previously mentioned curcumin, a natural product,whose fluorine-containing derivatives were developed into pro-teasome inhibitors [62], itself is a promising anticanceragent [181-183]. Curcumin has been the subject of a recent studywhere its amino acid conjugates (Table 8, 12c, (1E,6E)-1,7-Bis(4-glycinoyl-3-methoxyphenyl)hepta-1,6-diene-3,5-dionehydrochloride, 12d, (1E,6E)-1,7-Bis(4-alaninoyl-3-methoxy-phenyl)hepta-1,6-diene-3,5-dione hydrochloride and 12e, (1E,6E)-1,7-Bis(4-valinoyl-3-methoxyphenyl)1,6-diene-3,5-dione

Table 7. Survey of metal complexes and known therapeutic classes of compounds (continued).

Compound reference

number

2-D Compound structure Description Inhibition

activity*

Refs.

13e A,B,C 20S (CT-L) 7.5 µM [174]

*Inhibition activity reported as IC50 unless otherwise noted.

A: Biochemical assay; B: Cellular assay; C: CT-L activity assayed; D: T-L activity assayed; E: PGPH activity assayed; F: Covalent inhibitor; G: Noncovalent inhibitor;

H: Peptide; I: Peptidomimetic; J: Structure determined, no data reported.

Recent advances in proteasome inhibitor discovery

Expert Opin. Drug Discov. (2013) 8(5) 557

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 22: Recent advances in proteasome inhibitor discovery

hydrochloride) demonstrated a potent antiproliferative effectagainst a prostate cancer cell line [184].Krunic et al. evaluated two cyclic peptides isolated from cya-

nobacteria, scytonemides A and B, and demonstrated that in a

biochemical assay scytonemide A (Table 8, 12f) inhibited 20Sproteasome on the nanomolar scale. The compound, however,did not perform as well in a human cancer cell line, which wasspeculated to be due to the metabolic instability of the

Table 8. Survey of newly emerging classes of compounds.

Compound

reference

number

2-D Compound structure Description Inhibition

activity*

Refs.

Newly emerging classes of compounds12a See reference for structure A,C,D,E,I (T-L) 0.038 ±

0.0065 µM(PGPH) 0.035 ±0.007 µM(CT-L) 0.028 ±0.0025 µM

[178]

12b A,B,C,D,E (PGPH) 0.036 µM (A)(PGPH) 0.058 µM (B)(CT-L) 0.028 ±0.0025 µM

[178]

12c A,C (CT-L) 70% inhib.at 50 µM

[184]

12d A,C (CT-L) 70% inhib.at 50 µM

[184]

12e A,C (CT-L) 38 -- 71%inhib. at 5 -- 10 µM

[184]

12f A,C (CT-L) 0.096 µM [185]

*Inhibition activity reported as IC50 unless otherwise noted.

A: Biochemical assay; B: Cellular assay; C: CT-L activity assayed; D: T-L activity assayed; E: PGPH activity assayed; F: Covalent inhibitor; G: Noncovalent inhibitor;

H: Peptide; I: Peptidomimetic; J: Structure determined, no data reported.

Y. Pevzner et al.

558 Expert Opin. Drug Discov. (2013) 8(5)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 23: Recent advances in proteasome inhibitor discovery

Table 8. Survey of newly emerging classes of compounds (continued).

Compound

reference

number

2-D Compound structure Description Inhibition

activity*

Refs.

12g A,B,C,D,E (CT-L) 2.2 ±0.3 µM (A)(CT-L) 1.9 µM

[188]

12h A,C,D,E (T-L) 9.1 µM(PGPH) 6.5 µM

[188]

12i A,B,C,D,E (CT-L) 2.5 µM(PGPH) 1.6 µM

[189]

12j A,C (CT-L) 1 � 10-3 µM [190]

12k A,C (CT-L) 7 � 10-3 µM [190]

12l A,B,C,D,E (CT-L) 0.64 µM (B)(PGPH) 0.15 µM (B)

[192]

*Inhibition activity reported as IC50 unless otherwise noted.

A: Biochemical assay; B: Cellular assay; C: CT-L activity assayed; D: T-L activity assayed; E: PGPH activity assayed; F: Covalent inhibitor; G: Noncovalent inhibitor;

H: Peptide; I: Peptidomimetic; J: Structure determined, no data reported.

Recent advances in proteasome inhibitor discovery

Expert Opin. Drug Discov. (2013) 8(5) 559

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 24: Recent advances in proteasome inhibitor discovery

compound [185]. It was concluded, however, that improvementsmay be possible via chemical modifications [185]. As a follow-up to a study, where in search for novel scaffolds for proteasomeinhibition by assessing several naturally occurring acids, litho-cholic acid was identified as a potential candidate [186,187] Dangand associates synthesized and evaluated a series of lithocholicacid derivatives [188]. One of the resulting compounds(Table 8, 12g, 3a-O-(3,3-Tetramethyleneglutaryl)-lithocholicacid) inhibited CT-L activity of the 20S proteasome atIC50 = 2.2 µM in a biochemical assay as well as exhibited protea-some inhibition in a cellular assay. Another compound (Table 8,12h, 3a-O-(3-Methylenecarboxy-benzylcarbonyl)-lithocholic acid)inhibited CT-L, T-L and PGPH activities at IC50 = 1.9 µM,IC50 = 9.1 µM and IC50 = 6.5 µM respectively [188].Lansdell et al. evaluated a series of oroidin-derived

alkaloids for proteasome inhibitory activity in both biochem-ical and cellular assays and found them to inhibit the CT-L and PGPH activities of both the human 20S proteasomeand 20S immunoproteasome cores through irreversible bind-ing in the low nanomolar range. The most potent inhibitor(Table 8, 12i (-)-Palauamine) inhibited the CT-L activity atIC50 = 2.5 µM and PGPH activity at IC50 = 1.6 µM aswell as exhibiting inhibition of protein degradation incells [189].While in the process of generating a series of sterically

constrained oxazoline derivatives, Dunn and colleagues seren-dipitously discovered a series of (2S, 3R)-2-amino-3-hydroxy-butyric acid derivatives by performing a fluorogenic substrateassay followed by an intracellular activity assay [190]. Theexperiment led to potent leads (Table 8, 12j and 12k) thateventually resulted in development of a promising inhibitordelanzomib [57].Margarucci et al., in an investigation of targets of a marine

natural product with known anti-inflammatory properties [191],petrosaspongiolide M (PM), performed a chemical proteomicsstudy and identified the proteasome as a target [192]. Furtheranalysis of inhibitory potential using biochemical and cellularassays indicated PM to be a potent inhibitor with inhibitionof CT-L activity at IC50 = 0.64 µM and PGPH activity atIC50 = 0.15 µM in cells. (Table 8, 12l, petrosaspongiolideM) [192]. In a later study, Margarucci and colleagues have tested

a series of compounds belonging to petrosaspongiolide family aswell as several synthetic analogs of PM [193]. This resulted inanother compound (Table 8, 12m) that compared to PM in a puri-fied 20S activity assay at IC50 = 0.07 µM vs IC50 = 0.085 µMfor CT-L activity and IC50 = 0.07 µM vs IC50 = 1.05 µM forPGPH activity as well as showed activity in cells [193].

The variety of compounds showing promise when it comesto proteasome inhibition highlights the importance of contin-ued inquiry and interest in novel compounds and compoundfamilies that come from a variety of sources. The proteasomeis an awe-inspiring piece of molecular machinery and to tameit requires looking far beyond a small number of commonscaffolds and chemical moieties. Constantly looking for the“next big thing,” although, seemingly fruitless in some cases,is a strategy of considerable importance when it comes totargeting the proteasome.

5. Expert opinion

What should be immediately obvious is the diversity of chem-ical structures that can inhibit the proteasome. In this review,we catalogued more than a dozen different classes of mole-cules. Interestingly, activities of the classes display a commonrange from one group to another with low micromolar tomid-nanomolar being the frame of inhibitory potency; at leastas far as IC50s are concerned. A common motif in drug dis-covery is the screening-to-optimization approach, which isalso discussed in this manuscript. Clearly, the proteasome isamenable to inhibition by a wide variety of scaffolds andmotifs. This is likely due to several factors: i) the active sitessignificantly differ in their recognition pockets, ii) the activesites are very large as they accommodate peptides allowingsubstantial surface area for inhibitor binding, and iii) the cat-alytic THR1, common to each of the proteasome’s threeactivities, is a ready target for chemical modification. Whilethese factors can contribute to difficulty in isolating a givencompound’s mode of action, the process of identifying alead compound is relatively straight forward and such effortstend to be successful. However, characterizing and optimizinga compound (in regards to mechanism, specificity, andpotency) can be difficult. This leads to a situation where

Table 8. Survey of newly emerging classes of compounds (continued).

Compound

reference

number

2-D Compound structure Description Inhibition

activity*

Refs.

12m A,B,C,D,E (CT-L) 0.07 µM (B)(PGPH) 0.07 µM (B)

[193]

*Inhibition activity reported as IC50 unless otherwise noted.

A: Biochemical assay; B: Cellular assay; C: CT-L activity assayed; D: T-L activity assayed; E: PGPH activity assayed; F: Covalent inhibitor; G: Noncovalent inhibitor;

H: Peptide; I: Peptidomimetic; J: Structure determined, no data reported.

Y. Pevzner et al.

560 Expert Opin. Drug Discov. (2013) 8(5)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 25: Recent advances in proteasome inhibitor discovery

identification of an inhibitor is relatively easy compared tocreating a lead candidate suitable for clinical trials.

Several major issues present themselves when designingproteasome inhibitors. The first is whether to target the 20Sor 26S complex. Assays examining the 26S are more difficultthan those looking at the 20S particle [9]. However, it is the26S that is found within the cell and, as has been shownwith the HIV protease inhibitor ritonavir, there are occasionswhere an inhibitor of the 20S can actually be an activator ofthe 26S via a not yet fully understood mechanism [165,194,195].The next question is whether or not to target one or moreof the activities of the proteasome. The chymotrypsin-like activity is a common target, especially in anticancerresearch, as inhibition of this activity is strongly correlatedwith induction of apoptosis in cancer cells. However, inhibi-tion of all three activities may be necessary for potency andto fully interrupt protein degradation. To this end, modelingwill be an important strategic approach since the three sitesare similar in their catalytic regions but differ greatly withintheir substrate recognition regions, as should be anticipated.

Tissue targeting, particularly in regards to cancer, is a majorissue in proteasome inhibition. Bortezomib, the first clinicallyapproved proteasome inhibitor, demonstrates significant tox-icity. Many of the off-target effects are likely derived fromthe reactive “warhead” of the compound, but it should alsobe appreciated that many tissues rely on proteasome activity.It has been reported that normal cells are “resistant” to apo-ptosis caused by proteasome inhibition. However, this doesnot render those cells immune to proteasome inhibition orresistant to other off-target effects that may arise due to thepresence of warheads or the need for higher or longer dosesof inhibitor. Therefore, tissue targeting and high potencymay be more important and will provide better protectionfrom toxic effects. Obviously, increased potency is desirablein order to reduce load on a patient but this is offset by thetrend that covalent or irreversible inhibitors, while morepotent, have reactive groups that increase the risk of off-target effects. Thus, tissue targeting may be the primarymeans of avoiding toxicity especially in cancer therapeutics.After all, if an inhibitor is going to kill cancer cells and istargeted specifically to cancer tissues then toxicity fromsecondary action is less consequential.

With this in mind, different avenues to specifically targetcells or tissues of therapeutic interest should be carefullyexplored. In the case of cancer, its hallmark of accumulatingcopper seems an obvious path to targeted therapy by coppercomplexing compounds. The ligands of such complexes alonecould be designed to have minimal toxicity in the absence ofcopper. In addition, the concentration of copper would dic-tate the concentration of active complexes; thus, reducingconcentration of active compounds in healthy cells to belowtoxic levels. Moreover, a copper complex carrying a reactivegroup would require a lesser dose of the drug upon deliveryreducing the concentrations of active complexes in healthycells even further.

Promising results achieved with low-toxicity naturalcompounds and their derivatives should also be noted.Even though these compounds may not lead us to thenext big thing in proteasome inhibition as a therapy, theiruse as a preventative measure vis-a-vis diet can help lowerincidence of disease. This is extremely valuable in thelong term.

In addition to developing known tried and true scaffolds,the approach of screening large libraries should not be over-looked. The diversity of proteasome inhibitors is evident inthis review, however chemical space is infinitely large andcasting the net wide will, undoubtedly, yield novel com-pounds that may develop into promising clinical candidates.Use of modern high-throughput techniques in combinationwith virtual screening will allow researchers to interrogate amuch larger body of compounds.

Development and discovery of proteasome inhibitors hasbeen steadily gaining momentum. As more investigators puttheir efforts toward conquering this target and strike that per-fect balance of potency, toxicity, and bioavailability, the nextfew years may result in more successful clinical candidatesand therapeutics.

Declaration of interest

This work was supported by the Chemical Biology Corefacility at the Moffitt Cancer Center and Research Instituteand the University of South Florida College of Artsand Sciences.

Recent advances in proteasome inhibitor discovery

Expert Opin. Drug Discov. (2013) 8(5) 561

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 26: Recent advances in proteasome inhibitor discovery

BibliographyPapers of special note have been highlighted as

either of interest (�) or of considerable interest(��) to readers.

1. Rubin DM, Finley D. The proteasome:

a protein-degrading organelle? Curr Biol

1995;5:854-4

2. Baumeister W, Walz J, Proteolysis C.

The proteasome: paradigm review of a

self-compartmentalizing protease. Cell

1998;92:367-80

3. Ciechanover A, Orian A, Schwartz AL.

Ubiquitin-mediated proteolysis:

biological regulation via destruction.

Bioessays 2000;22(5):442-51

4. Kerscher O, Felberbaum R,

Hochstrasser M. Modification of proteins

by ubiquitin and ubiquitin-like proteins.

Annu Rev Cell Dev Biol 2006;22:159-80

5. Elsasser S, Gali RR, Schwickart M, et al.

Proteasome subunit Rpn1 binds

ubiquitin-like protein domains.

Nat Cell Biol 2002;4(9):725-30

6. Pickart CM, Cohen RE. Proteasomes

and their kin: proteases in the machine

age. Nat Rev Mol Cell Biol

2004;5(3):177-87

7. Groll M, Ditzel L, L€owe J, et al.

Structure of 20S proteasome from yeast

at 2.4 A resolution. Nature (London)

1997;386(6624):463-71

8. Borissenko L, Groll M. 20S proteasome

and its inhibitors: crystallographic

knowledge for drug development.

Chem Rev 2007;107(3):687-717

9. Tsvetkov L, Nanjundan M, Domino M,

Daniel KG. The ubiquitin-proteasome

system and assays to determine responses

to inhibitors. Expert Opin Drug Discov

2010;5(12):1221-36

10. Kisselev AF, van der Linden WA,

Overkleeft HS. Proteasome inhibitors:

an expanding army attacking a unique

target. Chem Biol 2012;19(1):99-115

11. Kloetzel PM, Ossendorp F. Proteasome

and peptidase function in MHC-class-I-

mediated antigen presentation.

Curr Opin Immunol 2004;16(1):76-81

12. Murata S, Takahama Y, Tanaka K.

Thymoproteasome: probable role in

generating positively selecting peptides.

Curr Opin Immunol 2008;20(2):192-6

13. Rajkumar SV, Richardson PG,

Hideshima T, Anderson KC. Proteasome

inhibition as a novel therapeutic target in

human cancer. J Clin Oncol

2005;23(3):630-9

14. An W, Hwang S, Trepel J, et al.

Protease inhibitor-induced apoptosis:

accumulation of wt p53, p21WAF1/

CIP1, and induction of apoptosis are

independent markers of proteasome

inhibition. Leukemia 2000;14(7):1276

15. Almond J, Cohen G, et al. The

proteasome: a novel target for cancer

chemotherapy. Leukemia 2002;16(4):433

16. Paul S. Dysfunction of the

ubiquitin-proteasome system in multiple

disease conditions: therapeutic

approaches. Bioessays

2008;30(11-12):1172-84

17. Willis MS, Townley-Tilson WHD,

Kang EY, et al. Sent to destroy the

ubiquitin proteasome system regulates

cell signaling and protein quality control

in cardiovascular development and

disease. Circ Res 2010;106(3):463-78

18. Yu X, Kem DC. Proteasome inhibition

during myocardial infarction.

Cardiovasc Res 2010;85(2):312-20

19. Flechner SM, Fatica R, Askar M, et al.

The role of proteasome inhibition with

bortezomib in the treatment of

antibody-mediated rejection after

kidney-only or kidney-combined organ

transplantation. Transplantation

2010;90(12):1486

20. Gante J. Peptidomimeticstailored enzyme

inhibitors. Angew Chem Int Ed Engl

1994;33(17):1699-720

21. Kisselev AF, Goldberg AL. Proteasome

inhibitors: from research tools to drug

candidates. Chem Biol 2001;8(8):739-58

22. Food US, Administration D. About the

Center for Drug Evaluation and Research

-- Velcade (bortezomib) is Approved for

Initial Treatment of Patients with

Multiple Myeloma.

2009. Available from: http://www.fda.

gov/AboutFDA/CentersOffices/

OfficeofMedicalProductsandTobacco/

CDER/ucm094633.htm

23. Food US, Administration D. About the

Center for Drug Evaluation and Research

-- FDA approves bortezomib (Velcade)

for the treatment of patients with mantle

cell lymphoma who have received at least

one prior therapy. 2009. Available from:

http://www.fda.gov/AboutFDA/Centers

Offices/OfficeofMedicalProductsand

Tobacco/CDER/ucm094929.htm

24. Mujtaba T, Dou Q. Advances in the

understanding of mechanisms and

therapeutic use of bortezomib.

Discov Med 2011;12(67):471-80

25. Adams J, et al. Potential for proteasome

inhibition in the treatment of cancer.

Drug Discov Today 2003;8(7):307

26. Zhu Y, Zhao X, Zhu X, et al. Design,

synthesis, biological evaluation, and

structure-activity relationship (SAR)

discussion of dipeptidyl boronate

proteasome inhibitors, part I:

comprehensive understanding of the SAR

of a-amino acid boronates. J Med Chem

2009;52(14):4192-9

27. Food US, Administration D. Approved

Drugs -- Carfilzomib.

2012. Available from: http://www.fda.

gov/Drugs/InformationOnDrugs/

ApprovedDrugs/ucm312945.htm

28. Fostier K, De Becker A, Schots R.

Carfilzomib: a novel treatment in

relapsed and refractory multiple

myeloma. Onco Targets Ther

2012;5:237

29. McConkey DJ, Zhu K. Mechanisms of

proteasome inhibitor action and

resistance in cancer. Drug Resist Updat

2008;11(4):164-79

30. Walters WP, Stahl MT, Murcko MA.

Virtual screening-an overview.

Drug Discov Today 1998;3(4):160-78

31. Shoichet BK. Virtual screening of

chemical libraries.

Nature;2004;432(7019):862-5

32. Kitchen DB, Decornez H, Furr JR,

Bajorath J. Docking and scoring in

virtual screening for drug discovery:

methods and applications. Nat Rev

Drug Discov 2004;3(11):935-49

33. NCI/NIH DTP. DTP -- Diversity Set

Information. Available from: http://dtp.

nci.nih.gov/branches/dscb/diversity.html

34. Daniel KG, Gupta P, Harbach RH,

et al. Organic copper complexes as a new

class of proteasome inhibitors and

apoptosis inducers in human cancer

cells. Biochem Pharmacol

2004;67(6):1139-51

35. Lavelin I, Beer A, Kam Z, et al.

Discovery of novel proteasome inhibitors

using a high-content cell-based screening

system. PLoS One 2009;4(12):e8503

36. Lawrence HR, Kazi A, Luo Y, et al.

Synthesis and biological evaluation of

Y. Pevzner et al.

562 Expert Opin. Drug Discov. (2013) 8(5)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 27: Recent advances in proteasome inhibitor discovery

naphthoquinone analogs as a novel class

of proteasome inhibitors.

Bioorg Med Chem 2010;18(15):5576-92

37. Ge Y, Kazi A, Marsilio F, et al.

Discovery and synthesis of

hydronaphthoquinones as novel

proteasome inhibitors. J Med Chem

2012;55(5):1978-98

38. Xu K, Xiao Z, Tang YB, et al. Design

and synthesis of naphthoquinone

derivatives as antiproliferative agents and

20S proteasome inhibitors. Bioorg Med

Chem Lett 2012;22(8):2772-4

39. Demasi M, Felicio A, Pacheco A, et al.

Studies on terrein as a new class of

proteasome inhibitors. J Braz Chem Soc

2010;21(2):299-305

40. Blackburn C, Gigstad KM, Hales P,

et al. Characterization of a new series of

non-covalent proteasome inhibitors with

exquisite potency and selectivity for the

20S beta5-subunit. Biochem J

2010;430(Pt 3):461.. This article is a good example of how

compound library screening can lead

to identification of novel potent

inhibitors. A multistep approach starts

with a library of 350,000 compounds,

utilizes a number of assays and ligand

optimization techniques.

41. Ausseil F, Samson A, Aussagues Y, et al.

High-throughput bioluminescence

screening of ubiquitin-proteasome

pathway inhibitors from chemical and

natural sources. J Biomol Screen

2007;12(1):106-16

42. Long C, Beck J, Cantagrel F, et al.

Proteasome Inhibitors from Neoboutonia

melleri. J Nat Prod 2011;75(1):34-47

43. Basse N, Montes M, Marechal X, et al.

Novel organic proteasome inhibitors

identified by virtual and in vitro

screening. J Med Chem

2009;53(1):509-13

44. Gallastegui N, Beck P, Arciniega M,

et al. Hydroxyureas as noncovalent

proteasome inhibitors. Angew Chem

Int Ed 2012;51(1):247-9

45. Zhu Y, Yao S, Xu B, et al. Design,

synthesis and biological evaluation of

tripeptide boronic acid proteasome

inhibitors. Bioorg Med Chem

2009;17(19):6851-61

46. Zhu Y, Zhu X, Wu G, et al. Synthesis,

in vitro and in vivo biological evaluation,

docking studies, and structure-activity

relationship (SAR) discussion of

dipeptidyl boronic acid proteasome

inhibitors composed of beta-amino acids.

J Med Chem 2010;53(5):1990-9. A comprehensive look at the

structure--activity relationship of a

series of boronic acid-containing

inhibitors is presented in this study. In

addition to synthesis, a thorough

activity evaluation and computational

modeling help gain important insight

into the potential of this family of

compounds as proteasome inhibitors.

47. Zhu Y, Wu G, Zhu X, et al. Synthesis,

in vitro and in vivo biological evaluation,

and comprehensive understanding of

structure--activity relationships of

dipeptidyl boronic acid proteasome

inhibitors constructed from beta-amino

acids. J Med Chem 2010;53:8619-26

48. Momose I, Sekizawa R, Hashizume H,

et al. Tyropeptins A and B, new

proteasome inhibitors produced by

Kitasatospora sp. MK993-dF2. I.

Taxonomy, isolation, physico-chemical

properties and biological activities.

J Antibiot 2001;54(12):997-1003

49. Momose I, Sekizawa R, Hirosawa S,

et al. Tyropeptins A and B, new

proteasome inhibitors produced by

Kitasatospora sp. MK993-dF2. II.

Structure determination and synthesis.

J Antibiot 2001;54(12):1004-12

50. Watanabe T, Momose I, Abe M, et al.

Synthesis of boronic acid derivatives of

tyropeptin: proteasome inhibitors.

Bioorg Med Chem Lett

2009;19(8):2343-5

51. Watanabe T, Abe H, Momose I, et al.

Structure-activity relationship of boronic

acid derivatives of tyropeptin: Proteasome

inhibitors. Bioorg Med Chem Lett

2010;20(19):5839-42

52. Asai A, Hasegawa A, Ochiai K, et al.

Belactosin A, a novel antitumor

antibiotic acting on cyclin/CDK

mediated cell cycle regulation, produced

by Streptomyces sp. ChemInform

2000;31:24

53. Asai A, Tsujita T, Sharma SV, et al.

A new structural class of proteasome

inhibitors identified by microbial

screening using yeast-based assay.

Biochem Pharmacol 2004;67(2):227-34

54. Nakamura H, Watanabe M, Ban HS,

et al. Synthesis and biological evaluation

of boron peptide analogues of Belactosin

C as proteasome inhibitors. Bioorg Med

Chem Lett 2009;19(12):3220-4

55. Lei M, Zhao X, Wang Z, Zhu Y.

Pharmacophore modeling, docking

studies, and synthesis of novel dipeptide

proteasome inhibitors containing boron

atoms. J Chem Inf Model

2009;49(9):2092-100

56. Iqbal M, Messina McLaughlin PA,

Dunn D, et al. Proteasome inhibitors for

cancer therapy. Bioorg Med Chem

2012;20:2362-68

57. Dorsey BD, Iqbal M, Chatterjee S, et al.

Discovery of a potent, selective, and

orally active proteasome inhibitor for the

treatment of cancer. J Med Chem

2008;51(4):1068-72

58. Begue JP, Bonnet-Delpon D. Recent

advances (1995-2005) in fluorinated

pharmaceuticals based on natural

products. J fluorine Chem

2006;127(8):992-1012

59. Hagmann WK. The many roles for

fluorine in medicinal chemistry.

ChemInform 2008;39:45

60. Isanbor C, OHagan D. Fluorine in

medicinal chemistry: a review of

anti-cancer agents. J Fluorine Chem

2006;127(3):303-19

61. Begue JP, Bonnet-Delpon D. Wiley

Online Library; 2007

62. Padhye S, Yang H, Jamadar A, et al.

New difluoro Knoevenagel condensates

of curcumin, their Schiff bases and

copper complexes as proteasome

inhibitors and apoptosis inducers in

cancer cells. Pharm Res

2009;26(8):1874-80

63. Kirk KL. Selective fluorination in drug

design and development: an overview of

biochemical rationales. Curr Top

Med Chem 2006;6(14):1447-56

64. B€ohm HJ, Banner D, Bendels S, et al.

Fluorine in medicinal chemistry.

ChemBioChem 2004;5(5):637-43

65. Formicola L, Marechal X, Basse N, et al.

Novel fluorinated pseudopeptides as

proteasome inhibitors. Bioorg Med

Chem Lett 2009;19(1):83-6

66. Abas F, Lajis NH, Shaari K, et al.

A labdane diterpene glucoside from the

rhizomes of curcuma mangga.

J Nat Prod 2005;68(7):1090-3

67. Bisht S, Feldmann G, Soni S, et al.

Polymeric nanoparticle-encapsulated

curcumin (nanocurcumin): a novel

strategy for human cancer therapy.

J Nanobiotechnology 2007;5(3):1-18

Recent advances in proteasome inhibitor discovery

Expert Opin. Drug Discov. (2013) 8(5) 563

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 28: Recent advances in proteasome inhibitor discovery

68. Jagetia GC, Rajanikant G. Role of

curcumin, a naturally occurring phenolic

compound of turmeric in accelerating the

repair of excision wound, in mice

whole-body exposed to various doses of

g-radiation. J Surg Res

2004;120(1):127-38

69. Milacic V, Banerjee S,

Landis-Piwowar KR, et al. Curcumin

inhibits the proteasome activity in

human colon cancer cells in vitro and in

vivo. Cancer Res 2008;68(18):7283-92

70. Geurink PP, Liu N, Spaans MP, et al.

Incorporation of fluorinated

phenylalanine generates highly specific

inhibitor of proteasomes

chymotrypsin-like sites. J Med Chem

2010;53(5):2319-23

71. L€owe J, Stock D, Jap B, et al. Crystal

structure of the 20S proteasome from the

archaeon T. acidophilum at

3.4 A resolution. Science (New York,

NY). 1995;268(5210):533

72. Vinitsky A, Michaud C, Powers JC,

Orlowski M. Inhibition of the

chymotrypsin-like activity of the

pituitary multicatalytic proteinase

complex. Biochemistry

1992;31(39):9421-8

73. Momose I, Umezawa Y, Hirosawa S,

et al. Structure-based design of

derivatives of tyropeptin A as the potent

and selective inhibitors of mammalian

20S proteasome. Bioorg Med Chem Lett

2005;15(7):1867-71

74. Loidl G, Groll M, Musiol HJ, et al.

Bifunctional inhibitors of the trypsin-like

activity of eukaryotic proteasomes.

Chem Biol 1999;6(4):197-204

75. Ma Y, Chen B, Liu D, et al.

MG132 treatment attenuates cardiac

remodeling and dysfunction following

aortic banding in rats via the NF-kB/TGFb1 pathway. Biochem Pharmacol

2011;81(10):1228-36

76. Donkor I. A survey of calpain inhibitors.

Curr Med Chem 2000;7(12):1171-88

77. Tsubuki S, Saito Y, Tomioka M, et al.

Differential inhibition of calpain and

proteasome activities by peptidyl

aldehydes of di-leucine and tri-leucine.

J Biochem 1996;119(3):572-6

78. Sherwood SW, Kung AL, Roitelman J,

et al. In vivo inhibition of cyclin B

degradation and induction of cell-cycle

arrest in mammalian cells by the neutral

cysteine protease inhibitor

N-acetylleucylleucylnorleucinal. Proc Natl

Acad Sci 1993;90(8):3353-7

79. Jullig M, Zhang W, Ferreira A, Stott N.

MG132 induced apoptosis is associated

with p53-independent induction of

pro-apoptotic Noxa and transcriptional

activity of beta-catenin. Apoptosis

2006;11(4):627-41

80. Han YH, Moon HJ, You BR, Park WH.

The effect of MG132, a proteasome

inhibitor on HeLa cells in relation to cell

growth, reactive oxygen species and

GSH. Oncol Rep 2009;22(1):215-21

81. Ma Y, Xu B, Fang Y, et al. Synthesis and

SAR study of novel peptide aldehydes as

inhibitors of 20S proteasome. Molecules

2011;16(9):7551-64. This study is a good example how an

insight gained from a previously

published crystallographic study can

serve as a good starting point for

further development of a family of

compounds using a combination of

experimental and

computational methods.

82. Ettari R, Bonaccorso C, Micale N, et al.

Development of novel peptidomimetics

containing a vinyl sulfone moiety as

proteasome inhibitors. ChemMedChem

2011;6(7):1228-37

83. Vivier M, Rapp M, Galmier MJ, et al.

New aldehyde and vinylsulfone

proteasome inhibitors for targeted

melanoma therapy. Eur J Med Chem

2011;46(11):5705-10

84. Screen M, Britton M, Downey SL, et al.

Nature of pharmacophore influences

active site specificity of proteasome

inhibitors. J Biol Chem

2010;285(51):40125-34. This article demonstrates the potential

for tunability of proteasome inhibitors

toward the preference of specific

binding pockets or proteolytic

activities. In addition, a combination

of biochemical and cellular evaluations

helps gain important insight into the

inhibitory effect of such tuning.

85. Baldisserotto A, Marastoni M, Gavioli R,

Tomatis R. New cyclic peptide

proteasome inhibitors. Bioorg Med

Chem Lett 2009;19(7):1966-9

86. Chen W, Mou K, Xu B, et al. Capillary

electrophoresis for screening of 20S

proteasome inhibitors. Anal Biochem

2009;394(1):62-7

87. Garcıa-Echeverrıa C. Peptide and

peptide-like modulators of 20S

proteasome enzymatic activity in cancer

cells. Int J Pept Res Ther

2006;12(1):49-64

88. Powers JC, Asgian JL, Ekici OD, et al.

Irreversible inhibitors of serine, cysteine,

and threonine proteases. Chem Rev

2002;102(12):4639-750

89. Baldisserotto A, Franceschini C,

Scalambra F, et al. Synthesis and

proteasome inhibition of N-allyl vinyl

ester-based peptides. J Pept Sci

2010;16(11):659-63

90. Demo SD, Kirk CJ, Aujay MA, et al.

Antitumor activity of PR-171, a novel

irreversible inhibitor of the proteasome.

Cancer Res 2007;67(13):6383-91

91. Bennett M, Kirk C, et al. Development

of proteasome inhibitors in oncology and

autoimmune diseases. Curr Opin Drug

Discov Dev 2008;11(5):616

92. Kuhn DJ, Chen Q, Voorhees PM, et al.

Potent activity of carfilzomib, a novel,

irreversible inhibitor of the

ubiquitin-proteasome pathway, against

preclinical models of multiple myeloma.

Blood 2007;110(9):3281-90

93. Oconnor O, Orlowski R, Alsina M,

et al. Multicenter phase I studies to

evaluate the safety, tolerability, and

clinical response to intensive dosing with

the proteasome inhibitor PR-171 in

patients with relapsed or refractory

hematological malignancies. Blood

2006;108(11):687A-8A

94. Stapnes C, Døskeland AP, Hatfield K,

et al. The proteasome inhibitors

bortezomib and PR-171 have

antiproliferative and proapoptotic effects

on primary human acute myeloid

leukaemia cells. Br J Haematol

2007;136(6):814-28

95. Orlowski R, Stewart K, Vallone M, et al.

Safety and antitumor efficacy of the

proteasome inhibitor carfilzomib (PR-

171) dosed for five consecutive days in

hematologic malignancies:

phase 1 results. BLOOD-New York

2007;110(11):409

96. Alsina M, Trudel S, Vallone M, et al.

Phase 1 single agent antitumor activity of

twice weekly consecutive day dosing of

the proteasome inhibitor carfilzomib

(PR-171) in hematologic malignancies.

Blood (New York) 2007;110(11):411

97. Zhou HJ, Aujay MA, Bennett MK, et al.

Design and synthesis of an orally

bioavailable and selective peptide

Y. Pevzner et al.

564 Expert Opin. Drug Discov. (2013) 8(5)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 29: Recent advances in proteasome inhibitor discovery

epoxyketone proteasome inhibitor (PR-

047). J Med Chem 2009;52(9):3028-38

98. Marco ML, Legac J, Lindow SE.

Pseudomonas syringae genes induced

during colonization of leaf surfaces.

Environ Microbiol 2005;7(9):1379-91

99. Oka M, Nishiyama Y, Ohta S, et al.

Glidobactins A, B and C, new antitumor

antibiotics. I. Production, isolation,

chemical properties and biological

activity. J Antibiot (Tokyo)

1988;41(10):1331

100. Clerc J, Groll M, Illich DJ, et al.

Synthetic and structural studies on

syringolin A and B reveal critical

determinants of selectivity and potency of

proteasome inhibition. Proc Nat

Acad Sci 2009;106(16):6507-12

101. Clerc J, Schellenberg B, Groll M, et al.

Convergent synthesis and biological

evaluation of syringolin A and derivatives

as eukaryotic 20S proteasome inhibitors.

Eur J Org Chem

2010;2010(21):3991-4003

102. Archer CR, Groll M, Stein ML, et al.

Activity enhancement of synthetic

syrbactin proteasome inhibitor hybrid

and biological evaluation in tumor cells.

Biochemistry 2012;51(34):6880-8

103. van der Linden WA, Willems LI,

Shabaneh TB, et al. Discovery of a

potent and highly b1 specific proteasome

inhibitor from a focused library of

urea-containing peptide vinyl sulfones

and peptide epoxyketones.

Org Biomol Chem 2012;10(1):181-94

104. Coleman C, Rocetes J, Park D, et al.

Syringolin A, a new plant elicitor from

the phytopathogenic bacterium

Pseudomonas syringae pv. syringae,

inhibits the proliferation of

neuroblastoma and ovarian cancer cells

and induces apoptosis. Cell Prolif

2006;39(6):599-609

105. Groll M, Schellenberg B, Bachmann AS,

et al. A plant pathogen virulence factor

inhibits the eukaryotic proteasome by a

novel mechanism. Nature

2008;452(7188):755-8

106. Clerc J, Li N, Krahn D, et al. The

natural product hybrid of Syringolin

A and Glidobactin A synergizes

proteasome inhibition potency with

subsite selectivity. Chem Commun

2011;47(1):385-7

107. Bonfili L, Cecarini V, Amici M, et al.

Natural polyphenols as proteasome

modulators and their role as anti-cancer

compounds. FEBS J

2008;275(22):5512-26

108. Chen D, Chen MS, Cui QC, et al.

Structure-proteasome-inhibitory activity

relationships of dietary flavonoids in

human cancer cells. Front Biosci

2007;12:1935-45

109. Chen D, Daniel KG, Chen MS, et al.

Dietary flavonoids as proteasome

inhibitors and apoptosis inducers in

human leukemia cells.

Biochem Pharmacol

2005;69(10):1421-32

110. Chang TL. Inhibitory effect of flavonoids

on 26S proteasome activity. J Agric

Food Chem 2009;57(20):9706-15

111. Shim SH. 20S proteasome inhibitory

activity of flavonoids isolated from

Spatholobus suberectus. Phytother Res

2011;25(4):615-18

112. Guo S, Lu J, Subramanian A,

Sonenshein GE. Microarray-assisted

pathway analysis identifies

mitogen-activated protein kinase

signaling as a mediator of resistance to

the green tea polyphenol epigallocatechin

3-gallate in her-2/neu-overexpressing

breast cancer cells. Cancer Res

2006;66(10):5322-9

113. Sun CL, Yuan JM, Koh WP, Mimi CY.

Green tea, black tea and breast cancer

risk: a meta-analysis of epidemiological

studies. Carcinogenesis

2006;27(7):1310-15

114. Sartippour MR, Heber D, Ma J, et al.

Green tea and its catechins inhibit breast

cancer xenografts. Nutr Cancer

2001;40(2):149-56

115. Landau JM, Wang ZY, Yang GY, et al.

Inhibition of spontaneous formation of

lung tumors and rhabdomyosarcomas in

A/J mice by black and green tea.

Carcinogenesis 1998;19(3):501-7

116. Horie N, Hirabayashi N, Takahashi Y,

et al. Synergistic effect of green tea

catechins on cell growth and apoptosis

induction in gastric carcinoma cells.

Biol Pharm Bull 2005;28(4):574-9

117. Bettuzzi S, Brausi M, Rizzi F, et al.

Chemoprevention of human prostate

cancer by oral administration of green tea

catechins in volunteers with high-grade

prostate intraepithelial neoplasia:

a preliminary report from a one-year

proof-of-principle study. Cancer Res

2006;66(2):1234-40

118. Kemberling J, Hampton JA, Keck RW,

et al. Inhibition of bladder tumor growth

by the green tea derivative

epigallocatechin-3-gallate. J Urol

2003;170(3):773-6

119. Smith DM, Daniel KG, Wang Z, et al.

Docking studies and model development

of tea polyphenol proteasome inhibitors:

applications to rational drug design.

Proteins 2003;54(1):58-70

120. Nam S, Smith DM, Dou QP. Ester

bond-containing tea polyphenols potently

inhibit proteasome activity in vitro and

in vivo. J Biol Chem

2001;276(16):13322-30

121. Okushio K, Suzuki M, Matsumoto N,

et al. Methylation of tea catechins by rat

liver homogenates.

Biosci Biotechnol Biochem

1999;63(2):430-2

122. Lu H, Meng X, Yang CS. Enzymology

of methylation of tea catechins and

inhibition of

catechol-O-methyltransferase by

(-)-epigallocatechin gallate.

Drug Metab Dispos 2003;31(5):572-9

123. Huo C, Yang H, Cui QC, et al.

Proteasome inhibition in human breast

cancer cells with high catechol-O-

methyltransferase activity by green tea

polyphenol EGCG analogs.

Bioorg Med Chem 2010;18(3):1252-8

124. Daniel KG, Landis-Piwowar KR,

Chen D, et al. Methylation of green tea

polyphenols affects their binding to and

inhibitory poses of the proteasome

beta5 subunit. Int J Mol Med

2006;18(4):625

125. Kanwar J, Mohammad I, Yang H, et al.

Computational modeling of the potential

interactions of the proteasome

b5 subunit and catechol-O-

methyltransferase-resistant EGCG

analogs. Int J Mol Med 2010;26(2):209

126. Bonfili L, Cuccioloni M,

Mozzicafreddo M, et al. Identification of

an EGCG oxidation derivative with

proteasome modulatory activity.

Biochimie 2011;93(5):931-40

127. Feling RH, Buchanan GO, Mincer TJ,

et al. Salinosporamide A: a highly

cytotoxic proteasome inhibitor from a

novel microbial source, a marine

bacterium of the new genus Salinospora.

Angew Chem Int Ed 2003;42(3):355-7

128. Fenical W, Jensen PR, Palladino MA,

et al. Discovery and development of the

Recent advances in proteasome inhibitor discovery

Expert Opin. Drug Discov. (2013) 8(5) 565

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 30: Recent advances in proteasome inhibitor discovery

anticancer agent salinosporamide

A (NPI-0052). Bioorg Med Chem

2009;17(6):2175-80

129. Lam KS, Lloyd GK, Neuteboom STC,

et al. From natural product to clinical

trials: NPI-0052 (salinosporamide A), a

marine actinomycete-derived anticancer

agent. In: Buss AD, Butler MS, editors,

Natural products chemistry for

drug discovery. 1st edition. Royal

Publishing, Cambridge, UK; 2009

130. Nguyen H, Ma G, Gladysheva T, et al.

Bioinspired total Synthesis and human

proteasome inhibitory activity of

(-)-salinosporamide A,(-)-

homosalinosporamide A, and derivatives

obtained via organonucleophile promoted

bis-cyclizations. J Org Chem

2011;76(1):2

131. Rachid S, Huo L, Herrmann J, et al.

Mining the cinnabaramide biosynthetic

pathway to generate novel proteasome

inhibitors. Chembiochem

2011;12(6):922-31

132. Sallam AA, Ramasahayam S, Meyer SA,

Sayed KAE. Design, synthesis, and

biological evaluation of dibromotyrosine

analogues inspired by marine natural

products as inhibitors of human prostate

cancer proliferation, invasion, and

migration. Bioorg Med Chem

2010;18(21):7446-57

133. Mudit M, Khanfar M, Muralidharan A,

et al. Discovery, design, and synthesis of

anti-metastatic lead phenylmethylene

hydantoins inspired by marine natural

products. Bioorg Med Chem

2009;17(4):1731-8

134. Koguchi Y, Kohno J, Nishio M, et al.

TMC-95A, B, C, and D, novel

proteasome inhibitors produced by

apiospora montagnei sacc. TC 1093.

Taxonomy, production, isolation, and

biological activities. Chem Inform

2000;31(26):no-o

135. Groll M, Koguchi Y, Huber R, Kohno J.

Crystal structure of the 20 S proteasome:

TMC-95A complex: a non-covalent

proteasome inhibitor. J Mol Biol

2001;311(3):543-8

136. Lin S, Danishefsky SJ. The total

synthesis of proteasome inhibitors

TMC-95A and TMC-95B: discovery of a

new method to generate cis-propenyl

amides. Angew Chem Int Ed

2002;41(3):512-15

137. Inoue M, Sakazaki H, Furuyama H,

Hirama M. Total Synthesis of

TMC-95A. Angew Chem Int Ed

2003;42(23):2654-7

138. Albrecht BK, Williams RM. A concise,

total synthesis of the TMC-95A/B

proteasome inhibitors. Proc Nat Acad

Sci USA 2004;101(33):11949-54

139. Kaiser M, Groll M, Renner C, et al. The

core structure of TMC-95A Is a

promising lead for reversible proteasome

inhibition. Angew Chem Int Ed

2002;41(5):780-3

140. Lin S, Danishefsky SJ. Synthesis of the

functionalized macrocyclic core of

proteasome inhibitors TMC-95A and B.

Angew Chemie Int Ed

2001;40(10):1967-70

141. Berthelot A, Piguel S, Le Dour G,

Vidal J. Synthesis of macrocyclic peptide

analogues of proteasome inhibitor

TMC-95A. J Org Chem

2003;68(25):9835-8

142. Groll M, Gallastegui N, Marechal X,

et al. 20S proteasome inhibition:

designing noncovalent linear peptide

mimics of the natural product

TMC-95A. Chem Med Chem

2010;5(10):1701-5

143. Basse N, Piguel S, Papapostolou D, et al.

Linear TMC-95-based proteasome

inhibitors. J Med Chem

2007;50(12):2842-50

144. Marechal X, Pujol A, Richy N, et al.

Noncovalent inhibition of 20S

proteasome by pegylated dimerized

inhibitors. Eur J Med Chem

2012;52:322-7

145. Milacic V, Chen D, Ronconi L, et al.

A novel anticancer gold (III)

dithiocarbamate compound inhibits the

activity of a purified 20S proteasome and

26S protea some in human breast cancer

cell cultures and xenografts. Cancer Res

2006;66(21):10478-86

146. Milacic V, Dou QP. The tumor

proteasome as a novel target for gold

(III) complexes: implications for breast

cancer therapy. Coord Chem Rev

2009;253(11):1649-60

147. Cvek B, Milacic V, Taraba J, Dou QP.

Ni (II), Cu (II), and Zn (II)

diethyldithiocarbamate complexes show

various activities against the proteasome

in breast cancer cells. J Med Chem

2008;51(20):6256-8

148. Frezza M, Hindo SS, Tomco D, et al.

Comparative activities of nickel (II) and

zinc (II) complexes of asymmetric [NN

O] ligands as 26S proteasome inhibitors.

Inorg Chem 2009;48(13):5928-37

149. Daniel KG, Chen D, Orlu S, et al.

Clioquinol and pyrrolidine

dithiocarbamate complex with copper to

form proteasome inhibitors and apoptosis

inducers in human breast cancer cells.

Breast Cancer Res 2005;7(6):R897-908

150. Milacic V, Jiao P, Zhang B, et al. Novel

8-hydroxylquinoline analogs induce

copper-dependent proteasome inhibition

and cell death in human breast cancer

cells. Int J Oncol 2009;35(6):1481-91.. The concept of selective targeting of

cancer cells via the formation of

copper complexes is well presented in

this study. Otherwise innocuous

compound upon forming a copper

complex inside copper-rich cancer cells

becomes an effective proteasome

inhibitor exhibiting anticancer effects.

151. Hindo SS, Frezza M, Tomco D, et al.

Metals in anticancer therapy: copper (II)

complexes as inhibitors of the 20S

proteasome. Eur J Med Chem

2009;44(11):4353-61

152. Zuo J, Bi C, Fan Y, et al. Cellular and

computational studies of proteasome

inhibition and apoptosis induction in

human cancer cells by amino acid schiff

base-copper complexes. J Inorg Biochem

2013;118:83-93

153. Rackham O, Nichols SJ, Leedman PJ,

et al. A gold (I) phosphine complex

selectively induces apoptosis in breast

cancer cells: implications for anticancer

therapeutics targeted to mitochondria.

Biochem Pharmacol

2007;74(7):992-1002

154. Geraki K, Farquharson M, Bradley D.

Concentrations of Fe, Cu and Zn in

breast tissue: a synchrotron XRF study.

Phys Med Biol 2002;47(13):2327

155. Nayak SB, Bhat VR, Upadhyay D,

Udupa SL. Copper and ceruloplasmin

status in serum of prostate and colon

cancer patients. Indian J

Physiol Pharmacol 2003;47(1):108-10

156. Diez M, Arroyo M, Cerdan F, et al.

Serum and tissue trace metal levels in

lung cancer. Oncology 1989;46(4):230-4

157. Yoshida D, Ikeda Y, Nakazawa S.

Quantitative analysis of copper, zinc and

copper/zinc ratio in selected human brain

tumors. J Neurooncol 1993;16(2):109-15

158. Schoof S, Pradel G, Aminake MN, et al.

Antiplasmodial thiostrepton derivatives:

Y. Pevzner et al.

566 Expert Opin. Drug Discov. (2013) 8(5)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 31: Recent advances in proteasome inhibitor discovery

proteasome inhibitors with a dual mode

of action. Angew Chem Int Ed

2010;49(19):3317-21

159. Kashiwada Y, Hashimoto F,

Cosentino LM, et al. Betulinic acid and

dihydrobetulinic acid derivatives as

potent anti-HIV agents. J Med Chem

1996;39(5):1016

160. Dogra N, Mukhopadhyay T. Impairment

of the ubiquitin-proteasome pathway by

methyl N-(6-Phenylsulfanyl-1H-

benzimidazol-2-yl) carbamate leads to a

potent cytotoxic effect in tumor cells

A novel antiproliferative agent with a

potential therapeutic implication.

J Biol Chem 2012;287(36):30625-40. This study is a good representative of a

drug repurposing approach. A known

therapeutic fenbendazole is shown to

exhibit antiproliferative effect in cancer

cells as well as inhibition of CT-L

activity of the proteasome.

161. Santoro AM, Lo Giudice MC, Durso A,

et al. Cationic porphyrins are reversible

proteasome inhibitors. J Am Chem Soc

2012;134(25):10451-7

162. Schlitzer M. Malaria chemotherapeutics

part I: history of antimalarial drug

development, currently used therapeutics,

and drugs in clinical development.

Chem Med Chem 2007;2(7):944-86

163. Nesbitt G, Fox P. Clinical evaluation of

Panolog Cream used to treat canine and

feline dermatoses. Vet Med Small

Anim Clin 1981;76(4):535

164. McConkey GA, Rogers MJ,

McCutchan TF. Inhibition of

Plasmodium falciparum protein synthesis.

J Biol Chem 1997;272(4):2046-9

165. Schmidtke G, Holzhutter HG, Bogyo M,

et al. How an inhibitor of the HIV-I

protease modulates proteasome activity.

J Biol Chem 1999;274(50):35734-40

166. Kanamoto T, Kashiwada Y, Kanbara K,

et al. Anti-human immunodeficiency

virus activity of YK-FH312 (a betulinic

acid derivative), a novel compound

blocking viral maturation.

Antimicrob Agents Chemother

2001;45(4):1225-30

167. Qian K, Kim SY, Hung HY, et al. New

betulinic acid derivatives as potent

proteasome inhibitors. Bioorg Med

Chem Lett 2011;21(19):5944-7

168. Eckert J, Conraths F, Tackmann K, et al.

Echinococcosis: an emerging or

re-emerging zoonosis? Int J Parasitol

2000;30(12):1283-94

169. Ammann RW, Ilitsch N, Marincek B,

Freiburghaus AU. Effect of

chemotherapy on the larval mass and the

long-term course of alveolar

echinococcosis. Hepatology

1994;19(3):735-42

170. Muller E, Akovbiantz A, Ammann R,

et al. Treatment of human

echinococcosis with mebendazole.

Preliminary observations in 28 patients.

Hepatogastroenterology 1982;29(6):236

171. Dougherty TJ, Henderson BW,

Gomer CJ, et al. Photodynamic therapy.

J Natl Cancer Inst 1998;90(12):889-905

172. Smith KM, Kadish KM, Guilard R. The

porphyrin handbook. Elsevier, San

Diego, California, USA; 1999

173. Cen D, Brayton D, Shahandeh B, et al.

Disulfiram facilitates intracellular Cu

uptake and induces apoptosis in human

melanoma cells. J Med Chem

2004;47(27):6914-20

174. Chen D, Cui QC, Yang H, Dou QP.

Disulfiram, a clinically used

anti-alcoholism drug and copper-binding

agent, induces apoptotic cell death in

breast cancer cultures and xenografts via

inhibition of the proteasome activity.

Cancer Res 2006;66(21):10425-33

175. Conticello C, Martinetti D, Adamo L,

et al. Disulfiram, an old drug with new

potential therapeutic uses for human

hematological malignancies. Int J Cancer

2012;31(9):2197-203

176. Oprea T, Mestres J. Drug repurposing:

far beyond new targets for old drugs.

AAPS J 2012;14:1-5

177. Nickeleit I, Zender S, Sasse F, et al.

Argyrin A reveals a critical role for the

tumor suppressor protein p27(kip1)

in mediating antitumor activities in

response to proteasome inhibition.

Cancer Cell 2008;14(1):23-35

178. Bulow L, Nickeleit I, Girbig AK, et al.

Synthesis and biological ucharacterization

of argyrin F. ChemMedChem

2010;5(6):832-6

179. Stauch B, Simon B, Basile T, et al.

Elucidation of the structure and

intermolecular interactions of a reversible

cyclic-peptide inhibitor of the

proteasome by NMR spectroscopy and

molecular modeling. Angew Chem

2010;122(23):4026-30

180. Baldisserotto A, Ferretti V, Destro F,

et al. a, b-unsaturated N-acylpyrrole

peptidyl derivatives: new proteasome

inhibitors. J Med Chem

2010;53(17):6511-15

181. Hatcher H, Planalp R, Cho J, et al.

Curcumin: from ancient medicine to

current clinical trials. Cell Mol Life Sci

2008;65(11):1631-52

182. Aggarwal BB, Sundaram C, Malani N,

Ichikawa H. Curcumin: the Indian solid

gold. The molecular targets and

therapeutic uses of curcumin in health

and disease. Adv Exp Med Biol

2007;595:1-75

183. Landis-Piwowar KR, Milacic V, Chen D,

et al. The proteasome as a potential

target for novel anticancer drugs and

chemosensitizers. Drug Resist Updat

2006;9(6):263-73

184. Wan SB, Yang H, Zhou Z, et al.

Evaluation of curcumin acetates and

amino acid conjugates as proteasome

inhibitors. Int J Mol Med

2010;26(4):447

185. Krunic A, Vallat A, Mo S, et al.

Scytonemides A and B, cyclic peptides

with 20S proteasome inhibitory activity

from the cultured cyanobacterium

Scytonema hofmanii. J Nat Prod

2010;73(11):1927

186. Huang L, Yu D, Ho P, et al. Synthesis

and proteasome inhibition of

glycyrrhetinic acid derivatives.

Bioorg Med Chem Lett

2008;16(14):6696-701

187. Huang L, Ho P, Chen CH. Activation

and inhibition of the proteasome by

betulinic acid and its derivatives.

FEBS Lett 2007;581(25):4955-9

188. Dang Z, Lin A, Ho P, et al. Synthesis

and proteasome inhibition of lithocholic

acid derivatives. Bioorg Med Chem Lett

2011;21(7):1926-8

189. Lansdell TA, Hewlett NM,

Skoumbourdis AP, et al. Palauamine

and related oroidin alkaloids

dibromophakellin and

dibromophakellstatin inhibit the

human 20s proteasome. J Nat Prod

2012;75(5):980-5

190. Dunn D, Iqbal M, Husten J, et al.

Serendipity in discovery of proteasome

inhibitors. Bioorg Med Chem Lett

2012;22:3503-5

191. Garcia-Pastor P, Randazzo A,

Gomez-Paloma L, et al. Effects of

Recent advances in proteasome inhibitor discovery

Expert Opin. Drug Discov. (2013) 8(5) 567

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.

Page 32: Recent advances in proteasome inhibitor discovery

petrosaspongiolide M, a novel

phospholipase A2 inhibitor, on acute and

chronic inflammation. J Pharmacol

Exp Ther 1999;289(1):166-72

192. Margarucci L, Monti MC, Tosco A,

et al. Chemical proteomics discloses

petrosapongiolide M, an

antiinflammatory marine sesterterpene, as

a proteasome inhibitor. Angew Chem

Int Ed 2010;49(23):3960-3

193. Margarucci L, Tosco A, De Simone R,

et al. Modulation of proteasome

machinery by natural and synthetic

analogues of the marine bioactive

compound petrosaspongiolide m.

ChemBioChem 2012;13(7):982-6.. Emergence of a novel scaffold with a

potential for proteasome for inhibition

and its further development is

described in a series of studies by these

authors. This article is a good

representation and a validation of the

approach of looking outside of the

familiar scaffolds and venturing out to

find novel classes of compounds.

194. Kisselev AF, Goldberg AL. Monitoring

activity and inhibition of 26S

proteasomes with fluorogenic peptide

substrates. Methods Enzymol

2005;398:364-78

195. Gaedicke S, Firat-Geier E,

Constantiniu O, et al. Antitumor effect

of the human immunodeficiency virus

protease inhibitor ritonavir induction of

tumor-cell apoptosis associated with

perturbation of proteasomal proteolysis.

Cancer Res 2002;62(23):6901-8

AffiliationYuri Pevzner1,2, Rainer Metcalf2,

Melanie Kantor2, Desiree Sagaro2 &

Kenyon Daniel†3

†Author for correspondence1University of South Florida,

Chemistry, 4202 E Fowler Ave,

Tampa, FL 33612, USA2Moffitt Cancer Center and Research Institute,

Screening and Modeling Unit,

Chemical Biology Core,

12902 Magnolia Dr., Tampa,

FL 33612, USA3Moffitt Cancer Center, Screening and Modeling

Unit, Chemical Biology Core,

12902 Magnolia Dr., Tampa,

FL 33612, USA

E-mail: [email protected]

Y. Pevzner et al.

568 Expert Opin. Drug Discov. (2013) 8(5)

Exp

ert O

pin.

Dru

g D

isco

v. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Reg

ina

on 0

5/01

/13

For

pers

onal

use

onl

y.


Recommended