CORE Metadata, citation and similar papers at core.ac.uk

Provided by Elsevier - Publisher Connector

Critical Reviews in /Hematology 87 (2013) 306–322

The role of the ubiquitin system in lymphoma

a,∗ b a c

K. Stephen Suh , Takemi Tanaka , Sreeja Sarojini , Ginah Nightingale ,

a a a

Rajendra Gharbaran , Andrew Pecora , Andre Goy

a

John Theurer Center, Hackensack University Medical Center, Hackensack, NJ 07601, United States

b

Thomas Jefferson University, Philadelphia, PA 19107, United States

c

Jefferson School of Pharmacy, Thomas Jefferson University, Philadelphia, PA 19130, United States

Accepted 14 February 2013

Contents

1. Introduction ...... 306

2. Signaling pathways regulated by the UPS ...... 308

3. Current therapeutic options for lymphoma ...... 309

3.1. Class A compounds ...... 310

3.2. Class B compounds ...... 310

3.3. Class C compounds ...... 314

3.4. Class D compounds ...... 315

3.5. Class E compounds ...... 315

4. Unfolded protein response (UPR) ...... 316

5. ...... 316

6. Conclusion ...... 318

Reviewers ...... 318

References ...... 318

Biographies...... 322

Abstract

The ubiquitin-proteasome system (UPS) maintains the integrity of cellular processes by controlling protein degradation pathways. The

role of the UPS in proliferation, , differentiation, DNA repair, protein folding, and apoptosis is well documented, and a wide range

of protein activities in these signaling pathways can be manipulated by UPS inhibitors, which include many anti-cancer agents. Naturally

occurring and synthetic drugs designed to target the UPS are currently used for hematological , including lymphoma. These drugs

largely interfere with the E1 and E2 regions of the 26S proteasome, blocking proteasomal activity and promoting apoptosis by enhancing

activities of the extrinsic (death receptors, Trail, Fas) and intrinsic (caspases, Bax, Bcl2, , nuclear factor-kappa B, p27) cell death programs.

This review focuses on recent clinical developments concerning UPS inhibitors, signaling pathways that are affected by down-regulation of

UPS activities, and apoptotic mechanisms promoted by drugs in this class that are used to treat lymphoma.

© 2013 Elsevier Ireland Ltd. Open access under CC BY-NC-ND license.

Keywords: Ubiquitin proteasome system; Lymphoma; ; Carfilzomib; Marizomib; CEP-18770; Clinical trials-UPS inhibitors

1. Introduction

The homeostasis of cellular proteins is important for

Corresponding author at: The Genomics and Biomarkers Program, John

maintaining the integrity and health of the cell, and active

Theurer Cancer Center, Hackensack University Medical Center, 40 Prospect

protein degradation by the ubiquitin-proteasome system

Avenue, Hackensack, NJ 07601, United States. Tel.: +1 201 996 8214;

(UPS) is the major pathway through which this cellular

fax: +1 201 336 8776.

E-mail address: [email protected] (K.S. Suh). balance is achieved (Fig. 1). This process is pivotal in

1040-8428 © 2013 Elsevier Ireland Ltd. Open access under CC BY-NC-ND license. http://dx.doi.org/10.1016/j.critrevonc.2013.02.005

K.S. Suh et al. / Critical Reviews in Oncology/Hematology 87 (2013) 306–322 307

Fig. 1. Protein degradation pathway by ubiquitin-proteasome system.

cell cycle control, transcriptional regulation, signal trans- inhibitors that have been developed directly target the 20S

duction, antigen presentation, inflammation, ER-mediated proteasome.

degradation, membrane trafficking, receptor endocytosis, The UPS plays an important role in regulating the cell

apoptosis, and development [1–4]. Ubiquitin (Ub), a 76- cycle. Proteins such as cyclins A, B, D, and E, CDK inhibitor

residue protein, is attached to substrate proteins via multiple p27, p21, transcription factor E2F, retinoblastoma (Rb), and



adenosine-5 -triphosphate (ATP)-dependent processes by E1 tumor suppressor p53 are regulated by proteasome-mediated

(Ub-activating), E2 (Ub-conjugating), and E3 (Ub-ligating) proteolysis. The blockage of cell cycle progression with pro-

enzymes. These enzymes act in harmoniously coordinated teasome inhibitors is currently used against various forms of

fashion via the ubiquitin moiety to achieve specific targeting cancer. NF- B, which participates in immune and inflam-

and regulation of oligomerization, degradation, and post- matory responses, apoptosis, and cell proliferation, is also

translational modification [5]. are abundant in regulated by the UPS. UPS-mediated protein degradation also

the cytosol, and consist of multiple proteins that form the generates antigenic peptides for presentation on MHC class I

20S and 19S subcomponent complexes. Proteasomal pro- molecules [7]. Consistent with this function, the aldehyde

tein degradation generally requires polyubiquitination, and class of proteasome inhibitors partly inhibits presentation

ubiquitinated proteins are first recognized by the 19S cap of antigenic peptides [8]. The UPS is also manipulated

complex, which unfolds the substrate in an ATP-dependent and co-opted by certain viruses, including avian leuko-

process and translocates it into the proteolytic chamber of sis virus, human immunodeficiency virus type 1, simian

the 20S complex to be degraded [6]. The ubiquitin conju- immunodeficiency virus, Moloney murine leukemia virus,

gation cascade begins with activation of the carboxyl group and Epstein–Barr virus [9–11]. UPS is also involved in viral

of Gly-76 of ubiquitin by E1. Activated ubiquitin binds to assembly; proteolytic viral maturation is inhibited upon treat-

E1 and is transferred to a Cys residue of E2, which either ment of infected cells with proteasome-specific inhibitors

singly or in cooperation with E3 shuttles ubiquitin to a pro- [12,13]. Proteasome inhibitors are broadly categorized into

tein substrate. Subsequently, different combinations of E2 two groups (synthetic analogs and natural products), and their

and E3 enzymes add a polyubiquitin chain to provide selec- structural properties and mechanisms of action have been

tive tagging for degradation (Fig. 1). This process is used reviewed in detail.

to degrade misfolded or aged proteins, to regulate the cell Given the importance of proteasomal protein degradation

cycle through cyclin degradation, and to promote immune in various intracellular processes, inhibitors of this pathway

responses through antigenic peptide processing. Thus, com- will continue to serve as both molecular probes of major

ponents of the UPS pathway are attractive molecular targets cellular networks as well as potential therapeutic agents

for therapeutic intervention. For a variety of reasons, most for various human diseases [14]. This review focuses on

308 K.S. Suh et al. / Critical Reviews in Oncology/Hematology 87 (2013) 306–322

proteasome inhibitory mechanisms that are related to the cells in vitro and in patients with one of several types of

treatment of lymphoma, detailing current preclinical and clin- lymphoma [26–28]. Geminin inhibits DNA replication dur-

ical developments and describing apoptotic mechanisms that ing S, G2, and M phases by preventing the incorporation of

may underlie the antitumor activities of the inhibitors. MCM proteins into the prereplication complex, thus playing

an important role in DNA replication licensing. Geminin ubi-

quitination by the anaphase-promoting complex/cyclosome

2. Signaling pathways regulated by the UPS (APC/C) ubiquitin-protein ligase and subsequent degradation

by the 26S proteasome permits DNA replication to proceed

Numerous regulatory protein participants in cellular [29]. Unbalanced expression of geminin is associated with

processes undergo proteolytic degradation. The UPS has B-cell lymphomagenesis [30] and genomic instability, espe-

been targeted as part of the chemotherapeutic regimen cially in MCL with p53/ARF inactivation [31]. In addition,

for solid tumors and hematological malignancies, such as the overexpression of geminin has been proposed to play a

non-Hodgkin’s lymphoma (NHL). The roles of proteins or role in the pathology of acute leukemia [32].

molecular targets that are proteolytically degraded by the 26S Cyclins have been directly implicated in the pathogenesis

proteasome of the UPS in basic cellular activities (Fig. 1), in of lymphoma. Cyclin A is required for entry into mitosis and

lymphomagenesis, and in tumor progression are described S-phase progression, and is ubiquitinated and targeted for

below. proteasomal degradation by APC/C during late M to early

Cell cycle progression requires programmed and peri- G1 phase [33]. Bcl-1 (CCND1 and PRAD1) has been impli-

odic expression and degradation of specific proteins [15]. cated in the pathogenesis of multiple types of NHL, and its

Cyclins, cyclin-dependent kinases, and their inhibitors are protein product, cyclin D1, plays an important role in the

target proteins degraded by the 26S proteasome in an orderly transition from G1 to in response to mitogens. Over-

and sequential manner to regulate the cell cycle [16]. G1/S expression of cyclin D1 shortens the G1 phase and reduces

phase progression requires coordination of positive regula- the dependency of the cell on extrinsic mitogenic signals.

tors (cyclins, cyclin-dependent kinases [CDKs], CDK-cyclin Defects in the proteasomal degradation of cyclin D lead to

complexes, E2F, and Cdc6) and negative regulators (CDK its accumulation within the cell and contributes to prolif-

inhibitors (CKIs) of the Cip/Kip and INK4 families). Dys- eration; the prototypical NHL with dysregulation of cyclin

regulation of these proteins may lead to tumor progression. D is MCL [34]. Cyclin E is also required for the onset of

Proteasomal degradation of p27 (Cip/Kip inhibitor) promotes DNA replication during late G1 phase and early S phase,

cell cycle progression from G0 to G1 [17], consequently ini- and is targeted for ubiquitination and subsequent proteaso-

tiating the early onset of S phase [18]. For example, high mal degradation [35,36]. Cyclin E overexpression has been

levels of cyclin E and low levels of p27 (Kip1) expres- observed in lymphoma [37], particularly DLBCL, and its

sion have been associated with malignant lymphomas in overexpression has been associated with poor response to

humans [19], and the loss of p27 (Kip1) decreases sur- standard treatment and inferior outcomes [38]. Of the non-

vival in murine Myc transgenic lymphoma models [20]. cyclin factors, E2F1 is required for the completion of the

An increase in proteasomal degradation of p27 was also G1-to-S-phase transition, and is degraded by the 26S protea-

associated with decreased overall survival in mantle cell some at the S-to-G2-phase transition after dissociating from

lymphoma (MCL) in a study of 157 NHL patients with lym- the Rb protein [39]. It has been proposed that dysregula-

phoproliferative disorders [21]. A more recent study of 671 tion of E2F plays an oncogenic role in sporadic Burkitt’s

diffuse large B cell lymphoma (DLBCL) patients treated lymphoma [40] and in lymphomagenesis in murine models

with rituximab and CHOP (, doxoru- [41]. The Cdc6 protein is a component of the prereplication

bicin/hydroxydaunomycin, /oncovin, prednisone) complex required to initiate DNA unwinding and replication

(R-CHOP) or without rituximab (CHOP) showed that low before S phase [42]. During completion of DNA replication

expression of p27 (Kip1) and increased Skp correlated in S phase, Cdc6 dissociates from DNA-bound replication

with poor overall and progression-free survival rates [22]. complexes and is ubiquitinated and targeted for proteasomal

Similarly, p19INK4d is governed by ubiquitination and degradation by the APC/C [43]. Cdc6 is also overexpressed

subsequent proteasomal degradation during cell cycle pro- in a subset of MCL, in which it is associated with poor clin-

gression [23]. Genetic defects in the tumor suppressor ical outcome [31], and is differentially expressed in primary

INK4a/ARF locus accelerate lymphomagenesis in an Emu- cutaneous large B-cell lymphoma [44]. In addition, a Cdc6

myc transgenic murine model, and cytogenetic alterations of G1321A polymorphism is associated with decreased risk of

the INK4a/ARF locus are directly related to drug responses NHL [45]. These observations suggest that a failure of the

in primary tumors [24]. The prognostic significance of these UPS to effectively maintain optimal levels of intracellular

functional deletions is made evident by the poor progno- cyclins can result in hematological malignancies.

sis of subsets of MCL patients with higher levels of cyclin Terminal differentiation of eukaryotic cells requires

D1, which has been attributed to deletion of the INK4a/ARF withdrawal from the cell cycle, which is accomplished

locus (CDKN2A) [25]. Dysregulated expression or inacti- by the down-regulation of CDK activities during the G1

vation of p21 promotes cell cycle progression in lymphoma phase [46]. This mainly occurs in mammalian cells through

K.S. Suh et al. / Critical Reviews in Oncology/Hematology 87 (2013) 306–322 309

Ink4a

binding of CDKs to the CKIs of the INK4 family (p16 , blocking proteasomal degradation may enhance the activities

Ink4b Ink4c INK4d

p15 , p18 , and p19 ) and the Cip/Kip family of pro-apoptotic proteins rather than those of anti-apoptotic

WAF1/Cip1 Kip1 Kip2

(p21 , p27 , and p57 ) [47]. More recent stud- proteins. Emerging evidence regarding the proteins that con-

WAF1/Cip1

ies suggest that proteasomal degradation of p21 and stitute the apoptotic core machinery and upstream signaling

other cell cycle regulators, such as cyclin D1 and Cdc25A molecules has revealed multiple new opportunities for ther-

phosphatase, can promote differentiation of certain mouse apeutic intervention, some of which are already under

and human cells [48,49]. In aggregate, the data indicate investigation in clinical trials. With respect to core apopto-

that the 26S proteasome plays a complex role in cellular sis targets, for example, nuclease-resistant (phosphothioate)

differentiation [50]. antisense oligonucleotides directed against BCL-2 mRNA

Heat shock proteins (HSP110, HSP90, HSP70, and are currently in phase III trials for patients with multiple

HSP27) perform a crucial role as cytosolic molecular chap- myeloma (MM), and are in phase II trials for patients with

erones in regulating the balance between protein synthesis NHL, B-cell chronic lymphocytic leukemia (B-CLL), and

and degradation, stabilizing regulation of signal transduction acute myeloid leukemia. Small-molecule compounds that

pathways, and protecting cells from apoptosis in response mimic the BH3 domains of pro-apoptotic proteins have also

to cellular stress [51]. The role of HSP90 in tumorigenesis been described [60–62].

has been studied in vitro in lymphoma and leukemia models. The stability and activation of transcription factors are

HSP90 is commonly upregulated in high-grade or large-cell also regulated by proteasomal degradation. Dysregulated

NHL types of B- and T-cell lymphomas [52,53]. Immuno- expression of c-Myc is associated with many human can-

histochemical data also show that among B-cell lymphomas, cers including Burkitt’s lymphoma, and proteolysis of c-Myc

HSP90 is moderately to strongly expressed in Burkitt’s lym- is mediated by the UPS. Protein levels of c-Myc are over-

phoma (5/5, 100%) and in subsets of follicular lymphoma expressed up to 6-fold in Burkitt’s lymphoma-derived cell

(17/28, 61%), DLBCL (27/46, 59%), nodal marginal zone B- lines, and blocking of proteasomal degradation may enhance

cell lymphoma (6/16, 38%), plasma cell (14/39, c-Myc-mediated apoptosis. Aberrant, dysregulated NF- B

36%), small lymphocytic lymphoma/chronic lymphocytic activation is directly associated with several lymphoid malig-

leukemia (3/9, 33%), MCL (12/38, 32%), and lymphoplas- nancies, and blockage of proteasomal degradation of its

macytic lymphoma/Waldenstrom macroglobulinemia (3/10, inhibitory I B proteins ablates the survival signals medi-

30%) [54]. These observations identify a role of HSP90 in ated by NF- B and substantially shifts the balance to

the pathology of lymphomas, and suggest that HSP90 is a pro-apoptotic proteins [63,64].

viable therapeutic target of HSP inhibitors in lymphomas. In addition to the UPS, autophagy machinery is also

The proteasomal degradation pathway plays a role in the important for protein degradation in cells via a lysosome-

induction and suppression of apoptosis in eukaryotic cells dependent degradative pathway [65]. When proteasomes are

[55]. Among molecules that are regulated by proteasomal inhibited, autophagy is triggered as a compensatory mech-

degradation are members of the Bcl-2 family of anti-apoptotic anism of protein degradation through activation of the ER

and pro-apoptotic proteins. The anti-apoptotic Bcl-2 and stress-induced IRE1 pathway; thus, both systems are func-

Bcl-XL proteins are integral mitochondrial proteins that tionally coupled in protein degradation. Recent research

share sequence homology in all four Bcl-2 homology (BH1 investigating the role of autophagy in myc-induced lym-

TAM

to BH4) domains but vary in both structure and function. phoma derived from p53ER cells demonstrated the

Upon apoptotic stimuli, these proteins block apoptosis and significance of autophagy inhibitors (chloroquine) in com-

preserve mitochondrial integrity [34]. Proteasomal degrada- bination with other therapies for successfully inducing

tion of Bcl-2 results in release of proapoptotic signals and apoptosis in malignant cells [66]. Similarly, in leukemia,

consequent promotion of apoptosis [56]. Another class of deregulation of autophagy points to the fact that autophagy

anti-apoptotic proteins includes the inhibitor of apoptosis inhibitors could be included in combination therapies for

(IAP) family (XIAP and c-IAP2), which inhibits the activa- suppressing cancer progression [65].

tion and enzymatic activity of caspases by targeting them for

ubiquitination [56,57]. IAPs auto-ubiquitinate via the RING 3. Current therapeutic options for lymphoma

finger domain, and subsequent 26S proteasomal degrada-

tion initiates IAP binding to ubiquitin-conjugating enzymes To treat NHL, the combination regi-

(UbCs, E2) [58]. In these cases, inhibition of proteasomal men CHOP is most commonly used [67]. Other treatment

degradation of IAP family members enhances proliferative combinations for NHL include common chemotherapy

potential of the treated cells. However, the mechanism under- agents such as , , ,

lying this effect has not been well demonstrated. Similarly, , , fludarabine, and , and

pro-apoptotic members of the Bcl family (Bax, Bak, Bad, radioimmunotherapeutic agents including tositumomab and

Bim, Bik, and Bid) also undergo proteasomal degradation ibritumomab. For Hodgkin’s lymphoma (HL), which can be

[59], and inhibition of proteasomal degradation signifi- cured in about 75% of cases overall and 90% of cases in

cantly increases apoptotic potential in treated cells. Thus, younger patients [68], a combination

310 K.S. Suh et al. / Critical Reviews in Oncology/Hematology 87 (2013) 306–322

of ABVD (adriamycin, , vinblastine, ) non-small cell lung carcinoma [80]. More recently, accumu-

is most commonly used. Radiation therapy may be used alone lating evidence has shown that the efficacy of bortezomib

for treating HL, or may be combined with chemotherapy and can be enhanced through combination with chemother-

with stem cell transplantation after failure of primary therapy apy. When combined with -based chemotherapy,

[69]. For MCL, which is clinically aggressive with poor prog- bortezomib resulted in an 83% higher response rate in

nosis, rituximab and ASCT (Allogeneic/Autologous Stem relapsed/refractory activated B-cell-like DLBCL, and in a

Cell Transplant) prolongs progression-free survival. How- 13% higher response rate in relapsed/refractory germinal cen-

ever, for patients who are not fit for intensive treatment, ter B-cell-like DLBCL [81]. In addition, a combination of

less intensive treatment with various combinations of ritux- bortezomib with in a phase II clinical trial pro-

imab and other compounds, such as rituximab with CHOP or duced improvements in the ORR from 9% (bortezomib alone)

with is recommended, and for relapsed MCL, to 44% (bortezomib/irinotecan) in advanced gastric cancer

ASCT can be effective if a compatible donor is available (n = 28) [82]. More recently, in a phase II trial of 63 refractory

(reviewed in Cortelazzo et al. [70]). In addition to con- and relapsing FL patients, bortezomib combined with ben-

ventional combination chemotherapy, proteasome inhibitors damustine and rituximab resulted in an 88% ORR (including

have been used to treat lymphomas. This class of compounds a 53% CR), with toxicity within manageable limits [83]. In

consists of synthetic and natural products and can be catego- a separate phase II clinical trial of 94 untreated advanced

rized into five different classes (A to E) based on chemical stage (III/IV) FL patients, bortezomib added to rituximab,

structure. cyclophosphamide, vincristine, and prednisone resulted in

an intent-to-treat outcome of 46 of 94 patients (49%; 95%

3.1. Class A compounds confidence interval [CI] of 38.8–59.0%) who achieved a

CR or unconfirmed CR, and 32 of 94 patients (34%) who

Class A compounds are peptide aldehydes and include achieved a partial response, for an overall response rate of

MG 132 and MG 115. These compounds primarily inhibit 83% (95% CI of 75.4–90.6%) [84]. In this study (Table 1),

chymotrypsin-like activity through the formation of a cova- no grade 4 neurotoxicity was observed, although 5 patients

lent bond between the aldehyde and an N-terminal threonine developed largely reversible grade 3 neurotoxicity. Other

moiety in a reversible manner. The specificity of class A combinations that have shown encouraging results in hemato-

compounds is relatively low. Well-known peptide aldehyde logical malignancies include thalidomide and its derivatives,

inhibitors have been further refined for specificity by using flavopiridol (cyclin inhibitor), rapamycin (mTOR inhibitor),

peptide vinyl sulfones and peptide-boronic acid derivatives. and , doxorubicin, , and prednisone

These efforts have led to the development of highly potent [85,86]. In fact, a multicenter phase II trial of 29 evaluable

and specific drugs like PS-341 (bortezomib) [71] and CEP- relapsed/refractory indolent and mantle cell NHL patients

2

18770, which is currently being clinically evaluated (Table 1) treated with 90 mg/m of bendamustine on days 1 and 4;

2 2

(Fig. 2). 375 mg/m rituximab on day 1, and 1.3 mg/m bortezomib

on days 1, 4, 8, and 11, on six 28-day cycles resulted in

3.2. Class B compounds a 47% 2-year progression-free survival rate (15/29 patients

achieved complete response) [87]. In another phase II clin-

Class B compounds are synthetic peptide boronates, ical trial of 16 relapsed and chemotherapy-refractory MCL

and including benzamide, alpha-ketoamide, bortezomib, and patients treated with bortezomib combined with rituximab

Cbz-Leu-Leu-Leu-boronic acid. Bortezomib binds with high and dexamethasone (treatment cycle consisted of bortezomib

2

affinity and specificity (yet reversibly) to the threonine [1.3 mg/m on days 1, 4, 8, and 11; six 21-day cycles], rit-

2

hydroxyl groups in the chymotrypsin-like active site of the uximab [375 mg/m on day 1], and dexamethasone [40 mg

proteasome ␤5 subunit and inhibits the peptidyl-glutamyl orally on days 1–4]) resulted in 13 patients (81.3%) achiev-

peptide-hydrolyzing activity of the ␤1 subunit. Bortezomib ing responses and 7 patients achieving complete responses

has shown promising activity and has led to durable responses (43.8%) [88]. In addition, in chemotherapy-naïve low-grade

as a single agent in relapsed or refractory MCL patients and NHL, bortezomib combined with rituximab, dexamethasone,

2

MM patients [72,73]. In a phase II trial (n = 202), the over- and cyclophosphamide (bortezomib given at 1.6 mg/m , on

all response rate (ORR) and complete response rate (CR) days 1, 8, 15, and 22 of every 35-day cycle; rituximab given

2

of patients with MM treated with bortezomib were 35% at 375 mg/m on the same days as bortezomib during cycle

and 4%, respectively, with a median overall survival of 16 1 and then only on day 1 in subsequent cycles; dexametha-

months [74]. Similarly, heavily pre-treated MCL patients sone given orally at 40 mg on days 1, 2, 8, 9, 15, 16, 22, and

2

(n = 36) responded to bortezomib favorably with an ORR of 23; and cyclophosphamide administered orally at 400 mg/m

46.5% and a CR of 16% [75]. While somewhat less success- on days 1–4) resulted in an ORR of 90% (CR of 54%),

ful, encouraging responses have also been seen in patients with a 75% (n = 9) survival rate at a 22-month follow-up

with DLBCL, follicular lymphoma (FL) [76], Waldenstrom [89]. In this study, there were no grade 3 or 4 periph-

macroglobulinemia [77], and in patients with a few solid eral neuropathies. In addition, results from in vitro studies

tumors including sarcoma [78], renal cell carcinoma [79], and with cells derived from primary effusion lymphomas showed

K.S. Suh et al. / Critical Reviews in Oncology/Hematology 87 (2013) 306–322 311 Status Completed Completed Completed Completed Completed Completed Completed Completed Recruiting

NCT NCT00030875 [154] NA NCT00038571 NCT00038571 NCT00636792 NCT00374699 [155] NCT00295932

ORR 46% 31% 58% 41% 19% 88% NA 75% NA

months

months months months months

years

TTP 10 15 11–24+ 24 24 NA NA 4 NA

PR NA NA 37% 18% 5% 35% NA 5% NA

CR NA 8% 7% 18% 15% 53% NA 95% NA

2 1 2 2 2 1–2 1 2

Phase Phase Phase Phase Phase Phase2 Phase Phase Phase Phase

(patients)

155 20 n 55 115 30 26 73 33/60 27/60 toxicity,

hematologic constipation,

neuropathy neuropathy peripheral

myalgia/edema

neutropenia, neutropenia, anorexia

nausea, peripheral peripheral toxicity Neuropathy, dyspnea, Neurological Toxicity Lymphopenia, thrombocytopenia NA NA Thrombocytopenia, fatigue, Thrombocytopenia, fatigue, neuropathy, Asthenia, Pancytopenia, thrombocytopenia, neutropenia 2 2 8, 8, 4, 1,

a a

21 of 8, and

22

4, 4, IV oral

of and 1,

the

1 each

4, 6 2

for for

22 5

mg/m

1, 1, mg/m Total 1, days

phase

days

IV 2–6, and

of (at 1,

is dose

5. every

for in

days 21 day1 therapy. 3-week 2

on

3 and

375 day 8. Days 15 days days

was

was courses

IV

mg/m

days days

cycles cycles

on

days on 50–90 8 of and administered days

8,

on

IV IV on 6 6 and

21 21 15,

cycle; on on

each 1 and 1.6 cycle

to 4,

every 1 1, on ) )

given i.v. on

8, repeats of of 2 and IV 2 2

duration

of

was cycle.

2 2 2 3,

bortezomib up

11 1

1,

with every every 2,

11

rituximab lymphoma days days of

determined

days for 35-day

21-day

each

and mg/m mg/m 11 11 11 bortezomib Bortezomib Bortezomib

in

mg/m mg/m mg/m

on on days a

cycle

8 and

on

of

[B] administered each of prednisone levels MTD cycles bendamustine and administered rituximab 4 (1.5 (1.5 Dose maximum maximum on on cycle; and Treatment 1.3 1.3 Bortezomib IV IV R-CHOP Cyclophosphamide 8, treatment [A] 4, days I) NA 1.5 cycles, and and cycle cycles. administered

inhibitors lymphoma and cell

NHL

for

NHL

NK/T

DLBCL

indolent

B-cell proteasome

treatment

RR MCL, R/R-FL Condition MCL MCL Indolent-NHL T-cellor R/R Lymphomas MCL in

major

the

of CHOP R- R-CP

Inhibitors + + +

trials

1

Bendamustine Rituximab

+ CHOP +

Bortezomib Bortezomib Bortezomib Bortezomib Bortezomib Bortezomib Bortezomib Table Proteasome PI Bortezomib Clinical

312 K.S. Suh et al. / Critical Reviews in Oncology/Hematology 87 (2013) 306–322 Recruiting Completed Recruiting Recruiting Completed Recruiting Recruiting Completed Status

NCT00572637 NCT00893464 NCT00667082 NCT01111188 [109] NCT01276717 [156] NCT00396864 NCT

).

NA 3.5% NA NA NA ORR NA NA NA

NA NA NA NA NA NA NA TTP NA

NA NA 3.5% NA NA NA PR NA 3.5% http://clinicaltrials.gov/ (

trial

NA NA NA 3.5% NA None NA CR NA

clinical 1 1 1 1 1 1 1 1

of

Phase Phase Phase Phase Phase Phase Phase Phase Phase

number

NCT,

(patients)

55 34 29 50 40 30 24 30 n rate;

response

and

status balance

overall

of

fatigue

mental loss

ORR,

nausea NA NA NA NA NA Diarrhea, NA Toxicity change, Fatigue, progression; 8, 9

at a and 15,

9, to

a

2 will cycle

dose

×

8, 9, 2.4,

to 8, by

days

cycles. 2

a

of

infusion 1, 8,

dose 6 3,

time

while up

of on

be each

11

1.2, mg/m

days

2,

to by 2,

no

and

min daily

15 at 17. of

1,

days cycles. Starting mouth 1, cycles. 28 0.1 weekly

continuously days, up TTP,

and will

30 15 days bolus

2

followed 18

without

with on 13 by 13 8, and Bortezomib and

oral IV cohorts

28

,

days 2 28 days

2 2 2 days 11,

toxicities dose

4, 8,

every

cycle, cycle and 16

days 14 , mg/m

1, 1, on for

× 4.4, period 0332991 15, 15, Every

mg/m 12 response; 3

every

administered mg/m mg/m mg/m

Maximum 28-day limiting Starting be day vorinostat 20 20 21-day 4.6, for 10, Days Days daily 11, daily IV NPI-0052 16, treatment. PD Every Carfilzomib 0.7 15 0.125 intravenous Dose 15 maximum administered partial

PR,

lymphoma

for

MCL

response;

treatment Relapsed Lymphoma R/Lymphoma NHL Lymphoma MCL/MM R/R-Lymphoma Lymphoma Condition

in

complete )

CR,

Vorinostat PD0332991 :

+ Inhibitors +

Vorinostat

Continued ( +

1

18770

0052

CEP MLN9708 NPI-0052 NPI-0052 NPI Carfilzomib Carfilzomib Proteasome PI Bortezomib Abbreviations Table

K.S. Suh et al. / Critical Reviews in Oncology/Hematology 87 (2013) 306–322 313

Fig. 2. Chemical structures of major proteasome inhibitors in the treatment of lymphoma bortezomib, CEP 18770, marizomib, ixizomib and carfilzomib.

that treatment with bortezomib in combination with doxoru- assessed. Bortezomib was well tolerated with no grade 4

bicin and Taxol was schedule-dependent, and was additive toxicity. The most common grade 3 hematologic toxicities

with tumor necrosis factor-related apoptosis-inducing ligand were neutropenia (2 patients, 17%) and thrombocytopenia

(TRAIL) and synergistic with dexamethasone [90]. A recent (2 patients, 17%). However, no grade 3 or higher anemia

report demonstrated that cytarabine interferes with DNA was recorded. The most common non-hematologic toxic-

polymerases and incorporates into the genome during DNA ity was sensory neuropathy, which was observed in 50% of

replication, consequently arresting cell cycle progression patients. One patient (8%) reported grade 3 sensory neuropa-

[91]. Furthermore, cytarabine shows sequence-dependent thy but had complete recovery within six weeks. There was no

synergism with bortezomib in MCL [92]. A combination observation of GI toxicity or fatigue, perhaps due to inherent

of rituximab and bortezomib for treatment of MCL, FL, differences between diseases or differences in prior therapy.

and marginal zone lymphoma is under clinical development The safety profile of bortezomib for use in lymphoma

[93]. treatment appears to be similar to previous experiences in

The safety of bortezomib has been demonstrated by clini- MM. During the first phase II clinical trial, the Study of

cal trials (Table 4). One such study was a phase II, multicenter Uncontrolled Managed with Proteasome

clinical trial of patients with relapsed or refractory MCL Inhibition Therapy (SUMMIT), bortezomib was adminis-

2

(PINNACLE study). In total, 152 patients were treated with tered at 1.3 mg/m IV on days 1, 4, 8, and 11 of a 21-day

2

bortezomib administered at 1.3 mg/m intravenously (IV) on cycle for a maximum of eight cycles [96]. Grade 3 toxicities

days 1, 4, 8, and 11 of a 21-day cycle for up to 17 cycles included thrombocytopenia (28%), peripheral neuropathy

[94]. The most common non-hematologic adverse effects of (12%), fatigue (12%), and neutropenia (11%). A second

grade 3 or higher were diarrhea (11 patients, 7%), fatigue (19 phase II trial, the Clinical Response and Efficacy Study of

patients, 12%), and peripheral neuropathy (20 patients, 13%). Bortezomib in the Treatment of Relapsing Multiple Myeloma

The median onset of neuropathy was estimated at 12 weeks. (CREST) (Table 2), was an open-label, randomized trial

However, data on reversibility was not collected. The most of 54 patients who were previously treated for MM [97].

common hematologic toxicity was thrombocytopenia, which Patients were randomized to receive bortezomib at a dose of

2 2

occurred in 17 (11%) patients. Five (3%) patients died from 1.3 mg/m or 1 mg/m IV using the same dosing schedule as

bortezomib-associated causes, including non-neutropenic the SUMMIT trial (Table 2). The incidence of drug-related

sepsis and respiratory failure. The most common adverse adverse effects was considerably lower for patients receiv-

2 2

effects leading to treatment discontinuation were peripheral ing 1 mg/m versus 1.3 mg/m . The most common adverse

neuropathy (10%) and fatigue (6%). The safety of bortezomib effects were peripheral neuropathy (58% vs. 19%), diarrhea

was evaluated in another phase II clinical trial in patients (65% vs. 25%), nausea (62% vs. 46%), and vomiting (38%

with relapsed or refractory cutaneous T-cell lymphoma vs. 14%). The relatively common occurrence of peripheral

[95]. Patients were treated with bortezomib administered at sensory neuropathy associated with bortezomib necessitates

2

1.3 mg/m IV on days 1, 4, 8, and 11 of a 21-day cycle for up early detection, and recommendations for dose modifications

to six cycles. Fifteen patients were registered and 12 were are detailed in Table 3 [98].

314 K.S. Suh et al. / Critical Reviews in Oncology/Hematology 87 (2013) 306–322

Table 2

Combined safety data for pivotal phase II clinical trial results for patients with multiple myeloma at standard doses.

Study Dose Patients Toxicity ≥ 20% Overall (%)/Grade 3 (%)

2

1.3 mg/m 202 Nausea and vomiting 55%/6% and 27%/8%

Diarrhea 44%/7%

Fatigue 41%/12%

SUMMIT trial [96]

Thrombocytopenia 40%/28%

Peripheral neuropathy 31%/12%

Anemia 21%/8%

2

1.3 mg/m 28 Nausea 41%/0%

2

1 mg/m 26 Diarrhea 39%/4%

CREST trial [97] Fatigue 56%/7%

Thrombocytopenia 28%/22%

Peripheral neuropathy 35%/9%

Table 3

Prescribing information: recommended dose modifications for bortezomib-related neuropathic pain and/or peripheral sensory or motor neuropathy [97].

Severity of peripheral neuropathy Modification of dose and regimen

Grade 1 (asymptomatic; deep tendon reflex loss or No action needed

paresthesia) without pain or loss of function

2

Grade 1 with pain or Grade 2 (moderate symptoms; limiting Reduce bortezomib dose to 1 mg/m

instrumental activities of daily living)

Grade 2 with pain or Grade 3 (severe symptoms; self-care Withhold bortezomib therapy until symptoms of toxicity resolve; after symptoms resolve,

2

limiting activities of daily living) reinitiate bortezomib at 0.7 mg/m and change treatment schedule to once weekly

Grade 4 (life-threatening consequences with urgent Discontinue bortezomib treatment

intervention indicated; permanent sensory loss)

A number of conclusions and recommendations can be during cycles 1–2. Nausea and vomiting may require the use

drawn from these trials. Both of these myeloma trials con- of anti-emetics; diarrhea may be controlled with antidiar-

firm that thrombocytopenia is not associated with serious rheal medications, and constipation can be managed with

bleeding events (Table 2), although one patient developed stool softeners and laxatives (Table 4).

an episode of gastrointestinal bleeding with grade 3 throm- There have been a number of attempts to identify biomark-

−1

bocytopenia (platelet count < 50,000 mL ). Anemia and ers to predict responses to bortezomib to spare a population

neutropenia associated with bortezomib are usually not prob- of patients that is unlikely to respond favorably to the drug.

lematic. Fatigue usually occurs during cycles 1–2 and may A few studies showed that antitumor activity of bortezomib

persist for several cycles before subsiding. Treatment should is greater against tumors with an NF-␬B mutation and cyclin

be withheld at the onset of any treatment-associated grade D overexpression (JNCI). Such pre-selection may potentially

3 fatigue. Once symptoms resolve, bortezomib therapy may improve ORR while sparing unnecessary exposure to therapy.

be reinitiated with a 25% dose reduction. Bortezomib does

not appear to be directly cytotoxic to most normal bone 3.3. Class C compounds

marrow cells or to destroy progenitor cells [99]. Complete

blood counts should be performed prior to bortezomib dose Class C compounds are the first natural proteaso-

administration. Dose adjustments for bortezomib-associated mal inhibitors discovered and contain ␤-lactone structures.

hematologic toxicity are detailed in Table 4. Gastrointestinal More recent experiments have demonstrated that lacta-

adverse events occur frequently, with the most common being cystin blocks cell-cycle progression, inhibits degradation

nausea, diarrhea, constipation, and vomiting. These gastroin- of KIP1, and induces apoptosis of activated B-CLL cells

testinal disturbances can occur at any time during treatment, with little effect on resting B-CLL cells [3]. NPI-0052

but in clinical trials they have occurred most frequently (marizomib; Fig. 2) was originally isolated from marine

Table 4

Prescribing information: recommended dose modifications for bortezomib-related hematologic toxicities [97].

Hematologic toxicity Modification of dose

−3

Platelets ≤30,000 mm or absolute neutrophil count Withhold bortezomib; if several bortezomib doses in consecutive cycles are withheld, reduce

−3 2 2 2

≤750 mm on bortezomib treatment day(s) (except day 1) dose 1 level (1.3 mg/m /dose reduced to 1 mg/m /dose; 1 mg/m /dose reduced to 2 0.7 mg/m /dose)

−3 2

Grade 4 hematological toxicity: platelets ≤25,000 mm or Withhold until toxicity resolved; may reinitiate with a 25% dose reduction (1.3 mg/m /dose

−3 2 2 2

absolute neutrophil count ≤500 mm on bortezomib reduced to 1 mg/m /dose; 1 mg/m /dose reduced to 0.7 mg/m /dose)

treatment day(s)

K.S. Suh et al. / Critical Reviews in Oncology/Hematology 87 (2013) 306–322 315

bacteria. It resembles lactacystin, and is used largely for exhibited grade 3 thrombocytopenia, thus establishing

2

cancer treatment. However, unlike bortezomib, NPI-0052 15 mg/m as the maximum tolerated dose (Table 3). Anti-

2

irreversibly inhibits the three major enzymatic activities tumor responses were observed at doses ≥11 mg/m . More

(chymotrypsin-, trypsin-, and caspase-like) of the 20S pro- than one-third of patients experienced grade 1 or 2 non-

teasome [71,100]. When compared to bortezomib, NPI-0052 hematologic toxicities including diarrhea, fatigue, and nausea

exhibits an improved therapeutic ratio and significant activity in the absence of grade 3–4 peripheral neuropathy, whereas

in hematologic malignancies and solid tumors, includ- 48% (n = 14) of patients experienced grade 3–4 toxicity, thus

ing bortezomib-resistant MM. To overcome inherent and establishing the reduced incidence of neuropathy associ-

acquired bortezomib resistance and for broader anti-cancer ated with carfilzomib [109]. Co-administration of carfilzomib

effects, NPI-0052, with its structural and pharmacological (subtoxic or minimally toxic levels) in combination with

uniqueness, is being tested both as a single agent and as com- the histone deacetylase inhibitor vorinostat potentiates the

bined with biologics and targeted therapeutic agents (Table 4) activities of carfilzomib including mitochondrial injury, cell

[101]. This marine-derived, orally active, broad-spectrum cycle arrest, and caspase activation, leading to apoptosis in

proteasome inhibitor has shown potent antitumor activity in both germinal-center B cell-like DLBCL and activated B

leukemia in vitro and in a leukemic mouse model in vivo cell-like DLBCL [110]. Given the promising results of com-

[102,103]. Interestingly, combining NPI-0052 with either binatorial drug administration with proteasome inhibitors and

MS-275 or valproic acid (VPA) induced greater levels of cell histone deacetylase inhibitors, a phase I clinical trial has been

death than the combination of bortezomib with these histone established for carfilzomib (PR-171) in combination with

deacetylase (HDAC) inhibitors [102]. A phase II clinical trial vorinostat (SAHA) in patients with relapsed/refractory B cell

(n = 30) demonstrated that NPI-0052 was well tolerated and lymphomas [111] (Table 1).

inhibited caspase- and trypsin-like activities with 51% and

72% efficacy, respectively, which led to stable disease in 31%

of cases in MCL, HL, FL, and other solid tumors [104]. Simi- 3.5. Class E compounds

lar to bortezomib, NPI-0052 increased cytotoxic effects when

used in combination with 5-fluorouracil, CTP-11, Avastin Class E compounds are macrocyclic vinyl ketones

(bevacizumab), leucovorin, or in tumor xenograft that irreversibly inhibit proteasomal activity. While small-

models (Fig. 2) [105]. Due to the greater potency of NPI-0052 molecule inhibitors in general suffer from poor pharma-

compared to bortezomib in increasing apoptotic potential in cokinetics due to rapid renal clearance, more potent and

CLL and in preclinical settings [106], further clinical devel- sustained effects can be expected from these newly developed

opments are expected. irreversible proteasomal inhibitors.

As a part of developing combination therapy for hemato-

3.4. Class D compounds logical malignancies, the BH3- mimetic ABT-737 was found

to exhibit synergistic potential in combination with borte-

Class D proteasomal inhibitors have epoxyketone struc- zomib against CLL, MCL, and DLBCL. Among these, MCL

tures. The efficacy of the class D compound carfilzomib is showed the highest sensitivity toward ABT-737 and DLBCL

under investigation in clinical trials (Table 1). Carfilzomib showed the least [112]. Currently ABT-737 is in a clinical trial

(Fig. 2) is an irreversible proteasomal inhibitor that primarily in oral form as ABT-263, which exhibits high-affinity binding

inhibits chymotrypsin-, trypsin-, and caspase-like activities to Bcl-2 and Bcl-xL, thus inducing apoptosis by preventing

of the proteasome. It has equal potency but greater selectiv- the sequestration of proapoptotic molecules in hematological

ity for the chymotrypsin-like activity of the proteasome than malignancies, including MM [113,114]. Similarly, the anti-

bortezomib, causing stronger cytotoxic effects and enhanc- tumor effects and sensitivity of tumor cells to ABT-737 have

ing cell cycle arrest and apoptosis in vitro. In animal studies, been assessed in six MCL cell lines and in primary MCL cells

carfilzomib-mediated inhibition of proteasome activity was (n = 13); the results showed that ABT-737 induces apoptosis

high low

found to occur in a dose-dependent manner in all tissues in MCL cells that express a Bcl-2 /Mcl-1 profile [115].

examined, with the exception of the brain [71]. Epoxomicin This study shows that down-regulation of Mcl-1 enhances

is a selective, irreversible inhibitor of the chymotrypsin-like the effects of ABT-737, and indicates that the Bcl-2/Mcl-1

activity of the proteasome, which promotes proapoptotic profile of the tumor should guide the use of ABT-737 in the

activity in anaplastic large-cell lymphoma cell lines [107] treatment of MCL.

and in Bcl-2-overexpressing recombinant Jurkat cells in vitro In addition to the use of drugs that inhibit proteasomal

[108]. Carfilzomib is being evaluated in early clinical trials activity, other enzymes such as E3 ligase or de-ubiquitylating

and in a phase I trial in patients with refractory and relapsed enzymes may be additional, alternative molecular targets

hematologic malignancies, including MM and MCL. In the for effectively inhibiting the proteasomal degradation path-

latter trial (n = 29), carfilzomib was administered every day way [116]. The ubiquitin-like molecules represent another

for 5 consecutive days every 14 days at 1.2, 2.4, 4.6, 4.4, family of enzymes of interest for drug development. Numer-

2

11, 15, and 20 mg/m . No dose-limiting toxicities were ous substrates for SUMO and NEDD8 have been identified

2 2

observed up to 15 mg/m . At 20 mg/m , two of five patients as drugable targets by proteomics approaches [71]. Major

316 K.S. Suh et al. / Critical Reviews in Oncology/Hematology 87 (2013) 306–322

proteasomal inhibitors currently being evaluated in clinical a recent study analyzing 18 MCL samples including cell

trials are listed in Table 1. lines, Roué et al. demonstrated that loss of sensitivity of

Similarly, HDAC inhibitors have been reported to have a MCL cells to proteasome inhibitors is related to the up

synergistic effect in MCL therapy when combined with pro- regulation of prosurvival chaperone BiP/Grp78 and to the

teasomal inhibitors, where the combination induces higher stabilization of this protein by increased chaperoning activ-

cytotoxicity and cell death. Studies on interactions of SAHA ity of Hsp90 (Heat shock protein 90 kDa). Knocking down

(HDAC inhibitor) with bortezomib in MCL reveal that the of BiP/Grp78 coupled with Hsp90 inhibitor IPI-504 (class:

combination of both drugs synergistically induces apoptosis ansamycin) in combination with bortezomib resulted in apo-

in MCL cells [117]. ptosis of MCL cells. Results of this study further indicate that

in MCL cultures and tumors the combination of bortezomib

and IPI-504 can overcome both intrinsic and acquired resis-

4. Unfolded protein response (UPR) tance via dissociation of Hsp90/BiP complexes, BiP/Grp78

depletion, inhibition of UPR, and thus, enhanced cell death,

Cellular stress can cause disruption in peptide processing suggesting the effectiveness of this combination in treat-

in endoplasmic reticulum either by altering Calcium levels ment of patients with MCL refractory to bortezomib therapy

or by interfering with glycosylation and disulfide bond for- [124].

mation, leading to the induction of a cascade of intracellular

stress signaling—the UPR (unfolded protein response)—due

to the accumulation of the unfolded proteins. Induction of 5. Apoptosis

the UPR results in a reduction in general protein synthesis,

an increase in clearing misfolded proteins, and a restoration Proteasome inhibitors mediate cell cycle arrest and apo-

of peptide processing in the ER lumen. IRE-1 and ATF6 ptosis by causing dysregulation of a number of proapoptotic

can induce UPR since they are ER proximal stress sensing proteins in lymphoma cells [125]. Earlier studies reported

molecules [118,119]. In stressed normal cells, UPR ame- the accumulation of p53 in cells treated with proteasome

liorates protein misfolding and helps the cells to survive inhibitors, causing up regulation of the p53-inducible gene

the stress. However, in tumorigenesis, UPR plays a signif- products p21 and Mdm-2[126]. In normal cells, the RING

icant role in maintaining malignancy and chemoresistance domain of Mdm2 or Hdm2 (the human counterpart of Mdm2)

of malignant cells by upregulating the expression of XBP1, binds to the tumor suppressor p53, and an E3 ubiquitin ligase

inducing ATF4 and CHOP (C/EBP homologous protein), and then targets p53 to enhance its rapid degradation [127]. Upon

activating ATF6 [120]. proteasome inhibitor treatment, p53-Mdm2 binding is inter-

Recent studies have revealed that proteasomal inhibitors rupted; p53 becomes phosphorylated, and initiates proapop-

(e.g., bortezomib) induce endoplasmic reticular stress and totic signaling [128]. Disruption of the p53–Mdm2 interac-

UPR in MM cells. The intensity of the response to borte- tion leads to defective apoptosis [129], and overexpression of

zomib depends on levels of XBP1 [121]. Also, proteasomal wild type p53 increases sensitivity to proteasome inhibitors

inhibitors cause accumulation of unfolded proteins in tumor in Burkitt’s lymphoma, suggesting that effective apoptosis

cells, and trigger proapoptotic signaling leading to cell death mediated by proteasome inhibitors requires an intact p53

[120]. In MM cells, bortezomib and tunicamycin activate pathway [130]. In contrast, recent reports indicate that borte-

ER stress specific PERK (PKR-like ER kinase) in less than zomib induces apoptosis by mitotic catastrophe independent

an hour. Similarly, constitutive expression of physiologic of p53 activity in B-cell lymphoma cells in vitro [72].

UPR genes is necessary in MM cells since they produce Proteasome inhibitors activate apoptosis in Lymphoma

significant amounts of immunoglobulin which causes them cells. Two major pathways are involved in apoptosis-

to be inherently sensitive to proteasome inhibitors, espe- associated caspase activation in mammalian cells: the

cially bortezomib, which selectively induces apoptosis in extrinsic death receptor pathway, which is regulated by mem-

MM cells. However; MM cells constantly express ER stress bers of the death receptor superfamily, and the intrinsic

survival proteins necessary to support their role as secre- mitochondrial pathway, which is associated with extracel-

tory cells; hence, they have a lower threshold for proteasome lular cues and internal insults [131]. The active form of

inhibitor-induced UPR induction and ER stress-induced apo- caspase-3 is necessary to cleave the other initiator caspases,

ptosis [122]. Moreover, as detailed analysis of the role of which ultimately leads to the cleavage of many cellular pro-

UPR in balancing apoptosis of cancer cells, dormancy, and tein substrates and apoptosis. It has recently been shown that

invasive growth and increasing susceptibility of tumors to caspase-3 is degraded by the 26S proteasome. Proteasome

chemotherapeutic agents has shown (reviewed earlier by Ma inhibitors induce the accumulation of caspase-3 subunits and

and Hendershot [123]), prolonged activation of UPR could enhance apoptosis induced by overexpression of pro-caspase-

result in apoptosis. 3 [132]. Another indication that caspase-3 is targeted by

Similarly, inhibition of UPR plays a significant role ubiquitination for degradation is that monoubiquitination or

in reducing the resistance of MCL cells to proteasome diubiquitination of caspase-3 subunits was seen after protea-

inhibitors, thereby increasing the efficacy of treatment. In some inhibitor treatment of cells [133,134].

K.S. Suh et al. / Critical Reviews in Oncology/Hematology 87 (2013) 306–322 317

TRAIL has the ability to induce apoptosis in cancer cells pathways in the cell including (1) the inhibition of NF- B,

with little or no toxicity to normal cells. Death receptor 5 which contributes to many features of the malignant phe-

(DR5; also called TRAIL-R2, Apo2, TRICK2, or KILLER) notype, (2) maintenance of p27 and p21 levels, which are

is a receptor for TRAIL. In fact, preclinical studies in mice cell cycle-dependent kinase inhibitors, (3) accumulation of

provided the first evidence that the soluble form of recom- proapoptotic proteins in the mitochondria, and (4) effects on

binant TRAIL would suppress the growth of human tumor cytokines and cell adhesion molecules [147]. The UPS tar-

xenografts with no apparent systemic toxicity [135]. DR5 gets the p27 protein in MCL, whereas, in FL and aggressive

mediates TRAIL-induced apoptosis through the interaction lymphoma, NF-␬B is targeted.

with adapter proteins, such as FADD, and through caspase Proteins like p21 and p27 are members of the Cip/Kip

activation [136]. Ubiquitin ligases bind to caspases, resulting family of cyclin-dependent kinase inhibitors that promote

in the degradation of these caspases and possibly tumori- cell cycle progression at the G1-S phase junction by inac-

genesis. However, proteasome inhibitors can induce DR5 tivating cyclin/cdk complexes [147,148]. These proteins are

expression and sensitize tumor cells to TRAIL-induced apo- substrates for the UPS, and drugs that affect this pathway

ptosis [137–139]. could lead to the accumulation of regulatory proteins that

Proteasome inhibitors also induce the activation of Bax, a could cause cell cycle arrest. Most cases of MCL (91/112)

member of the proapoptotic Bcl-2 family, which is required and DLBCL (12/19) have been shown to have lost expres-

for apoptosis [140]. As a result of apoptotic signaling and sion of p27 [21,149], but other subtypes of NHL such as

caspase activation, the BH3 domain of Bax dimerizes and small lymphocytic lymphoma and extranodal marginal zone

becomes resistant to proteasomal degradation. Bax then lymphomas have not been found to have any loss of p27

translocates into the mitochondria, which leads to the release expression [149].

of proapoptotic mitochondrial factors such as cytochrome c Molecular and in vivo protein degradation assays showed

resulting in apoptosis [139]. that MCL exhibited normal p27 mRNA expression but

Proteins known as ␤-transducin repeat-containing pro- increased p27 protein loss. Correlation of the p53 and p27

teins (␤-TrCPs) play a role as substrates of E3 ubiquitin defects with clinical data from patients with MCL indicates

ligases that control stability of important regulators of the cell that patients with both defects exhibit a significant reduc-

cycle and signal transduction [141]. One of the major mech- tion in overall survival. The explanations for the loss of p27

anisms of the anticancer effects of proteasomal inhibitors is might be (1) the possible sequestration of p27 by cyclins

thought to be the suppression of prosurvival NF-␬B due to sta- D1 and D3, which are overexpressed in MCL and (2) an

bilization of its inhibitor I␬B [142]. Proteasomal degradation over-accumulation of Skp2, which is a part of the p27Kip1

of I␬B requires phosphorylation-dependent ubiquitination, ubiquitin ligase (E3) combination that plays a significant role

which is mediated by ␤-TrCPs [143,144]. Thus, ␤-TrCP func- in the degradation of the CDK inhibitor [147,149]. High

tion is essential for the induction of NF-␬B transcriptional levels of Skp2 correlate with greater E3 enzyme activity,

activities, which play a key role in proliferation and survival which ultimately leads to proteasome-mediated degradation

of cancer cells and are often augmented in human cancers of the target protein p27. In large B-cell lymphomas and

[141]. blastic MCL, elevated Skp2 levels were significantly asso-

NF- B activation contributes to many aspects of tumor ciated with a low p27Kip1 level, indicating that increased

development, such as accelerated cell cycle progression, cell proteasome-mediated degradation of p27Kip1 contributes

proliferation, tumor initiation, and metastasis. As a major to the malignant phenotype [149], an observation in MCL

antiapoptotic factor, NF-␬B is involved significantly in the that provides a powerful rationale for the use of proteasome

resistance of tumors to chemotherapy and radiation. Constitu- inhibitors in lymphoma [147].

tive activation of NF-␬B is seen in many human malignancies, FL is characterized by the translocation of the Bcl-2 proto-

including [145]. Furthermore, ␤-TrCP1 and oncogene from chromosome 18q21 to the immunoglobulin

␤-TrCP2 proteins seem to play a redundant role in ubiquitin- heavy chain locus at chromosome 14q32 [150]. This

ation and degradation of I␬B. Also, expression of ␤-TrCP2 is translocation t(14:18) leads to the overexpression of the anti-

induced in human breast lines and primary tumor apoptotic Bcl-2 protein, which is driven by NF-␬B, protecting

samples [143,146]. cells from apoptosis. Furthermore, NF-␬B overexpression

Targeting ␤-TrCP for inhibition with an RNAi approach is seen in lymphoma cells carrying t(14:18), and it has

or forced expression of a dominant-negative ␤-TrCP mutant been demonstrated that cell lines expressing an I␬B␣ super-

suppresses growth and survival of human breast cancer cells. repressor exhibit decreased levels of Bcl-2 protein, indicating

In addition, inhibiting ␤-TrCP augments the antiproliferative a role for NF-␬B in cells harboring this translocation. Since

effects of anticancer drugs such as doxorubicin, tamoxifen, proteasome inhibitors are employed in FL, a relationship

and on human mammary tumor cells. These data between NF-␬B and the apoptotic pathways is suggested in

indicate that targeting ␤-TrCP is a likely beneficial approach FL [147,151]. Also, limited effects of proteasome inhibitors

for anticancer therapies [141]. have been seen in DLBCL, whereas some evidence suggests

Bortezomib is capable of inhibiting the 26S protea- that NF-␬B helps to define the chemotherapy-resistant post-

some, and has the ability to target a number of different GC B-cell lymphomas [152].

318 K.S. Suh et al. / Critical Reviews in Oncology/Hematology 87 (2013) 306–322

In hematological malignancies, increased levels of TNF-␣ generation proteasome inhibitors such as carfilzomib, mari-

result in poor prognosis, and this in turn heightens the risk zomib, MLN9708, CEP-18770, and ONYX-0912 along with

of MM and other cancers such as hepatocellular carcinoma, targeted agents such as heat shock protein inhibitors, histone

gastric carcinoma, and breast and bladder cancer. Promyelo- deacetylase inhibitors, and AKT inhibitors may also drive

cytic leukemia protein (PML) is a known molecular marker lymphoma therapy in a promising direction. Bortezomib

for cancer, and since PML regulates cell growth, DNA repair, in combination with widely used chemotherapeutic drugs

and cell death, patients with normal expression of PML and such as dexamethasone, rituximab, , ,

P53 have a better survival chance compared to patients with melphalan, doxorubicin, thalidomide, , and

poor expression of PML and P53 [153]. etoposide is being evaluated under clinical trials for MCL,

MM, WM (Waldenstrom macroglobulinemia), and other

hematological malignancies (http://www.clinicaltrials.gov/).

6. Conclusion However, there are challenges and limitations regarding

toxicity profiles of proteasome inhibitors when used in com-

Multiple therapeutic targets based on proteasomal inhibi- bination therapy such as peripheral neuropathy, fatigue,

tion have been identified in hematological malignancies such nausea, anxiety, insomnia, and transient cognitive changes.

as MCL, NHL, Hodgkin’s lymphoma, MM, CLL, FL, ALL, The future of proteasome inhibition in lymphoma therapy

and DLBCL. As alternatives to conventional combination resides in successfully managing these challenges, improv-

chemotherapy, proteasome inhibitors have been used to treat ing safety and tolerability, and decreasing chemotherapy

lymphomas as single agents or in combination with HDAC resistance for the betterment and prolonged survival of

inhibitors or BH3 family inhibitors. Proteasome inhibitors patients.

consist of synthetic and natural products, and belong to five

different classes based on chemical structure (A to E). It has

been shown that proteasome inhibitors enhance antiprolif- Reviewers

erative, proapoptotic, antitumor, and antiangiogenic effects

of conventional therapeutic agents in many hematologi- Samir Parekh, Albert Einstein Cancer Center, Jack adn

cal malignancies. Among proteasome inhibitors, bortezomib Pearl Resnick Campus, 1300 Morris Park Avenue, Bronx,

(a class B drug) was the first to receive FDA approval NY 10461, United States.

– against MCL (for patients who had received one prior Andrés J. Ferreri, M.D., Fondazione IRCCS, START

therapy) and MM (as an initial treatment). Substantial clin- Project, Via Venezian 1, IT-20133 Milan, Italy.

ical benefits of bortezomib treatment have been observed

in patients with MCL (relapsed or refractory). Further-

more, bortezomib (intravenous or subcutaneous infusion) References

was successful in enhancing the antitumor activities of radio-

therapy, immunotherapy, and chemotherapy, and was well

[1] Nalepa G, Rolfe M, Harper JW. Drug discovery in the ubiquitin-

tolerated with minimal added toxicities. Similarly, protea- proteasome system. Nature Reviews Drug Discovery 2006;5:

some inhibitors such as CEP-18770 (oral administration), 596–613.

[2] Nandi D, Tahiliani P, Kumar A, Chandu D. The ubiquitin-proteasome

carfilzomib (infusion), and marizomib (oral or intravenous),

system. Journal of Biosciences 2006;31:137–55.

and MLN9708 (intravenous or oral) are also currently being

[3] Adams J. The development of proteasome inhibitors as anticancer

clinically evaluated for various hematological malignan-

drugs. Cancer Cell 2004;5:417–21.

cies. Thus, the inhibition of tumor proteasomal activity has [4] Ross CA, Pickart CM. The ubiquitin-proteasome pathway in Parkin-

son’s proved to be highly significant in treating lymphomas and disease and other neurodegenerative diseases. Trends in Cell

Biology 2004;14:703–11.

other malignancies, and has shown potential impact on vari-

[5] Varshavsky A. Regulated protein degradation. Trends in Biochemical

ous transcription factors including NF-␬B, IkB, and c-Myc.

Sciences 2005;30:283–6.

Taken together, proteasome inhibitors comprise a group of

[6] Schmidt M, Hanna J, Elsasser S, Finley D. Proteasome-associated

promising therapeutics for future anti cancer therapy; they proteins: regulation of a proteolytic machine. Biological Chemistry

2005;386:725–37.

may be used as single agents or used in combination to

[7] Loureiro J, Ploegh HL. Antigen presentation and the ubiquitin-

enhance the antitumor effects of conventional drugs.

proteasome system in host-pathogen interactions. Advances in

Since mechanisms of proteasome inhibition are dif-

Immunology 2006;92:225–305.

ferent among the proteasome inhibitors as described

[8] Borissenko L, Groll M. Diversity of proteasomal missions: fine tuning

earlier, a combination therapy with multiple PIs which of the immune response. Biological Chemistry 2007;388:947–55.

[9] Gao G,

are chemically different should be interesting in arresting Luo H. The ubiquitin-proteasome pathway in viral infections.

Canadian Journal of Physiology and Pharmacology 2006;84:5–14.

tumorigenesis and managing lymphoma patient outcomes.

[10] Shackelford J, Pagano JS. Targeting of host-cell ubiquitin pathways

Combinations of proteasome inhibitors with novel cyto-

by viruses. Essays in Biochemistry 2005;41:139–56.

toxic and conventional agents are currently being evaluated

[11] Ambagala AP, Solheim JC, Srikumaran S. Viral interference with

in pre-clinical and clinical trials to improve patient MHC class I antigen presentation pathway: the battle continues. Vet-

erinary Immunology and Immunopathology 2005;107:1–15.

responses and outcomes. Various combinations of second

K.S. Suh et al. / Critical Reviews in Oncology/Hematology 87 (2013) 306–322 319

[12] Watanabe H, Tanaka Y, Shimazu Y, et al. Cell-specific inhibition of [32] Zhang KH, Li GL, Liu ZZ. Expressions of cell cycle associated factors

paramyxovirus maturation by proteasome inhibitors. Microbiology geminin and cdt1 in patients with acute leukemia. Zhongguo Shi Yan

and Immunology 2005;49:835–44. Xue Ye Xue Za Zhi 2011;19:578–81.

[13] Ott DE, Coren LV, Sowder 2nd RC, Adams J, Nagashima K, Schu- [33] Bastians H, Topper LM, Gorbsky GL, Ruderman JV. Cell cycle-

bert U. Equine infectious anemia virus and the ubiquitin-proteasome regulated proteolysis of mitotic target proteins. Molecular Biology

system. Journal of Virology 2002;76:3038–44. of the Cell 1999;10:3927–41.

[14] Kisselev AF, Goldberg AL. Proteasome inhibitors: from research tools [34] Hachem A, Gartenhaus RB. Oncogenes as molecular targets in lym-

to drug candidates. Chemistry and Biology 2001;8:739–58. phoma. Blood 2005;106:1911–23.

[15] Myung J, Kim KB, Crews CM. The ubiquitin-proteasome path- [35] Singer JD, Gurian-West M, Clurman B, Roberts JM. Cullin-3 targets

way and proteasome inhibitors. Medicinal Research Reviews cyclin E for ubiquitination and controls S phase in mammalian cells.

2001;21:245–73. Genes and Development 1999;13:2375–87.

[16] Yew PR. Ubiquitin-mediated proteolysis of vertebrate G1- and [36] Winston JT, Chu C, Harper JW. Culprits in the degradation of cyclin

S-phase regulators. Journal of Cellular Physiology 2001;187: E apprehended. Genes and Development 1999;13:2751–7.

1–10. [37] Bai M, Tsanou E, Agnantis NJ, et al. Expression of cyclin D3

[17] Hara T, Kamura T, Nakayama K, Oshikawa K, Hatakeyama S, and cyclin E and identification of distinct clusters of proliferation

Nakayama K. Degradation of p27(Kip1) at the G(0)-G(1) transition and apoptosis in diffuse large B-cell lymphomas. Histology and

mediated by a Skp2-independent ubiquitination pathway. Journal of Histopathology 2003;18:449–57.

Biological Chemistry 2001;276:48937–43. [38] Tzankov A, Gschwendtner A, Augustin F, et al. Diffuse large B-cell

[18] Tyers M, Jorgensen P. Proteolysis and the cell cycle: with this RING lymphoma with overexpression of cyclin e substantiates poor standard

I do thee destroy. Current Opinion in Genetics and Development treatment response and inferior outcome. Clinical Cancer Research

2000;10:54–64. 2006;12:2125–32.

[19] Geisen C, Karsunky H, Yucel R, Moroy T. Loss of p27(Kip1) [39] Marti A, Wirbelauer C, Scheffner M, Krek W. Interaction

cooperates with cyclin E in T-cell lymphomagenesis. Oncogene between ubiquitin-protein ligase SCFSKP2 and E2F-1 underlies

2003;22:1724–9. the regulation of E2F-1 degradation. Nature Cell Biology 1999;1:

[20] Martins CP, Berns A. Loss of p27(Kip1) but not p21(Cip1) decreases 14–9.

survival and synergizes with MYC in murine lymphomagenesis. [40] Molina-Privado I, Rodriguez-Martinez M, Rebollo P, et al. E2F1

EMBO Journal 2002;21:3739–48. expression is deregulated and plays an oncogenic role in sporadic

[21] Chiarle R, Budel LM, Skolnik J, et al. Increased proteasome Burkitt’s lymphoma. Cancer Research 2009;69:4052–8.

degradation of cyclin-dependent kinase inhibitor p27 is associated [41] Rempel RE, Mori S, Gasparetto M, et al. A role for E2F activi-

with a decreased overall survival in mantle cell lymphoma. Blood ties in determining the fate of Myc-induced lymphomagenesis. PLoS

2000;95:619–26. Genetics 2009;5:e1000640.

[22] Seki R, Ohshima K, Nagafuji K, et al. Rituximab in combination [42] DePamphilis ML. Review: nuclear structure and DNA replication.

with CHOP chemotherapy for the treatment of diffuse large B cell Journal of Structural Biology 2000;129:186–97.

lymphoma in Japan: a retrospective analysis of 1,057 cases from [43] Petersen BO, Wagener C, Marinoni F, et al. Cell cycle- and cell

Kyushu Lymphoma Study Group. International Journal of Hemato- growth-regulated proteolysis of mammalian CDC6 is dependent on

logy 2010;91:258–66. APC-CDH1. Genes and Development 2000;14:2330–43.

[23] Thullberg M, Bartek J, Lukas J. Ubiquitin/proteasome-mediated [44] Hoefnagel JJ, Vermeer MH, Jansen PM, et al. Primary cutaneous

degradation of p19INK4d determines its periodic expression during marginal zone B-cell lymphoma: clinical and therapeutic features in

the cell cycle. Oncogene 2000;19:2870–6. 50 cases. Archives of Dermatology 2005;141:1139–45.

[24] Helmrich A, Lee S, O’Brien P, et al. Recurrent chromosomal aber- [45] Xiong XD, Qiu FE, Fang JH, et al. Association analysis between

rations in INK4a/ARF defective primary lymphomas predict drug the Cdc6 G1321A polymorphism and the risk for non-Hodgkin

responses in vivo. Oncogene 2005;24:4174–82. lymphoma and hepatocellular carcinoma. Mutation Research

[25] Dunphy CH. Gene expression profiling data in lymphoma and 2009;662:10–5.

leukemia: review of the literature and extrapolation of pertinent clin- [46] Zavitz KH, Zipursky SL. Controlling cell proliferation in dif-

ical applications. Archives of Pathology and Laboratory Medicine ferentiating tissues: genetic analysis of negative regulators of

2006;130:483–520. G1 S-phase progression. Current Opinion in Cell Biology 1997;9:

[26] Quintanilla-Martinez L, Kremer M, Keller G, et al. p53 Mutations in 773–81.

nasal natural killer/T-cell lymphoma from Mexico: association with [47] Sherr CJ, Roberts JM. CDK inhibitors: positive and negative

large cell morphology and advanced disease. American Journal of regulators of G1-phase progression. Genes and Development

Pathology 2001;159:2095–105. 1999;13:1501–12.

[27] Sanchez-Beato M, Saez AI, Navas IC, et al. Overall survival in [48] Bernardi R, Liebermann DA, Hoffman B. Cdc25A stability is

aggressive B-cell lymphomas is dependent on the accumulation of controlled by the ubiquitin-proteasome pathway during cell cycle pro-

alterations in p53, p16, and p27. American Journal of Pathology gression and terminal differentiation. Oncogene 2000;19:2447–54.

2001;159:205–13. [49] Negishi Y, Ui N, Nakajima M, et al. p21Cip-1/SDI-1/WAF-1 gene

[28] Visco C, Canal F, Parolini C, et al. The impact of P53 and is involved in chondrogenic differentiation of ATDC5 cells in vitro.

P21(waf1) expression on the survival of patients with the germinal Journal of Biological Chemistry 2001;276:33249–56.

center phenotype of diffuse large B-cell lymphoma. Haematologica [50] Lavabre-Bertrand T, Henry L, Carillo S, et al. Plasma proteasome level

2006;91:687–90. is a potential marker in patients with solid tumors and hemopoietic

[29] McGarry TJ, Kirschner MW. Geminin, an inhibitor of DNA replica- malignancies. Cancer 2001;92:2493–500.

tion, is degraded during mitosis. Cell 1998;93:1043–53. [51] Valbuena JR, Rassidakis GZ, Lin P, et al. Expression of heat-

[30] Obermann EC, Eward KL, Dogan A, et al. DNA replication shock protein-90 in non-Hodgkin’s lymphomas. Modern Pathology

licensing in peripheral B-cell lymphoma. Journal of Pathology 2005;18:1343–9.

2005;205:318–28. [52] Shringarpure R, Catley L, Bhole D, et al. Gene expression analysis

[31] Pinyol M, Salaverria I, Bea S, et al. Unbalanced expression of of B-lymphoma cells resistant and sensitive to bortezomib. British

licensing DNA replication factors occurs in a subset of mantle cell Journal of Haematology 2006;134:145–56.

lymphomas with genomic instability. International Journal of Cancer [53] Robles AI, Wright MH, Gandhi B, et al. Schedule-dependent

2006;119:2768–74. synergy between the heat shock protein 90 inhibitor

320 K.S. Suh et al. / Critical Reviews in Oncology/Hematology 87 (2013) 306–322

17-(dimethylaminoethylamino)-17-demethoxygeldanamycin [75] O’Connor OA, Wright J, Moskowitz C, et al. Phase II clinical expe-

and doxorubicin restores apoptosis to p53-mutant lymphoma cell rience with the novel proteasome inhibitor bortezomib in patients

lines. Clinical Cancer Research 2006;12:6547–56. with indolent non-Hodgkin’s lymphoma and mantle cell lymphoma.

[54] Valbuena JR, Gualco G, Espejo-Plascencia I, Medeiros LJ. Classi- Journal of Clinical Oncology 2005;23:676–84.

cal Hodgkin lymphoma arising in the rectum. Annals of Diagnostic [76] Goy A, Younes A, McLaughlin P, et al. Phase II study of proteasome

Pathology 2005;9:38–42. inhibitor bortezomib in relapsed or refractory B-cell non-Hodgkin’s

[55] Grimm LM, Osborne BA. Apoptosis and the proteasome. Results and lymphoma. Journal of Clinical Oncology 2005;23:667–75.

Problems in Cell Differentiation 1999;23:209–28. [77] Chen CIKC, White D, Voralia M, et al. Bortezomib is active in

[56] Naujokat C, Hoffmann S. Role and function of the 26S pro- patients with untreated or relapsed Waldenstrom’s macroglobuline-

teasome in proliferation and apoptosis. Laboratory Investigation mia: a phase II study of the National Cancer Institute of Canada

2002;82:965–80. Clinical Trials Group. Journal of Clinical Oncology 2007;25:1570–5.

[57] Suzuki Y, Nakabayashi Y, Nakata K, Reed JC, Takahashi R. X- [78] Maki RGKA, Scheu K, Yamada J, et al. A multicenter Phase II

linked inhibitor of apoptosis protein (XIAP) inhibits caspase-3 and study of bortezomib in recurrent or metastatic sarcomas. Cancer

-7 in distinct modes. Journal of Biological Chemistry 2001;276: 2005;103:1431–8.

27058–63. [79] Davis NBTD, Ansari RH, Ryan CW, et al. Phase II trial of PS-341

[58] Yang Y, Fang S, Jensen JP, Weissman AM, Ashwell JD. Ubiquitin in patients with renal cell cancer: a University of Chicago phase II

protein ligase activity of IAPs and their degradation in proteasomes consortium study. Journal of Clinical Oncology 2004;22:115–9.

in response to apoptotic stimuli. Science 2000;288:874–7. [80] Stevenson J, Nho CW, Johnson SW, et al. Effects of Bortezomib

[59] Li B, Dou QP. Bax degradation by the ubiquitin/proteasome- (PS-341) on NF-␬B activation in peripheral blood mononuclear cells

dependent pathway: involvement in tumor survival and progression. (PBMCs) of advanced non-small lung cancer (NSCLC) patients:

Proceedings of the National Academy of Sciences of the United States a phase II/pharmacodynamic trial. Journal of Clinical Oncology,

of America 2000;97:3850–5. 2004 ASCO Annual Meeting Proceedings (Post-Meeting Edition)

[60] Degterev A, Boyce M, Yuan J. The channel of death. Journal of Cell 2004;22:7145.

Biology 2001;155:695–8. [81] Dunleavy K, Pittaluga S, Czuczman MS, et al. Differential efficacy of

[61] Degterev A, Lugovskoy A, Cardone M, et al. Identification of small- bortezomib plus chemotherapy within molecular subtypes of diffuse

molecule inhibitors of interaction between the BH3 domain and Bcl- large B-cell lymphoma. Blood 2009;113:6069–76.

xL. Nature Cell Biology 2001;3:173–82. [82] Kozuch PS, Rocha-Lima CM, Dragovich T, et al. Bortezomib with or

[62] Tzung SP, Kim KM, Basanez G, et al. Antimycin A mimics a without irinotecan in relapsed or refractory colorectal cancer: results

cell-death-inducing Bcl-2 homology domain 3. Nature Cell Biology from a randomized phase II study. Journal of Clinical Oncology

2001;3:183–91. 2008;26:2320–6.

[63] Davis RE, Brown KD, Siebenlist U, Staudt LM. Constitutive nuclear [83] Fowler N, Kahl BS, Lee P, et al. Bortezomib, bendamustine, and

factor kappaB activity is required for survival of activated B cell- rituximab in patients with relapsed or refractory follicular lym-

like diffuse large B cell lymphoma cells. Journal of Experimental phoma: the phase II VERTICAL study. Journal of Clinical Oncology

Medicine 2001;194:1861–74. 2011;29:3389–95.

[64] Stoffel A. The NF-kappaB signalling pathway: a therapeutic target [84] Sehn LH, MacDonald D, Rubin S, et al. Bortezomib ADDED to R-

in lymphoid malignancies? Expert Opinion on Therapeutic Targets CVP is safe and effective for previously untreated advanced-stage

2005;9:1045–61. follicular lymphoma: a phase II study by the National Cancer Insti-

[65] Ekiz HA, Can G, Baran Y. Role of autophagy in the progression and tute of Canada Clinical Trials Group. Journal of Clinical Oncology

suppression of leukemias. Critical Reviews in Oncology/Hematology 2011;29:3396–401.

2012;81:275–85. [85] Williams ME, Densmore JJ. Biology and therapy of mantle cell lym-

[66] Amaravadi RK, Yu D, Lum JJ, et al. Autophagy inhibition enhances phoma. Current Opinion in Oncology 2005;17:425–31.

therapy-induced apoptosis in a Myc-induced model of lymphoma. [86] Ludwig H, Hajek R, Tothova E, et al. Thalidomide-dexamethasone

Journal of Clinical Investigation 2007;117:326–36. compared with melphalan-prednisolone in elderly patients with mul-

[67] Mihelic R, Kaufman J, Lonial S, Flowers C. Maintenance therapy in tiple myeloma. Blood 2009;113:3435–42.

lymphoma. Clinical Lymphoma & Myeloma 2007;7:507–13. [87] Friedberg JW, Kelly JL, Neuberg D, et al. Phase II study of a

[68] Cashen AF, Bartlett NL. Therapy of relapsed Hodgkin lymphoma. TLR-9 agonist (1018 ISS) with rituximab in patients with relapsed

Blood Reviews 2007;21:233–43. or refractory follicular lymphoma. British Journal of Haematology

[69] Fuchs R, Loffler M, Pfreundschuh M, et al. Prognosis of high 2009;146:282–91.

dose chemotherapy/autologous bone marrow transplantation candi- [88] Lamm W, Kaufmann H, Raderer M, et al. Bortezomib combined

dates not receiving this treatment after failure of primary therapy of with rituximab and dexamethasone is an active regimen for patients

Hodgkin’s disease. Leukemia and Lymphoma 1994;14:79–89. with relapsed and chemotherapy-refractory mantle cell lymphoma.

[70] Cortelazzo S, Ponzoni M, Ferreri AJ, Dreyling M. Mantle cell lym- Haematologica 2011;96:1008–14.

phoma. Critical Reviews in Oncology/Hematology 2012;82:78–101. [89] Nabhan C, Villines D, Dalal N, Tolzien K, Kozolff M, Starr A. Borte-

[71] Piccinini M, Tazartes O, Mezzatesta C, et al. Proteasomes are a zomib (Velcade), rituximab, cyclophosphamide, and dexamethasone

target of the anti-tumour drug vinblastine. Biochemical Journal combination regimen is active as front-line therapy of low-grade

2001;356:835–41. non-Hodgkin lymphoma. Clinical Lymphoma, Myeloma & Leukemia

[72] Perez-Galan P, Roue G, Villamor N, Montserrat E, Campo E, Colomer 2012;12:26–31.

D. The proteasome inhibitor bortezomib induces apoptosis in mantle- [90] An J, Sun Y, Fisher M, Rettig MB. Antitumor effects of

cell lymphoma through generation of ROS and Noxa activation bortezomib (PS-341) on primary effusion lymphomas. Leukemia

independent of p53 status. Blood 2006;107:257–64. 2004;18:1699–704.

[73] Paoluzzi L, O’Connor OA. Mechanistic rationale and clinical evi- [91] Tobias SC, Borch RF. Synthesis and biological evaluation of

dence for the efficacy of proteasome inhibitors against indolent and a cytarabine phosphoramidate prodrug. Molecular Pharmaceutics

mantle cell lymphomas. BioDrugs 2006;20:13–23. 2004;1:112–6.

[74] Blade JSD, Reece D. Criteria for evaluating disease response and [92] Hutter G, Rieken M, Pastore A, et al. The proteasome inhibitor borte-

progression in patients with multiple myeloma treated by high-dose zomib targets cell cycle and apoptosis and acts synergistically in a

therapy and haemopoietic stem cell transplantation. British Journal sequence-dependent way with chemotherapeutic agents in mantle cell

of Haematology 1998;102:1115–23. lymphoma. Annals of Hematology 2012;91:847–56.

K.S. Suh et al. / Critical Reviews in Oncology/Hematology 87 (2013) 306–322 321

[93] O’Connor OA. Marked clinical activity of the proteasome inhibitor [114] Yecies D, Carlson NE, Deng J, Letai A. Acquired resistance to ABT-

bortezomib in patients with follicular and mantle-cell lymphoma. Clin 737 in lymphoma cells that up-regulate MCL-1 and BFL-1. Blood

Lymphoma Myeloma 2005;6:191–9. 2010;115:3304–13.

[94] Goy ABS, Kahl B. Bortezomib in patients with relapsed or refractory [115] Touzeau C, Dousset C, Bodet L, et al. ABT-737 induces apoptosis in

mantle cell lymphoma (MCL): preliminary results of the PINNACLE mantle cell lymphoma cells with a Bcl-2high/Mcl-1low profile and

study. In: Program and abstracts of the 41st annual meeting of the synergizes with other antineoplastic agents. Clinical Cancer Research

American Society of Clinical Oncology. 2005. 2011;17:5973–81.

[95] Zinzani PL, Musuraca G, Tani M, et al. Phase II trial of proteasome [116] Rossi M, De Laurenzi V, Munarriz E, et al. The ubiquitin-protein

inhibitor bortezomib in patients with relapsed or refractory cutaneous ligase Itch regulates p73 stability. EMBO Journal 2005;24:836–48.

T-cell lymphoma. Journal of Clinical Oncology 2007;25:4293–7. [117] Heider U, von Metzler I, Kaiser M, et al. Synergistic interaction of

[96] Richardson PG, Barlogie B, Berenson J, et al. A phase 2 study of the histone deacetylase inhibitor SAHA with the proteasome inhibitor

bortezomib in relapsed, refractory myeloma. New England Journal bortezomib in mantle cell lymphoma. European Journal of Haema-

of Medicine 2003;348:2609–17. tology 2008;80:133–42.

[97] Jagannath S, Barlogie B, Berenson J, et al. A phase 2 study of two [118] DuRose JB, Tam AB, Niwa M. Intrinsic capacities of molecular

doses of bortezomib in relapsed or refractory myeloma. British Jour- sensors of the unfolded protein response to sense alternate forms

nal of Haematology 2004;127:165–72. of endoplasmic reticulum stress. Molecular Biology of the Cell

[98] Millenium Pharmaceuticals, Cambridge, MA. Product information. 2006;17:3095–107.

Velcade (bortezomib); 2012. [119] Kim I, Xu W, Reed JC. Cell death and endoplasmic reticulum stress:

[99] Fitzgerald MFC, Webb I, Schenkein D, Esseltine D, Weich N. Nor- disease relevance and therapeutic opportunities. Nature Reviews Drug

mal hematopoietic stem cell function in mice following treatment Discovery 2008;7:1013–30.

with bortezomib. Biology of Blood and Marrow Transplantation [120] Li X, Zhang K, Li Z. Unfolded protein response in cancer: the physi-

2003;9:193. cian’s perspective. Journal of Hematology & Oncology 2011;4:8.

[100] Joazeiro CA, Anderson KC, Hunter T. Proteasome inhibitor drugs on [121] Ling SC, Lau EK, Al-Shabeeb A, et al. Response of myeloma to

the rise. Cancer Research 2006;66:7840–2. the proteasome inhibitor bortezomib is correlated with the unfolded

[101] Potts BC, Albitar MX, Anderson KC, et al. Marizomib, a protea- protein response regulator XBP-1. Haematologica 2012;97:64–72.

some inhibitor for all seasons: preclinical profile and a framework for [122] Obeng EA, Carlson LM, Gutman DM, Harrington Jr WJ, Lee KP,

clinical trials. Current Cancer Drug Targets 2011;11:254–84. Boise LH. Proteasome inhibitors induce a terminal unfolded protein

[102] Miller CP, Ban K, Dujka ME, et al. NPI-0052, a novel proteasome response in multiple myeloma cells. Blood 2006;107:4907–16.

inhibitor, induces caspase-8 and ROS-dependent apoptosis alone [123] Ma Y, Hendershot LM. The role of the unfolded protein response

and in combination with HDAC inhibitors in leukemia cells. Blood in tumour development: friend or foe? Nature Reviews Cancer

2007;110:267–77. 2004;4:966–77.

[103] Bichi R, Shinton SA, Martin ES, et al. Human chronic lympho- [124] Roue G, Perez-Galan P, Mozos A, et al. The Hsp90 inhibitor IPI-504

cytic leukemia modeled in mouse by targeted TCL1 expression. overcomes bortezomib resistance in mantle cell lymphoma in vitro

Proceedings of the National Academy of Sciences of the United States and in vivo by down-regulation of the prosurvival ER chaperone

of America 2002;99:6955–60. BiP/Grp78. Blood 2011;117:1270–9.

[104] Townsend A, Millward M, Price T, et al. Clinical trial of NPI-0052 in [125] Bogner C, Ringshausen I, Schneller F, et al. Inhibition of the protea-

advanced malignancies including lymphoma and leukemia (advanced some induces cell cycle arrest and apoptosis in mantle cell lymphoma

malignancies arm). Journal of Clinical Oncology 2009;27:3582. cells. British Journal of Haematology 2003;122:260–8.

[105] Cusack Jr JC, Liu R, Xia L, et al. NPI-0052 enhances tumoricidal [126] Lopes UG, Erhardt P, Yao R, Cooper GM. p53-dependent induction of

response to conventional cancer therapy in a colon cancer model. apoptosis by proteasome inhibitors. Journal of Biological Chemistry

Clinical Cancer Research 2006;12:6758–64. 1997;272:12893–6.

[106] Ruiz S, Krupnik Y, Keating M, Chandra J, Palladino M, McConkey [127] de Rozieres S, Maya R, Oren M, Lozano G. The loss of mdm2 induces

D. The proteasome inhibitor NPI-0052 is a more effective inducer p53-mediated apoptosis. Oncogene 2000;19:1691–7.

of apoptosis than bortezomib in lymphocytes from patients with [128] Sun Y. p53 and its downstream proteins as molecular targets of cancer.

chronic lymphocytic leukemia. Molecular Cancer Therapeutics Molecular Carcinogenesis 2006;45:409–15.

2006;5:1836–43. [129] MacPherson D, Kim J, Kim T, et al. Defective apoptosis and B-cell

[107] Slupianek A, Skorski T. NPM/ALK downregulates p27Kip1 in lymphomas in mice with p53 point mutation at Ser 23. EMBO Journal

a PI-3K-dependent manner. Experimental Hematology 2004;32: 2004;23:3689–99.

1265–71. [130] Yu D, Carroll M, Thomas-Tikhonenko A. p53 status dictates responses

[108] Nencioni A, Wille L, Dal Bello G, et al. Cooperative cytotox- of B lymphomas to monotherapy with proteasome inhibitors. Blood

icity of proteasome inhibitors and tumor necrosis factor-related 2007;109:4936–43.

apoptosis-inducing ligand in chemoresistant Bcl-2-overexpressing [131] Hengartner MO. The biochemistry of apoptosis. Nature

cells. Clinical Cancer Research 2005;11:4259–65. 2000;407:770–6.

[109] Jain S, Diefenbach C, Zain J, O’Connor OA. Emerging role of carfil- [132] Earnshaw WC, Martins LM, Kaufmann SH. Mammalian caspases:

zomib in treatment of relapsed and refractory lymphoid neoplasms structure, activation, substrates, and functions during apoptosis.

and multiple myeloma. Core Evidence 2011;6:43–57. Annual Review of Biochemistry 1999;68:383–424.

[110] Dasmahapatra G, Lembersky D, Kramer L, et al. The pan-HDAC [133] Cryns V, Yuan J. Proteases to die for. Genes and Development

inhibitor vorinostat potentiates the activity of the proteasome inhibitor 1998;12:1551–70.

carfilzomib in human DLBCL cells in vitro and in vivo. Blood [134] Porn-Ares MI, Saido TC, Andersson T, Ares MP. Oxidized

2010;115:4478–87. low-density lipoprotein induces calpain-dependent cell death and ubi-

[111] http://clinicaltrials.gov/ct2/show/NCT01276717 quitination of caspase 3 in HMEC-1 endothelial cells. Biochemical

[112] Paoluzzi L, Gonen M, Bhagat G, et al. The BH3-only mimetic ABT- Journal 2003;374:403–11.

737 synergizes the antineoplastic activity of proteasome inhibitors in [135] Walczak H, Miller RE, Ariail K, et al. Tumoricidal activity of tumor

lymphoid malignancies. Blood 2008;112:2906–16. necrosis factor-related apoptosis-inducing ligand in vivo. Nature

[113] Kline MP, Rajkumar SV, Timm MM, et al. ABT-737, an inhibitor Medicine 1999;5:157–63.

of Bcl-2 family proteins, is a potent inducer of apoptosis in multiple [136] LeBlanc HN, Ashkenazi A. Apo2L/TRAIL and its death and decoy

myeloma cells. Leukemia 2007;21:1549–60. receptors. Cell Death and Differentiation 2003;10:66–75.

322 K.S. Suh et al. / Critical Reviews in Oncology/Hematology 87 (2013) 306–322

[137] Lenz HJ. Clinical update: proteasome inhibitors in solid tumors. Can- with relapsed and relapsed/refractory multiple myeloma (MM). Blood

cer Treatment Reviews 2003;29(Suppl 1):41–8. 2009;114. Abstr 431.

[138] He Q, Huang Y, Sheikh MS. Proteasome inhibitor MG132 upregulates

death receptor 5 and cooperates with Apo2L/TRAIL to induce apopto-

sis in Bax-proficient and -deficient cells. Oncogene 2004;23:2554–8.

Biographies

[139] Zhang HG, Wang J, Yang X, Hsu HC, Mountz JD. Regula-

tion of apoptosis proteins in cancer cells by ubiquitin. Oncogene

2004;23:2009–15.

K. Stephen Suh, PhD, directs cancer research program

[140] Ding WX, Ni HM, Chen X, Yu J, Zhang L, Yin XM. A coordinated

action of Bax, PUMA, and p53 promotes MG132-induced mitochon- (Genomics and Biomarkers Program) conducting biomarker

dria activation and apoptosis in colon cancer cells. Molecular Cancer discoveries related to early detection, predicting clinical

Therapeutics 2007;6:1062–9.

outcome and patient selection for optimizing personalized

[141] Tang W, Li Y, Yu D, Thomas-Tikhonenko A, Spiegelman VS, Fuchs

medicine at the John Theurer Cancer Center of Hackensack

SY. Targeting beta-transducin repeat-containing protein E3 ubiquitin

University Medical Center in Hackensack, New Jersey. He

ligase augments the effects of antitumor drugs on breast cancer cells.

Cancer Research 2005;65:1904–8. is a molecular and cell biologist with expertise in cancer

[142] Ma MH, Yang HH, Parker K, et al. The proteasome inhibitor PS-341 signaling pathways and animal modeling.

markedly enhances sensitivity of multiple myeloma tumor cells to

chemotherapeutic agents. Clinical Cancer Research 2003;9:1136–44. Takemi Tanaka, PhD, is associate professor in the Depart-

[143] Fuchs SY, Spiegelman VS, Kumar KG. The many faces of beta- ment of Pharmaceutical Sciences of Thomas Jefferson

TrCP E3 ubiquitin ligases: reflections in the magic mirror of cancer.

University and specializes in nanoparticle-based drug deliv-

Oncogene 2004;23:2028–36.

ery, angiogenesis and metastasis of cancer by using molecular

[144] Fuchs SY. The role of ubiquitin-proteasome pathway in oncogenic

biological techniques and animal models.

signaling. Cancer Biology and Therapy 2002;1:337–41.

[145] Garg A, Aggarwal BB. Nuclear transcription factor-kappaB as a target

Sreeja Sarojini, PhD, is a postdoctoral fellow in the

for cancer drug development. Leukemia 2002;16:1053–68.

Biomarkers and Genomics Program at the John Theurer Can-

[146] Spiegelman VS, Tang W, Chan AM, et al. Induction of homologue

of Slimb ubiquitin ligase receptor by mitogen signaling. Journal of cer Center of Hackensack University Medical Center and

Biological Chemistry 2002;277:36624–30. investigates on topics related to biological response of radia-

[147] O’Connor OA. Targeting histones and proteasomes: new strate-

tion and chemotherapeutic drugs on cancer cells.

gies for the treatment of lymphoma. Journal of Clinical Oncology

2005;23:6429–36. Ginah Nightingale, PharmD, is an assistant professor in

[148] Harper JW, Adami GR, Wei N, Keyomarsi K, Elledge SJ. The p21

the Department of Clinical Pharmacy of Thomas Jefferson

Cdk-interacting protein Cip1 is a potent inhibitor of G1 cyclin-

University and specializes in toxicity and therapeutics against

dependent kinases. Cell 1993;75:805–16.

B-cell malignancies.

[149] Lim MS, Adamson A, Lin Z, et al. Expression of Skp2, a p27(Kip1)

ubiquitin ligase, in malignant lymphoma: correlation with p27(Kip1)

Rajendara Gharbaran, PhD, is a postdoctoral fellow in

and proliferation index. Blood 2002;100:2950–6.

the Biomarkers and Genomics Program in the John Theurer

[150] Tsujimoto Y, Gorham J, Cossman J, Jaffe E, Croce CM. The t(14;18)

chromosome translocations involved in B-cell neoplasms result from Cancer Center of Hackensack University Medical Center and

mistakes in VDJ joining. Science 1985;229:1390–3. investigates on topics related to Hodgkins lymphoma, drug

[151] Heckman CA, Mehew JW, Boxer LM. NF-kappaB activates Bcl-2

response and discovery of predictive biomarkers for clinical

expression in t(14;18) lymphoma cells. Oncogene 2002;21:3898–908. outcome.

[152] Rosenwald A, Wright G, Wiestner A, et al. The proliferation

gene expression signature is a quantitative integrator of oncogenic

Andrew Pecora, MD, is a vice president of cancer services

events that predicts survival in mantle cell lymphoma. Cancer Cell

and Innovation officer of the John Theurer Cancer Center of

2003;3:185–97.

the Hackensack University Medical Center. He is a certified

[153] Malaguarnera L, Cristaldi E, Malaguarnera M. The role of immu-

nity in elderly cancer. Critical Reviews in Oncology/Hematology hematologist/oncologist and specializes in stem cells, blood

2010;74:40–60. and marrow transplantation, cellular medicine and immunol-

[154] Kane RC, Dagher R, Farrell A, et al. Bortezomib for the treatment of ogy.

mantle cell lymphoma. Clinical Cancer Research: An Official Jour-

nal of the American Association for Cancer Research 2007;13(18 Pt Andre Goy, MD, is chairman and director of Clinical and

1):5291–4, http://dx.doi.org/10.1158/1078-0432.CCR-07-0871.

Translational Cancer Research in the John Theurer Cancer

[155] Furman RR, Martin P, Ruan J, et al. Phase 1 trial of borte-

Center of the Hackensack University Medical Center. He

zomib plus R-CHOP in previously untreated patients with

is the chief of lymphoma division and an internationally

aggressive non-Hodgkin lymphoma. Cancer 2010;116(23):5432–9,

http://dx.doi.org/10.1002/cncr.25509. renowned clinician and researcher on all types of lymphoma,

[156] Richardson PG, Hofmeister C, Jakubowiak A, et al. Phase 1 clinical and he specializes on developing novel targeted therapies for

trial of the novel structure proteasome inhibitor NPI-0052 in patients lymphoma.