With the approval by the U.S. Food and Drug Administration of bortezomib for the treatment of multiple myeloma and mantle cell lymphoma, the proteasome was clinically validated as a target in oncology. The proteasome is part of a complex cellular pathway that controls the specificity and rate of degradation of the majority of proteins in the cell. The search for additional drug targets in the proteasomal pathway is ongoing. In parallel, the next generation of proteasome inhibitors, exhibiting some properties distinct from that of bortezomib, are currently being studied in clinical trials. The key question will be whether these distinctions can improve upon the clinical efficacy and safety standards established by bortezomib and refine our understanding of the mechanism by which proteasome inhibitors are effective in the treatment of cancer. Clin Cancer Res; 18(1); 15–20. ©2011 AACR.

The ubiquitin–proteasome protein degradation pathway

Cells exert tight control over the synthesis and elimination of important proteins. One of the cell's most effective tools for the rapid elimination of proteins is the ubiquitin–proteasome pathway (Fig. 1; ref. 1). Proteins to be eliminated are marked by the covalent attachment of multiple copies of the protein ubiquitin. These polyubiquitin-tagged proteins then bind to the lid of the proteasome, where the tag is removed, and the protein is unfolded and threaded through the core of the proteasome complex. In the core, the proteins are digested by 3 threonine proteases to release small peptide fragments. These fragments are processed by intracellular peptidases to yield amino acids, which are then recycled into new proteins (2). Inhibition of proteasome function leads to the accumulation of polyubiquitin-tagged proteins and the withdrawal of the cell from the cell cycle, followed by the induction of apoptosis (programmed cell death) in susceptible cells (3–5). Although many potential therapeutic targets exist in the ubiquitin–proteasome pathway (6), the proteasome itself has been the only target to date for which successful drugs have been developed.

Figure 1.

Protein degradation by the ubiquitin–proteasome pathway. A chain of 4 or more ubiquitin molecules are attached by the action of a series of ubiquitin ligases (E1, E2, E3) to 1 or more lysine residues on the target protein to be degraded. The ubiquitin–protein complex is transported to the proteasome, where the ubiquitin chain is removed, allowing the target protein to be unfolded by an ATP-dependent process and translocated to the interior of the proteasome, where it is degraded by 3 threonine (Thr) proteases to yield peptide fragments.

Figure 1.

Protein degradation by the ubiquitin–proteasome pathway. A chain of 4 or more ubiquitin molecules are attached by the action of a series of ubiquitin ligases (E1, E2, E3) to 1 or more lysine residues on the target protein to be degraded. The ubiquitin–protein complex is transported to the proteasome, where the ubiquitin chain is removed, allowing the target protein to be unfolded by an ATP-dependent process and translocated to the interior of the proteasome, where it is degraded by 3 threonine (Thr) proteases to yield peptide fragments.

Close modal

Clinical validation of the proteasome as a therapeutic target

Cancer cells are more sensitive to proteasome inhibition and apoptotic induction than nontransformed cells for reasons that are not entirely understood (7, 8), and proteasome inhibitors have shown single-agent activity in several different animal tumor models.

Clinical validation of the proteasome as a cancer therapeutic target was established by bortezomib (Velcade; Millennium Pharmaceuticals/Takeda Pharmaceuticals; 9). Bortezomib, the first proteasome inhibitor to enter clinical trials, was granted accelerated approval by the U.S. Food and Drug Administration (FDA) in 2003 after showing impressive single-agent responses in patients with relapsed and refractory myeloma (10–13). Bortezomib has since been approved for the treatment of mantle cell lymphoma (MCL; refs. 14–17), as well as for the treatment of newly diagnosed multiple myeloma (18, 19). Although bortezomib is an effective treatment, proteasome inhibitors with reduced toxicity, improved efficacy, and oral bioavailability are still needed (20, 21). Inhibitors that have efficacy in other tumor types are also needed (22). These objectives have spawned the development of several “next-generation” proteasome inhibitors, of which 5 have entered clinical development since the approval of bortezomib.

Next-generation proteasome inhibitors in clinical development

The proteasome inhibitors currently in the clinic are derived from 3 structural classes: dipeptide boronic acids [represented by bortezomib (23), CEP-18770 (24), and MLN9708 (25)]; β-lactones (represented by NPI-0052, a marine microbial natural product related to omuralide; ref. 26); and peptide epoxyketones [represented by carfilzomib (PR-171; ref. 27), a tetrapeptide epoxyketone related to the natural product epoxomicin (28) and ONX-0912, a tripeptide analogue (29)].

Each inhibitor class reacts with the proteasome N-terminal threonine active sites by a distinct mechanism (30). Peptide boronic acids form a slowly reversible tetrahedral adduct with the γ-OH group of the catalytic threonine (23, 31). For the β-lactone NPI-0052, attack of the lactone ring by the catalytic γ-OH results in formation of an ester bond and an intramolecular rearrangement that makes the inhibition irreversible (32–34). For peptide epoxyketones, the peptide portion binds to the substrate-binding pocket of the proteasome, allowing the epoxyketone to interact stereospecifically with both the γ-OH and the α-amino groups of the catalytic threonine to form 2 covalent bonds, making the inhibition irreversible and selective (35).

Interestingly, proteasome activity recovers at the same rate with irreversible inhibitors as with slowly reversible inhibitors (27, 36), presumably because of induction of de novo proteasome synthesis (37).

The 3 threonine proteases are defined by their substrate selectivity: chymotrypsin-like–β5 subunit (CT-L); trypsin like–β2 subunit (T-L); and postacidic or caspase-like–β1 subunit (C-L). The proteasome inhibitors in clinical development have the greatest potency for the CT-L active site of the proteasome (24, 27, 36, 38, 39), but they differ in their activity against the other catalytic sites. The development of compounds with different selectivity profiles will help address the question of what combination of proteasome site inhibition provides maximal antitumor effects and the best therapeutic index.

Preclinical antitumor activity

NPI-0052.

NPI-0052 is active against a variety of tumor cell types (40, 41), including primary plasma cells from bortezomib-resistant patients with multiple myeloma. When NPI-0052 and bortezomib were compared head to head in xenograft studies using the twice-weekly (days 1 and 4) bortezomib clinical-dosing schedule, the activity of the 2 compounds was comparable in a multiple myeloma model (38, 42), whereas bortezomib displayed better efficacy than NPI-0052 in a prostate cancer model (36). In a disseminated lymphoma model, once-weekly bortezomib had superior efficacy to twice-weekly NPI-0052 (36, 40).

NPI-0052 is orally active, as are CEP-18770, MLN9708, and ONX-0912. However, unlike any of the other proteasome inhibitors (27, 36, 39, 43), NPI-0052 penetrates the blood–brain barrier and inhibits brain proteasome CT-L activity by >90% (36). Abnormalities in the ubiquitin–proteasome system have been linked to neurodegenerative diseases (44, 45), raising potential safety concerns around long-term treatment with NPI-0052.

CEP-18770.

CEP-18770 has in vitro antitumor activity similar to that of bortezomib in hematologic and solid tumor cell lines, as well as in primary plasma cells from patients with multiple myeloma. In addition, CEP-18770 had improved activity relative to bortezomib in a subcutaneous multiple myeloma xenograft model (RPMI-8226 cells) using a twice-weekly dosing schedule on days 1 and 4, and it had comparable activity in a disseminated multiple myeloma model using ARP-1 cells (39).

Carfilzomib.

Carfilzomib (PR-171) has in vitro activity across a range of tumor cell types, including multiple myeloma cells that are resistant to bortezomib (27, 46, 47). Carfilzomib is more active than bortezomib when cells are treated for short periods that mimic in vivo single-dose exposure; however, both compounds have greater activity when cells are exposed for prolonged periods of time (27, 46). To achieve prolonged inhibition in vivo, carfilzomib was dosed daily and was well tolerated at doses that inhibited more than 80% of proteasome CT-L activity in blood and tissues. This finding is in contrast to bortezomib, in which a twice-weekly clinical-dosing schedule on days 1 and 4, which allows full recovery of proteasome activity between doses, was selected, in part, because of excessive toxicity associated with more frequent dosing schemes (9). In xenograft studies, dosing of carfilzomib for 2 consecutive days (days 1 and 2) on a weekly schedule was superior to either twice-weekly dosing on days 1 and 4 or once-weekly dosing (27), supporting the in vitro observation that delaying complete proteasome recovery results in superior antitumor responses.

MLN9708.

The boronate MLN9708 has a similar profile to that of bortezomib in active site selectivity and potency in cytotoxicity assays, but it has a shorter proteasome dissociation half-life. In vivo, MLN9708 disseminates more widely to tissues than does bortezomib, resulting in greater blood and plasma exposures at the maximum tolerated dose and greater pharmacodynamic activity and efficacy in tumors. In contrast to bortezomib, MLN9708 has oral bioavailability, and it is efficacious and tolerated when dosed daily (25).

ONX-0912.

ONX-0912 (PR-047), an analogue of carfilzomib, was developed as an orally bioavailable peptide epoxyketone proteasome inhibitor (29). ONX-0912 has similar potency to carfilzomib in cytotoxicity assays and has equivalent antitumor activity to i.v.-administered carfilzomib in multiple human tumor xenograft and mouse syngeneic models. It is well tolerated with repeated daily oral administration at doses that result in >80% proteasome inhibition in most tissues (48).

Mechanism of action

Proteasome inhibitors block the global turnover of proteins, but there may be 1 or more key proteins with turnover that, when blocked, causes caspase activation and apoptosis in cancer cells. It has been suggested that blocking the prosurvival NF-κB pathway, by blocking the turnover of the inhibitory protein I-κB, is key for inducing apoptosis. It has also been shown that the proapoptotic factor NOXA, which interacts with the antiapoptotic factor Mcl-1 as well as other antiapoptotic factors in the Bcl-2 family and causes the release of cyctochrome c into the cytosol and activation of apoptosis, is induced in tumor cells upon treatment with proteasome inhibitors. In multiple myeloma, the tumor cells secrete great quantities of monoclonal antibody (M protein) and are dependent on the induction of the unfolded protein response (UPR) pathway to reduce the accumulation of misfolded proteins in the endoplasmic reticulum; otherwise the cells undergo apoptosis. Proteasome inhibition interferes with the UPR pathways and blocks the destruction of misfolded proteins by the proteasome (49, 50).

Clinical experience with proteasome inhibitors

Bortezomib approvals in multiple myeloma and mantle cell lymphoma.

Bortezomib has single-agent efficacy in multiple myeloma, resulting in 30% to 40% partial response rates in patients who have relapsed from prior therapies (a partial response is ≥50% decrease in M protein, the monoclonal antibody secreted by transformed plasma cells and present in blood and urine of patients with myeloma). This finding enabled rapid approval of bortezomib by the FDA in 2003 (11, 12). The combination of bortezomib and pegylated liposomal doxorubicin was shown to be superior to single-agent bortezomib (51), leading to the FDA approval of this combination in 2007. In newly diagnosed multiple myeloma, the addition of bortezomib to the standard therapy of melphalan and prednisone (MP) improved response rate (71% vs. 35%) and progression-free survival (24 months vs. 16.6 months), and it conferred an overall survival advantage relative to MP alone. On the basis of this study, bortezomib received FDA approval in 2008 in newly diagnosed patients (20, 21). In a phase I and II study in first-line patients with myeloma, bortezomib addition to lenalidomide and dexamethasone gave a 100% response rate, with 74% of responses a very good partial response or better (a very good partial response is ≥90% decrease in M protein), showing that the combination of these drugs is highly effective (52).

Bortezomib also has single-agent activity in relapsed MCL. A single-arm phase II study, in which the response rate was 31% and the duration of response was 9.3 months, was the basis for approval of bortezomib to treat MCL (15, 16).

Clinical pharmacodynamics and safety of bortezomib.

In patients treated with bortezomib, inhibition of CT-L activity in blood reaches a plateau at 65% to 70% (10), considerably less than the >90% proteasome inhibition seen in animal studies (43). The bortezomib clinical-dosing schedule (days 1, 4, 8, and 11 of a 21-day cycle) allows for complete recovery of proteasome activity between doses. The most notable toxicities observed with bortezomib include hematologic toxicities (thrombocytopenia and neutropenia), peripheral neuropathy, and gastrointestinal toxicities. Bortezomib-induced thrombocytopenia is transient in nature with recovery seen between dosing cycles (20); a block of platelet budding from megakaryocytes has been proposed as the underlying mechanism. Dose-dependent peripheral neuropathy (frequently painful) was reported in the initial phase II bortezomib trials, with an incidence of 37% at the recommended 1.3 mg/m2 dose (14% grade 3; refs. 9, 21, 53). This toxicity is now managed by dose reduction or withholding doses, but the impact of these modifications on the antimyeloma activity of bortezomib is not fully known.

Carfilzomib.

The most advanced of the next-generation proteasome inhibitors is carfilzomib, which entered the clinic in 2005. Single-agent responses were seen in 2 phase I studies with consecutive-day dosing schedules (54, 55) in which the drug was well tolerated and induced >80% proteasome inhibition in blood. On the basis of these data, a large phase II single-arm trial was conducted in patients with myeloma who had failed all available therapies, including bortezomib. Despite the fact that patients had a median of 5 prior lines of multidrug therapy, 24% of patients had a partial response and 37% of patients had a minimal response (≥25% decrease in M protein) or better to carfilzomib. In responding patients, the median overall survival was 20.7 months (56). Transient thrombocytopenia was observed (similar to that noted with bortezomib), but peripheral neuropathy rates were greatly reduced relative to bortezomib, suggesting that neuropathy may be an off-target side effect (57). An even higher response rate of 53% partial response was seen in patients with relapsed myeloma who had not received prior bortezomib. In newly diagnosed patients treated with a combination of carfilzomib, lenalidomide, and dexamethasone, the complete response rate (absence of detectable M protein and ≤5% myeloma plasma cells in bone marrow) was 61%, and 83% of patients had a very good partial response or better (58). Carfilzomib is currently in a randomized phase III registration trial in patients with relapsed myeloma, comparing carfilzomib with lenalidomide and dexamethose to lenalidomide and dexamethasone alone.

NPI-0052 and CEP-18770.

NPI-0052 entered the clinic with a phase I trial in 2006 in patients with solid tumors and in patients with lymphoma. Stable disease was observed in some patients, and proteasome inhibition in blood exceeded that observed with bortezomib (59). Additional phase I trials with NPI-0052 in myeloma and other malignancies are currently ongoing.

CEP-18770 entered the clinic in 2007 in a phase I study in patients with solid tumors and in patients with lymphoma to whom the drug was administered on the same schedule as bortezomib (days 1, 4, 8, and 11 of a 21-day cycle). Phase I and II trials in relapsed refractory myeloma, with CEP-18770 given alone or in combination with lenalidomide and dexamethasone, are currently ongoing.

MLN9708 and ONX-0912.

In a series of phase I trials initiated in 2009, MLN9708 had substantial oral bioavailability and was well tolerated when dosed on a once-weekly or twice-weekly schedule. In relapsed and refractory patients who had received prior bortezomib, interim data from a dose-escalation study reported at the International Myeloma Workshop in May 2011 showed that 2 out of 35 patients had partial responses. A phase I trial with ONX-0912 in patients with solid tumors started in 2010, and interim results reported at the American Society of Clinical Oncology in 2011 showed that, at doses that were well tolerated, >80% proteasome inhibition in blood could be achieved on a once-daily dosing schedule for 5 consecutive days (60).

The approvals of bortezomib for the treatment of multiple myeloma and MCL have provided clinical validation of the proteasome as a therapeutic target. The next generation of proteasome inhibitors exhibits both similarities and differences relative to bortezomib in terms of mechanism, selectivity, and preclinical antitumor activity. The results of clinical trials with these new agents will increase our understanding of the role of the proteasome in cancer cells and, it is hoped, expand the role that proteasome inhibitors will play in the treatment of cancer.

S.M. Molineaux: past employment, Onyx Pharmaceuticals (Proteolix); spouse was formerly a consultant for Onyx Pharmaceuticals.

1.
Ciechanover
A
. 
Intracellular protein degradation: from a vague idea thru the lysosome and the ubiquitin-proteasome system and onto human diseases and drug targeting
.
Cell Death Differ
2005
;
12
:
1178
90
.
2.
Bochtler
M
,
Ditzel
L
,
Groll
M
,
Hartmann
C
,
Huber
R
. 
The proteosome
.
Abstract Annu Rev Biophys Struct
1999
;
28
:
2985
317
.
3.
Driscoll
JJ
,
Dechowdhury
R
. 
Therapeutically targeting the SUMOylation, Ubiquitination and proteasome pathways as a novel anticancer strategy
.
Target Oncol
2010
;
5
:
281
9
.
4.
Nalepa
G
,
Rolfe
M
,
Harper
JW
. 
Drug discovery in the ubiquitin-proteasome system
.
Nat Rev Drug Discov
2006
;
5
:
596
613
.
5.
Sterz
J
,
von Metzler
I
,
Hahne
JC
,
Lamottke
B
,
Rademacher
J
,
Heider
U
, et al
The potential of proteasome inhibitors in cancer therapy
.
Expert Opin Investig Drugs
2008
;
17
:
879
95
.
6.
Sun
Y
. 
Targeting E3 ubiquitin ligases for cancer therapy
.
Cancer Biol Ther
2003
;
2
:
623
9
.
7.
Orlowski
RZ
,
Kuhn
DJ
. 
Proteasome inhibitors in cancer therapy: lessons from the first decade
.
Clin Cancer Res
2008
;
14
:
1649
57
.
8.
Adams
J
. 
The proteasome: a suitable antineoplastic target
.
Nat Rev Cancer
2004
;
4
:
349
60
.
9.
Bross
PF
,
Kane
R
,
Farrell
AT
,
Abraham
S
,
Benson
K
,
Brower
ME
, et al
Approval summary for bortezomib for injection in the treatment of multiple myeloma
.
Clin Cancer Res
2004
;
10
:
3954
64
.
10.
Orlowski
RZ
,
Stinchcombe
TE
,
Mitchell
BS
,
Shea
TC
,
Baldwin
AS
,
Stahl
S
, et al
Phase I trial of the proteasome inhibitor PS-341 in patients with refractory hematologic malignancies
.
J Clin Oncol
2002
;
20
:
4420
7
.
11.
Jagannath
S
,
Barlogie
B
,
Berenson
J
,
Siegel
D
,
Irwin
D
,
Richardson
PG
, et al
A phase 2 study of two doses of bortezomib in relapsed or refractory myeloma
.
Br J Haematol
2004
;
127
:
165
72
.
12.
Richardson
PG
,
Barlogie
B
,
Berenson
J
,
Singhal
S
,
Jagannath
S
,
Irwin
D
, et al
A phase 2 study of bortezomib in relapsed, refractory myeloma
.
N Engl J Med
2003
;
348
:
2609
17
.
13.
Richardson
PG
,
Sonneveld
P
,
Schuster
MW
,
Irwin
D
,
Stadtmauer
EA
,
Facon
T
, et al
Assessment of Proteasome Inhibition for Extending Remissions (APEX) Investigators
. 
Bortezomib or high-dose dexamethasone for relapsed multiple myeloma
.
N Engl J Med
2005
;
352
:
2487
98
.
14.
Belch
A
,
Kouroukis
CT
,
Crump
M
,
Sehn
L
,
Gascoyne
RD
,
Klasa
R
, et al
A phase II study of bortezomib in mantle cell lymphoma: the National Cancer Institute of Canada Clinical Trials Group trial IND.150
.
Ann Oncol
2007
;
18
:
116
21
.
15.
Fisher
RI
,
Bernstein
SH
,
Kahl
BS
,
Djulbegovic
B
,
Robertson
MJ
,
de Vos
S
, et al
Multicenter phase II study of bortezomib in patients with relapsed or refractory mantle cell lymphoma
.
J Clin Oncol
2006
;
24
:
4867
74
.
16.
Goy
A
,
Younes
A
,
McLaughlin
P
,
Pro
B
,
Romaguera
JE
,
Hagemeister
F
, et al
Phase II study of proteasome inhibitor bortezomib in relapsed or refractory B-cell non-Hodgkin's lymphoma
.
J Clin Oncol
2005
;
23
:
667
75
.
17.
O'Connor
OA
,
Wright
J
,
Moskowitz
C
,
Muzzy
J
,
MacGregor-Cortelli
B
,
Stubblefield
M
, et al
Phase II clinical experience with the novel proteasome inhibitor bortezomib in patients with indolent non-Hodgkin's lymphoma and mantle cell lymphoma
.
J Clin Oncol
2005
;
23
:
676
84
.
18.
Mateos
MV
,
Hernández
JM
,
Hernández
MT
,
Gutiérrez
NC
,
Palomera
L
,
Fuertes
M
, et al
Bortezomib plus melphalan and prednisone in elderly untreated patients with multiple myeloma: results of a multicenter phase 1/2 study
.
Blood
2006
;
108
:
2165
72
.
19.
San
Miguel JF
,
Schlag
R
,
Khuageva
N
,
Shpilberg
O
,
Dimopoulos
M
,
Kropff
M
, et al
MMY-3002: a phase 3 study comparing bortezomib-melphalan-prednisone (VMP) with melphalan-prednisone (MP) in newly diagnosed multiple myeloma
.
Blood
(
ASH Annual Meeting Abstracts
) 
2007
;
110
:
76
.
20.
Lonial
S
,
Waller
EK
,
Richardson
PG
,
Jagannath
S
,
Orlowski
RZ
,
Giver
CR
, et al
SUMMIT/CREST Investigators
. 
Risk factors and kinetics of thrombocytopenia associated with bortezomib for relapsed, refractory multiple myeloma
.
Blood
2005
;
106
:
3777
84
.
21.
Richardson
PG
,
Briemberg
H
,
Jagannath
S
,
Wen
PY
,
Barlogie
B
,
Berenson
J
, et al
Frequency, characteristics, and reversibility of peripheral neuropathy during treatment of advanced multiple myeloma with bortezomib
.
J Clin Oncol
2006
;
24
:
3113
20
.
22.
Milano
A
,
Iaffaioli
RV
,
Caponigro
F
. 
The proteasome: a worthwhile target for the treatment of solid tumours?
Eur J Cancer
2007
;
43
:
1125
33
.
23.
Adams
J
,
Behnke
M
,
Chen
S
,
Cruickshank
AA
,
Dick
LR
,
Grenier
L
, et al
Potent and selective inhibitors of the proteasome: dipeptidyl boronic acids
.
Bioorg Med Chem Lett
1998
;
8
:
333
8
.
24.
Dorsey
BD
,
Iqbal
M
,
Chatterjee
S
,
Menta
E
,
Bernardini
R
,
Bernareggi
A
, et al
Discovery of a potent, selective, and orally active proteasome inhibitor for the treatment of cancer
.
J Med Chem
2008
;
51
:
1068
72
.
25.
Kupperman
E
,
Lee
EC
,
Cao
Y
,
Bannerman
B
,
Fitzgerald
M
,
Berger
A
, et al
Evaluation of the proteasome inhibitor MLN9708 in preclinical models of human cancer
.
Cancer Res
2010
;
70
:
1970
80
.
26.
Feling
RH
,
Buchanan
GO
,
Mincer
TJ
,
Kauffman
CA
,
Jensen
PR
,
Fenical
W
. 
Salinosporamide A: a highly cytotoxic proteasome inhibitor from a novel microbial source, a marine bacterium of the new genus salinospora
.
Angew Chem Int Ed Engl
2003
;
42
:
355
7
.
27.
Demo
SD
,
Kirk
CJ
,
Aujay
MA
,
Buchholz
TJ
,
Dajee
M
,
Ho
MN
, et al
Antitumor activity of PR-171, a novel irreversible inhibitor of the proteasome
.
Cancer Res
2007
;
67
:
6383
91
.
28.
Hanada
M
,
Sugawara
K
,
Kaneta
K
,
Toda
S
,
Nishiyama
Y
,
Tomita
K
, et al
Epoxomicin, a new antitumor agent of microbial origin
.
J Antibiot(Tokyo)
1992
;
45
:
1746
52
.
29.
Zhou
HJ
,
Aujay
MA
,
Bennett
MK
,
Dajee
M
,
Demo
SD
,
Fang
Y
, et al
Design and synthesis of an orally bioavailable and selective peptide epoxyketone proteasome inhibitor (PR-047)
.
J Med Chem
2009
;
52
:
3028
38
.
30.
Borissenko
L
,
Groll
M
. 
20S proteasome and its inhibitors: crystallographic knowledge for drug development
.
Chem Rev
2007
;
107
:
687
717
.
31.
Groll
M
,
Berkers
CR
,
Ploegh
HL
,
Ovaa
H
. 
Crystal structure of the boronic acid-based proteasome inhibitor bortezomib in complex with the yeast 20S proteasome
.
Structure
2006
;
14
:
451
6
.
32.
Groll
M
,
Potts
BC
. 
Proteasome structure, function, and lessons learned from beta-lactone inhibitors
.
Curr Top Med Chem
2011 Aug 9
.
33.
Groll
M
,
Huber
R
,
Potts
BC
. 
Crystal structures of Salinosporamide A (NPI-0052) and B (NPI-0047) in complex with the 20S proteasome reveal important consequences of beta-lactone ring opening and a mechanism for irreversible binding
.
J Am Chem Soc
2006
;
128
:
5136
41
.
34.
Dick
LR
,
Cruikshank
AA
,
Grenier
L
,
Melandri
FD
,
Nunes
SL
,
Stein
RL
. 
Mechanistic studies on the inactivation of the proteasome by lactacystin: a central role for clasto-lactacystin beta-lactone
.
J Biol Chem
1996
;
271
:
7273
6
.
35.
Groll
M
,
Kim
KB
,
Kairies
N
,
Huber
R
,
Crews
CM
. 
Crystal structure of epoxomicin:20S proteasome reveals a molecular basis of alpha',beta'-epoxyketone proteasome inhibitors
.
J Am Chem Soc
2000
;
122
:
1237
8
.
36.
Williamson
MJ
,
Blank
JL
,
Bruzzese
FJ
,
Cao
Y
,
Daniels
JS
,
Dick
LR
, et al
Comparison of biochemical and biological effects of ML858 (salinosporamide A) and bortezomib
.
Mol Cancer Ther
2006
;
5
:
3052
61
.
37.
Meiners
S
,
Heyken
D
,
Weller
A
,
Ludwig
A
,
Stangl
K
,
Kloetzel
PM
, et al
Inhibition of proteasome activity induces concerted expression of proteasome genes and de novo formation of Mammalian proteasomes
.
J Biol Chem
2003
;
278
:
21517
25
.
38.
Chauhan
D
,
Catley
L
,
Li
G
,
Podar
K
,
Hideshima
T
,
Velankar
M
, et al
A novel orally active proteasome inhibitor induces apoptosis in multiple myeloma cells with mechanisms distinct from Bortezomib
.
Cancer Cell
2005
;
8
:
407
19
.
39.
Piva
R
,
Ruggeri
B
,
Williams
M
,
Costa
G
,
Tamagno
I
,
Ferrero
D
, et al
CEP-18770: A novel, orally active proteasome inhibitor with a tumor-selective pharmacologic profile competitive with bortezomib
.
Blood
2008
;
111
:
2765
75
.
40.
Potts
CB
,
Albitar
XM
,
Anderson
CK
,
Baritaki
S
,
Berkers
C
,
Bonavida
B
, et al
Marizomib, a proteasome inhibitor for all seasons: preclinical profile and a framework for clinical trials
.
Curr Cancer Drug Targets
2011
;
11
:
254
84
.
41.
Ruiz
S
,
Krupnik
Y
,
Keating
M
,
Chandra
J
,
Palladino
M
,
McConkey
D
. 
The proteasome inhibitor NPI-0052 is a more effective inducer of apoptosis than bortezomib in lymphocytes from patients with chronic lymphocytic leukemia
.
Mol Cancer Ther
2006
;
5
:
1836
43
.
42.
Singh
AV
,
Palladino
MA
,
Lloyd
GK
,
Potts
BC
,
Chauhan
D
,
Anderson
KC
. 
Pharmacodynamic and efficacy studies of the novel proteasome inhibitor NPI-0052 (marizomib) in a human plasmacytoma xenograft murine model
.
Br J Haematol
2010
;
149
:
550
9
.
43.
Adams
J
,
Palombella
VJ
,
Sausville
EA
,
Johnson
J
,
Destree
A
,
Lazarus
DD
, et al
Proteasome inhibitors: a novel class of potent and effective antitumor agents
.
Cancer Res
1999
;
59
:
2615
22
.
44.
Ardley
HC
,
Hung
CC
,
Robinson
PA
. 
The aggravating role of the ubiquitin-proteasome system in neurodegeneration
.
FEBS Lett
2005
;
579
:
571
6
.
45.
Ross
CA
,
Pickart
CM
. 
The ubiquitin-proteasome pathway in Parkinson's disease and other neurodegenerative diseases
.
Trends Cell Biol
2004
;
14
:
703
11
.
46.
Kuhn
DJ
,
Chen
Q
,
Voorhees
PM
,
Strader
JS
,
Shenk
KD
,
Sun
CM
, et al
Potent activity of carfilzomib, a novel, irreversible inhibitor of the ubiquitin-proteasome pathway, against preclinical models of multiple myeloma
.
Blood
2007
;
110
:
3281
90
.
47.
Stapnes
C
,
Døskeland
AP
,
Hatfield
K
,
Ersvaer
E
,
Ryningen
A
,
Lorens
JB
, et al
The proteasome inhibitors bortezomib and PR-171 have antiproliferative and proapoptotic effects on primary human acute myeloid leukaemia cells
.
Br J Haematol
2007
;
136
:
814
28
.
48.
Chauhan
D
,
Singh
AV
,
Aujay
M
,
Kirk
CJ
,
Bandi
M
,
Ciccarelli
B
, et al
A novel orally active proteasome inhibitor ONX 0912 triggers in vitro and in vivo cytotoxicity in multiple myeloma
.
Blood
2010
;
116
:
4906
15
.
49.
Chen
D
,
Frezza
M
,
Schmitt
S
,
Kanwar
J
,
P Dou
Q
. 
Bortezomib as the first proteasome inhibitor anticancer drug: current status and future perspectives
.
Curr Cancer Drug Targets
2011
;
11
:
239
53
.
50.
Chari
A
,
Mazumder
A
,
Jagannath
S
. 
Proteasome inhibition and its therapeutic potential in multiple myeloma
.
Biologics
2010
;
4
:
273
87
.
51.
Orlowski
RZ
,
Nagler
A
,
Sonneveld
P
,
Bladé
J
,
Hajek
R
,
Spencer
A
, et al
Randomized phase III study of pegylated liposomal doxorubicin plus bortezomib compared with bortezomib alone in relapsed or refractory multiple myeloma: combination therapy improves time to progression
.
J Clin Oncol
2007
;
25
:
3892
901
.
52.
Richardson
PG
,
Weller
E
,
Lonial
S
,
Jakubowiak
J
,
Jagannath
S
,
Raje
NS
, et al
Lenalidomide, bortezomib, and dexamethasone combination therapy in patients with newly diagnosed multiple myeloma
.
Blood
2010
;
116
:
679
86
.
53.
Cavaletti
G
,
Jakubowiak
AJ
. 
Peripheral neuropathy during bortezomib treatment of multiple myeloma: a review of recent studies
.
Leuk Lymphoma
2010
;
51
:
1178
87
.
54.
Alsina
M
,
Trudel
S
,
Vallone
M
,
Molineaux
C
,
Kunkel
L
,
Goy
A
. 
Phase 1 single agent antitumor activity of twice weekly consecutive day dosing of the proteasome inhibitor carfilzomib (PR-171) in hematologic malignancies
.
Blood
(
ASH Annual Meeting Abstracts
) 
2007
;
110
:
411
.
55.
Orlowski
RZ
,
Stewart
K
,
Vallone
M
,
Molineaux
C
,
Kunkel
L
,
Gericitano
J
, et al
Safety and antitumor efficacy of the proteasome inhibitor carfilzomib (PR-171) dosed for five consecutive days in hematologic malignancies: phase 1 results
.
Blood
(
ASH Annual Meeting Abstracts
) 
2007
;
110
:
409
.
56.
Siegel
DSD
,
Martin
T
,
Wang
M
,
Vij
R
,
Jakubowiak
AJ
,
Jagannath
S
, et al
Results of PX-171-003-A1, an open-label, single-arm, phase 2 (ph 2) study of carfilzomib (CFZ) in patients (pts) with relapsed and refractory multiple myeloma (MM)
.
Blood
(
ASH Annual Meeting Abstracts
) 
2010
;
116
:
985
.
57.
Vij
R
,
Kaufman
JL
,
Jakubowiak
AJ
,
Stewart
AK
,
Jagannath
S
,
Kukreti
V
, et al
carfilzomib: high single agent response rate with minimal neuropathy even in high-risk patients
.
Blood
2010
;
116
:
1938
.
58.
Jakubowiak
AJ
,
Dytfeld
D
,
Jagannath
S
,
Vesole
DH
,
Anderson
TB
,
Nordgren
BK
, et al
Carfilzomib, lenalidomide, and dexamethasone in newly diagnosed multiple myeloma: initial results of phase I/II MMRC trial
.
Blood
2010
;
116
:
862
.
59.
Aghajanian
CA
,
Hamlin
P
,
Gordon
DS
,
Hong
M
,
Naing
A
,
Younes
A
, et al
Phase I study of the novel proteasome inhibitor NPI-0052 in patients with lymphoma and solid tumors
.
J Clin Oncol
26
, 
2008
(
suppl; abstr 3574
).
60.
Papadopoulos
KP
,
Mendelson
DS
,
Tolcher
AW
,
Patnaik
A
,
Burris
HA
,
Rasco
DW
, et al
A phase I, open-label, dose-escalation study of the novel oral proteasome inhibitor (PI) ONX 0912 in patients with advanced refractory or recurrent solid tumors
.
J Clin Oncol
29
, 
2011
(
suppl; abstr 3075
).