This spotlight review focuses on the second-generation proteasome inhibitor carfilzomib, which was recently approved by the US Food and Drug Administration for treatment of relapsed and refractory multiple myeloma patients who have received at least 2 prior therapies, including bortezomib and an immunomodulatory agent, and have demonstrated disease progression on or within 60 days of the completion of the last therapy. This review focuses on clinical trial data leading to drug approval and provides advice for treating physicians who are now accessing this drug for patients.

Proteasome inhibition is highly effective treatment for multiple myeloma (MM), Waldenström macroglobulinemia, low-grade non-Hodgkin lymphomas, and primary amyloidosis.1  Almost 10 years ago, the US Food and Drug Administration (FDA) approved the dipeptidyl boronic acid derivative proteasome inhibitor bortezomib (BTZ) for the treatment of refractory MM and subsequently frontline therapy for MM, thus opening the door to a new era of improved MM therapy. Indeed, disease-free and overall survival for most MM patients has been significantly extended. Nevertheless primary or secondary BTZ resistance2  is common, and treatment is often limited by BTZ-induced, dose-limiting side effects, mostly consequent to peripheral neuropathy.3  Although less frequent BTZ administration at lower doses and using subcutaneous delivery4,5  may contribute to a lowered BTZ neuropathy incidence and severity without seemingly compromising efficacy, new proteasome inhibitors—the “second generation”—have now been developed and are aimed at being potentially more efficacious and less toxic. The FDA recently granted accelerated approval for carfilzomib (CFZ) injection for the treatment of patients with MM who have received at least 2 prior therapies, including BTZ and an immunomodulatory agent, and who have demonstrated disease progression on or within 60 days of the completion of last therapy. This spotlight review focuses on the data leading to drug approval and provides helpful management tips for treating physicians.

Ubiquitin-proteasome pathway inhibition in MM treatment

Proteasomes are present in all eukaryotic cells.6,7  They degrade proteins8,9  and influence a multitude of cellular processes,10-16  including proliferation and DNA repair.17-19  Proteasome inhibition leads to an unfolded protein stress response by accumulation of misfolded proteins in the cell,20-22  inhibits NF-κB,23  and thus induces cell-cycle arrest24,25  and apoptosis.26-28  The (malignant) plasma cell in particular is susceptible to proteasome inhibition, even to small changes,29  because of its inherent function in Ab production.22,30-32 

The proteasome

The constitutive 26S proteasome consists of protein-recognizing 19S regulatory particles and a 20S proteolytic core33  that carries 3 protein-specific29  catalytic sites: the chymotrypsin, trypsin, and caspase-like sites.34,35  Blocking the chymotrypsin-like site is most effective in cellular growth inhibition in vitro, but coinhibition of other proteasome subunits further increases overall growth retardation.35-38  Unique immunoproteasomes exist in cells of immune or hematopoietic origin, where the catalytic sites differ from the constitutive proteasomes.36  These play an important role in generating antigens for MHC class I presentation.39  Both constitutive and immunoproteasomes are expressed in MM cells and are targeted by the available inhibitors BTZ and CFZ. It has not been fully elucidated whether the anticancer effects depend on inhibition of one or both proteasome types because study results are ambiguous.40-43 

CFZ-specific inhibition of the proteasome

CFZ (also known as PR-171) is a cell-permeable tetrapeptide epoxyketone analog of epoxomicin.44,45  It primarily inhibits the chymotrypsin-like site of the proteasome (Figure 1); in high doses, it shows additional inhibitory effects on the trypsin-like and caspase-like sites.46  CFZ forms stable and irreversible adducts exclusively with the proteasome but not with other proteases.45-48  BTZ, in contrast, forms (slowly) reversible and less specific adducts predominantly with the chymotrypsin-like and the caspase-like site, but also with a multitude of serine proteases,49-51  potentially contributing to some of the neurotoxicity.

Figure 1

CFZ-induced proteasome inhibition.

Figure 1

CFZ-induced proteasome inhibition.

Close modal

CFZ pharmacodynamics and pharmacokinetics

CFZ penetrates all tissues but the brain extensively. It is largely metabolized extrahepatically and is rapidly cleared from the circulation by biliary and renal excretion (t1/2 = 15-30 minutes): less than 1% is excreted intact.46,52-54  Unlike BTZ, CFZ is not primarily metabolized by hepatic cytochrome P450,55,56  and therefore plasma levels are minimally dependent on liver function and concomitant medication.57  Because of irreversible binding of CFZ, proteasome function after therapy can only be regained by de novo proteasome synthesis.52  Consecutive daily dosing of CFZ is optimal for inhibition of the proteasome in preclinical studies, thus leading to the eventual clinical schedule.46 

CFZ resistance

A significant number of MM patients present with either primary or secondary proteasome inhibitor resistance. Interestingly, some BTZ-resistant cell lines41  and patients58  are responsive to CFZ therapy and some CFZ-resistant cell lines remain sensitive to BTZ treatment.59,60  Differences in underlying resistance mechanisms have not been convincingly elucidated.2,61-67  Recently, up-regulation of P-glycoprotein could be demonstrated after CFZ, suggesting that drug transport may contribute to resistance.68,69 

CFZ phase 1 clinical trials

In the first phase 1 trial, CFZ was administered in escalating doses on 5 consecutive days, followed by a 9-day rest period in a 14-day cycle. Sustained proteasome inhibition was achieved; common side effects were low-grade fatigue and nausea, but no significant peripheral neuropathy was observed. Two CFZ-associated dose-limiting toxicities (DLT) occurred (thrombocytopenia and febrile neutropenia). The minimal effective dose (MED) was determined to be 11 mg/m2 and 15 mg/m2 the maximal tolerated dose (MTD).52 

In a second phase 1 trial, CFZ was given on a 4-week cycle, on days 1, 2, 8, 9, 15, and 16 to 37 patients with hematologic malignancies. Five responses were seen in 26 MM patients at doses above 15 mg/m2 and an MTD was not reached. The highest dose administered was 27 mg/m2. Of all of the patients included, 3 had serious adverse events (SAEs) during phase one (sepsis, elevated liver enzymes, and chemical pancreatitis), all considered possibly related to CFZ. Five patients in the expansion cohort had 9 SAEs, of which 1 was considered to be CFZ related (hypoxia); 1 patient with a preexisting grade 2 renal impairment developed grade 3 renal failure. Although 50% of the patients entered the study with preexisting peripheral neuropathy, no aggravation to grade 3/4 was reported.54 

CFZ phase 2 clinical trials

PX 171-003 was the primary trial used for registration purposes and addressed CFZ single-agent activity in a cohort of particularly poor prognosis and unmet medical need patients. Therefore, 266 heavily pretreated MM patients (82% had > 4 lines of therapy and 80% were double refractory) were treated with single-agent CFZ in the twice-weekly regimen (see previous paragraph) with a dose-reduced (20 mg/m2) first cycle to abrogate potential tumor-lysis syndrome. Dosing in cycle 2 was escalated to 27 mg/m2. The overall response rate (ORR) was 23.7%, with a median duration of response of 7.8 months, a progression-free survival of 3.7 months, and an overall survival of 15.6 months. Clinical benefit was seen in one-third of the patients. Outcome was not influenced by adverse cytogenetics, renal impairment, disease stage, or Eastern Cooperative Oncology Group (ECOG) performance score. Five of the 24 deaths in the 266 patients on trial were considered possibly related to CFZ treatment, including 2 cardiac arrests. Drug-related AEs of all grades were most frequently fatigue (37%), nausea (34%), and thrombocythemia. Hematologic AE grade 3/4 included anemia (24%), thrombocytopenia (29%), lymphopenia (20%), and neutropenia (11%). Nonhematologic AEs grade 3/4 included pneumonia (9%), hyponatremia (8.3%), fatigue (7.5%), and hypophosphatemia (6.0%). One-third of the patients experienced mild to moderate dyspnea without detectable lung injury, possibly because of extensive hydration patients received. Although 77% of patients had grade 1/2 peripheral neuropathy at study inclusion, treatment-emergent peripheral neuropathy was uncommon (12.4%) and considered to be CFZ related in only 8.3% of patients. Acute renal failure qualifying for an severe AE occurred in 5 of 266 patients and was considered likely to be related to CFZ.70 

Other supportive clinical trials for registration included a phase 2 trial in 129 relapsed but BTZ-naive MM patients. This trial showed an impressive single-agent ORR of 52%.71 Table 1 lists trials including ORR data and Table 2 lists trials including survival data.

The impact of renal insufficiency in CFZ treatment was investigated in a phase 2 trial in 39 relapsed MM patients with varying renal impairment. Toxicities were manageable and independent of renal status; dose adjustment was not necessary.72,73 

In the PX-171-010 trial, the long-term safety of CFZ treatment was assessed. No cumulative toxicities, including late development of peripheral neuropathy or significant emerging renal dysfunction, were observed.74 

The extension of the infusion time from 2-10 minutes to 30 minutes allowed tolerance of higher doses of CFZ. DLTs were seen at a dose of 70 mg/m2 and at 56 mg/m2, an impressive single-agent ORR of 60% was achieved. Grade 3/4 toxicities were thrombocytopenia (38%), anemia (21%), and hypertension (13%).75 

CFZ clinical trials combination therapy

A phase 1 trial (PX-171-006) in relapsed MM patients examined CFZ with standard-dose lenalidomide and low-dose dexamethasone in a 28-day cycle (no DLTs were observed and the ORR was 78%).76  A phase 1/2 trial of this combination was then performed in newly diagnosed patients. CFZ was dose escalated in this study and a 36 mg/m2 dose was found to be tolerable. After a median of 12 cycles of therapy, the ORR was 98%, with an impressive stringent complete remission rate of 42%.77 

CFZ plus melphalan and prednisone (CMP) for elderly patients with newly diagnosed MM was administered on the usual schedule twice weekly with a 12-day rest in a 42-day cycle. The MTD was 36 mg/m2 and 2 DLTs (fever and hypotension) occurred. The regimen was judged to be safe and effective. An ORR of 92% could be determined in an interim analysis.78 

Other reported alkylator combination regimens include CYCLONE (cyclophosphamide, CFZ, thalidomide, and dexamethasone) for newly diagnosed MM patients, which, again, demonstrated efficacy, with an ORR of 100% and deep responses in many patients.79 

CFZ practical guidance

The FDA-approved label dose and the dose being tested in phase 3 clinical trials is CFZ 20 mg/m2 in the first 28-day cycle and, if tolerated in cycle 1, escalating to 27 mg/m2 for cycle 2 and beyond. The current vial size of 60 mg is well suited to the 27 mg/m2 dose. Higher doses up to at least 45 mg/m2 were tolerable in limited phase 1/2 testing, but data on these doses are still accumulating and this treatment is not currently used outside of clinical trials.

CFZ is administered intravenously over 2-10 minutes on consecutive days each week for 3 weeks (days 1, 2, 8, 9, 15, and 16), followed by a 12-day rest period (days 17-28). If signs of dose intolerance occur, a dose reduction down to 20 mg/m2 or an extension of infusion time up to 30 minutes can be considered.

The staged dose escalation between cycles 1 and 2 reflects concerns from phase 1 testing about possible tumor lysis, early infusion reaction with fever and dyspnea, and increases in creatinine in the presence of high tumor burden and dehydration. Therefore, current advice is to maintain adequate fluid volume status throughout treatment and to monitor blood chemistries closely, particularly in cycle 1. In our practice, this currently means obtaining laboratory reports on days 1, 2, 3, 8, and 15 in cycle 1 and on days 1, 8, and 15 thereafter. Before each dose in cycle 1, it is currently recommended—and is our practice—to give 250 mL of IV fluid before each dose if the patient can tolerate such fluid challenge. In cycle 2 and beyond, oral hydration is likely adequate. After hydration and routine allopurinol, tumor lysis on clinical trials was relatively uncommon. However, concerns have been voiced that the high incidence of dyspnea and, less frequently, cardiac events observed may in part reflect fluid overload rather than drug toxicity. This has led to a relaxation of hydration guidelines in ongoing clinical trials so that aggressive oral hydration for 24 hours before the first dose rather than IV fluids and CFZ dose escalation beginning on day 8 instead of cycle 2 are being used. Data from these studies are not yet available. Even in these studies, IV hydration is recommended for patients considered at high risk for tumor lysis.

Infusion reactions characterized by a spectrum of systemic symptoms were observed in early clinical trials. These were most evident with the first few doses, so the advice is to premedicate in cycle 1 with dexamethasone 4 mg before all doses of CFZ. Beyond cycle 1, dexamethasone prophylaxis can also be stopped if the patient tolerates the drug.

As with BTZ treatment, shingles or hepatitis reactivation has been reported so prophylaxis is recommended.

In conclusion, although the use of tumor-lysis prophylaxis, premedication with dexamethasone, and dose escalation in cycle 2 may seem cumbersome, many of these preventative measures are being relaxed in ongoing trials and as results emerge, more convenient supportive care regimens may be recommended. Even now, we would advise caution with overly aggressive hydration in elderly, cardiac-,compromised, or hypertensive patients. Although the consecutive daily dosing schedule may be less convenient, it is important to note that patient compliance has generally been high in our experience and a consensus opinion of physician clinical trial participants is that CFZ is a well tolerated drug in the majority of patients and offers the unique benefit of minimal neuropathy.

Summary

CFZ is a potent proteasome inhibitor and effective therapy in MM with an advantageous side effect profile characterized by low rates of peripheral neuropathy and potential use in other diseases as Waldenström macroglobulinemia, lymphoma, amyloidosis, and autoimmune diseases. In MM, CFZ is a welcome addition to BTZ, alkylators, corticosteroids, and the immunomodulatory drugs thalidomide and lenalidomide in the therapeutic arsenal. The results of ongoing phase 2 and multiple phase 3 trials will help to further define the role of CFZ in MM therapy and will help to establish the best dosing, schedule, and supportive care management that benefit these patients.

K.M.K. is supported by a research grant (KO 4604/1-1) from the Deutsche Forschungsgemeinschaft (Germany). Onyx Pharmaceuticals and Millennium Pharmaceuticals provided research support for the clinical trials.

Contribution: K.M.K. and A.K.S. analyzed the data and wrote the manuscript.

Conflict-of-interest disclosure: K.M.K. declares no competing financial interests. A.K.S. is a consultant for Onyx Pharmaceuticals (funds to institution), Millennium Pharmaceuticals, Janssen, and Celgene.

Correspondence: A. Keith Stewart, Mayo Clinic Collaborative Research Bldg, 13400 E Shea Blvd, Scottsdale, AZ 85259-5494; e-mail: stewart.keith@mayo.edu.

1
Rajkumar
 
SV
Richardson
 
PG
Hideshima
 
T
Anderson
 
KC
Proteasome inhibition as a novel therapeutic target in human cancer.
J Clin Oncol
2005
, vol. 
23
 
3
(pg. 
630
-
639
)
2
Richardson
 
PG
Barlogie
 
B
Berenson
 
J
, et al. 
A phase 2 study of bortezomib in relapsed, refractory myeloma.
N Engl J Med
2003
, vol. 
348
 
26
(pg. 
2609
-
2617
)
3
Richardson
 
PG
Briemberg
 
H
Jagannath
 
S
, et al. 
Frequency, characteristics, and reversibility of peripheral neuropathy during treatment of advanced multiple myeloma with bortezomib.
J Clin Oncol
2006
, vol. 
24
 
19
(pg. 
3113
-
3120
)
4
Moreau
 
P
Pylypenko
 
H
Grosicki
 
S
, et al. 
Subcutaneous versus intravenous administration of bortezomib in patients with relapsed multiple myeloma: a randomised, phase 3, non-inferiority study.
Lancet Oncol
2011
, vol. 
12
 
5
(pg. 
431
-
440
)
5
Bringhen
 
S
Larocca
 
A
Rossi
 
D
, et al. 
Efficacy and safety of once-weekly bortezomib in multiple myeloma patients.
Blood
2010
, vol. 
116
 
23
(pg. 
4745
-
4753
)
6
Coux
 
O
Tanaka
 
K
Goldberg
 
AL
Structure and functions of the 20S and 26S proteasomes.
Annu Rev Biochem
1996
, vol. 
65
 (pg. 
801
-
847
)
7
Hershko
 
A
Ciechanover
 
A
The ubiquitin system.
Annu Rev Biochem
1998
, vol. 
67
 (pg. 
425
-
479
)
8
Peters
 
JM
Proteasomes: protein degradation machines of the cell.
Trends Biochem Sci
1994
, vol. 
19
 
9
(pg. 
377
-
382
)
9
Ciechanover
 
A
Schwartz
 
AL
The ubiquitin-proteasome pathway: the complexity and myriad functions of proteins death.
Proc Natl Acad Sci U S A
1998
, vol. 
95
 
6
(pg. 
2727
-
2730
)
10
Rock
 
KL
Gramm
 
C
Rothstein
 
L
, et al. 
Inhibitors of the proteasome block the degradation of most cell proteins and the generation of peptides presented on MHC class I molecules.
Cell
1994
, vol. 
78
 
5
(pg. 
761
-
771
)
11
Craiu
 
A
Akopian
 
T
Goldberg
 
A
Rock
 
KL
Two distinct proteolytic processes in the generation of a major histocompatibility complex class I-presented peptide.
Proc Natl Acad Sci U S A
1997
, vol. 
94
 
20
(pg. 
10850
-
10855
)
12
King
 
RW
Deshaies
 
RJ
Peters
 
JM
Kirschner
 
MW
How proteolysis drives the cell cycle.
Science
1996
, vol. 
274
 
5293
(pg. 
1652
-
1659
)
13
Terrell
 
J
Shih
 
S
Dunn
 
R
Hicke
 
L
A function for monoubiquitination in the internalization of a G protein-coupled receptor.
Mol Cell
1998
, vol. 
1
 
2
(pg. 
193
-
202
)
14
Polo
 
S
Sigismund
 
S
Faretta
 
M
, et al. 
A single motif responsible for ubiquitin recognition and monoubiquitination in endocytic proteins.
Nature
2002
, vol. 
416
 
6879
(pg. 
451
-
455
)
15
Zhou
 
W
Zhu
 
P
Wang
 
J
, et al. 
Histone H2A monoubiquitination represses transcription by inhibiting RNA polymerase II transcriptional elongation.
Mol Cell
2008
, vol. 
29
 
1
(pg. 
69
-
80
)
16
Garrett
 
IR
Chen
 
D
Gutierrez
 
G
, et al. 
Selective inhibitors of the osteoblast proteasome stimulate bone formation in vivo and in vitro.
J Clin Invest
2003
, vol. 
111
 
11
(pg. 
1771
-
1782
)
17
Nijman
 
SM
Huang
 
TT
Dirac
 
AM
, et al. 
The deubiquitinating enzyme USP1 regulates the Fanconi anemia pathway.
Mol Cell
2005
, vol. 
17
 
3
(pg. 
331
-
339
)
18
Huang
 
TT
D'Andrea
 
AD
Regulation of DNA repair by ubiquitylation.
Nat Rev Mol Cell Biol
2006
, vol. 
7
 
5
(pg. 
323
-
334
)
19
Huang
 
TT
Nijman
 
SM
Mirchandani
 
KD
, et al. 
Regulation of monoubiquitinated PCNA by DUB autocleavage.
Nat Cell Biol
2006
, vol. 
8
 
4
(pg. 
339
-
347
)
20
Patil
 
C
Walter
 
P
Intracellular signaling from the endoplasmic reticulum to the nucleus: the unfolded protein response in yeast and mammals.
Curr Opin Cell Biol
2001
, vol. 
13
 
3
(pg. 
349
-
355
)
21
Kaufman
 
RJ
Stress signaling from the lumen of the endoplasmic reticulum: coordination of gene transcriptional and translational controls.
Genes Dev
1999
, vol. 
13
 
10
(pg. 
1211
-
1233
)
22
Obeng
 
EA
Carlson
 
LM
Gutman
 
DM
Harrington
 
WJ
Lee
 
KP
Boise
 
LH
Proteasome inhibitors induce a terminal unfolded protein response in multiple myeloma cells.
Blood
2006
, vol. 
107
 
12
(pg. 
4907
-
4916
)
23
Hideshima
 
T
Chauhan
 
D
Richardson
 
P
, et al. 
NF-kappa B as a therapeutic target in multiple myeloma.
J Biol Chem
2002
, vol. 
277
 
19
(pg. 
16639
-
16647
)
24
Brewer
 
JW
Diehl
 
JA
PERK mediates cell-cycle exit during the mammalian unfolded protein response.
Proc Natl Acad Sci U S A
2000
, vol. 
97
 
23
(pg. 
12625
-
12630
)
25
Brewer
 
JW
Hendershot
 
LM
Sherr
 
CJ
Diehl
 
JA
Mammalian unfolded protein response inhibits cyclin D1 translation and cell-cycle progression.
Proc Natl Acad Sci U S A
1999
, vol. 
96
 
15
(pg. 
8505
-
8510
)
26
Zinszner
 
H
Kuroda
 
M
Wang
 
X
, et al. 
CHOP is implicated in programmed cell death in response to impaired function of the endoplasmic reticulum.
Genes Dev
1998
, vol. 
12
 
7
(pg. 
982
-
995
)
27
McCullough
 
KD
Martindale
 
JL
Klotz
 
LO
Aw
 
TY
Holbrook
 
NJ
Gadd153 sensitizes cells to endoplasmic reticulum stress by down-regulating Bcl2 and perturbing the cellular redox state.
Mol Cell Biol
2001
, vol. 
21
 
4
(pg. 
1249
-
1259
)
28
Harding
 
HP
Zhang
 
Y
Bertolotti
 
A
Zeng
 
H
Ron
 
D
Perk is essential for translational regulation and cell survival during the unfolded protein response.
Mol Cell
2000
, vol. 
5
 
5
(pg. 
897
-
904
)
29
Kisselev
 
AF
Callard
 
A
Goldberg
 
AL
Importance of the different proteolytic sites of the proteasome and the efficacy of inhibitors varies with the protein substrate.
J Biol Chem
2006
, vol. 
281
 
13
(pg. 
8582
-
8590
)
30
Bianchi
 
G
Oliva
 
L
Cascio
 
P
, et al. 
The proteasome load versus capacity balance determines apoptotic sensitivity of multiple myeloma cells to proteasome inhibition.
Blood
2009
, vol. 
113
 
13
(pg. 
3040
-
3049
)
31
Cenci
 
S
van Anken
 
E
Sitia
 
R
Proteostenosis and plasma cell pathophysiology.
Curr Opin Cell Biol
2011
, vol. 
23
 
2
(pg. 
216
-
222
)
32
Meister
 
S
Schubert
 
U
Neubert
 
K
, et al. 
Extensive immunoglobulin production sensitizes myeloma cells for proteasome inhibition.
Cancer Res
2007
, vol. 
67
 
4
(pg. 
1783
-
1792
)
33
Arrigo
 
AP
Tanaka
 
K
Goldberg
 
AL
Welch
 
WJ
Identity of the 19S ‘prosome’ particle with the large multifunctional protease complex of mammalian cells (the proteasome).
Nature
1988
, vol. 
331
 
6152
(pg. 
192
-
194
)
34
Orlowski
 
M
Wilk
 
S
Catalytic activities of the 20 S proteasome, a multicatalytic proteinase complex.
Arch Biochem Biophys
2000
, vol. 
383
 
1
(pg. 
1
-
16
)
35
Heinemeyer
 
W
Fischer
 
M
Krimmer
 
T
Stachon
 
U
Wolf
 
DH
The active sites of the eukaryotic 20 S proteasome and their involvement in subunit precursor processing.
J Biol Chem
1997
, vol. 
272
 
40
(pg. 
25200
-
25209
)
36
Britton
 
M
Lucas
 
MM
Downey
 
SL
, et al. 
Selective inhibitor of proteasome's caspase-like sites sensitizes cells to specific inhibition of chymotrypsin-like sites.
Chem Biol
2009
, vol. 
16
 
12
(pg. 
1278
-
1289
)
37
Mirabella
 
AC
Pletnev
 
AA
Downey
 
SL
, et al. 
Specific cell-permeable inhibitor of proteasome trypsin-like sites selectively sensitizes myeloma cells to bortezomib and carfilzomib.
Chem Biol
2011
, vol. 
18
 
5
(pg. 
608
-
618
)
38
Arendt
 
CS
Hochstrasser
 
M
Identification of the yeast 20S proteasome catalytic centers and subunit interactions required for active-site formation.
Proc Natl Acad Sci U S A
1997
, vol. 
94
 
14
(pg. 
7156
-
7161
)
39
Kloetzel
 
PM
Antigen processing by the proteasome.
Nat Rev Mol Cell Biol
2001
, vol. 
2
 
3
(pg. 
179
-
187
)
40
Parlati
 
F
Lee
 
SJ
Aujay
 
M
, et al. 
Carfilzomib can induce tumor cell death through selective inhibition of the chymotrypsin-like activity of the proteasome.
Blood
2009
, vol. 
114
 
16
(pg. 
3439
-
3447
)
41
Kuhn
 
DJ
Hunsucker
 
SA
Chen
 
Q
Voorhees
 
PM
Orlowski
 
M
Orlowski
 
RZ
Targeted inhibition of the immunoproteasome is a potent strategy against models of multiple myeloma that overcomes resistance to conventional drugs and nonspecific proteasome inhibitors.
Blood
2009
, vol. 
113
 
19
(pg. 
4667
-
4676
)
42
Ho
 
YK
Bargagna-Mohan
 
P
Wehenkel
 
M
Mohan
 
R
Kim
 
KB
LMP2-specific inhibitors: chemical genetic tools for proteasome biology.
Chem Biol
2007
, vol. 
14
 
4
(pg. 
419
-
430
)
43
Singh
 
AV
Bandi
 
M
Aujay
 
MA
, et al. 
PR-924, a selective inhibitor of the immunoproteasome subunit LMP-7, blocks multiple myeloma cell growth both in vitro and in vivo.
Br J Haematol
2011
, vol. 
152
 
2
(pg. 
155
-
163
)
44
Hanada
 
M
Sugawara
 
K
Kaneta
 
K
, et al. 
Epoxomicin, a new antitumor agent of microbial origin.
J Antibiot (Tokyo)
1992
, vol. 
45
 
11
(pg. 
1746
-
1752
)
45
Meng
 
L
Mohan
 
R
Kwok
 
BH
Elofsson
 
M
Sin
 
N
Crews
 
CM
Epoxomicin, a potent and selective proteasome inhibitor, exhibits in vivo antiinflammatory activity.
Proc Natl Acad Sci U S A
1999
, vol. 
96
 
18
(pg. 
10403
-
10408
)
46
Demo
 
SD
Kirk
 
CJ
Aujay
 
MA
, et al. 
Antitumor activity of PR-171, a novel irreversible inhibitor of the proteasome.
Cancer Res
2007
, vol. 
67
 
13
(pg. 
6383
-
6391
)
47
Sin
 
N
Kim
 
KB
Elofsson
 
M
, et al. 
Total synthesis of the potent proteasome inhibitor epoxomicin: a useful tool for understanding proteasome biology.
Bioorg Med Chem Lett
1999
, vol. 
9
 
15
(pg. 
2283
-
2288
)
48
Groll
 
M
Kim
 
KB
Kairies
 
N
Huber
 
R
Crews
 
CM
Crystal Structure of epoxomicin:20s proteasome reveals a molecular basis for selectivity of alpha', beta'-epoxyketone proteasome inhibitors.
J Am Chem Soc
2000
, vol. 
122
 
6
(pg. 
1237
-
1238
)
49
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
, vol. 
14
 
3
(pg. 
451
-
456
)
50
Adams
 
J
Behnke
 
M
Chen
 
S
, et al. 
Potent and selective inhibitors of the proteasome: dipeptidyl boronic acids.
Bioorg Med Chem Lett
1998
, vol. 
8
 
4
(pg. 
333
-
338
)
51
Dorsey
 
BD
Iqbal
 
M
Chatterjee
 
S
, et al. 
Discovery of a potent, selective, and orally active proteasome inhibitor for the treatment of cancer.
J Med Chem
2008
, vol. 
51
 
4
(pg. 
1068
-
1072
)
52
O'Connor
 
OA
Stewart
 
AK
Vallone
 
M
, et al. 
A phase 1 dose escalation study of the safety and pharmacokinetics of the novel proteasome inhibitor carfilzomib (PR-171) in patients with hematologic malignancies.
Clin Cancer Res
2009
, vol. 
15
 
22
(pg. 
7085
-
7091
)
53
Yang
 
J
Wang
 
Z
Fang
 
Y
, et al. 
Pharmacokinetics, pharmacodynamics, metabolism, distribution, and excretion of carfilzomib in rats.
Drug Metab Dispos
2011
, vol. 
39
 
10
(pg. 
1873
-
1882
)
54
Alsina
 
M
Trudel
 
S
Furman
 
RR
, et al. 
A phase I single-agent study of twice-weekly consecutive-day dosing of the proteasome inhibitor carfilzomib in patients with relapsed or refractory multiple myeloma or lymphoma.
Clin Cancer Res
2012
, vol. 
18
 
17
(pg. 
4830
-
4840
)
55
Uttamsingh
 
V
Lu
 
C
Miwa
 
G
Gan
 
LS
Relative contributions of the five major human cytochromes P450, 1A2, 2C9, 2C19, 2D6, and 3A4, to the hepatic metabolism of the proteasome inhibitor bortezomib.
Drug Metab Dispos
2005
, vol. 
33
 
11
(pg. 
1723
-
1728
)
56
Wang
 
Z
Yang
 
J
Fang
 
Y
, et al. 
In vitro and In vivo drug-drug interaction studies of carfilzomib.
AAPS J
2011
, vol. 
13
 pg. 
T2419
 
57
Venkatakrishnan
 
K
Rader
 
M
Ramanathan
 
RK
, et al. 
Effect of the CYP3A inhibitor ketoconazole on the pharmacokinetics and pharmacodynamics of bortezomib in patients with advanced solid tumors: a prospective, multicenter, open-label, randomized, two-way crossover drug-drug interaction study.
Clin Ther
2009
, vol. 
31
 
Pt 2
(pg. 
2444
-
2458
)
58
Vij
 
R
Siegel
 
DS
Jagannath
 
S
, et al. 
An open-label, single-arm, phase 2 study of single-agent carfilzomib in patients with relapsed and/or refractory multiple myeloma who have been previously treated with bortezomib.
Br J Haematol
2012
, vol. 
158
 
6
(pg. 
739
-
748
)
59
Berenson
 
JR
Yellin
 
O
Dichmann
 
R
, et al. 
A phase I/II study of carfilzomib (CFZ) as a replacement for bortezomib (BTZ) for multiple myeloma (MM) patients (Pts) progressing while receiving a BTZ-containing combination regimen.
ASCO Annual Meeting Abstracts
2012
, vol. 
30
 
15 Suppl
pg. 
8098
 
60
Kuhn
 
DJ
Orlowski
 
RZ
Bjorklund
 
CC
Second generation proteasome inhibitors: carfilzomib and immunoproteasome-specific inhibitors (IPSIs).
Curr Cancer Drug Targets
2011
, vol. 
11
 
3
(pg. 
285
-
295
)
61
Lu
 
S
Yang
 
J
Song
 
X
, et al. 
Point mutation of the proteasome beta5 subunit gene is an important mechanism of bortezomib resistance in bortezomib-selected variants of Jurkat T cell lymphoblastic lymphoma/leukemia line.
J Pharmacol Exp Ther
2008
, vol. 
326
 
2
(pg. 
423
-
431
)
62
Fuchs
 
D
Berges
 
C
Opelz
 
G
Daniel
 
V
Naujokat
 
C
Increased expression and altered subunit composition of proteasomes induced by continuous proteasome inhibition establish apoptosis resistance and hyperproliferation of Burkitt lymphoma cells.
J Cell Biochem
2008
, vol. 
103
 
1
(pg. 
270
-
283
)
63
Ri
 
M
Iida
 
S
Nakashima
 
T
, et al. 
Bortezomib-resistant myeloma cell lines: a role for mutated PSMB5 in preventing the accumulation of unfolded proteins and fatal ER stress.
Leukemia
2010
, vol. 
24
 
8
(pg. 
1506
-
1512
)
64
Oerlemans
 
R
Franke
 
NE
Assaraf
 
YG
, et al. 
Molecular basis of bortezomib resistance: proteasome subunit beta5 (PSMB5) gene mutation and overexpression of PSMB5 protein.
Blood
2008
, vol. 
112
 
6
(pg. 
2489
-
2499
)
65
Ruckrich
 
T
Kraus
 
M
Gogel
 
J
, et al. 
Characterization of the ubiquitin-proteasome system in bortezomib-adapted cells.
Leukemia
2009
, vol. 
23
 
6
(pg. 
1098
-
1105
)
66
Zhang
 
L
Littlejohn
 
JE
Cui
 
Y
Cao
 
X
Peddaboina
 
C
Smythe
 
WR
Characterization of bortezomib-adapted I-45 mesothelioma cells.
Mol Cancer
2010
, vol. 
9
 pg. 
110
 
67
Gareau
 
C
Fournier
 
MJ
Filion
 
C
, et al. 
p21(WAF1/CIP1) upregulation through the stress granule-associated protein CUGBP1 confers resistance to bortezomib-mediated apoptosis.
PLoS One
2011
, vol. 
6
 
5
pg. 
e20254
 
68
Gutman
 
D
Morales
 
AA
Boise
 
LH
Acquisition of a multidrug-resistant phenotype with a proteasome inhibitor in multiple myeloma.
Leukemia
2009
, vol. 
23
 
11
(pg. 
2181
-
2183
)
69
Verbrugge
 
SE
Assaraf
 
YG
Dijkmans
 
BA
, et al. 
Inactivating PSMB5 mutations and P-glycoprotein (multidrug resistance-associated protein/ATP-binding cassette B1) mediate resistance to proteasome inhibitors: ex vivo efficacy of (immuno)proteasome inhibitors in mononuclear blood cells from patients with rheumatoid arthritis.
J Pharmacol Exp Ther
2012
, vol. 
341
 
1
(pg. 
174
-
182
)
70
Siegel
 
DS
Martin
 
T
Wang
 
M
, et al. 
A phase 2 study of single-agent carfilzomib (PX-171-003-A1) in patients with relapsed and refractory multiple myeloma.
Blood
2012
, vol. 
120
 
14
(pg. 
2817
-
2825
)
71
Vij
 
R
Wang
 
M
Kaufman
 
JL
, et al. 
An open-label, single-arm, phase 2 (PX-171-004) study of single-agent carfilzomib in bortezomib-naive patients with relapsed and/or refractory multiple myeloma.
Blood
2012
, vol. 
119
 
24
(pg. 
5661
-
5670
)
72
Badros
 
AZ
Vij
 
R
Martin
 
T
, et al. 
Phase I study of carfilzomib in patients (Pts) with relapsed and refractory multiple myeloma (MM) and varying degrees of renal insufficiency [abstract].
Blood (ASH Annual Meeting Abstracts)
2009
, vol. 
114
 
22
pg. 
3877
 
73
Badros
 
AZ
Vij
 
R
Martin
 
T
, et al. 
Phase II study of carfilzomib in patients with relapsed/refractory multiple myeloma and renal insufficiency.
ASCO Annual Meeting Abstracts
2010
, vol. 
28
 
15 Suppl
pg. 
8128
 
74
Jagannath
 
S
Vij
 
R
Kaufman
 
JL
, et al. 
Long-term treatment and tolerability of the novel proteasome inhibitor carfilzomib (CFZ) in patients with relapsed and/or refractory multiple myeloma (R/R MM) [abstract].
Blood (ASH Annual Meeting Abstracts)
2010
, vol. 
116
 
21
pg. 
1953
 
75
Papadopoulos
 
KP
Lee
 
P
Singhal
 
S
, et al. 
A phase 1b/2 study of prolonged infusion carfilzomib in patients with relapsed and/or refractory (R/R) multiple myeloma: updated efficacy and tolerability from the completed 20/56mg/m2 expansion cohort of PX-171-007 [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
, vol. 
118
 
21
pg. 
2930
 
76
Wang
 
M
Bensinger
 
W
Martin
 
T
, et al. 
Interim results from PX-171-006, a phase (Ph) II multicenter dose-expansion study of carfilzomib (CFZ), lenalidomide (LEN), and low-dose dexamethasone (loDex) in relapsed and/or refractory multiple myeloma (R/R MM).
ASCO Annual Meeting Abstracts
2011
, vol. 
29
 
15 Suppl
pg. 
8025
 
77
Jakubowiak
 
AJ
Dytfeld
 
D
Griffith
 
KA
, et al. 
A phase 1/2 study of carfilzomib in combination with lenalidomide and low-dose dexamethasone as a frontline treatment for multiple myeloma.
Blood
2012
, vol. 
120
 
9
(pg. 
1801
-
1809
)
78
Kolb
 
B
Hulin
 
C
Caillot
 
D
, et al. 
Phase I/II study of carfilzomib plus melphalan-prednisone (CMP) in elderly patients with de novo multiple myeloma.
ASCO Annual Meeting Abstracts
2012
, vol. 
30
 
15 Suppl
pg. 
8009
 
79
Mikhael
 
J
Reeder
 
CB
Libby
 
EN
, et al. 
A phase I/II trial of cyclophosphamide, carfilzomib, thalidomide, and dexamethasone (CYCLONE) in patients with newly diagnosed multiple myeloma.
ASCO Annual Meeting Abstracts
2012
, vol. 
30
 
15 Suppl
pg. 
8010
 
Sign in via your Institution