Skip to main content
Erschienen in: Cancer and Metastasis Reviews 4/2017

Open Access 25.10.2017

(Immuno)proteasomes as therapeutic target in acute leukemia

verfasst von: Jacqueline Cloos, Margot SF Roeten, Niels E Franke, Johan van Meerloo, Sonja Zweegman, Gertjan JL Kaspers, Gerrit Jansen

Erschienen in: Cancer and Metastasis Reviews | Ausgabe 4/2017

Abstract

The clinical efficacy of proteasome inhibitors in the treatment of multiple myeloma has encouraged application of proteasome inhibitor containing therapeutic interventions in (pediatric) acute leukemia. Here, we summarize the positioning of bortezomib, as first-generation proteasome inhibitor, and second-generation proteasome inhibitors in leukemia treatment from a preclinical and clinical perspective. Potential markers for proteasome inhibitor sensitivity and/or resistance emerging from leukemia cell line models and clinical sample studies will be discussed focusing on the role of immunoproteasome and constitutive proteasome (subunit) expression, PSMB5 mutations, and alternative mechanisms of overcoming proteolytic stress.

1 Introduction

Hematological malignancies comprise of many subgroups including chronic and acute leukemia, lymphoma, and multiple myeloma (MM). In this review, we focus exclusively on acute leukemia, which can be divided into two major subgroups: acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML). In children, the majority of leukemia cases are ALL [1], while AML is more prevalent in adults with leukemia [2]. With a 5-year overall survival (OS) of 83–94% [3], the prognosis of pediatric ALL is considerably better as compared to adults (5-year OS 15–35%, depending on age) [4]. A similar difference in prognosis between children and adults is seen in AML with a 5-year OS of, respectively, 65–70% and 10–45% (depending on age) [5]. The main reasons for treatment failure in both children and adults are intrinsic or acquired drug resistance in a subset of leukemia cells that are responsible for refractory disease or the development of relapse, which have a dismal prognosis. Since the (emergence of) drug resistance is one of the limiting factors that impacts long-term efficacy of anti-leukemic drugs, the search for new anti-leukemic drugs with novel mechanisms of action is an ongoing challenge.
Most anti-leukemic drugs are targeted against DNA replication to interfere with abundant cell proliferation (Fig. 1). For leukemia cells to expand, they also rely on a very high protein turnover. In normal cells, with normal chromosome numbers and normal protein balance, protein homeostasis is maintained mainly by the ubiquitin-proteasome system (UPS) [6]. Besides rapid cell growth, leukemia cells also feature many chromosomal and molecular aberrations, including chromosomal translocations (e.g., t(8:21), Inv(16)), hypo- and hyper-diploidy, activating mutations (e.g., FLT3/ITD, cKIT), and splicing defects, the latter leading to many different protein isoforms [7, 8]. Together, this leads to an aberrant protein expression, which imposes an inherent heavy burden on the UPS. These considerations set the stage for therapeutic interventions of UPS-targeting with proteasome inhibitors (PIs) [9], of which bortezomib (BTZ) as the first prototypical PI proved successful in therapy-refractory multiple myeloma (MM) [10]. Currently, first line treatment of MM includes BTZ and extends to clinical evaluations of next-generation PIs. Updates of PI treatment in MM have been subject of several recent dedicated reviews [6, 11, 12].
This review will primarily focus on the application of PIs in acute leukemia, in particular refractory disease. Since many novel drugs with different mechanisms of action are currently available, it is crucial to select those patients for certain PI who will benefit from the treatment. Therefore, it is of clinical relevance to understand the mechanism of action of PIs in leukemia and identify parameters that can help to define (non)-responsiveness to PIs. To this end, this review provides a comprehensive overview on the molecular mechanisms of action and resistance to PIs in leukemia as well as current applications of PIs in clinical trials in leukemia patients.

2 Proteasome inhibitors in leukemia

In the context of hematological cells, it is of importance to recognize that the proteasome composition is highly skewed for > 70% towards immunoproteasomes (iP) over constitutive proteasomes (cP) [13, 14]. The three catalytically active β-subunits (β1, β2, and β5) of the constitutive proteasome and the immunoproteasome counterparts β1i, β2i, and β5i harbor caspase-like, trypsin-like, and chymotrypsin-like proteolytic activity, respectively (Fig. 2).
iP expression is markedly induced upon stimulation by inflammatory cytokines such as IFNγ and TNFα [16] (Fig. 2a, b). One of the primary functions of iP is to broaden the spectrum of generating antigenic peptides for presentation on MHC class I molecules [15, 17], but also additional functions for iPs have been defined, e.g., clearance of polyubiquitinated protein aggregates emerging under inflammatory oxidative stress conditions [15, 1823] (Fig. 2c). Given the abundance of iP in leukemia cells, selective targeting of iP is an attractive treatment option [24].
BTZ and next-generation PIs [25, 26] have been evaluated in preclinical and clinical studies as potential anti-leukemia drugs. An overview of their properties is provided in Table 1. BTZ is a reversible PI primarily targeting the β5 catalytic active subunit of the proteasome. Next-generation PIs differ from BTZ by being irreversible inhibitors (e.g., carfilzomib (CFZ)), favoring oral administration (e.g., ixazomib (IXA)), attenuating hematological and neurological side effects and overcoming BTZ-associated resistance modalities [4345]. Moreover, these next-generation PIs display selectivity for cP and iP, and subunits other than β5 [27, 28, 46, 47]. Initial ex vivo activity studies of BTZ and next-generation inhibitors of cP (CFZ, ONX-0912) and iP (ONX-0914) revealed considerable inter-patient variabilities but overall greater potency in ALL than AML cells [48]. Moreover, BTZ, CFZ, and ONX-0912 were 3–10-fold more potent than the iP inhibitor ONX-914. Interestingly, this study also showed that a higher ratio of immunoproteasome over constitutive proteasome levels in leukemia cells was a good indicator for PI sensitivity. An overview of selected preclinical studies of several PIs in ALL and AML cell lines and primary cells is summarized in Table 2. Overall, these data show the relevance of preclinical studies to unravel the individual specific mechanisms of action of PIs related to apoptosis induction. In addition, these data reveal promising combination strategies for improvement of successful PI therapy.
Table 1
Overview of current proteasome inhibitors
Class
Compounds
Binding to proteasome
Specificity and mechanisms
Peptide aldehydes
MG-132, ALLnL, ALLnM, LLnV, PSI
Reversible
Interact with the catalytic threonine residue of the proteasome.
Peptide boronates
Bortezomib, MG-262, PS273
CEP-18770 (delanzomib)
MLN9708/MLN2238 (ixazomib citrate/ixazomib)
Reversible
Selective proteasome inhibitors. Interact with the catalytic threonine residue of the proteasome.
Peptide vinyl sulfones
NLVS, YLVS
Irreversible
Interact with β-subunits of the proteasome.
Peptide epoxyketones
Dihydroeponemycin
Epoxomycin
PR-171 (carfilzomib)
PR-047 (ONX 0912, oprozomib)
Irreversible
Selective proteasome inhibitors. Bind specifically to β5-subunit of the proteasome.
 
PR-957 (ONX 0914)
PR-924
 
Selective immune proteasome inhibitors. Bind to immune β-subunits of the proteasome.
β-Lactones
Lactacystin
Irreversible
Relatively specific but weak proteasome inhibitors. Binds to β-subunits of the proteasome.
 
NPI-0052 (marizomib)
Irreversible
Binds to β-subunits of the proteasome.
Abbreviations: MG-132 Carbobenzoxy-L-leucyl-L-leucyl-leucinal, ALLnL N-acetyl-L-leucyl-L-leucyl-L-norleucinal, ALLnM N-acetyl-L-leucyl-L-leucyl-Lmethioninal, LLnV N-Carbobenzoxy-L-leucyl-L-norvalinal, PSI N-carbobenzoxy-L-isoleucyl-L-γ-t-butyl-L-glutamyl-L-alanyl-L-leucinal, Leu-Leu-vinyl sulfone, MG-262N-benzyloxycarbonyl-L-leucyl-L-leucyl-L-leucyl boronic acid. See for details [13, 2742]
Table 2
Selection of preclinical studies of proteasome inhibitors in leukemia
Proteasome inhibitors
Leukemic cells
Study results and mechanisms involved
Refs.
Several
AML cell line HL60
Induction of apoptosis. Increase of p27Kip1. Activation of cysteine proteases.
[49]
PSI
CML, AML, and ALL cell lines
Induction of apoptosis in all cell lines. Enhanced taxol and cisplatinum cytotoxicity. PSI was more active on leukemic than on normal CD34+ bone marrow progenitors.
[50]
Lactacystin
AML cell line U937
Lactacystin combined with PKC activator bryostatin enhanced apoptosis.
[51]
Lactacystin, MG-132
Primary CLL cells
Induction of apoptosis in both GC sensitive and resistant cells. Activation of cysteine proteases. Apoptosis is blocked by caspase antagonist zVADfmk. Inhibition of NF-κB.
[52]
MG-132, LLnL, lactacystin
AML and ALL cell lines, primary AML cells
Synergistic interactions between PI and cyclin-dependent kinase inhibitors flavopiridol and roscovitine. Downregulation of XIAP, p21CIP1, and Mcl-1.
[53]
Bortezomib
Primary CLL cells
Induction of apoptosis associated with release of SMAC and cytochrome c.
[54]
Bortezomib
CML, AML, and ALL cell lines
Synergistic with flavopiridol. Blockade of the IκB/NF-κB pathway. Activation of the SAPK/JNK cascade. Reduction in activity of STAT3 and STAT5.
[55]
Bortezomib
Primary CLL cells
Dose-dependent cytotoxicity of bortezomib. Additive effect with purine nucleoside analogues cladribine and fludaribine. CLL cells more sensitive than normal lymphocytes.
[56]
Bortezomib
AML and ALL cell lines, primary pediatric AML and ALL cells
Lymphoblastoid, CML and AML cell lines. Bortezomib induced apoptosis and acted at least additive with dexamethasone, vincristine, asparaginase, cytarabine, doxorubicin, geldanamycin, HA14.1, and trichostatin A.
[57]
Bortezomib
AML cell lines
Synergistic with tipifarnib. The combination overcomes cell adhesion-mediated drug resistance.
[58]
Bortezomib
Pediatric ALL xenocraft model
In vitro and in vivo activity of bortezomib against primary pediatric ALL cells in a xenocraft mouse model.
[59]
Bortezomib, PSI
CML and AML cell lines
PSI enhanced toxicity of daunoblastin, taxol, cisplatinum, and bortezomib. PSI and bortezomib suppressed clonogenic potential of AML and CML more than that of normal bone marrow (NBM) progenitors. Bortezomib inhibited the clonogenic potential of CML and NBM more effectively.
[60]
Carfizomib
Primary AML and ALL cells
Inhibits proliferation and induces apoptosis AML, inhibits proliferation in ALL.
[61]
Carfilzomib, bortezomib
AML cell lines and primary AML cells
Synergistic effect on proteotoxic stress together with the protease inhibitors ritonavir, nelfinavir, saquinavir, and lopinavir.
[62]
Carfilzomib, bortezomib
ALL cell lines in vitro and in xenograft model
Proteasome inhibitors evoke latent tumor suppression programs in pro-B MLL leukemia through MLL-AF4.
[63]
Carfilzomib
MM, AML, Burkitt’s lymphoma cell lines
Induces proapoptotic sequelae, including proteasome substrate accumulation, Noxa and caspase 3/7 induction, and phospho-eIF2α suppression.
[13]
Marizomib
ALL, AML, and CML cell lines and in xenograft model
Induces caspase-8 and ROS-dependent apoptosis alone and in combination with HDAC inhibitors.
[64, 65]
Marizomib, bortezomib
AML and ALL cell lines
Anti-leukemic activity, synergistic in combination with bortezomib.
[31]
ONX 0914
AML and ALL cell lines
Growth inhibition, proteasome inhibitor-induced apoptosis, activation of PARP cleavage and accumulation of polyubiquitinated proteins.
[16]
PR-924
AML and ALL cell lines
Growth inhibition, immune proteasome inhibition, apoptosis, activation of PARP cleavage.
[29]
Ixazomib
Primary CLL cells
Annexin-V staining, PARP1 and caspase-3 cleavage and an increase in mitochondrial membrane permeability, apoptosis was only partially blocked by the pan-caspase inhibitor z-VAD-fmk.
[66]
Updated from Franke et al. [67]
Abbreviations: PSI N-carbobenzoxy-L-isoleucyl-L-γ-t-butyl-L-glutamyl-L-alanyl-L-leucinal, LLnV N-Carbobenzoxy-L-leucyl-L-norvalinal, LLnL N-acetylleucylleucylnorleucinal, MG-132 Carbobenzoxy-L-leucyl-L-leucyl-leucinal, GC glucocorticoid, PKC protein kinase C

3 Markers for PI (BTZ) sensitivity/resistance in leukemia cell lines

As with any new treatment strategy, selection of patients who will benefit from the treatment is essential. With respect to PIs, studies with leukemia cell lines can help to define markers for response, long-term efficacy, and emergence of resistance to PIs. Resistance mechanisms often reveal critical processes such as targeted and compensatory mechanisms that leukemia cells harbor to overcome the effects of PIs. A large number of studies have followed the approach of exposing leukemia cells to stepwise increasing concentrations of PIs (mostly BTZ) and characterize cells with acquired resistance [6871]. Figure 3 depicts an overview of molecular mechanisms of resistance in PI/BTZ-resistant leukemia cell lines [44, 46, 72]. The most common mechanisms are discussed below.

3.1 Upregulation of proteasomal subunits

Upregulation of the primary target is a well-known response of cells exposed to drugs. Likewise, upregulation of proteasomal subunits, and the β5 subunit in particular, is frequently observed in BTZ-resistant leukemia cell lines (reviewed in [44, 46]) indicating its role in BTZ resistance. Consistently, CGH analysis of BTZ-resistant cell lines revealed amplification of the PSMB5 gene with variability in size and extent of the amplification of chromosome 6 [73]. Remarkably, immunoproteasome subunit levels are often found to be decreased in BTZ-resistant leukemia cell lines [16, 68, 71]. The resulting decrease in immuno- / constitutive proteasome ratio and alterations in subunit composition has been linked to a diminished BTZ sensitivity [74, 75]. Interferon gamma (IFNγ) is an efficient inducer of immunoproteasome expression and partly restored BTZ sensitivity in BTZ-resistant leukemia cells [16]. Beyond BTZ-resistant cells, IFNγ also enhanced BTZ sensitivity in a panel of B cell lines by 50% [76]. Together, these data underscore the importance of immuno- and constitutive proteasome subunit composition in BTZ sensitivity and resistance in leukemia cells.

3.2 PSMB5 mutations

Molecular analysis of the proteasome subunits of PI-resistant leukemia cell lines revealed several mutations in exon 2 of the PSBM5 gene encoding the highly conserved binding pocket region for PIs within the β5 subunit of the proteasome. This highly conserved region appears to be a mutation “hot spot” when cells are exposed to BTZ for a prolonged period. Figure 4 shows an overview of PSMB5 mutations in PI-resistant cell lines, including non-hematological malignancies.
All mutations result in amino acid alteration in, or in the close vicinity of, the PI binding pocket [82]. In fact, except for the A247G mutation which introduces a Thr21Ala substitution, all mutations result in amino acid alterations in the S1 pocket of the β5 subunit with Ala49 being the most affected amino acid. This specificity pocket is a highly conserved part of the subunit and responsible for recognizing the peptide bond of the substrate. It is also the site that has to be cleaved and determines the specificity as well as facilitates the binding of the P1 side chain of BTZ [82]. In silico analysis provided evidence for hindered binding of BTZ by the majority of the mutations, including the Thr21Ala amino acid alteration positioned outside of the S1 pocket but within the BTZ binding pocket [82]. The only exceptions were the Met45Ile and Met45Val substitutions, which do not directly interact with BTZ. However, Met45 is known to contribute to the specificity of the S1 pocket and upon binding, Met45 undergoes a conformational change and shifts the direction of its side chain towards Cys52 vicinity. Alterations in this amino acid might therefore hinder this conformational change and contribute as well to decreased BTZ binding. Docking the LLVY-AMC substrate in silico in the crystal structure of the β5 subunit supported the hypothesis of decreased binding of the substrate in the mutated binding pocket.
There are two strong indications that PSMB5 mutations are related to PI resistance. First, from marine biology studies, characterization of proteasome subunits of the PI Salinosporamide A (NPI-0052, Marizomib) producing actinobacterium Salinispora tropica revealed the same Ala49Val and Met45Phe mutations in the β5 subunit homologue as in BTZ-resistant leukemia cells. In S. tropica, these mutations conferred “self-resistance” to Salinosporamide A [80]. Second, acquired resistance to the immunoproteasome inhibitor in PR-924 was not associated with any mutation(s) in immunoproteasome subunits, but rather provoked PSMB5 mutations, i.e., Met45Ile, Ala49Thr, and Met45Val [29]. Taken together, inhibition of the β5 subunit is essential for the anti-leukemia effect of PIs and PSMB5 mutations emerging after prolonged PI treatment attenuate the inhibitory potency and confer resistance in leukemia cells. PSMB5 mutations have thus far not been identified in patients receiving PI therapy and it has been challenged whether these mutations hold clinical relevance for PI resistance. However, until now, longstanding treatment with the first-generation PI was hardly applied in clinical practice. This may change with the introduction of second-generation and oral PI’s allowing for long-term treatment.

3.3 Alternative protein disposal

Gene expression profiling of BTZ-resistant leukemia cells identified myristoylated alanine-rich C-kinase substrate (MARCKS) as being highly upregulated. MARCKS is an 80-kDa protein that is involved in multiple exocytosis pathways (reviewed in [83]). In fact, in BTZ-resistant leukemia cells, MARCKs protein co-localized with intracellular vesicles that contained polyubiquitinated proteins and which were formed upon exposing cells to increasing concentrations of BTZ. The ubiquitin-containing vesicles have been described before [84], and in co-cultures, it was shown that after extrusion of the vesicles, they were taken up in proteasome-proficient acceptor cells [73]. MARCKs may thus contribute to BTZ resistance by facilitating exocytosis-mediated extrusion of polyubiquitinated proteins to overcome proteolytic stress imposed by BTZ. MARCKs upregulation is not restricted to BTZ-resistant leukemia cells, but was also observed in leukemia cells with acquired resistance to second-generation PI, e.g., Salinosporamide A (Marizomib) and the immunoproteasome inhibitor PR924.
Another alternative protein disposal that is often related to drug resistance is autophagy. This process has been linked to BTZ resistance in several tumor models [8591], although it was not indicated in MARCKs overexpressing PI-resistant leukemia cells [73]. Inhibition of autophagy through different mechanisms including calpain inhibitor [86], downregulation of heat shock protein B8 (HSPB8) [87], B cell lymphoma 2-interacting mediator of cell death (BIM) upregulation [90], tipifarnib [85], or (hydroxy)chloroquine [92, 93] increased BTZ sensitivity in a MM model. In addition, histone deacetylase (HDAC) inhibitors can block autophagy by disrupting aggresome formation, the process that precedes autophagy [94, 95]. Interestingly, the E3 ligase, TRAF6, links the NFkB pathway to autophagy [96] and it was shown that BTZ-resistant AML cells were sensitized by downregulating TRAF 6 [97]. Despite the potential impact of autophagy on BTZ resistance, the extent of involvement of this process in leukemia cells needs further exploration.

3.4 Activation of pro-survival pathways

Analogous to resistance to many anti-leukemia drugs, PI resistance has also been associated with the activation of pro-survival pathways. The most extensively described is the association with the NFκB survival pathway (for AML reviewed in [98]). As PI result in the stability of the inhibitor of NFκB (IκB), this survival pathway is inhibited by exposure to PI. Intrinsically resistant leukemia cells (e.g., stem cells) have a constitutively activated NFκB pathway and the combination of BTZ with NFκB inhibition by the IKK inhibitor BMS-345541, enhanced the kill of AML stem cells [99]. Although the interaction between NFκB and PI resistance is clearly established for MM and other tumor types [100104], the data of relevance for leukemia is limited. Besides NFκB, the pro-survival pathways via AKT/mTOR [105107] and insulin-like growth factor 1 (IGF-1) [108110] have also been described to confer BTZ resistance and may affect the interaction of leukemia cells with (stromal) cells in their micro-environment.

3.5 X-box binding protein 1 (XBP1)

X-box binding protein 1 (XBP1) is a transcription factor involved in the unfolded protein response. In addition, it is important for the differentiation of plasma cells. In this respect, XBP1 expression has been linked to BTZ resistance [111] and survival [112] in MM. In leukemia, this pathway may also be related to PI resistance; however, data are currently lacking. Due to the role of XBP1 in B cell differentiation, this factor may be particularly relevant for B-ALL and warrants further study.

3.6 Drug efflux transporters

Drug efflux transporters of the ATP-binding cassette protein family have been explored for their role in PI resistance. P-glycoprotein (Pgp, ABCB1), as one of its main representatives only marginally contributes to BTZ efflux [68, 113], but is of relevance for second-generation PIs such as CFZ and the immunoproteasome inhibitor ONX-0914 as these PIs are bona fide substrates for Pgp [114, 115]. Notably, BTZ can downregulate Pgp expression and this manner indirectly attenuate drug resistance [116]. There is no evidence that BTZ and other second-generation PIs are substrates for other drug efflux transporters, e.g., multidrug resistance-associated protein 1–5 (MRP1–5, ABCC1–5) or breast cancer resistance protein (BCRP, ABCG2); hence, a role in PI resistance is not anticipated [114].

4 Markers for PI (BTZ) sensitivity/resistance in primary leukemia samples

Although in vitro models are valuable tools to identify possible mechanisms of BTZ resistance, assessment of the relevance for the clinic requires validation in ex vivo studies using primary patient samples. Preferably, this is studied in add-on studies of clinical trials that include BTZ in the treatment protocol. The potential role of three markers for PI (BTZ) sensitivity and/or resistance in leukemia samples is discussed below.

4.1 Proteasome levels and subunit composition

Add-on studies of clinical trials in leukemia are limited and most data for PIs are obtained from MM studies. These studies demonstrated besides proteasome expression levels, the proteasome subunit composition is important for response [117, 118]. Although upregulation of the proteasome was not related to resistance in mantle cell lymphoma (MCL) patients [119], studies in leukemia indicated a possible correlation between higher 20S protein expression and BTZ sensitivity [120]. In particular, studies by Niewerth et al. (2013) showed that lower β5 subunit expression correlated with increased ex vivo sensitivity for proteasome inhibitors in pediatric AML and ALL samples. In addition, the sensitivity for BTZ in AML cells inversely correlated with the ratios between immunoproteasome subunits over constitutive subunits, specifically β1i/β1 and β2i/β2, and a trend for β5i/β5. ALL cells showed higher sensitivity to BTZ as compared to AML cells. Although for ALL no significant correlations were revealed with BTZ sensitivity, they featured a higher β2i/β2 ratio and trends of a higher β1i/β1 ratio as compared to AML samples. Together, these data support the notion that a relative high immuno- / constitutive proteasome ratio promotes BTZ sensitivity. These data were confirmed in AML and ALL samples obtained from two pediatric clinical COG trials (AAML07P1 and AALL07P1) in which BTZ treatment was incorporated [14, 48]. After further validation in larger studies, assessment of immunoproteasome over constitutive proteasome ratios may be used as biomarkers of response to PIs.

4.2 PSMB5 mutations

PSMB5 mutations observed in BTZ-resistant hematological cell lines were as yet not identified in clinical samples. The mutations found in the cell line models do not represent SNPs found in the general population. Wang et al. sequenced the PSMB5 gene in a large cohort of healthy persons and 61 MM patients after BTZ treatment [121]. No SNPs were found in the exon 2 of the PSMB5 gene neither in the general population nor in MM patients. Of interest, polymorphisms that influenced PSMB5 gene expression were observed, but these did not correlate with BTZ response. It should be taken into account that sequence analysis in one third of the MM patients was performed only on whole blood and not on isolated malignant plasma cells, which may have influenced the sensitivity of the analysis. In addition, it was not stated how many resistant patients were included. More recently, Lichter et al. sequenced the PSMB5 gene in blood samples of MM patients included in the APEX trial in which patients were treated with either BTZ or dexamethasone [122]. No PSMB5 mutations were identified in this group. Although sample size of this group was limited, the data suggest that PSMB5 mutations do not represent a leading cause of acquired BTZ resistance in investigated MM protocols. Whether this holds true for leukemia or MM with long-term BTZ maintenance therapy is yet to be determined. Lastly, as a preliminary account, Barrio et al. (2016) reported PSMB5 mutations in subclones of CD138+ cells of a single MM patient after a therapy [123]. Since the subclonal frequencies were low, this poses analytical challenges to detect these subclones. Also in leukemia, it is established that there is substantial oligoclonality in mutational status of cells within the leukemia. The cells with the specific mutations tendering growth advantage are probably selected to grow out and develop a (drug resistant) relapse [124], so also in leukemia patients subclonal analysis may reveal additional mutations.

4.3 MARCKS

Upregulation of MARCKs protein expression emerged as a marker for BTZ- and second-generation PI-resistant leukemia cell lines [73]. To test whether this upregulation might be a prognostic marker for clinical BTZ resistance, MARCKs expression was examined in 44 primary ALL patient samples obtained from the clinical COG trial AALL07P1 using combination chemotherapy including BTZ. A trend (p = 0.07) was seen in the inverse correlation between MARCKS expression and clinical response. Since the samples were obtained in the setting of a clinical trial using combination therapy, a direct correlation between BTZ response and MARCKS expression cannot be made. However, these findings are consistent with data from Micallef et al. who showed MARCKS protein upregulation in a small group of BTZ-resistant MM patients [125] and Yang et al. who identified MARCKS upregulation as a resistance marker in primary MM samples [126]. These studies encourage a prospective validation of the possible prognostic role of MARCKS in leukemia.

5 Clinical trials with proteasome inhibitors in leukemia

The efficacy and safety of PIs in MM have already been extensively reviewed [127]. For leukemia, an overview of phase I/II clinical studies of PI as single agent and combination therapy in adult and pediatric leukemia is presented in Table 3. An overview of ongoing phase II/III clinical trials with BTZ (combination) therapy involving pediatric leukemia patients and adult leukemia is shown in Table 4 and Table 5, respectively. In addition, ongoing clinical trials with second-generation PIs (mainly CFZ and IXA) are depicted in Table 6. The outcome for clinical efficacy included stable disease, progressive disease, complete/partial remission, and mortality. In all studies, infections and neutropenia were common adverse drug reactions (ADRs). Neurologic ADRs were also common in all studies with BTZ, including neuropathy. Interestingly, BTZ might also have some protective effects as well, as it has been reported to prevent muscle wasting [148], which can be induced by cancer cachexia [149] but this has not yet been reported in a clinical setting. Two studies were conducted with CFZ [143, 144] and less ADRs were found in these studies including the absence of neurologic ADRs. Moreover, no dose-limiting toxicities (DLT) were found indicating a better safety profile. Importantly, the clinical response to CFZ in these studies was also better than reported for BTZ treatment.
Table 3
Published clinical studies of proteasome inhibitors in leukemia
Study drugs
Cohort
Number
Phase
Study results and mechanisms involved
Refs.
BTZ
Several hematologic malignancies
27
I
Bortezomib was given twice weekly for 4 weeks every 6 weeks. The MTD was 1.04 mg/m2. CR in 1 MM patient. PR in 1 patient with MCL and 1 with FL.
[128]
BTZ
Refractory or relapsed acute leukemia
15
I
Bortezomib was given twice weekly for 4 weeks every 6 weeks. The MTD was 1.25 mg/m2. No ≥grade 3 toxicities. 5 patients showed hematological improvement. No CR achieved.
[129]
BTZ, PegLD
AML, MM, and NHL
42
I
Bortezomib was given on days 1, 4, 8, and 11 and PedLD on day 4. MTD of BTZ 1.3 mg/m2. No significant pharmacokinetic and pharmacodynamic interactions between bortezomib and PegLD. 16 of 22 MM patients achieved CR, near-CR or PR. 1 CR and 1 PR in NHL patients. 2 of 2 AML patients achieved a PR.
[130]
BTZ
Recurrent childhood ALL, AML, blastic phase CML, M3
12
I
Bortezomib was administered twice weekly for 2 weeks followed by a 1-week rest. MTD of bortezomib was 1.3 mg/m2/dose. 5 patients were fully evaluable. DLT’s occurred in 2 patients at the 1.7 mg/m2 dose level. No OR achieved.
[131]
BTZ, IDA, AraC
AML
31
I
Addition of BTZ to AML induction chemotherapy. Bortezomib added on days 1, 4, 8, and 11. 19 CR, 3 CRp, 2 PR and 7 no response. BTZ was well-tolerated up to 1.5 mg/m2.
[132]
BTZ, VCR, DEX, PegAspa, DOX
Recurrent childhood ALL
10
I
Combination of bortezomib (1.3 mg/m2) with ALL induction therapy is active with acceptable toxicity. 6 patients achieved CR.
[133]
BTZ, VCR, DEX, PegAspa, DOX
Recurrent childhood ALL
22
II
14 patients achieved CR, and 2 achieved CRp, 3 patients died from bacterial infections, 2 of 2 included T cell ALL patients did not respond.
[134]
BTZ, tipifarnib
Relapsed or refractory ALL(26) or AML (1)
27
I
Combination well tolerated. 2 patients achieved CRp and 5 SD.
[135]
BTZ, DNR, AraC
AML (age > 65)
95
I/II
Combination was tolerated. 62 patients achieved CR and 4 patients CRp.
[136]
BTZ, 17-AAG
Relapsed or refractory AML
11
I
The combination of 17-AAG and BTZ led to toxicity without measurable response in patients with relapsed or refractory AML.
[137]
BTZ, DAC
Poor-risk AML
19
I
Combination was tolerable and active in this cohort of AML patients; 7 of 19 patients had CR or CRi. 5 of 10 patients > 65 years had CR.
[138]
BTZ, LEN
14 MDS/CMML
9 AML
23
I
MTD of BTZ 1.3 mg/m2 was tolerable in this regimen. Responses were seen in patients with MDS and AML. Two fatal infections occurred.
[139]
BTZ, IDA
Relapsed AML (7) or AML > 60 years (13)
20
I
4 patients achieved complete remission. 1 treatment-related death. Overall the combination was well tolerated.
[140]
BTZ, AZA
Relapsed or refractory AML
23
I
Dose of 1.3 mg/m2 BTZ was reached without dose-limiting toxicities. 5 out of 23 patients achieved CR.
[141]
BTZ, MIDO vs BTZ, MIDO, DHAD, etoposide, AraC
Relapsed/refractory AML
21
I
56.5% CR rate and 82.5% overall response rate (CR + CR with incomplete neutrophil or platelet count recovery). Combination is active but is associated with expected drug-related toxicities. DLTs were peripheral neuropathy, decrease in ejection fraction and diarrhea.
[142]
CFZ + dexamethasone
Refractory or relapsed acute leukemia
18
I
CFZ was given twice weekly for 4 weeks with a maximal of 6 cycles. Prior to CFZ dexamethasone was given. The MTD was not established, because no DLTs were observed (36–46 mg/m2). PR in 2/10 patients and 4/10 SD.
[143]
CFZ + dexamethasone
Previously treated patient with CLL or SLL
19
I
CFZ was given twice weekly for 3 weeks in a 28-day cycle. Prior to CFZ dexamethasone was given. No DLTs observed and all available patients for evaluation had SD (including patients with Del(17p13.1) and fludarabine-resistant disease.
[144]
BTZ, cytarabine, idarubicin vs BTZ, cytarabine etoposide
Children with relapsed, refractory, or secondary AML
37
II
BTZ, 1 or 1.3 mg/m2, was given at days 1, 4, and 8 in combination with idarubicin and cytarabine (arm A) or with etoposide and high dose cytarabine (arm B). Hypokalemia incidence was high, 17%. Four deaths occur, 3 infectious deaths and one from PD. Both arms failed to meet predetermined efficacy thresholds (CRi was not included). Arm A: CR = 21.4%, CRp + CRi = 35.6%, PR = 14.3%. Arm B: CR = 34.8%, CRp + CRi = 13% and one death.
[145]
BTZ
Relapsed/refractory ATL
15
II
BTZ, 1.3 mg/m2, was given at days 1, 4, 8, and 11. After stage 1, all patients discontinued treatment (PD = 11, AEs = 3) and the study was terminated because BTZ was not considered promising enough as a single agent. 12 patients had Gr 3/4 drug-related AEs of which 2 Gr3/4 peripheral neuropathy. Overall responses: PR = 1, SD = 5. ORR = 6.7%, PFS = 38 days (8–122).
[146]
BTZ, DEX, DOX vs BTZ, DEX, cyclophosphamide
Newly diagnosed primary plasma cell leukemia
39
II
Four alternating cycles of BTZ (1.3 mg/m2 on days 1, 4, 8, and 11), DEX plus DOX, or cyclophosphamide was given. 35 patients completed the 4 cycles. ORR = 69%, CR = 10%, VGPR = 26%, PR = 23%. 10 were refractory to the induction phase, and 2 deaths due to sepsis occur. 25 patients underwent HDM/ASCT and 1 a syngeneic allograft. After ASCT: ORR = 92% CR = 34%, VGPR = 38%, PR = 16%, PD = 8%. In the intention-to-treat population, the median PFS = 15.1 months and overall survival = 36.3 months.
[147]
Updated from Franke et al. [67]
Abbreviations: Study outcome: MTD maximum tolerated dose, DLT dose-limiting toxicities, CR complete response, CRi incomplete remission, CRp CR with incomplete platelet recovery, PR partial response, OR objective response, SD stable disease, PFS progression-free survival, EFS event-free survival, OS overall survival. Malignancies: MCL mantle cell lymphoma, FL follicular lymphoma, NHL non-Hodgkin lymphoma. Drugs: 17-AAG 17-N-Allylamino-17-Demethoxygeldanamycin, AraC cytarabine, AZA azacitidine, BTZ bortezomib, CFZ carfilzomib, DAC decitabine, DEX dexamethasone, DHAD mitoxantrone, DNR daunorubicin, DOX doxorubicin, IDA idarubicin, LEN lenalidomide, PegLD pegylated liposomal doxorubicin, PegAspa pegylated L-asparaginase, VCR vincristine
Table 4
Ongoing and unpublished clinical trials of bortezomib in acute leukemia which include pediatric patients
Study drugs
Time period
Number
Phase
Cohort
Age
Sponsor
Clinical trial identifier
BTZ + intensive reinduction chemotherapy
Mar 2009
Sept 2014
60
II
Relapsed ALL
1–31
National Cancer Institute (USA)
NCT00873093
BTZ, DEX, VCR, MTX
Sep 2009
Jul 2014
24
II
Relapsed/refractory ALL
0.5–19
Erasmus Medical Center (Rotterdam, The Netherlands)
NTR1881a
BTZ, ATO
May 2013
May 2018
30
II
Relapsed acute promyelocytic leukemia (APL)
1–75
Christian Medical College (Vellore, India)
NCT01950611
Standard leukemia chemotherapy ± BTZ
Apr 2014
Feb 2019
1400
III
T cell ALL or stages II–IV T cell lymphoblastic lymphoma
2–30
National Cancer Institute (USA)
NCT02112916
BTZ, SAHA + reinduction chemotherapy
Apr 2015
Apr 2019
30
II
Refractory or relapsed MLL rearranged leukemia
< 21
St Jude Children’s Research Hospital (Memphis, TN, USA)
NTC 02419755
BTZ, PANO + reinduction chemotherapy
Dec 2015
Apr 2019
40
II
Relapsed T cell leukemia or lymphoma
< 21
St Jude Children’s Research Hospital (Memphis, TN, USA)
NCT02518750
BTZ + induction chemotherapy
Oct 2015
Oct 2020
50
I/II
Infant leukemia and lymphoblastic lymphoma
< 1
St Jude Children’s Research Hospital (Memphis, TN, USA)
NCT02553460
BTZ + reinduction chemotherapy
July 2015
Apr 2019
20
II
Refractory or relapsed leukemia and lymphoblastic lymphoma
1–39
Children’s Mercy Hospital (Kansas City)
NCT02535806
BTZ + HR reinduction chemotherapy
Aug 2015
Aug 2018
250
II
High-risk (HR) relapsed ALL
< 18
Charité - Universitätsmedizin (Berlin, Germany)
EudraCT number: 2012–000810-12a
Updated from Franke et al. [67]
Abbreviations: Drugs: ATO arsenic trioxide, BTZ bortezomib, DEX dexamethasone, MTX methotrexate, PANO panobinostat, SAHA vorinostat, VCR vincristine
Table 5
Ongoing and unpublished clinical trials of proteasome inhibitors in adult acute leukemia
Study drugs
Time period
Number
Phase
Cohort
Age
Sponsor
Clinical trial ID
BTZ, DHAD, VP16, AraC
Jan 2006
Sept 2016
55
I/II
Relapsed/refractory acute leukemia
> 18
Thomas Jefferson University (PA, USA)
NCT00410423
BTZ, FLAG, IDA
Apr 2008
Jan 2013
40
I/II
Refractory or relapsed AML
> 18
PETHEMA Foundation
NCT00651781
BTZ, SAHA, SFN
Feb 2010
Sept 2016
38
I/II
Poor-risk AML
> 18
Indiana University (IN, USA)
NCT01534260
BTZ, BEL
May 2010
Feb 2014
24
I
Relapsed/refractory acute leukemia
> 18
Virginia Commonwealth University (VA, USA)
NCT01075425
BTZ, NFV
July 2010
Mar 2013
18
I
Relapsed or progressive advanced hematologic cancer
> 18
Swiss Group for Clinical Cancer Research (Switzerland)
NCT01164709
BTZ, DHAD, VP16, AraC
July 2010
May 2014
34
I
Relapsed/refractory AML
18–70
Case Comprehensive Cancer Center (OH, USA)
NCT01127009
Several drugs in randomization arms ± BTZ
June 2011
June 2017
1250
III
Initial AML
> 29
National Cancer Institute (USA)
NCT01371981
DAC vs BTZ, DAC
Nov 2011
June 2015
172
II
AML
> 60
National Cancer Institute (USA)
NCT01420926
BTZ, DOX, PegAspa, VCR, DEX, AraC, MTX
Mar 2013
July 2017
17
II
Relapsed/refractory ALL
> 18
National Cancer Institute (USA)
NCT01769209
BTZ, SFN, DAC
July 2013
Dec 2016
30
I
AML
> 60
National Cancer Institute (USA)
NCT01861314
BTZ, DOX
Mar 2015
Mar 2017
30
II
AML
18–80
University of California, Davis (CA, USA)
NCT01736943
BTZ, LEN
Mar 2015
Aug 2018
24
I
Relapsed AML and MDS after Alllo SCT
> 18
Massachusetts General Hospital (MA, USA)
NCT023121
Updated from Franke et al. [67]
Abbreviations: Drugs: 17-AAG 17-N-Allylamino-17-Demethoxygeldanamycin, AraC cytarabine, BEL belinostat, BTZ bortezomib, DAC decitabine, DEX dexamethasone, DHAD mitoxantrone, DNR daunorubicin, DOX doxorubicin, IDA idarubicin, FLAG fludarabine, Ara-C cytarabine, G-CSF granulocyte colony-stimulating factor, LEN lenalidomide, MTX methotrexate, NFV nelvinavir, PegLD pegylated liposomal doxorubicin, PegAspa pegylated L-asparaginase, SAHA vorinostat, SFN sorafenib, VCR vincristine, VP16 etoposide
Table 6
Ongoing clinical trials of second-generation proteasome inhibitors in acute leukemia
Study drugs
Time period
Number
Phase
Cohort
Age
Sponsor
Clinical trial ID
CFZ
Sept 2010
Jul 2015
18
I
Relapsed/refractory ALL and AML
> 18
Washington University School of Medicine (MO, USA)
NCT01137747
IXA, DHAD, VP16, AraC
May 2014
Nov 2017
30
I
Relapsed/refractory AML
18–70
Case Comprehensive Cancer Center; National Cancer Institute (NCI)
NCT02070458
IXA
Mar 2014
Mar 2016
16
II
Relapsed/refractory AML
> 18
Stanford university/National Cancer Institute (NCI)
NCT02030405
IXA, DHAD, VP16, AraC
Oct 2014
Nov 2018
30
I
Relapsed/refractory AML
18–70
Case Comprehensive Cancer Center (USA)
NCT 02070458
CFZ, DEX, DHAD, PegAspa, VCR
Dec 2014
Jul 2017
39
I/II
Relapsed/refractory AML
< 18
Onyx Therapeutics Inc. (CA, USA)
NCT02303821
CFZ, CYCLO, VP16
Jul 2015
Dec 2017
50
I
Relapsed leukemia and solid tumors
6–29
Phoenix Children’s Hospital (AZ, USA)
NCT 02512926
IXA + induction and consolidation chemotherapy
Nov 2015
Feb 2022
54
I
AML
> 60
Massachusetts General Hospital (MA, USA)
NCT02582359
Updated from Franke et al. [67]
Abbreviations: Drugs: AraC cytarabine, CFZ carfilzomib, CYCLO cyclophosphamide, DEX dexamethasone, DHAD mitoxantrone, IXA ixazomib, VCR vincristine, VP16 etoposide

6 Future perspectives

This overview of results of PI in leukemia reveals that treatment with PI as monotherapy may not give satisfactory clinical responses. To improve the employability of PI in leukemia, several factors are implied to be considered.

6.1 Combination strategies

For leukemia, chemotherapy commonly consists of cocktails of different drugs with different mechanisms of actions and different side effects to exert optimal treatment response with achievable dosages. For novel drugs, it is therefore essential that they can be combined with the most effective drugs currently used. Based on the mechanism of action, glucocorticoids are good candidate drugs to be combined with PI. BTZ and dexamethasone showed synergy in ex vivo combination studies in primary pediatric ALL samples [48] clinically BTZ was combined with induction therapy including dexamethasone, vincristine, PEG-asparaginase, and doxorubicin in relapsed pediatric ALL, showing promising CR rates [133, 134]. Currently, several clinical trials are ongoing (Tables 4 and 5), combining BTZ with several chemotherapy protocols which include the standard chemotherapeutics and novel strategies (heat shock protein inhibitors, HDAC inhibitors, and autophagy inhibitors). Based on preclinical data and expected toxicity profiles of the different drugs, the addition of PI to other chemotherapeutics seems a fruitful strategy. In addition, recently the nuclear transport (XPO1) inhibitor selinexor has been combined with BTZ [150] and is currently in a clinical trial for MM (NCT03110562). Whether this is also a relevant strategy for leukemia still has to be established.

6.2 Second-generation proteasome inhibitors

Despite the successful introduction of BTZ, several drawbacks such as resistance and toxic side effects led to development of second-generation proteasome inhibitors which are at several stages of clinical development (Table 6). Due to promising preclinical studies, the irreversible proteasome inhibitor CFZ has advanced rapidly into the clinic for MM as well as leukemia, and is supposed to be a promising alternative for BTZ and might even overcome BTZ resistance.
Recently, an oral formulation resembling BTZ, ixazomib (MLN9708), has emerged into the clinic, with two clinical trials investigating the efficacy of ixazomib in AML (NCT0230405 and NCT 02070458).
PR-957 (ONX 0914) and PR-924 represent members of a new class of proteasome inhibitors being directed specifically against the immunoproteasome [27, 30]. PR-924 demonstrated preclinical efficacy in leukemia and MM [29, 30]. In addition, ex vivo cytotoxicity of PR-957 was shown in primary leukemia samples [29]. Since leukemia cells, especially ALL cells, express high levels of immunoproteasome, it might be a good candidate for further clinical development in hematological malignancies.
Development of acquired resistance to the second-generation PIs [29, 31] has to be taken in account as well. Hence, combination therapy is also recommended for the treatment of leukemia with these novel PIs in order to circumvent toxicity and resistance.

6.3 Biomarkers of clinical response to proteasome inhibitors

The combination of preclinical research and ongoing clinical studies (e.g., add-on studies) will be needed to identify and confirm determinants of resistance and markers for clinical response in order to further personalize the treatment of acute leukemia with PI. Based on accumulating data, prediction of effectivity of PI lies in the composition of the proteasome, in particular the ratio between constitutive- and immunoproteasome subunits. This can either be assessed by measuring protein expression of the different subunits [14, 121] or their specific catalytic activities for which several assays are available [151, 152]. In addition, the activity assays can be used for pharmacodynamic monitoring of PI inhibition and duration of inhibition in PBMC’s [152, 153].
Since mutations in the genes encoding proteasome subunits have not been found in primary MM and acute leukemia patient samples either before or after treatment with PI, they are currently not considered potential biomarkers for resistance to proteasomes. However, when patients are treated for prolonged time periods such as the maintenance treatment of elderly MM patients with IXA, they might be acquired. Therefore, add-on studies measuring the mutational status in samples during treatment are currently performed. If mutations are identified, they may be used to monitor acquired resistance to PI. Selecting the patients that benefit from PI treatment and the recognition of PI resistance is indispensable for the optimal implementation of PI in acute leukemia treatment.

Acknowledgements

This research was supported by KIKA (children cancer free) and the Egbers Foundation.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
3.
Zurück zum Zitat Pui, C.-H., Carroll, W. L., Meshinchi, S., & Arceci, R. J. (2011). Biology, risk stratification, and therapy of pediatric acute leukemias: an update. Journal of Clinical Oncology, 29, 551–565.PubMedCrossRef Pui, C.-H., Carroll, W. L., Meshinchi, S., & Arceci, R. J. (2011). Biology, risk stratification, and therapy of pediatric acute leukemias: an update. Journal of Clinical Oncology, 29, 551–565.PubMedCrossRef
4.
Zurück zum Zitat Paul, S., Kantarjian, H., & Jabbour, E. J. (2016). Adult acute lymphoblastic leukemia. Mayo Clinic Proceedings, 91, 1645–1666.PubMedCrossRef Paul, S., Kantarjian, H., & Jabbour, E. J. (2016). Adult acute lymphoblastic leukemia. Mayo Clinic Proceedings, 91, 1645–1666.PubMedCrossRef
5.
Zurück zum Zitat Chaudhury, S. S., Morison, J. K., Gibson, B. E. S., & Keeshan, K. (2015). Insights into cell ontogeny, age, and acute myeloid leukemia. Experimental Hematology, 43, 745–755.PubMedCrossRef Chaudhury, S. S., Morison, J. K., Gibson, B. E. S., & Keeshan, K. (2015). Insights into cell ontogeny, age, and acute myeloid leukemia. Experimental Hematology, 43, 745–755.PubMedCrossRef
6.
Zurück zum Zitat Micel, L. N., Tentler, J. J., Smith, P. G., & Eckhardt, G. S. (2013). Role of ubiquitin ligases and the proteasome in oncogenesis: novel targets for anticancer therapies. Journal of Clinical Oncology, 31, 1231–1238.PubMedPubMedCentralCrossRef Micel, L. N., Tentler, J. J., Smith, P. G., & Eckhardt, G. S. (2013). Role of ubiquitin ligases and the proteasome in oncogenesis: novel targets for anticancer therapies. Journal of Clinical Oncology, 31, 1231–1238.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Tasian, S. K., & Hunger, S. P. (2017). Genomic characterization of paediatric acute lymphoblastic leukaemia: an opportunity for precision medicine therapeutics. British Journal of Haematology, 176, 867–882.PubMedCrossRef Tasian, S. K., & Hunger, S. P. (2017). Genomic characterization of paediatric acute lymphoblastic leukaemia: an opportunity for precision medicine therapeutics. British Journal of Haematology, 176, 867–882.PubMedCrossRef
8.
Zurück zum Zitat Prada-Arismendy, J., Arroyave, J. C., & Röthlisberger, S. (2017). Molecular biomarkers in acute myeloid leukemia. Blood Reviews, 31, 63–76.PubMedCrossRef Prada-Arismendy, J., Arroyave, J. C., & Röthlisberger, S. (2017). Molecular biomarkers in acute myeloid leukemia. Blood Reviews, 31, 63–76.PubMedCrossRef
9.
Zurück zum Zitat Adams, J. (2004). The proteasome: a suitable antineoplastic target. Nature Reviews. Cancer, 4, 349–360.PubMedCrossRef Adams, J. (2004). The proteasome: a suitable antineoplastic target. Nature Reviews. Cancer, 4, 349–360.PubMedCrossRef
10.
Zurück zum Zitat Anderson, K. C. (2012). The 39th David A. Karnofsky Lecture: bench-to-bedside translation of targeted therapies in multiple myeloma. Journal of Clinical Oncology, 30, 445–452.PubMedPubMedCentralCrossRef Anderson, K. C. (2012). The 39th David A. Karnofsky Lecture: bench-to-bedside translation of targeted therapies in multiple myeloma. Journal of Clinical Oncology, 30, 445–452.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Kapoor, P., Ramakrishnan, V., & Rajkumar, S. V. (2012). Bortezomib combination therapy in multiple myeloma. Seminars in Hematology, 49, 228–242.PubMedPubMedCentralCrossRef Kapoor, P., Ramakrishnan, V., & Rajkumar, S. V. (2012). Bortezomib combination therapy in multiple myeloma. Seminars in Hematology, 49, 228–242.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Wright, J. J. (2010). Combination therapy of bortezomib with novel targeted agents: an emerging treatment strategy. Clinical Cancer Research, 16, 4094–4104.PubMedCrossRef Wright, J. J. (2010). Combination therapy of bortezomib with novel targeted agents: an emerging treatment strategy. Clinical Cancer Research, 16, 4094–4104.PubMedCrossRef
13.
Zurück zum Zitat Parlati, F., Lee, S. J., Aujay, M., et al. (2009). Carfilzomib can induce tumor cell death through selective inhibition of the chymotrypsin-like activity of the proteasome. Blood, 114, 3439–3447.PubMedCrossRef Parlati, F., Lee, S. J., Aujay, M., et al. (2009). Carfilzomib can induce tumor cell death through selective inhibition of the chymotrypsin-like activity of the proteasome. Blood, 114, 3439–3447.PubMedCrossRef
14.
Zurück zum Zitat Niewerth, D., Kaspers, G. J. L., Jansen, G., et al. (2016). Proteasome subunit expression analysis and chemosensitivity in relapsed paediatric acute leukaemia patients receiving bortezomib-containing chemotherapy. Journal of Hematology & Oncology, 9, 82.CrossRef Niewerth, D., Kaspers, G. J. L., Jansen, G., et al. (2016). Proteasome subunit expression analysis and chemosensitivity in relapsed paediatric acute leukaemia patients receiving bortezomib-containing chemotherapy. Journal of Hematology & Oncology, 9, 82.CrossRef
15.
Zurück zum Zitat Verbrugge, S. E., Scheper, R. J., Lems, W. F., et al. (2015). Proteasome inhibitors as experimental therapeutics of autoimmune diseases. Arthritis Research & Therapy, 17, 17.CrossRef Verbrugge, S. E., Scheper, R. J., Lems, W. F., et al. (2015). Proteasome inhibitors as experimental therapeutics of autoimmune diseases. Arthritis Research & Therapy, 17, 17.CrossRef
16.
Zurück zum Zitat Niewerth, D., Kaspers, G. J. L., Assaraf, Y. G., et al. (2014). Interferon-γ-induced upregulation of immunoproteasome subunit assembly overcomes bortezomib resistance in human hematological cell lines. Journal of Hematology & Oncology, 7, 7.CrossRef Niewerth, D., Kaspers, G. J. L., Assaraf, Y. G., et al. (2014). Interferon-γ-induced upregulation of immunoproteasome subunit assembly overcomes bortezomib resistance in human hematological cell lines. Journal of Hematology & Oncology, 7, 7.CrossRef
17.
Zurück zum Zitat Basler, M., Kirk, C. J., & Groettrup, M. (2012). The immunoproteasome in antigen processing and other immunological functions. Current Opinion in Immunology, 25, 74–80.PubMedCrossRef Basler, M., Kirk, C. J., & Groettrup, M. (2012). The immunoproteasome in antigen processing and other immunological functions. Current Opinion in Immunology, 25, 74–80.PubMedCrossRef
18.
Zurück zum Zitat Seifert, U., Bialy, L. P., Ebstein, F., et al. (2010). Immunoproteasomes preserve protein homeostasis upon interferon-induced oxidative stress. Cell, 142, 613–624.PubMedCrossRef Seifert, U., Bialy, L. P., Ebstein, F., et al. (2010). Immunoproteasomes preserve protein homeostasis upon interferon-induced oxidative stress. Cell, 142, 613–624.PubMedCrossRef
19.
Zurück zum Zitat Miller, Z., Ao, L., Kim, K. B., & Lee, W. (2013). Inhibitors of the immunoproteasome: current status and future directions. Current Pharmaceutical Design, 19, 4140–4151.PubMedPubMedCentralCrossRef Miller, Z., Ao, L., Kim, K. B., & Lee, W. (2013). Inhibitors of the immunoproteasome: current status and future directions. Current Pharmaceutical Design, 19, 4140–4151.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Ho, Y. K., Bargagna-Mohan, P., Wehenkel, M., et al. (2007). LMP2-specific inhibitors: chemical genetic tools for proteasome biology. Chemistry & Biology, 14, 419–430.CrossRef Ho, Y. K., Bargagna-Mohan, P., Wehenkel, M., et al. (2007). LMP2-specific inhibitors: chemical genetic tools for proteasome biology. Chemistry & Biology, 14, 419–430.CrossRef
21.
Zurück zum Zitat Myung, J., Kim, K. B., Lindsten, K., et al. (2001). Lack of proteasome active site allostery as revealed by subunit-specific inhibitors. Molecular Cell, 7, 411–420.PubMedCrossRef Myung, J., Kim, K. B., Lindsten, K., et al. (2001). Lack of proteasome active site allostery as revealed by subunit-specific inhibitors. Molecular Cell, 7, 411–420.PubMedCrossRef
22.
Zurück zum Zitat Kuhn, D. J., Hunsucker, S. A., Chen, Q., et al. (2009). 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, 113, 4667–4676.PubMedPubMedCentralCrossRef Kuhn, D. J., Hunsucker, S. A., Chen, Q., et al. (2009). 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, 113, 4667–4676.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat de Bruin, G., Huber, E. M., Xin, B.-T., et al. (2014). Structure-based design of β1i or β5i specific inhibitors of human immunoproteasomes. Journal of Medicinal Chemistry, 57, 6197–6209.PubMedCrossRef de Bruin, G., Huber, E. M., Xin, B.-T., et al. (2014). Structure-based design of β1i or β5i specific inhibitors of human immunoproteasomes. Journal of Medicinal Chemistry, 57, 6197–6209.PubMedCrossRef
24.
Zurück zum Zitat Kuhn, D. J., & Orlowski, R. Z. (2012). The immunoproteasome as a target in hematologic malignancies. Seminars in Hematology, 49, 258–262.PubMedCrossRef Kuhn, D. J., & Orlowski, R. Z. (2012). The immunoproteasome as a target in hematologic malignancies. Seminars in Hematology, 49, 258–262.PubMedCrossRef
25.
Zurück zum Zitat Kisselev, A. F., van der Linden, W. A., & Overkleeft, H. S. (2012). Proteasome inhibitors: an expanding army attacking a unique target. Chemistry & Biology, 19, 99–115.CrossRef Kisselev, A. F., van der Linden, W. A., & Overkleeft, H. S. (2012). Proteasome inhibitors: an expanding army attacking a unique target. Chemistry & Biology, 19, 99–115.CrossRef
26.
Zurück zum Zitat Huber, E. M., Basler, M., Schwab, R., et al. (2012). Immuno- and constitutive proteasome crystal structures reveal differences in substrate and inhibitor specificity. Cell, 148, 727–738.PubMedCrossRef Huber, E. M., Basler, M., Schwab, R., et al. (2012). Immuno- and constitutive proteasome crystal structures reveal differences in substrate and inhibitor specificity. Cell, 148, 727–738.PubMedCrossRef
27.
Zurück zum Zitat Muchamuel, T., Basler, M., Aujay, M. A., et al. (2009). A selective inhibitor of the immunoproteasome subunit LMP7 blocks cytokine production and attenuates progression of experimental arthritis. Nature Medicine, 15, 781–787.PubMedCrossRef Muchamuel, T., Basler, M., Aujay, M. A., et al. (2009). A selective inhibitor of the immunoproteasome subunit LMP7 blocks cytokine production and attenuates progression of experimental arthritis. Nature Medicine, 15, 781–787.PubMedCrossRef
28.
Zurück zum Zitat Zhou, H.-J., Aujay, M. A., Bennett, M. K., et al. (2009). Design and synthesis of an orally bioavailable and selective peptide epoxyketone proteasome inhibitor (PR-047). Journal of Medicinal Chemistry, 52, 3028–3038.PubMedCrossRef Zhou, H.-J., Aujay, M. A., Bennett, M. K., et al. (2009). Design and synthesis of an orally bioavailable and selective peptide epoxyketone proteasome inhibitor (PR-047). Journal of Medicinal Chemistry, 52, 3028–3038.PubMedCrossRef
29.
Zurück zum Zitat Niewerth, D., van Meerloo, J., Jansen, G., et al. (2014). Anti-leukemic activity and mechanisms underlying resistance to the novel immunoproteasome inhibitor PR-924. Biochemical Pharmacology, 89, 43–51.PubMedCrossRef Niewerth, D., van Meerloo, J., Jansen, G., et al. (2014). Anti-leukemic activity and mechanisms underlying resistance to the novel immunoproteasome inhibitor PR-924. Biochemical Pharmacology, 89, 43–51.PubMedCrossRef
30.
Zurück zum Zitat Singh, A. V., Bandi, M., Aujay, M. A., et al. (2011). PR-924, a selective inhibitor of the immunoproteasome subunit LMP-7, blocks multiple myeloma cell growth both in vitro and in vivo. British Journal of Haematology, 152, 155–163.PubMedCrossRef Singh, A. V., Bandi, M., Aujay, M. A., et al. (2011). PR-924, a selective inhibitor of the immunoproteasome subunit LMP-7, blocks multiple myeloma cell growth both in vitro and in vivo. British Journal of Haematology, 152, 155–163.PubMedCrossRef
31.
Zurück zum Zitat Niewerth, D., Jansen, G., Riethoff, L. F., et al. (2014). Antileukemic activity and mechanism of drug resistance to the marine Salinispora tropica proteasome inhibitor salinosporamide A (Marizomib). Molecular Pharmacology, 86, 12–19.PubMedPubMedCentralCrossRef Niewerth, D., Jansen, G., Riethoff, L. F., et al. (2014). Antileukemic activity and mechanism of drug resistance to the marine Salinispora tropica proteasome inhibitor salinosporamide A (Marizomib). Molecular Pharmacology, 86, 12–19.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Adams, J., Palombella, V. J., & Elliott, P. J. (2000). Proteasome inhibition: a new strategy in cancer treatment. Investigational New Drugs, 18, 109–121.PubMedCrossRef Adams, J., Palombella, V. J., & Elliott, P. J. (2000). Proteasome inhibition: a new strategy in cancer treatment. Investigational New Drugs, 18, 109–121.PubMedCrossRef
33.
Zurück zum Zitat Almond, J. B., & Cohen, G. M. (2002). The proteasome: a novel target for cancer chemotherapy. Leukemia, 16, 433–443.PubMedCrossRef Almond, J. B., & Cohen, G. M. (2002). The proteasome: a novel target for cancer chemotherapy. Leukemia, 16, 433–443.PubMedCrossRef
34.
Zurück zum Zitat Adams, J., Behnke, M., Chen, S., et al. (1998). Potent and selective inhibitors of the proteasome: dipeptidyl boronic acids. Bioorganic & Medicinal Chemistry Letters, 8, 333–338.CrossRef Adams, J., Behnke, M., Chen, S., et al. (1998). Potent and selective inhibitors of the proteasome: dipeptidyl boronic acids. Bioorganic & Medicinal Chemistry Letters, 8, 333–338.CrossRef
35.
Zurück zum Zitat Lee, D. H., & Goldberg, A. L. (1998). Proteasome inhibitors: valuable new tools for cell biologists. Trends in Cell Biology, 8, 397–403.PubMedCrossRef Lee, D. H., & Goldberg, A. L. (1998). Proteasome inhibitors: valuable new tools for cell biologists. Trends in Cell Biology, 8, 397–403.PubMedCrossRef
36.
Zurück zum Zitat Kisselev, A. F., & Goldberg, A. L. (2001). Proteasome inhibitors: from research tools to drug candidates. Chemistry & Biology, 8, 739–758.CrossRef Kisselev, A. F., & Goldberg, A. L. (2001). Proteasome inhibitors: from research tools to drug candidates. Chemistry & Biology, 8, 739–758.CrossRef
37.
Zurück zum Zitat Adams, J. (2000). The proteasome: structure, function, and role in the cell. Cancer Treatment Reviews, 29(Suppl 1), 3–9. Adams, J. (2000). The proteasome: structure, function, and role in the cell. Cancer Treatment Reviews, 29(Suppl 1), 3–9.
38.
Zurück zum Zitat Chauhan, D., Hideshima, T., & Anderson, K. C. (2006). A novel proteasome inhibitor NPI-0052 as an anticancer therapy. British Journal of Cancer, 95, 961–965.PubMedPubMedCentralCrossRef Chauhan, D., Hideshima, T., & Anderson, K. C. (2006). A novel proteasome inhibitor NPI-0052 as an anticancer therapy. British Journal of Cancer, 95, 961–965.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Chauhan, D., Singh, A. V., Aujay, M., et al. (2010). A novel orally active proteasome inhibitor ONX 0912 triggers in vitro and in vivo cytotoxicity in multiple myeloma. Blood, 116, 4906–4915.PubMedPubMedCentralCrossRef Chauhan, D., Singh, A. V., Aujay, M., et al. (2010). A novel orally active proteasome inhibitor ONX 0912 triggers in vitro and in vivo cytotoxicity in multiple myeloma. Blood, 116, 4906–4915.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Chauhan, D., Tian, Z., Zhou, B., et al. (2011). In vitro and in vivo selective antitumor activity of a novel orally bioavailable proteasome inhibitor MLN9708 against multiple myeloma cells. Clinical Cancer Research, 17, 5311–5321.PubMedPubMedCentralCrossRef Chauhan, D., Tian, Z., Zhou, B., et al. (2011). In vitro and in vivo selective antitumor activity of a novel orally bioavailable proteasome inhibitor MLN9708 against multiple myeloma cells. Clinical Cancer Research, 17, 5311–5321.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Kuhn, D. J., Chen, Q., Voorhees, P. M., et al. (2007). Potent activity of carfilzomib, a novel, irreversible inhibitor of the ubiquitin-proteasome pathway, against preclinical models of multiple myeloma. Blood, 110, 3281–3290.PubMedPubMedCentralCrossRef Kuhn, D. J., Chen, Q., Voorhees, P. M., et al. (2007). Potent activity of carfilzomib, a novel, irreversible inhibitor of the ubiquitin-proteasome pathway, against preclinical models of multiple myeloma. Blood, 110, 3281–3290.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Piva, R., Ruggeri, B., Williams, M., et al. (2008). CEP-18770: a novel, orally active proteasome inhibitor with a tumor-selective pharmacologic profile competitive with bortezomib. Blood, 111, 2765–2775.PubMedCrossRef Piva, R., Ruggeri, B., Williams, M., et al. (2008). CEP-18770: a novel, orally active proteasome inhibitor with a tumor-selective pharmacologic profile competitive with bortezomib. Blood, 111, 2765–2775.PubMedCrossRef
43.
Zurück zum Zitat Dick, L. R., & Fleming, P. E. (2010). Building on bortezomib: second-generation proteasome inhibitors as anti-cancer therapy. Drug Discovery Today, 15, 243–249.PubMedCrossRef Dick, L. R., & Fleming, P. E. (2010). Building on bortezomib: second-generation proteasome inhibitors as anti-cancer therapy. Drug Discovery Today, 15, 243–249.PubMedCrossRef
44.
Zurück zum Zitat Niewerth, D., Jansen, G., Assaraf, Y. G., et al. (2015). Molecular basis of resistance to proteasome inhibitors in hematological malignancies. Drug Resistance Updates, 18, 18–35.PubMedCrossRef Niewerth, D., Jansen, G., Assaraf, Y. G., et al. (2015). Molecular basis of resistance to proteasome inhibitors in hematological malignancies. Drug Resistance Updates, 18, 18–35.PubMedCrossRef
45.
Zurück zum Zitat Ruschak, A. M., Slassi, M., Kay, L. E., & Schimmer, A. D. (2011). Novel proteasome inhibitors to overcome bortezomib resistance. Journal of the National Cancer Institute, 103, 1007–1017.PubMedCrossRef Ruschak, A. M., Slassi, M., Kay, L. E., & Schimmer, A. D. (2011). Novel proteasome inhibitors to overcome bortezomib resistance. Journal of the National Cancer Institute, 103, 1007–1017.PubMedCrossRef
46.
Zurück zum Zitat Kale, A. J., & Moore, B. S. (2012). Molecular mechanisms of acquired proteasome inhibitor resistance. Journal of Medicinal Chemistry, 55, 10317–10327.PubMedPubMedCentralCrossRef Kale, A. J., & Moore, B. S. (2012). Molecular mechanisms of acquired proteasome inhibitor resistance. Journal of Medicinal Chemistry, 55, 10317–10327.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Kirk, C. J. (2012). Discovery and development of second-generation proteasome inhibitors. Seminars in Hematology, 49, 207–214.PubMedCrossRef Kirk, C. J. (2012). Discovery and development of second-generation proteasome inhibitors. Seminars in Hematology, 49, 207–214.PubMedCrossRef
48.
Zurück zum Zitat Niewerth, D., Franke, N. E., Jansen, G., et al. (2013). Higher ratio immune vs. constitutive proteasome level as novel indicator of sensitivity of pediatric acute leukemia cells to proteasome inhibitors. Haematologica, 98, 1896–1904.PubMedPubMedCentralCrossRef Niewerth, D., Franke, N. E., Jansen, G., et al. (2013). Higher ratio immune vs. constitutive proteasome level as novel indicator of sensitivity of pediatric acute leukemia cells to proteasome inhibitors. Haematologica, 98, 1896–1904.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Drexler, H. C. (1997). Activation of the cell death program by inhibition of proteasome function. Proceedings of the National Academy of Sciences of the United States of America, 94, 855–860.PubMedPubMedCentralCrossRef Drexler, H. C. (1997). Activation of the cell death program by inhibition of proteasome function. Proceedings of the National Academy of Sciences of the United States of America, 94, 855–860.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Soligo, D., Servida, F., Delia, D., et al. (2001). The apoptogenic response of human myeloid leukaemia cell lines and of normal and malignant haematopoietic progenitor cells to the proteasome inhibitor PSI. British Journal of Haematology, 113, 126–135.PubMedCrossRef Soligo, D., Servida, F., Delia, D., et al. (2001). The apoptogenic response of human myeloid leukaemia cell lines and of normal and malignant haematopoietic progenitor cells to the proteasome inhibitor PSI. British Journal of Haematology, 113, 126–135.PubMedCrossRef
51.
Zurück zum Zitat Vrana, J. A., & Grant, S. (2001). Synergistic induction of apoptosis in human leukemia cells (U937) exposed to bryostatin 1 and the proteasome inhibitor lactacystin involves dysregulation of the PKC/MAPK cascade. Blood, 97, 2105–2114.PubMedCrossRef Vrana, J. A., & Grant, S. (2001). Synergistic induction of apoptosis in human leukemia cells (U937) exposed to bryostatin 1 and the proteasome inhibitor lactacystin involves dysregulation of the PKC/MAPK cascade. Blood, 97, 2105–2114.PubMedCrossRef
52.
Zurück zum Zitat Chandra, J., Niemer, I., Gilbreath, J., et al. (1998). Proteasome inhibitors induce apoptosis in glucocorticoid-resistant chronic lymphocytic leukemic lymphocytes. Blood, 92, 4220–4229.PubMed Chandra, J., Niemer, I., Gilbreath, J., et al. (1998). Proteasome inhibitors induce apoptosis in glucocorticoid-resistant chronic lymphocytic leukemic lymphocytes. Blood, 92, 4220–4229.PubMed
53.
Zurück zum Zitat Dai, Y., Rahmani, M., & Grant, S. (2003). Proteasome inhibitors potentiate leukemic cell apoptosis induced by the cyclin-dependent kinase inhibitor flavopiridol through a SAPK/JNK- and NF-kappaB-dependent process. Oncogene, 22, 7108–7122.PubMedCrossRef Dai, Y., Rahmani, M., & Grant, S. (2003). Proteasome inhibitors potentiate leukemic cell apoptosis induced by the cyclin-dependent kinase inhibitor flavopiridol through a SAPK/JNK- and NF-kappaB-dependent process. Oncogene, 22, 7108–7122.PubMedCrossRef
54.
Zurück zum Zitat Pahler, J. C., Ruiz, S., Niemer, I., et al. (2003). Effects of the proteasome inhibitor, bortezomib, on apoptosis in isolated lymphocytes obtained from patients with chronic lymphocytic leukemia. Clinical Cancer Research, 9, 4570–4577.PubMed Pahler, J. C., Ruiz, S., Niemer, I., et al. (2003). Effects of the proteasome inhibitor, bortezomib, on apoptosis in isolated lymphocytes obtained from patients with chronic lymphocytic leukemia. Clinical Cancer Research, 9, 4570–4577.PubMed
55.
Zurück zum Zitat Dai, Y., Rahmani, M., Pei, X. Y., et al. (2004). Bortezomib and flavopiridol interact synergistically to induce apoptosis in chronic myeloid leukemia cells resistant to imatinib mesylate through both Bcr/Abl-dependent and -independent mechanisms. Blood, 104, 509–518.PubMedCrossRef Dai, Y., Rahmani, M., Pei, X. Y., et al. (2004). Bortezomib and flavopiridol interact synergistically to induce apoptosis in chronic myeloid leukemia cells resistant to imatinib mesylate through both Bcr/Abl-dependent and -independent mechanisms. Blood, 104, 509–518.PubMedCrossRef
56.
Zurück zum Zitat Duechler, M., Shehata, M., Schwarzmeier, J. D., et al. (2005). Induction of apoptosis by proteasome inhibitors in B-CLL cells is associated with downregulation of CD23 and inactivation of Notch2. Leukemia, 19, 260–267.PubMedCrossRef Duechler, M., Shehata, M., Schwarzmeier, J. D., et al. (2005). Induction of apoptosis by proteasome inhibitors in B-CLL cells is associated with downregulation of CD23 and inactivation of Notch2. Leukemia, 19, 260–267.PubMedCrossRef
57.
Zurück zum Zitat Horton, T. M., Gannavarapu, A., Blaney, S. M., et al. (2006). Bortezomib interactions with chemotherapy agents in acute leukemia in vitro. Cancer Chemotherapy and Pharmacology, 58, 13–23.PubMedCrossRef Horton, T. M., Gannavarapu, A., Blaney, S. M., et al. (2006). Bortezomib interactions with chemotherapy agents in acute leukemia in vitro. Cancer Chemotherapy and Pharmacology, 58, 13–23.PubMedCrossRef
58.
Zurück zum Zitat Yanamandra, N., Colaco, N. M., Parquet, N. A., et al. (2006). Tipifarnib and bortezomib are synergistic and overcome cell adhesion-mediated drug resistance in multiple myeloma and acute myeloid leukemia. Clinical Cancer Research, 12, 591–599.PubMedCrossRef Yanamandra, N., Colaco, N. M., Parquet, N. A., et al. (2006). Tipifarnib and bortezomib are synergistic and overcome cell adhesion-mediated drug resistance in multiple myeloma and acute myeloid leukemia. Clinical Cancer Research, 12, 591–599.PubMedCrossRef
59.
Zurück zum Zitat Houghton, P. J., Morton, C. L., Kolb, E. A., et al. (2008). Initial testing (stage 1) of the proteasome inhibitor bortezomib by the pediatric preclinical testing program. Pediatric Blood & Cancer, 50, 37–45.CrossRef Houghton, P. J., Morton, C. L., Kolb, E. A., et al. (2008). Initial testing (stage 1) of the proteasome inhibitor bortezomib by the pediatric preclinical testing program. Pediatric Blood & Cancer, 50, 37–45.CrossRef
60.
Zurück zum Zitat Servida, F., Soligo, D., Delia, D., et al. (2005). Sensitivity of human multiple myelomas and myeloid leukemias to the proteasome inhibitor I. Leukemia, 19, 2324–2331.PubMedCrossRef Servida, F., Soligo, D., Delia, D., et al. (2005). Sensitivity of human multiple myelomas and myeloid leukemias to the proteasome inhibitor I. Leukemia, 19, 2324–2331.PubMedCrossRef
61.
Zurück zum Zitat Stapnes, C., Doskeland, A. P., Hatfield, K., et al. (2007). The proteasome inhibitors bortezomib and PR-171 have antiproliferative and proapoptotic effects on primary human acute myeloid leukaemia cells. British Journal of Haematology, 136, 814–828.PubMedCrossRef Stapnes, C., Doskeland, A. P., Hatfield, K., et al. (2007). The proteasome inhibitors bortezomib and PR-171 have antiproliferative and proapoptotic effects on primary human acute myeloid leukaemia cells. British Journal of Haematology, 136, 814–828.PubMedCrossRef
62.
Zurück zum Zitat Kraus, M., Muller-Ide, H., Ruckrich, T., et al. (2014). Ritonavir, nelfinavir, saquinavir and lopinavir induce proteotoxic stress in acute myeloid leukemia cells and sensitize them for proteasome inhibitor treatment at low micromolar drug concentrations. Leukemia Research, 38, 383–392.PubMedCrossRef Kraus, M., Muller-Ide, H., Ruckrich, T., et al. (2014). Ritonavir, nelfinavir, saquinavir and lopinavir induce proteotoxic stress in acute myeloid leukemia cells and sensitize them for proteasome inhibitor treatment at low micromolar drug concentrations. Leukemia Research, 38, 383–392.PubMedCrossRef
63.
Zurück zum Zitat Liu, H., Westergard, T. D., Cashen, A., et al. (2014). Proteasome inhibitors evoke latent tumor suppression programs in pro-B MLL leukemias through MLL-AF4. Cancer Cell, 25, 530–542.PubMedPubMedCentralCrossRef Liu, H., Westergard, T. D., Cashen, A., et al. (2014). Proteasome inhibitors evoke latent tumor suppression programs in pro-B MLL leukemias through MLL-AF4. Cancer Cell, 25, 530–542.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Miller, C. P., Rudra, S., Keating, M. J., et al. (2009). Caspase-8 dependent histone acetylation by a novel proteasome inhibitor, NPI-0052: a mechanism for synergy in leukemia cells. Blood, 113, 4289–4299.PubMedPubMedCentralCrossRef Miller, C. P., Rudra, S., Keating, M. J., et al. (2009). Caspase-8 dependent histone acetylation by a novel proteasome inhibitor, NPI-0052: a mechanism for synergy in leukemia cells. Blood, 113, 4289–4299.PubMedPubMedCentralCrossRef
65.
Zurück zum Zitat Miller, C. P., Ban, K., Dujka, M. E., et al. (2007). NPI-0052, a novel proteasome inhibitor, induces caspase-8 and ROS-dependent apoptosis alone and in combination with HDAC inhibitors in leukemia cells. Blood, 110, 267–277.PubMedPubMedCentralCrossRef Miller, C. P., Ban, K., Dujka, M. E., et al. (2007). NPI-0052, a novel proteasome inhibitor, induces caspase-8 and ROS-dependent apoptosis alone and in combination with HDAC inhibitors in leukemia cells. Blood, 110, 267–277.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Paulus, A., Masood, A., Miller, K. C., et al. (2014). The investigational agent MLN2238 induces apoptosis and is cytotoxic to CLL cells in vitro, as a single agent and in combination with other drugs. British Journal of Haematology, 165, 78–88.PubMedCrossRef Paulus, A., Masood, A., Miller, K. C., et al. (2014). The investigational agent MLN2238 induces apoptosis and is cytotoxic to CLL cells in vitro, as a single agent and in combination with other drugs. British Journal of Haematology, 165, 78–88.PubMedCrossRef
67.
Zurück zum Zitat Franke, N. E., Vink, J., Cloos, J., & Kaspers, G. J. (2008). Proteasome and protease inhibitors. In G. J. Kaspers, B. Coiffier, M. Heinrich, & E. Estey (Eds.), Innov. Leuk. Lymphoma Ther (pp. 469–489). New York: Informa Healthcare USA, Inc.. Franke, N. E., Vink, J., Cloos, J., & Kaspers, G. J. (2008). Proteasome and protease inhibitors. In G. J. Kaspers, B. Coiffier, M. Heinrich, & E. Estey (Eds.), Innov. Leuk. Lymphoma Ther (pp. 469–489). New York: Informa Healthcare USA, Inc..
68.
Zurück zum Zitat Oerlemans, R., Franke, N. E., Assaraf, Y. G., et al. (2008). Molecular basis of bortezomib resistance: proteasome subunit beta5 (PSMB5) gene mutation and overexpression of PSMB5 protein. Blood, 112, 2489–2499.PubMedCrossRef Oerlemans, R., Franke, N. E., Assaraf, Y. G., et al. (2008). Molecular basis of bortezomib resistance: proteasome subunit beta5 (PSMB5) gene mutation and overexpression of PSMB5 protein. Blood, 112, 2489–2499.PubMedCrossRef
69.
Zurück zum Zitat Lu, S., Yang, J., Chen, Z., et al. (2009). Different mutants of PSMB5 confer varying bortezomib resistance in T lymphoblastic lymphoma/leukemia cells derived from the Jurkat cell line. Experimental Hematology, 37, 831–837.PubMedCrossRef Lu, S., Yang, J., Chen, Z., et al. (2009). Different mutants of PSMB5 confer varying bortezomib resistance in T lymphoblastic lymphoma/leukemia cells derived from the Jurkat cell line. Experimental Hematology, 37, 831–837.PubMedCrossRef
70.
Zurück zum Zitat Ri, M., Iida, S., Nakashima, T., et al. (2010). Bortezomib-resistant myeloma cell lines: a role for mutated PSMB5 in preventing the accumulation of unfolded proteins and fatal ER stress. Leukemia, 24, 1506–1512.PubMedCrossRef Ri, M., Iida, S., Nakashima, T., et al. (2010). Bortezomib-resistant myeloma cell lines: a role for mutated PSMB5 in preventing the accumulation of unfolded proteins and fatal ER stress. Leukemia, 24, 1506–1512.PubMedCrossRef
71.
Zurück zum Zitat Franke, N. E., Niewerth, D., Assaraf, Y. G., et al. (2012). Impaired bortezomib binding to mutant beta5 subunit of the proteasome is the underlying basis for bortezomib resistance in leukemia cells. Leukemia, 26, 757–768.PubMedCrossRef Franke, N. E., Niewerth, D., Assaraf, Y. G., et al. (2012). Impaired bortezomib binding to mutant beta5 subunit of the proteasome is the underlying basis for bortezomib resistance in leukemia cells. Leukemia, 26, 757–768.PubMedCrossRef
72.
Zurück zum Zitat McConkey, D. J., & Zhu, K. (2008). Mechanisms of proteasome inhibitor action and resistance in cancer. Drug Resistance Updates, 11, 164–179.PubMedCrossRef McConkey, D. J., & Zhu, K. (2008). Mechanisms of proteasome inhibitor action and resistance in cancer. Drug Resistance Updates, 11, 164–179.PubMedCrossRef
73.
Zurück zum Zitat Franke, N. E., Kaspers, G. L., Assaraf, Y. G., et al. (2014). Exocytosis of polyubiquitinated proteins in bortezomib-resistant leukemia cells: a role for MARCKS in acquired resistance to proteasome inhibitors. Oncotarget, 7, 74779–74796. Franke, N. E., Kaspers, G. L., Assaraf, Y. G., et al. (2014). Exocytosis of polyubiquitinated proteins in bortezomib-resistant leukemia cells: a role for MARCKS in acquired resistance to proteasome inhibitors. Oncotarget, 7, 74779–74796.
74.
Zurück zum Zitat Busse, A., Kraus, M., Na, I. K., et al. (2008). Sensitivity of tumor cells to proteasome inhibitors is associated with expression levels and composition of proteasome subunits. Cancer, 112, 659–670.PubMedCrossRef Busse, A., Kraus, M., Na, I. K., et al. (2008). Sensitivity of tumor cells to proteasome inhibitors is associated with expression levels and composition of proteasome subunits. Cancer, 112, 659–670.PubMedCrossRef
75.
Zurück zum Zitat Fuchs, D., Berges, C., Opelz, G., et al. (2008). Increased expression and altered subunit composition of proteasomes induced by continuous proteasome inhibition establish apoptosis resistance and hyperproliferation of Burkitt lymphoma cells. Journal of Cellular Biochemistry, 103, 270–283.PubMedCrossRef Fuchs, D., Berges, C., Opelz, G., et al. (2008). Increased expression and altered subunit composition of proteasomes induced by continuous proteasome inhibition establish apoptosis resistance and hyperproliferation of Burkitt lymphoma cells. Journal of Cellular Biochemistry, 103, 270–283.PubMedCrossRef
76.
Zurück zum Zitat Frisan, T., Levitsky, V., & Masucci, M. G. (2000). Variations in proteasome subunit composition and enzymatic activity in B-lymphoma lines and normal B cells. International Journal of Cancer, 88, 881–888.PubMedCrossRef Frisan, T., Levitsky, V., & Masucci, M. G. (2000). Variations in proteasome subunit composition and enzymatic activity in B-lymphoma lines and normal B cells. International Journal of Cancer, 88, 881–888.PubMedCrossRef
77.
Zurück zum Zitat Lu, S., Chen, Z., Yang, J., et al. (2008). Overexpression of the PSMB5 gene contributes to bortezomib resistance in T-lymphoblastic lymphoma/leukemia cells derived from Jurkat line. Experimental Hematology, 36, 1278–1284.PubMedCrossRef Lu, S., Chen, Z., Yang, J., et al. (2008). Overexpression of the PSMB5 gene contributes to bortezomib resistance in T-lymphoblastic lymphoma/leukemia cells derived from Jurkat line. Experimental Hematology, 36, 1278–1284.PubMedCrossRef
78.
Zurück zum Zitat Verbrugge, S. E., Al, M., Assaraf, Y. G., et al. (2013). Overcoming bortezomib resistance in human B cells by anti-CD20 / rituximab-mediated complement-dependent cytotoxicity and epoxyketone-based irreversible proteasome inhibitors. Experimental Hematology & Oncology, 2, 2–12.CrossRef Verbrugge, S. E., Al, M., Assaraf, Y. G., et al. (2013). Overcoming bortezomib resistance in human B cells by anti-CD20 / rituximab-mediated complement-dependent cytotoxicity and epoxyketone-based irreversible proteasome inhibitors. Experimental Hematology & Oncology, 2, 2–12.CrossRef
79.
Zurück zum Zitat de Wilt, L. H. A. M., Jansen, G., Assaraf, Y. G., et al. (2012). Proteasome-based mechanisms of intrinsic and acquired bortezomib resistance in non-small cell lung cancer. Biochemical Pharmacology, 83, 207–217.PubMedCrossRef de Wilt, L. H. A. M., Jansen, G., Assaraf, Y. G., et al. (2012). Proteasome-based mechanisms of intrinsic and acquired bortezomib resistance in non-small cell lung cancer. Biochemical Pharmacology, 83, 207–217.PubMedCrossRef
80.
Zurück zum Zitat Kale, A. J., McGlinchey, R. P., Lechner, A., & Moore, B. S. (2011). Bacterial self-resistance to the natural proteasome inhibitor salinosporamide A. ACS Chemical Biology, 6, 1257–1264.PubMedPubMedCentralCrossRef Kale, A. J., McGlinchey, R. P., Lechner, A., & Moore, B. S. (2011). Bacterial self-resistance to the natural proteasome inhibitor salinosporamide A. ACS Chemical Biology, 6, 1257–1264.PubMedPubMedCentralCrossRef
81.
82.
Zurück zum Zitat Groll, M., Berkers, C. R., Ploegh, H. L., & Ovaa, H. (2006). Crystal structure of the boronic acid-based proteasome inhibitor bortezomib in complex with the yeast 20S proteasome. Structure, 14, 451–456.PubMedCrossRef Groll, M., Berkers, C. R., Ploegh, H. L., & Ovaa, H. (2006). Crystal structure of the boronic acid-based proteasome inhibitor bortezomib in complex with the yeast 20S proteasome. Structure, 14, 451–456.PubMedCrossRef
83.
Zurück zum Zitat Adler, K. B., Tuvim, M. J., & Dickey, B. F. (2013). Regulated mucin secretion from airway epithelial cells. Front Endocrinol (Lausanne), 4, 129. Adler, K. B., Tuvim, M. J., & Dickey, B. F. (2013). Regulated mucin secretion from airway epithelial cells. Front Endocrinol (Lausanne), 4, 129.
84.
Zurück zum Zitat Buschow, S. I., Liefhebber, J. M. P., Wubbolts, R., & Stoorvogel, W. (2005). Exosomes contain ubiquitinated proteins. Blood Cells Mol Dis, 35, 398–403.PubMedCrossRef Buschow, S. I., Liefhebber, J. M. P., Wubbolts, R., & Stoorvogel, W. (2005). Exosomes contain ubiquitinated proteins. Blood Cells Mol Dis, 35, 398–403.PubMedCrossRef
85.
Zurück zum Zitat David, E., Kaufman, J. L., Flowers, C. R., et al. (2010). Tipifarnib sensitizes cells to proteasome inhibition by blocking degradation of bortezomib-induced aggresomes. Blood, 116, 5285–5288.PubMedPubMedCentralCrossRef David, E., Kaufman, J. L., Flowers, C. R., et al. (2010). Tipifarnib sensitizes cells to proteasome inhibition by blocking degradation of bortezomib-induced aggresomes. Blood, 116, 5285–5288.PubMedPubMedCentralCrossRef
86.
Zurück zum Zitat Escalante, A. M., McGrath, R. T., Karolak, M. R., et al. (2013). Preventing the autophagic survival response by inhibition of calpain enhances the cytotoxic activity of bortezomib in vitro and in vivo. Cancer Chemotherapy and Pharmacology, 71, 1567–1576.PubMedPubMedCentralCrossRef Escalante, A. M., McGrath, R. T., Karolak, M. R., et al. (2013). Preventing the autophagic survival response by inhibition of calpain enhances the cytotoxic activity of bortezomib in vitro and in vivo. Cancer Chemotherapy and Pharmacology, 71, 1567–1576.PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Hamouda, M.-A., Belhacene, N., Puissant, A., et al. (2014). The small heat shock protein B8 (HSPB8) confers resistance to bortezomib by promoting autophagic removal of misfolded proteins in multiple myeloma cells. Oncotarget, 5, 6252–6266.PubMedPubMedCentralCrossRef Hamouda, M.-A., Belhacene, N., Puissant, A., et al. (2014). The small heat shock protein B8 (HSPB8) confers resistance to bortezomib by promoting autophagic removal of misfolded proteins in multiple myeloma cells. Oncotarget, 5, 6252–6266.PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat Hoang, B., Benavides, A., Shi, Y., et al. (2009). Effect of autophagy on multiple myeloma cell viability. Molecular Cancer Therapeutics, 8, 1974–1984.PubMedCrossRef Hoang, B., Benavides, A., Shi, Y., et al. (2009). Effect of autophagy on multiple myeloma cell viability. Molecular Cancer Therapeutics, 8, 1974–1984.PubMedCrossRef
89.
Zurück zum Zitat Milani, M., Rzymski, T., Mellor, H. R., et al. (2009). The role of ATF4 stabilization and autophagy in resistance of breast cancer cells treated with bortezomib. Cancer Research, 69, 4415–4423.PubMedCrossRef Milani, M., Rzymski, T., Mellor, H. R., et al. (2009). The role of ATF4 stabilization and autophagy in resistance of breast cancer cells treated with bortezomib. Cancer Research, 69, 4415–4423.PubMedCrossRef
90.
Zurück zum Zitat Chen, S., Zhang, Y., Zhou, L., et al. (2014). A Bim-targeting strategy overcomes adaptive bortezomib resistance in myeloma through a novel link between autophagy and apoptosis. Blood, 124, 2687–2697.PubMedPubMedCentralCrossRef Chen, S., Zhang, Y., Zhou, L., et al. (2014). A Bim-targeting strategy overcomes adaptive bortezomib resistance in myeloma through a novel link between autophagy and apoptosis. Blood, 124, 2687–2697.PubMedPubMedCentralCrossRef
91.
Zurück zum Zitat Milan, E., Perini, T., Resnati, M., et al. (2015). A plastic SQSTM1/p62-dependent autophagic reserve maintains proteostasis and determines proteasome inhibitor susceptibility in multiple myeloma cells. Autophagy, 11, 1161–1178.PubMedPubMedCentralCrossRef Milan, E., Perini, T., Resnati, M., et al. (2015). A plastic SQSTM1/p62-dependent autophagic reserve maintains proteostasis and determines proteasome inhibitor susceptibility in multiple myeloma cells. Autophagy, 11, 1161–1178.PubMedPubMedCentralCrossRef
92.
Zurück zum Zitat Jarauta, V., Jaime, P., Gonzalo, O., et al. (2016). Inhibition of autophagy with chloroquine potentiates carfilzomib-induced apoptosis in myeloma cells in vitro and in vivo. Cancer Letters, 382, 1–10.PubMedCrossRef Jarauta, V., Jaime, P., Gonzalo, O., et al. (2016). Inhibition of autophagy with chloroquine potentiates carfilzomib-induced apoptosis in myeloma cells in vitro and in vivo. Cancer Letters, 382, 1–10.PubMedCrossRef
93.
Zurück zum Zitat Baranowska, K., Misund, K., Starheim, K. K., et al. (2016). Hydroxychloroquine potentiates carfilzomib toxicity towards myeloma cells. Oncotarget, 7, 70845–70856.PubMedPubMedCentral Baranowska, K., Misund, K., Starheim, K. K., et al. (2016). Hydroxychloroquine potentiates carfilzomib toxicity towards myeloma cells. Oncotarget, 7, 70845–70856.PubMedPubMedCentral
94.
Zurück zum Zitat Hideshima, T., Bradner, J. E., Wong, J., et al. (2005). Small-molecule inhibition of proteasome and aggresome function induces synergistic antitumor activity in multiple myeloma. Proceedings of the National Academy of Sciences, 102, 8567–8572.CrossRef Hideshima, T., Bradner, J. E., Wong, J., et al. (2005). Small-molecule inhibition of proteasome and aggresome function induces synergistic antitumor activity in multiple myeloma. Proceedings of the National Academy of Sciences, 102, 8567–8572.CrossRef
95.
Zurück zum Zitat Nawrocki, S. T., Carew, J. S., Pino, M. S., et al. (2006). Aggresome disruption: a novel strategy to enhance bortezomib-induced apoptosis in pancreatic cancer cells. Cancer Research, 66, 3773–3781.PubMedCrossRef Nawrocki, S. T., Carew, J. S., Pino, M. S., et al. (2006). Aggresome disruption: a novel strategy to enhance bortezomib-induced apoptosis in pancreatic cancer cells. Cancer Research, 66, 3773–3781.PubMedCrossRef
96.
Zurück zum Zitat Sriram, S. M., Han, D. H., & Kim, S. T. (2011). Partners in crime: ubiquitin-mediated degradation and autophagy. Science Signaling, 4, jc4.PubMedCrossRef Sriram, S. M., Han, D. H., & Kim, S. T. (2011). Partners in crime: ubiquitin-mediated degradation and autophagy. Science Signaling, 4, jc4.PubMedCrossRef
97.
Zurück zum Zitat Fang, J., Rhyasen, G., Bolanos, L., et al. (2012). Cytotoxic effects of bortezomib in myelodysplastic syndrome/acute myeloid leukemia depend on autophagy-mediated lysosomal degradation of TRAF6 and repression of PSMA1. Blood, 120, 858–867.PubMedPubMedCentralCrossRef Fang, J., Rhyasen, G., Bolanos, L., et al. (2012). Cytotoxic effects of bortezomib in myelodysplastic syndrome/acute myeloid leukemia depend on autophagy-mediated lysosomal degradation of TRAF6 and repression of PSMA1. Blood, 120, 858–867.PubMedPubMedCentralCrossRef
98.
Zurück zum Zitat Zhou, J., Ching, Y. Q., & Chng, W.-J. (2015). Aberrant nuclear factor-kappa B activity in acute myeloid leukemia: from molecular pathogenesis to therapeutic target. Oncotarget, 6, 5490–5500.PubMedPubMedCentralCrossRef Zhou, J., Ching, Y. Q., & Chng, W.-J. (2015). Aberrant nuclear factor-kappa B activity in acute myeloid leukemia: from molecular pathogenesis to therapeutic target. Oncotarget, 6, 5490–5500.PubMedPubMedCentralCrossRef
99.
Zurück zum Zitat Bosman, M. C. J., Schuringa, J. J., Quax, W. J., & Vellenga, E. (2013). Bortezomib sensitivity of acute myeloid leukemia CD34(+) cells can be enhanced by targeting the persisting activity of NF-κB and the accumulation of MCL-1. Experimental Hematology, 41, 530–538.e1.PubMedCrossRef Bosman, M. C. J., Schuringa, J. J., Quax, W. J., & Vellenga, E. (2013). Bortezomib sensitivity of acute myeloid leukemia CD34(+) cells can be enhanced by targeting the persisting activity of NF-κB and the accumulation of MCL-1. Experimental Hematology, 41, 530–538.e1.PubMedCrossRef
100.
Zurück zum Zitat Markovina, S., Callander, N. S., O’Connor, S. L., et al. (2010). Bone marrow stromal cells from multiple myeloma patients uniquely induce bortezomib resistant NF-κB activity in myeloma cells. Molecular Cancer, 9, 176.PubMedPubMedCentralCrossRef Markovina, S., Callander, N. S., O’Connor, S. L., et al. (2010). Bone marrow stromal cells from multiple myeloma patients uniquely induce bortezomib resistant NF-κB activity in myeloma cells. Molecular Cancer, 9, 176.PubMedPubMedCentralCrossRef
101.
Zurück zum Zitat Markovina, S., Callander, N. S., O’Connor, S. L., et al. (2008). Bortezomib-resistant nuclear factor-κB activity in multiple myeloma cells. Molecular Cancer Research, 6, 1356–1364.PubMedPubMedCentralCrossRef Markovina, S., Callander, N. S., O’Connor, S. L., et al. (2008). Bortezomib-resistant nuclear factor-κB activity in multiple myeloma cells. Molecular Cancer Research, 6, 1356–1364.PubMedPubMedCentralCrossRef
102.
Zurück zum Zitat Hideshima, T., Ikeda, H., Chauhan, D., et al. (2009). Bortezomib induces canonical nuclear factor-κB activation in multiple myeloma cells. Blood, 114, 1046–1052.PubMedPubMedCentralCrossRef Hideshima, T., Ikeda, H., Chauhan, D., et al. (2009). Bortezomib induces canonical nuclear factor-κB activation in multiple myeloma cells. Blood, 114, 1046–1052.PubMedPubMedCentralCrossRef
103.
Zurück zum Zitat Jung, H. J., Chen, Z., Fayad, L., et al. (2012). Bortezomib-resistant nuclear factor κB expression in stem-like cells in mantle cell lymphoma. Experimental Hematology, 40, 107–118.e2.PubMedCrossRef Jung, H. J., Chen, Z., Fayad, L., et al. (2012). Bortezomib-resistant nuclear factor κB expression in stem-like cells in mantle cell lymphoma. Experimental Hematology, 40, 107–118.e2.PubMedCrossRef
104.
Zurück zum Zitat Yang, D. T., Young, K. H., Kahl, B. S., et al. (2008). Prevalence of bortezomib-resistant constitutive NF-kappaB activity in mantle cell lymphoma. Molecular Cancer, 7, 40.PubMedPubMedCentralCrossRef Yang, D. T., Young, K. H., Kahl, B. S., et al. (2008). Prevalence of bortezomib-resistant constitutive NF-kappaB activity in mantle cell lymphoma. Molecular Cancer, 7, 40.PubMedPubMedCentralCrossRef
105.
Zurück zum Zitat Kim, A., Park, S., Lee, J.-E., et al. (2012). The dual PI3K and mTOR inhibitor NVP-BEZ235 exhibits anti-proliferative activity and overcomes bortezomib resistance in mantle cell lymphoma cells. Leukemia Research, 36, 912–920.PubMedCrossRef Kim, A., Park, S., Lee, J.-E., et al. (2012). The dual PI3K and mTOR inhibitor NVP-BEZ235 exhibits anti-proliferative activity and overcomes bortezomib resistance in mantle cell lymphoma cells. Leukemia Research, 36, 912–920.PubMedCrossRef
106.
Zurück zum Zitat Liu, C.-Y., Shiau, C.-W., Kuo, H.-Y., et al. (2013). Cancerous inhibitor of protein phosphatase 2A determines bortezomib-induced apoptosis in leukemia cells. Haematologica, 98, 729–738.PubMedPubMedCentralCrossRef Liu, C.-Y., Shiau, C.-W., Kuo, H.-Y., et al. (2013). Cancerous inhibitor of protein phosphatase 2A determines bortezomib-induced apoptosis in leukemia cells. Haematologica, 98, 729–738.PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat Que, W., Chen, J., Chuang, M., & Jiang, D. (2012). Knockdown of c-Met enhances sensitivity to bortezomib in human multiple myeloma U266 cells via inhibiting Akt/mTOR activity. APMIS, 120, 195–203.PubMedCrossRef Que, W., Chen, J., Chuang, M., & Jiang, D. (2012). Knockdown of c-Met enhances sensitivity to bortezomib in human multiple myeloma U266 cells via inhibiting Akt/mTOR activity. APMIS, 120, 195–203.PubMedCrossRef
108.
Zurück zum Zitat Kuhn, D. J., Berkova, Z., Jones, R. J., et al. (2012). Targeting the insulin-like growth factor-1 receptor to overcome bortezomib resistance in preclinical models of multiple myeloma. Blood, 120, 3260–3270.PubMedPubMedCentralCrossRef Kuhn, D. J., Berkova, Z., Jones, R. J., et al. (2012). Targeting the insulin-like growth factor-1 receptor to overcome bortezomib resistance in preclinical models of multiple myeloma. Blood, 120, 3260–3270.PubMedPubMedCentralCrossRef
109.
Zurück zum Zitat Maiso, P., Ocio, E. M., Garayoa, M., et al. (2008). The insulin-like growth factor-I receptor inhibitor NVP-AEW541 provokes cell cycle arrest and apoptosis in multiple myeloma cells. British Journal of Haematology, 141, 470–482.PubMedCrossRef Maiso, P., Ocio, E. M., Garayoa, M., et al. (2008). The insulin-like growth factor-I receptor inhibitor NVP-AEW541 provokes cell cycle arrest and apoptosis in multiple myeloma cells. British Journal of Haematology, 141, 470–482.PubMedCrossRef
110.
Zurück zum Zitat Wu, K.-D., Zhou, L., Burtrum, D., et al. (2006). Antibody targeting of the insulin-like growth factor I receptor enhances the anti-tumor response of multiple myeloma to chemotherapy through inhibition of tumor proliferation and angiogenesis. Cancer Immunology, Immunotherapy, 56, 343–357.PubMedCrossRef Wu, K.-D., Zhou, L., Burtrum, D., et al. (2006). Antibody targeting of the insulin-like growth factor I receptor enhances the anti-tumor response of multiple myeloma to chemotherapy through inhibition of tumor proliferation and angiogenesis. Cancer Immunology, Immunotherapy, 56, 343–357.PubMedCrossRef
111.
Zurück zum Zitat Leung-Hagesteijn, C., Erdmann, N., Cheung, G., et al. (2013). Xbp1s-negative tumor B cells and pre-plasmablasts mediate therapeutic proteasome inhibitor resistance in multiple myeloma. Cancer Cell, 24, 289–304.PubMedPubMedCentralCrossRef Leung-Hagesteijn, C., Erdmann, N., Cheung, G., et al. (2013). Xbp1s-negative tumor B cells and pre-plasmablasts mediate therapeutic proteasome inhibitor resistance in multiple myeloma. Cancer Cell, 24, 289–304.PubMedPubMedCentralCrossRef
112.
Zurück zum Zitat Gambella, M., Rocci, A., Passera, R., et al. (2014). High XBP1 expression is a marker of better outcome in multiple myeloma patients treated with bortezomib. Haematologica, 99, e14–e16.PubMedPubMedCentralCrossRef Gambella, M., Rocci, A., Passera, R., et al. (2014). High XBP1 expression is a marker of better outcome in multiple myeloma patients treated with bortezomib. Haematologica, 99, e14–e16.PubMedPubMedCentralCrossRef
113.
Zurück zum Zitat Minderman, H., Zhou, Y., O’Loughlin, K. L., & Baer, M. R. (2007). Bortezomib activity and in vitro interactions with anthracyclines and cytarabine in acute myeloid leukemia cells are independent of multidrug resistance mechanisms and p53 status. Cancer Chemotherapy and Pharmacology, 60, 245–255.PubMedCrossRef Minderman, H., Zhou, Y., O’Loughlin, K. L., & Baer, M. R. (2007). Bortezomib activity and in vitro interactions with anthracyclines and cytarabine in acute myeloid leukemia cells are independent of multidrug resistance mechanisms and p53 status. Cancer Chemotherapy and Pharmacology, 60, 245–255.PubMedCrossRef
114.
Zurück zum Zitat Verbrugge, S. E., Assaraf, Y. G., Dijkmans, B. A., et al. (2012). 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. The Journal of Pharmacology and Experimental Therapeutics, 341, 174–182.PubMedCrossRef Verbrugge, S. E., Assaraf, Y. G., Dijkmans, B. A., et al. (2012). 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. The Journal of Pharmacology and Experimental Therapeutics, 341, 174–182.PubMedCrossRef
115.
Zurück zum Zitat Ao, L., Wu, Y., Kim, D., et al. (2012). Development of peptide-based reversing agents for p-glycoprotein-mediated resistance to carfilzomib. Molecular Pharmaceutics, 9, 2197–2205.PubMedPubMedCentralCrossRef Ao, L., Wu, Y., Kim, D., et al. (2012). Development of peptide-based reversing agents for p-glycoprotein-mediated resistance to carfilzomib. Molecular Pharmaceutics, 9, 2197–2205.PubMedPubMedCentralCrossRef
116.
Zurück zum Zitat O’Connor, R., Ooi, M. G., Meiller, J., et al. (2013). The interaction of bortezomib with multidrug transporters: implications for therapeutic applications in advanced multiple myeloma and other neoplasias. Cancer Chemotherapy and Pharmacology, 71, 1357–1368.PubMedPubMedCentralCrossRef O’Connor, R., Ooi, M. G., Meiller, J., et al. (2013). The interaction of bortezomib with multidrug transporters: implications for therapeutic applications in advanced multiple myeloma and other neoplasias. Cancer Chemotherapy and Pharmacology, 71, 1357–1368.PubMedPubMedCentralCrossRef
117.
Zurück zum Zitat Shaughnessy Jr., J. D., Qu, P., Usmani, S., et al. (2011). Pharmacogenomics of bortezomib test-dosing identifies hyperexpression of proteasome genes, especially PSMD4, as novel high-risk feature in myeloma treated with total therapy 3. Blood, 118, 3512–3524.PubMedPubMedCentralCrossRef Shaughnessy Jr., J. D., Qu, P., Usmani, S., et al. (2011). Pharmacogenomics of bortezomib test-dosing identifies hyperexpression of proteasome genes, especially PSMD4, as novel high-risk feature in myeloma treated with total therapy 3. Blood, 118, 3512–3524.PubMedPubMedCentralCrossRef
118.
Zurück zum Zitat Shuqing, L., Jianmin, Y., Chongmei, H., et al. (2011). Upregulated expression of the PSMB5 gene may contribute to drug resistance in patient with multiple myeloma when treated with bortezomib-based regimen. Experimental Hematology, 39, 1117–1118.PubMedCrossRef Shuqing, L., Jianmin, Y., Chongmei, H., et al. (2011). Upregulated expression of the PSMB5 gene may contribute to drug resistance in patient with multiple myeloma when treated with bortezomib-based regimen. Experimental Hematology, 39, 1117–1118.PubMedCrossRef
119.
Zurück zum Zitat Weniger, M. A., Rizzatti, E. G., Perez-Galan, P., et al. (2011). Treatment-induced oxidative stress and cellular antioxidant capacity determine response to bortezomib in mantle cell lymphoma. Clinical Cancer Research, 17, 5101–5112.PubMedPubMedCentralCrossRef Weniger, M. A., Rizzatti, E. G., Perez-Galan, P., et al. (2011). Treatment-induced oxidative stress and cellular antioxidant capacity determine response to bortezomib in mantle cell lymphoma. Clinical Cancer Research, 17, 5101–5112.PubMedPubMedCentralCrossRef
120.
Zurück zum Zitat Matondo, M., Bousquet-Dubouch, M. P., Gallay, N., et al. (2010). Proteasome inhibitor-induced apoptosis in acute myeloid leukemia: a correlation with the proteasome status. Leukemia Research, 34, 498–506.PubMedCrossRef Matondo, M., Bousquet-Dubouch, M. P., Gallay, N., et al. (2010). Proteasome inhibitor-induced apoptosis in acute myeloid leukemia: a correlation with the proteasome status. Leukemia Research, 34, 498–506.PubMedCrossRef
121.
Zurück zum Zitat Wang, L., Kumar, S., Fridley, B. L., et al. (2008). Proteasome beta subunit pharmacogenomics: gene resequencing and functional genomics. Clinical Cancer Research, 14, 3503–3513.PubMedPubMedCentralCrossRef Wang, L., Kumar, S., Fridley, B. L., et al. (2008). Proteasome beta subunit pharmacogenomics: gene resequencing and functional genomics. Clinical Cancer Research, 14, 3503–3513.PubMedPubMedCentralCrossRef
122.
Zurück zum Zitat Lichter, D. I., Danaee, H., Pickard, M. D., et al. (2012). Sequence analysis of β-subunit genes of the 20S proteasome in patients with relapsed multiple myeloma treated with bortezomib or dexamethasone. Blood, 120, 4513–4516.PubMedPubMedCentralCrossRef Lichter, D. I., Danaee, H., Pickard, M. D., et al. (2012). Sequence analysis of β-subunit genes of the 20S proteasome in patients with relapsed multiple myeloma treated with bortezomib or dexamethasone. Blood, 120, 4513–4516.PubMedPubMedCentralCrossRef
123.
Zurück zum Zitat Barrio, S., Stühmer, T., Teufel, E., et al. (2016). Parallel evolution of multiple PSMB5 mutations in a myeloma patient treated with bortezomib. Blood, 128, 3282. Barrio, S., Stühmer, T., Teufel, E., et al. (2016). Parallel evolution of multiple PSMB5 mutations in a myeloma patient treated with bortezomib. Blood, 128, 3282.
124.
Zurück zum Zitat Bachas, C., Schuurhuis, G. J., Reinhardt, D., et al. (2014). Clinical relevance of molecular aberrations in paediatric acute myeloid leukaemia at first relapse. British Journal of Haematology, 166, 902–910.PubMedCrossRef Bachas, C., Schuurhuis, G. J., Reinhardt, D., et al. (2014). Clinical relevance of molecular aberrations in paediatric acute myeloid leukaemia at first relapse. British Journal of Haematology, 166, 902–910.PubMedCrossRef
125.
Zurück zum Zitat Micallef, J., Dharsee, M., Chen, J., et al. (2010). Applying mass spectrometry based proteomic technology to advance the understanding of multiple myeloma. Journal of Hematology & Oncology, 3, 13.CrossRef Micallef, J., Dharsee, M., Chen, J., et al. (2010). Applying mass spectrometry based proteomic technology to advance the understanding of multiple myeloma. Journal of Hematology & Oncology, 3, 13.CrossRef
126.
Zurück zum Zitat Yang, Y., Chen, Y., Saha, M. N., et al. (2015). Targeting phospho-MARCKS overcomes drug resistance and induces antitumor activity in preclinical models of multiple myeloma. Leukemia, 29, 715–726.PubMedCrossRef Yang, Y., Chen, Y., Saha, M. N., et al. (2015). Targeting phospho-MARCKS overcomes drug resistance and induces antitumor activity in preclinical models of multiple myeloma. Leukemia, 29, 715–726.PubMedCrossRef
127.
Zurück zum Zitat Scott, K., Hayden, P. J., Will, A., et al. (2016). Bortezomib for the treatment of multiple myeloma. In K. Scott (Ed.), Cochrane Database Syst. Rev (p. CD010816). Chichester: John Wiley & Sons, Ltd. Scott, K., Hayden, P. J., Will, A., et al. (2016). Bortezomib for the treatment of multiple myeloma. In K. Scott (Ed.), Cochrane Database Syst. Rev (p. CD010816). Chichester: John Wiley & Sons, Ltd.
128.
Zurück zum Zitat Orlowski, R. Z., Stinchcombe, T. E., Mitchell, B. S., et al. (2002). Phase I trial of the proteasome inhibitor PS-341 in patients with refractory hematologic malignancies. Journal of Clinical Oncology, 20, 4420–4427.PubMedCrossRef Orlowski, R. Z., Stinchcombe, T. E., Mitchell, B. S., et al. (2002). Phase I trial of the proteasome inhibitor PS-341 in patients with refractory hematologic malignancies. Journal of Clinical Oncology, 20, 4420–4427.PubMedCrossRef
129.
Zurück zum Zitat Cortes, J., Thomas, D., Koller, C., et al. (2004). Phase I study of bortezomib in refractory or relapsed acute leukemias. Clinical Cancer Research, 10, 3371–3376.PubMedCrossRef Cortes, J., Thomas, D., Koller, C., et al. (2004). Phase I study of bortezomib in refractory or relapsed acute leukemias. Clinical Cancer Research, 10, 3371–3376.PubMedCrossRef
130.
Zurück zum Zitat Orlowski, R. Z., Voorhees, P. M., Garcia, R. A., et al. (2005). Phase 1 trial of the proteasome inhibitor bortezomib and pegylated liposomal doxorubicin in patients with advanced hematologic malignancies. Blood, 105, 3058–3065.PubMedCrossRef Orlowski, R. Z., Voorhees, P. M., Garcia, R. A., et al. (2005). Phase 1 trial of the proteasome inhibitor bortezomib and pegylated liposomal doxorubicin in patients with advanced hematologic malignancies. Blood, 105, 3058–3065.PubMedCrossRef
131.
Zurück zum Zitat Horton, T. M., Pati, D., Plon, S. E., et al. (2007). A phase 1 study of the proteasome inhibitor bortezomib in pediatric patients with refractory leukemia: a Children’s Oncology Group study. Clinical Cancer Research, 13, 1516–1522.PubMedCrossRef Horton, T. M., Pati, D., Plon, S. E., et al. (2007). A phase 1 study of the proteasome inhibitor bortezomib in pediatric patients with refractory leukemia: a Children’s Oncology Group study. Clinical Cancer Research, 13, 1516–1522.PubMedCrossRef
132.
Zurück zum Zitat Attar, E. C., De Angelo, D. J., Supko, J. G., et al. (2008). Phase I and pharmacokinetic study of bortezomib in combination with idarubicin and cytarabine in patients with acute myelogenous leukemia. Clinical Cancer Research, 14, 1446–1454.PubMedCrossRef Attar, E. C., De Angelo, D. J., Supko, J. G., et al. (2008). Phase I and pharmacokinetic study of bortezomib in combination with idarubicin and cytarabine in patients with acute myelogenous leukemia. Clinical Cancer Research, 14, 1446–1454.PubMedCrossRef
133.
Zurück zum Zitat Messinger, Y., Gaynon, P., Raetz, E., et al. (2010). Phase I study of bortezomib combined with chemotherapy in children with relapsed childhood acute lymphoblastic leukemia (ALL): a report from the therapeutic advances in childhood leukemia (TACL) consortium. Pediatric Blood & Cancer, 55, 254–259.CrossRef Messinger, Y., Gaynon, P., Raetz, E., et al. (2010). Phase I study of bortezomib combined with chemotherapy in children with relapsed childhood acute lymphoblastic leukemia (ALL): a report from the therapeutic advances in childhood leukemia (TACL) consortium. Pediatric Blood & Cancer, 55, 254–259.CrossRef
134.
Zurück zum Zitat Messinger, Y. H., Gaynon, P. S., Sposto, R., et al. (2012). Bortezomib with chemotherapy is highly active in advanced B-precursor acute lymphoblastic leukemia: Therapeutic Advances in Childhood Leukemia & Lymphoma (TACL) Study. Blood, 120, 285–290.PubMedCrossRef Messinger, Y. H., Gaynon, P. S., Sposto, R., et al. (2012). Bortezomib with chemotherapy is highly active in advanced B-precursor acute lymphoblastic leukemia: Therapeutic Advances in Childhood Leukemia & Lymphoma (TACL) Study. Blood, 120, 285–290.PubMedCrossRef
135.
Zurück zum Zitat Lancet, J. E., Duong, V. H., Winton, E. F., et al. (2011). A phase I clinical-pharmacodynamic study of the farnesyltransferase inhibitor tipifarnib in combination with the proteasome inhibitor bortezomib in advanced acute leukemias. Clinical Cancer Research, 17, 1140–1146.PubMedPubMedCentralCrossRef Lancet, J. E., Duong, V. H., Winton, E. F., et al. (2011). A phase I clinical-pharmacodynamic study of the farnesyltransferase inhibitor tipifarnib in combination with the proteasome inhibitor bortezomib in advanced acute leukemias. Clinical Cancer Research, 17, 1140–1146.PubMedPubMedCentralCrossRef
136.
Zurück zum Zitat Attar, E. C., Johnson, J. L., Amrein, P. C., et al. (2013). Bortezomib added to daunorubicin and cytarabine during induction therapy and to intermediate-dose cytarabine for consolidation in patients with previously untreated acute myeloid leukemia age 60 to 75 years: CALGB (Alliance) study 10502. Journal of Clinical Oncology, 31, 923–929.PubMedCrossRef Attar, E. C., Johnson, J. L., Amrein, P. C., et al. (2013). Bortezomib added to daunorubicin and cytarabine during induction therapy and to intermediate-dose cytarabine for consolidation in patients with previously untreated acute myeloid leukemia age 60 to 75 years: CALGB (Alliance) study 10502. Journal of Clinical Oncology, 31, 923–929.PubMedCrossRef
137.
Zurück zum Zitat Walker, A. R., Klisovic, R., Johnston, J. S., et al. (2013). Pharmacokinetics and dose escalation of the heat shock protein inhibitor 17-allyamino-17-demethoxygeldanamycin in combination with bortezomib in relapsed or refractory acute myeloid leukemia. Leukemia & Lymphoma, 54, 1996–2002.CrossRef Walker, A. R., Klisovic, R., Johnston, J. S., et al. (2013). Pharmacokinetics and dose escalation of the heat shock protein inhibitor 17-allyamino-17-demethoxygeldanamycin in combination with bortezomib in relapsed or refractory acute myeloid leukemia. Leukemia & Lymphoma, 54, 1996–2002.CrossRef
138.
Zurück zum Zitat Blum, W., Schwind, S., Tarighat, S. S., et al. (2012). Clinical and pharmacodynamic activity of bortezomib and decitabine in acute myeloid leukemia. Blood, 119, 6025–6031.PubMedPubMedCentralCrossRef Blum, W., Schwind, S., Tarighat, S. S., et al. (2012). Clinical and pharmacodynamic activity of bortezomib and decitabine in acute myeloid leukemia. Blood, 119, 6025–6031.PubMedPubMedCentralCrossRef
139.
Zurück zum Zitat Attar, E. C., Amrein, P. C., Fraser, J. W., et al. (2013). Phase I dose escalation study of bortezomib in combination with lenalidomide in patients with myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). Leukemia Research, 37, 1016–1020.PubMedPubMedCentralCrossRef Attar, E. C., Amrein, P. C., Fraser, J. W., et al. (2013). Phase I dose escalation study of bortezomib in combination with lenalidomide in patients with myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). Leukemia Research, 37, 1016–1020.PubMedPubMedCentralCrossRef
140.
Zurück zum Zitat Howard, D. S., Liesveld, J., Phillips, G. L., et al. (2013). A phase I study using bortezomib with weekly idarubicin for treatment of elderly patients with acute myeloid leukemia. Leukemia Research, 37, 1502–1508.PubMedPubMedCentralCrossRef Howard, D. S., Liesveld, J., Phillips, G. L., et al. (2013). A phase I study using bortezomib with weekly idarubicin for treatment of elderly patients with acute myeloid leukemia. Leukemia Research, 37, 1502–1508.PubMedPubMedCentralCrossRef
141.
Zurück zum Zitat Walker, A. R., Klisovic, R. B., Garzon, R., et al. (2014). Phase I study of azacitidine and bortezomib in adults with relapsed or refractory acute myeloid leukemia. Leukemia & Lymphoma, 55, 1304–1308.CrossRef Walker, A. R., Klisovic, R. B., Garzon, R., et al. (2014). Phase I study of azacitidine and bortezomib in adults with relapsed or refractory acute myeloid leukemia. Leukemia & Lymphoma, 55, 1304–1308.CrossRef
142.
Zurück zum Zitat Walker, A. R., Wang, H., Walsh, K., et al. (2016). Midostaurin, bortezomib and MEC in relapsed/refractory acute myeloid leukemia. Leukemia & Lymphoma, 57, 2100–2108.CrossRef Walker, A. R., Wang, H., Walsh, K., et al. (2016). Midostaurin, bortezomib and MEC in relapsed/refractory acute myeloid leukemia. Leukemia & Lymphoma, 57, 2100–2108.CrossRef
143.
Zurück zum Zitat Wartman, L. D., Fiala, M. A., Fletcher, T., et al. (2016). A phase I study of carfilzomib for relapsed or refractory acute myeloid and acute lymphoblastic leukemia. Leukemia & Lymphoma, 57, 728–730.CrossRef Wartman, L. D., Fiala, M. A., Fletcher, T., et al. (2016). A phase I study of carfilzomib for relapsed or refractory acute myeloid and acute lymphoblastic leukemia. Leukemia & Lymphoma, 57, 728–730.CrossRef
144.
Zurück zum Zitat Awan, F. T., Flynn, J. M., Jones, J. A., et al. (2015). Phase I dose escalation trial of the novel proteasome inhibitor carfilzomib in patients with relapsed chronic lymphocytic leukemia and small lymphocytic lymphoma. Leukemia & Lymphoma, 56, 2834–2840.CrossRef Awan, F. T., Flynn, J. M., Jones, J. A., et al. (2015). Phase I dose escalation trial of the novel proteasome inhibitor carfilzomib in patients with relapsed chronic lymphocytic leukemia and small lymphocytic lymphoma. Leukemia & Lymphoma, 56, 2834–2840.CrossRef
145.
Zurück zum Zitat Horton, T. M., Perentesis, J. P., Gamis, A. S., et al. (2014). A phase 2 study of bortezomib combined with either idarubicin/cytarabine or cytarabine/etoposide in children with relapsed, refractory or secondary acute myeloid leukemia: a report from the Children’s Oncology Group. Pediatric Blood & Cancer, 61, 1754–1760.CrossRef Horton, T. M., Perentesis, J. P., Gamis, A. S., et al. (2014). A phase 2 study of bortezomib combined with either idarubicin/cytarabine or cytarabine/etoposide in children with relapsed, refractory or secondary acute myeloid leukemia: a report from the Children’s Oncology Group. Pediatric Blood & Cancer, 61, 1754–1760.CrossRef
146.
Zurück zum Zitat Ishitsuka, K., Utsunomiya, A., Katsuya, H., et al. (2015). A phase II study of bortezomib in patients with relapsed or refractory aggressive adult T-cell leukemia/lymphoma. Cancer Science, 106, 1219–1223.PubMedPubMedCentralCrossRef Ishitsuka, K., Utsunomiya, A., Katsuya, H., et al. (2015). A phase II study of bortezomib in patients with relapsed or refractory aggressive adult T-cell leukemia/lymphoma. Cancer Science, 106, 1219–1223.PubMedPubMedCentralCrossRef
147.
Zurück zum Zitat Royer, B., Minvielle, S., Diouf, M., et al. (2016). Bortezomib, doxorubicin, cyclophosphamide, dexamethasone induction followed by stem cell transplantation for primary plasma cell leukemia: a prospective phase II study of the Intergroupe Francophone du Myélome. Journal of Clinical Oncology, 34, 2125–2132.PubMedCrossRef Royer, B., Minvielle, S., Diouf, M., et al. (2016). Bortezomib, doxorubicin, cyclophosphamide, dexamethasone induction followed by stem cell transplantation for primary plasma cell leukemia: a prospective phase II study of the Intergroupe Francophone du Myélome. Journal of Clinical Oncology, 34, 2125–2132.PubMedCrossRef
148.
Zurück zum Zitat Araujo, K. P. C., Bonuccelli, G., Duarte, C. N., et al. (2013). Bortezomib (PS-341) treatment decreases inflammation and partially rescues the expression of the dystrophin-glycoprotein complex in GRMD dogs. PLoS One, 8, e61367.PubMedPubMedCentralCrossRef Araujo, K. P. C., Bonuccelli, G., Duarte, C. N., et al. (2013). Bortezomib (PS-341) treatment decreases inflammation and partially rescues the expression of the dystrophin-glycoprotein complex in GRMD dogs. PLoS One, 8, e61367.PubMedPubMedCentralCrossRef
149.
Zurück zum Zitat Penna, F., Bonetto, A., Aversa, Z., et al. (2016). Effect of the specific proteasome inhibitor bortezomib on cancer-related muscle wasting. Journal of Cachexia, Sarcopenia and Muscle, 7, 345–354.PubMedCrossRef Penna, F., Bonetto, A., Aversa, Z., et al. (2016). Effect of the specific proteasome inhibitor bortezomib on cancer-related muscle wasting. Journal of Cachexia, Sarcopenia and Muscle, 7, 345–354.PubMedCrossRef
150.
Zurück zum Zitat Turner, J. G., Kashyap, T., Dawson, J. L., et al. (2016). XPO1 inhibitor combination therapy with bortezomib or carfilzomib induces nuclear localization of IκBα and overcomes acquired proteasome inhibitor resistance in human multiple myeloma. Oncotarget, 7, 78896–78909.PubMedPubMedCentral Turner, J. G., Kashyap, T., Dawson, J. L., et al. (2016). XPO1 inhibitor combination therapy with bortezomib or carfilzomib induces nuclear localization of IκBα and overcomes acquired proteasome inhibitor resistance in human multiple myeloma. Oncotarget, 7, 78896–78909.PubMedPubMedCentral
151.
Zurück zum Zitat de Bruin, G., Xin, B. T., Kraus, M., et al. (2016). A set of activity-based probes to visualize human (immuno)proteasome activities. Angewandte Chemie (International Ed. in English), 55, 4199–4203.CrossRef de Bruin, G., Xin, B. T., Kraus, M., et al. (2016). A set of activity-based probes to visualize human (immuno)proteasome activities. Angewandte Chemie (International Ed. in English), 55, 4199–4203.CrossRef
152.
Zurück zum Zitat Lee, S. J., Levitsky, K., Parlati, F., et al. (2016). Clinical activity of carfilzomib correlates with inhibition of multiple proteasome subunits: application of a novel pharmacodynamic assay. British Journal of Haematology, 173, 884–895.PubMedPubMedCentralCrossRef Lee, S. J., Levitsky, K., Parlati, F., et al. (2016). Clinical activity of carfilzomib correlates with inhibition of multiple proteasome subunits: application of a novel pharmacodynamic assay. British Journal of Haematology, 173, 884–895.PubMedPubMedCentralCrossRef
153.
Zurück zum Zitat de Groot, K. A., Tsang, A., Sjoe, M., Niewerth, D., et al. (2015). Pharmacodynamic monitoring of (immuno)proteasome inhibition during bortezomib treatment of a critically ill patient with lupus nephritis and myocarditis. Lupus Science & Medicine, 2, e000121.CrossRef de Groot, K. A., Tsang, A., Sjoe, M., Niewerth, D., et al. (2015). Pharmacodynamic monitoring of (immuno)proteasome inhibition during bortezomib treatment of a critically ill patient with lupus nephritis and myocarditis. Lupus Science & Medicine, 2, e000121.CrossRef
Metadaten
Titel
(Immuno)proteasomes as therapeutic target in acute leukemia
verfasst von
Jacqueline Cloos
Margot SF Roeten
Niels E Franke
Johan van Meerloo
Sonja Zweegman
Gertjan JL Kaspers
Gerrit Jansen
Publikationsdatum
25.10.2017
Verlag
Springer US
Erschienen in
Cancer and Metastasis Reviews / Ausgabe 4/2017
Print ISSN: 0167-7659
Elektronische ISSN: 1573-7233
DOI
https://doi.org/10.1007/s10555-017-9699-4

Weitere Artikel der Ausgabe 4/2017

Cancer and Metastasis Reviews 4/2017 Zur Ausgabe

OriginalPaper

Preface

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.