Next Article in Journal
Primary Cutaneous B-Cell Lymphoma: An Update on Pathologic and Molecular Features
Next Article in Special Issue
Histological Kidney Re-Evaluation after Daratumumab Monotherapy for AL Amyloidosis
Previous Article in Journal
What Is Genomic High-Risk Myeloma?
Previous Article in Special Issue
From the Light Chain Sequence to the Tissue Microenvironment: Contribution of the Mesangial Cells to Glomerular Amyloidosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Laboratory-Based Rationale for Targeting the Protein Homeostasis Network in AL Amyloidosis

1
Amyloidosis Program, Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 4 Blackfan Circle, Boston, MA 02115, USA
2
John C. Davis Myeloma and Amyloid Program, Tufts Medical Center, Tufts University School of Medicine, 800 Washington Street, Boston, MA 02111, USA
*
Author to whom correspondence should be addressed.
Hemato 2022, 3(2), 298-317; https://doi.org/10.3390/hemato3020022
Submission received: 21 March 2022 / Revised: 12 April 2022 / Accepted: 12 April 2022 / Published: 25 April 2022

Abstract

:
AL amyloidosis is an incurable plasma cell dyscrasia with limited therapeutic options. The pathogenetic mechanism in AL amyloidosis is the deposition of insoluble fibrillary aggregates of misfolded immunoglobulin (Ig) free light chains (FLC) and chaperone proteins in target organs. Therefore, AL amyloidosis is the prototypic, protein-toxicity hematologic disorder. Based on laboratory evidence of increased, constitutive proteotoxic stress, PCs are intrinsically vulnerable to agents that target proteins whose function is to guarantee that nascent polypeptides either reach a functional conformation or are disposed of (proteostasis network). The clinical efficacy of proteasome inhibitors (PIs), such as bortezomib, in the treatment of plasma cell (PC) disorders has provided proof of concept that disrupting protein homeostasis is an effective and generally safe therapeutic approach. Therefore, the intrinsic biology of PC offers us the opportunity to rationally develop therapies that target this distinct proteostasis vulnerability of PC dyscrasias. In this manuscript, we will review the laboratory rationale for the effectiveness of FDA-approved and investigational agents targeting protein homeostasis in AL amyloidosis and related PC disorders.

1. Introduction

AL amyloidosis is the most common form of the family of protein deposition disorders termed amyloidoses [1]. The shared fundamental pathogenetic mechanism of these conditions is the deposition in target organs of polymers of a misfolded protein, organized in fibrils, in association with chaperone proteins [2]. The amyloidogenic precursor protein defines the subtype of amyloidosis, determines the pattern of organ involvement and disease natural history, and ultimately dictates treatment approach [3]. In AL amyloidosis, the clonal Ig free light chain (FLC) is the precursor protein of amyloid fibrils. The underlying hematologic diathesis driving AL amyloidosis is generally a plasma cell (PC) disorder, and less frequently, it is a post-germinal center (GC) B cell lymphoma. In most instances, the PC clone responsible for the production of amyloidogenic FLC is relatively small and would otherwise be classified as a monoclonal gammopathy of undetermined significance (MGUS) in the absence of amyloid-driven end organ damage. However, circa 15% of patients with the PC cancer multiple myeloma (MM) will be diagnosed with overlapping AL amyloidosis during the course of their illness.
While AL amyloidosis and MM shared a common progenitor cell, a mutated, post-GC terminally differentiated B cell, the two conditions diverge in their primary pathogenetic mechanism. In fact, MM primarily causes organ damage as a consequence of the expansion of clonotypic PCs in the bone marrow with only occasional paraprotein-related damage as in the context of cast nephropathy or hyperviscosity [4]. Vice versa, fibrillogenic deposition of the secreted FLC is the pathogenetic mechanism in AL amyloidosis, making it the quintessential protein toxicity hematologic disorder.
Eukaryotic cells rely on a network of proteins (proteostasis network, PN) to maintain homeostasis and avoid toxicity related to the accumulation of unstable/misfolded or untimely protein species (Figure 1) [5]. In this effort, the PN integrates with adaptive stress responses such as the unfolded protein response (UPR) to restore protein equilibrium in the face of proteotoxicity [6]. Nevertheless, these redundant systems can become saturated if the proteotoxic stress is too overwhelming in duration or intensity and apoptosis then occurs [7]. The work of our groups and others has shown that normal and aberrant PCs depend on an intact PN for their survival, offering the opportunity to develop rationally designed therapies targeting our very own PC biology and turning it against PC disorders [8,9]. Our groups and others have previously shown that both MM and AL-PC suffer from baseline proteotoxic stress related to sustaining and imprecise protein synthesis. It is therefore intuitive to develop therapies that further exacerbate this balance in an effort to induce apoptosis. The classical example of the power of such a strategy is the selective anti-MM activity of proteasome inhibitors (PIs) [10]. Despite proteasomes being ubiquitously expressed in tissues, the first in class PI bortezomib proved selectively toxic against MM- and AL-PC with surprisingly limited side effects, owing to the level of proteotoxic stress experienced by these professional secretory cells [11,12]. The use of bortezomib in AL amyloidosis radically changed the natural history of this disease and profoundly impacted the survival of patients, providing proof that targeting proteostasis is clinically useful in AL amyloidosis.
In this review, we explore the laboratory-based rationale for targeting proteostasis in AL amyloidosis and discussed FDA-approved and investigational agents in advanced clinical development (Table 1). As preclinical models to study AL amyloidosis are scant, the majority of the data herein presented are extrapolated from MM models.

2. Targeting the Ubiquitin–Proteasome System (UPS)

2.1. Proteasome Inhibitors

Based on the very nature of normal and malignant PCs as immunoglobulin (or FLC)-synthesizing factories, these cells are dependent on an intact proteostasis network for survival [8,9]. Given the intrinsically flawed nature of protein synthesis, it is estimated that about one-third of newly synthesized proteins will be unable to reach native conformation and will need to be disposed of through the ubiquitin–proteasome system (UPS) [13]. This figure is significantly increased in cell types characterized by high synthetic function and/or malignant cells, explaining the disproportionate accumulation of polyubiquitinated proteins in AL or MM PCs [14,15,16,17,18]. For secretory and/or transmembrane proteins, this process involves retrotranslocation from the ER through the Sec61 translocon and proteasome-mediated degradation (ER-associated degradation or ERAD) [19]. Proteins along this pathway thus represent potentially relevant therapeutic targets to exacerbate proteotoxic stress in PC disorders. In fact, PIs proved remarkably effective in treating both AL and MM; however, the acquisition of resistance over time is inevitable for most patients, and a suboptimal response to bortezomib has been observed in t(11;14) AL amyloidosis patients, underscoring the need for the development of novel rationally informed therapeutics within the proteostasis network [20].
The proteasome is a 2.4 MDa, ATP-dependent, multicatalytic protein degradation organelle that acts in concert with the K48-ubiquitination of proteins to degrade cell-cycle asynchronous or misfolded proteins. The 20S barrel-shaped catalytic core of the proteasome is composed of one external α ring on each side and two inner β rings containing caspase-like (β1), trypsin-like (β2) and chymotrypsin-like (β5) catalytic activities. Interferon γ-inducible catalytic subunits LMP2 (β1i), MECL-1 (β2i) and LMP7 (β5i) can replace constitutive subunits to form the immunoproteasome in immune cells and facilitate the production of antigenic peptide precursors for MHC class I presentation [21,22,23]. On one or both sides of the 20S core sits a 19S regulatory cap that is responsible for the critical functions of binding, deubiquitylating, unfolding and delivering of K-48 polyubiquitinated (polyUb) cargo proteins to the catalytic core [24,25]. PIs were initially developed as tools to study the critical function of the proteasome and dissect the activity of each subunit in protein degradation [26,27]. PS-341, later named bortezomib, a reversible, boronic acid inhibitor of the β5 and to a lesser extent the β1 activities of the proteasome [26], proved remarkably active against MM in preclinical and phase I clinical studies, prompting its further development in late-stage clinical trials [28,29]. Bortezomib is thought to mediate MM activity via pleiotropic mechanisms targeting both the cancer cell directly as well as the tumor-supportive bone marrow microenvironment. Based on preclinical data, bortezomib is directly toxic to MM cells and modulates the BM microenvironment with anti-angiogeneic, pro-osteoblastic and anti-osteoclastic effects [30,31]. The initial hypothesis behind the extraordinary activity of bortezomib in MM, alongside the limited side effects observed in clinical trials, was inhibition of the canonical nuclear factor κB (NF-κB) pathway via stabilization of NF-κB inhibitors (IκB proteins). However, the treatment of MM cells with NF-κB inhibitors did not induce the same extent of cytotoxicity as bortezomib, indicating alternative mechanisms [32,33]. Subsequent studies showed that bortezomib impaired the DNA damage response and induced terminal unfolded protein (UPR) and heat shock responses (HSR), causing irreversible proteotoxic stress due to the accumulation of toxic polyUb protein species and intrinsic apoptosis [33,34,35]. Our group showed that the ratio between polyUb proteins and proteasome activity at baseline can serve as a predictive biomarker of response to PIs in MM [11]. Consequently, an increase in misfolded proteins or a decrease in proteasome capacity sensitizes to PIs, while decreased protein synthesis or increased proteasome capacity causes resistance [36].
In seminal AL amyloidosis preclinical studies, synthesis of the amyloidogenic Ig FLC was shown to be directly responsible for the striking sensitivity of AL PCs to PIs by increasing cellular stress levels and impairing proteostasis [37]. Clinical use of bortezomib-based regimens in AL amyloidosis completely changed the natural history of this disorder and resulted in high rates of deep responses across all stages of disease, translating into long-term remissions and providing an effective therapeutic strategy for patients ineligible for autologous stem cell transplant as well as effective induction and consolidation therapies for transplant-eligible patients [38,39,40]. The addition of bortezomib to melphalan and dexamethasone, the historical gold standard treatment for AL amyloidosis, boosted response rates without aggravating side effects [41]. Due to its efficacy and safety profile, the combination of bortezomib, cyclophosphamide, and dexamethasone (CyBorD) rapidly became standard of care in the frontline treatment of AL amyloidosis [42,43]. Bortezomib remains a critical component of AL amyloidosis treatment across all stages of disease with t(11;14) serving as a negative predictive marker of response [20,44]. Most recently, the quadruplet combination of CD38-targeting antibody daratumumab plus CyBorD was approved as frontline therapy of AL amyloidosis, the first (and thus far only) treatment approved for AL amyloidosis [45].
While highly active in MM and AL amyloidosis, acquired resistance to bortezomib is unavoidable over time, and an innate refractory state has been observed both in MM and AL amyloidosis [20]. Clinical resistance to PIs is an adverse prognostic factor in the treatment of PC disorders and a major clinical hurdle, prompting research efforts to develop strategies to overcome it. Carfilzomib, an epoxyketone PI, covalently binds to the β5 subunit of the proteasome, resulting in irreversible inhibition of this catalytic activity [26,46]. In preclinical studies, carfilzomib proved more potent than bortezomib, and its use as a single agent or in combination with other drugs resulted in durable responses in patients with relapsed/refractory MM, including bortezomib-refractory individuals [47,48,49]. However, a significant signal for carfilzomib-associated cardiovascular adverse events such as hypertension, heart failure, ischemia and arrhythmias emerged during larger clinical studies [50,51]. While the nature of these side effects has not been definitively clarified, there is reasonable concern that it may be related to the on-target effect of irreversible inhibition of the chymotrypsin-like activity of the proteasome. Although effective, a signal for vascular toxicity with renal and cardiac manifestations emerged in early-phase clinical trials of carfilzomib in AL amyloidosis [52,53]. In general, we recommend that carfilzomib administration to patients with AL amyloidosis be carefully pondered and generally reserved for a selected patient population with absent or minimal pre-existing cardiac, renal or small vessel involvement and limited therapeutic options. For instance, carfilzomib was proven to be an effective treatment strategy in patients with AL amyloidosis-related peripheral neuropathy that may preclude the safe use of bortezomib [54]. Finally, prescribers should be considerate in dose-escalating carfilzomib, as the cardiovascular signal in AL was observed for doses exceeding 36 mg/m2 [51,55].
Marizomib, a β-lactone-γ-lactam irreversible PI, was developed in an attempt to overcome bortezomib resistance by inhibiting all three catalytic activities of the proteasome. Supporting this rationale, preclinical studies in triple negative breast cancer and the concurrent inhibition of β2 and β5 proteasome subunits could overcome resistance to β5-only inhibitors bortezomib and carfilzomib, suggesting that the targeting of multiple proteasome subunits may be an effective strategy to overcome resistance to carfilzomib and bortezomib [56]. In vitro, marizomib proved more potent than bortezomib and effective in bortezomib-resistant MM models; however, central nervous system (CNS) side effects such as gait disturbances and visual hallucinations were observed during early-phase clinical studies in relapsed/refractory MM (RRMM), suggesting that marizomib crosses the blood–brain barrier [57,58]. As a consequence, its development in MM has been halted, but clinical trials are ongoing in primary CNS tumors, particularly glioblastoma multiforme, with a plan to launch a trial of marizomib in combination with pomalidomide and dexamethasone in CNS MM.
These data underscore the trade-off between the potency of proteasome inhibition and clinical tolerability, reminding us of the essential function of the UPS in normal tissues.
Recent seminal work has shown that cells are equipped with a proteasome stress response that leads to the transcription of proteasome subunits in the face of insufficient proteasome activity [59,60,61]. NFE2 Like BZIP Transcription Factor 1 (NFE2L1 or NRF1) and its homologue NFE2 Like BZIP Transcription Factor 2 (NFE2L2 or NRF2) are master regulators of baseline and stress-induced proteasome biogenesis. In homeostatic conditions, NRF1 is constitutively transcribed, translated, partially inserted in the endoplasmic reticulum (ER), deglycosylated by N-Glycanase 1 (NGLY1), retro-translocated via Valosin Containing Protein (VCP or p97), and degraded via the ERAD [59,62]. However, when proteasome activity is reduced as in the case of sublethal, partial inhibition via PI, cytosolic, deglycosylated NRF1 is cleaved by the aspartic protease DNA Damage Inducible 1 Homolog 2 (DDI2). Cleaved NRF1 is transcriptionally active, dimerizes with small MAF proteins and binds to antioxidant response elements (ARE) on the DNA, activating a transcriptional program that includes the transcription of proteasome subunits [62]. Recently, knock out (KO) of NGLY1, DDI2 or NRF1 was shown to be cytotoxic in MM cells, including in PI-acquired or innate resistant models, and it could overcome PI resistance, suggesting that this pathway is a promising molecular target in MM [63,64].
Finally, in an attempt to ease administration, there has been interest in developing orally bioavailable PIs. Ixazomib, an orally bioavailable boronate PI, is currently FDA approved as a second-line treatment for patients with relapsed and refractory MM, and it provides a convenient agent with ease of oral administration particularly suited for maintenance therapy and/or treatment of elderly patients [57,65,66]. In a randomized, phase III study of ixazomib plus dexamethasone versus physician’s choice in patients with relapsed/refractory AL amyloidosis, the investigational arm failed to improve the overall response rate or depth of response [67]. Nevertheless, a trend was observed in the prolongation of survival/major organ deterioration with the caveat of shorter duration of treatment in the control arm. These data suggest that the long-term administration of ixazomib plus dexamethasone might be a feasible approach to prolong organ and/or patient survival.

2.2. E3 Ub Ligase

The conjugation of multiple Ub moieties on a target protein is accomplished by the sequential activity of Ub activating (E1), conjugating (E2) and ligase (E3) enzymes [68]. There are more than 600 known E3 Ub ligases, and they dictate substrate specificity, thus representing appealing targets for drug development.
Among the substrates of Cullin-RING E3 ligases are tumor suppressor proteins, suggesting that their inhibition may be an effective cancer therapeutic [69]. Cullin-RING E3 ligases undergo NEDD8 Ubiquitin Like Modifier (NEDD8)-mediated neddylation, which is a post-translational modification that is essential for their activation. Consequently, blocking NEDD8 function should result in the blockade of Cullin-RING E3 function [70]. NEDD8-activating enzyme inhibitors showed promising results in preclinical MM models; however, their clinical development has been unsuccessful thus far due to lack of activity (MLN4924) or emergence of dose-limiting toxicities (TAS4464) [71,72].
Recent seminal work unraveled a new mechanism of function of the immunomodulatory drug (IMiD) lenalidomide. Lenalidomide was shown to bind to Cereblon, a component of a E3 ubiquitin ligase complex, and to redirect its function to the selective degradation of transcription factors Ikaros family zinc finger proteins 1 and 3 (IKZF1 and IKZF3) [73,74]. This observation paved the way for the development of a novel class of drugs called proteolysis-targeting chimeric molecules (PROTACs) to redirect the physiologic function of E3 Ub ligases for the degradation of specific targets of interest, specifically oncogenes [75]. KPG-818 is a small molecule investigational compound that binds cereblon with high affinity and leads to the reduced production of inflammatory cytokines and levels of IKZF1 and IKZF3 [76]. KPG-818 was effective in preclinical models of lymphoma and multiple myeloma and is currently being evaluated in phase 1 clinical trial in hematologic malignancies.

2.3. Deubiquitylating Enzymes/Ubiquitin Receptors

Deubiquitylating enzymes (DUBs) are a large family of enzymes whose function is the removal of ubiquitin (Ub) moieties from target proteins and its recycling, thus contributing to maintaining proteostasis and guaranteeing the availability of free Ub for the posttranslational modification of proteins participating in critical cellular processes [77,78]. Given the role of Ub in targeting protein substrate for proteasome-mediated degradation and in consideration of the exceptional activity of PIs in PC disorders, the inhibition of DUB may be an appealing therapeutic strategy in these diseases [79]. ADRM1 26S Proteasome Ubiquitin Receptor (ADRM1 or RPN13) is a 19S-associated Ub receptor that on the one end binds polyUb proteins with high affinity and on the other recruits DUBs to hydrolyze Ub moieties from the proteasome substrate, thus allowing engagement by the 20S and downstream degradation [80]. RPN13 is highly expressed in MM, and its inhibition leads to apoptosis in MM cells and animal models in the absence of direct 20S proteasome inhibition [81]. Similarly, the inhibition of proteasome-associated DUBs UCHL5, ubiquitin specific protease 14 (USP14) and Ubiquitin Specific Peptidase 7 (USP7) lead to the accumulation of polyUb proteins in the absence of direct inhibition of the catalytic activity of the proteasome, resulting in apoptosis [82,83,84].
Unfortunately, VLX1570, a first in human small molecule inhibitor of UCHL5, caused fatal pulmonary toxicity in early-phase clinical studies, with consequent halting of clinical trials [83]. Drug development efforts are ongoing to identify tolerable and effective compounds targeting cancer-associated DUBs.

2.4. ERAD

The cytosolic AAA-ATPase VCP organizes as a homo-hexameric channel that binds and extracts misfolded proteins from the ER and delivers them to the proteasome in the process known as ERAD [19]. Inhibition of VCP in combination with PIs was shown to be cytotoxic in preclinical MM models; however, clinical development of the first in class VCP inhibitor CB-5083 was halted due to the surfacing of retinal toxicity [85,86]. Given the strong biology underlying targeting VCP in MM, research efforts are ongoing to develop inhibitors with improved therapeutic index.

3. Alternative Proteolytic Pathways

Autophagy

Autophagy is an evolutionary conserved mechanism that delivers cytoplasmic content, such as organelles, misfolded proteins and intracellular pathogens, to the lysosome for degradation and recycling [87]. It can therefore be considered an alternative proteolytic pathway to the UPS. Autophagy plays a dual role of guaranteeing energy supply by recycling macromolecules and maintaining protein homeostasis by selective auto-digestion of cellular material. Autophagy was shown to play a pivotal role in the physiologic process of PC differentiation and in ensuring the survival of bone marrow-resident, long-lived PC [37,88,89]. In this context, autophagy is necessary to support ER expansion and Ig synthesis [88]. Extensive research has proven that a close crosstalk exists between UPS, endoplasmic reticulum (ER) stress, heat shock response (HSR), and autophagy that critically contributes to the quality control of newly synthesized proteins and proteostasis in eukaryotic cells [90,91,92,93]. Preclinical studies have shown that autophagic selectivity is conferred by specific receptor/adapter proteins that recognize and mediate the engulfment of the substrate in the autophagosome. Recently, the prototypical autophagic receptor, Sequestosome 1 (SQSTM1 or p62), has been implicated in the maintenance of homeostasis in MM by the clearance of redundant misfolded proteins [89]. SQSTM1-deficient PC displayed reduced intracellular ATP, elevated levels of ER stress and died prematurely, in keeping with a role of autophagy in cell homeostasis [88].
Autophagy was originally identified as a pro-survival mechanism in MM by reducing protein-related stress. However, recent studies show that depending on the context, autophagy can promote MM cell death through a non-apoptotic, caspase 10-dependent mechanism known as autophagy-dependent cell death [94,95]. These seemingly opposite roles of autophagy in determining cell fate suggest that both its pharmacologic inhibition (dysregulation of proteostasis) or its activation (induction of autophagic cell death) could promote anti-MM effect. Consistently, pharmacologic modulation of this pathway in combination with PI resulted in highly variable effects, ranging from synergism to antagonism [96]. For instance, the autophagy inhibitors 3-methyladeninne and chloroquine were shown to induce cytotoxicity in MM cell lines when used alone, but when combined with bortezomib, they resulted in an antagonistic response [95,97,98]. However, in a different study, chloroquine synergistically augmented carfilzomib cytotoxicity and was able to overcome carfilzomib resistance in vitro [99]. Vice versa, the activation of SQSTM1 yielded specific PI resistance by mediating a plastic adaptive response to proteasome stress, thus representing a novel, potential molecular target to overcome PI resistance [89,97]. It is therefore not surprising that the clinical translation of these inconsistent observations has been difficult. In early-phase clinical trials, a modest clinical benefit was observed by combining chloroquine with the PI carfilzomib [100].
Similar to MM, AL amyloidosis cells are exquisitely dependent on autophagy for their survival. AL plasma cells show distinctive organellar features such as ER expansion, perinuclear mitochondria, expression patterns consistent with cellular oxidative stress and reduced autophagic control of organelle homeostasis [37]. This study also reported that AL plasma cells have a unique PI susceptibility that correlates with defective autophagy and altered organelle homeostasis [37].
Adding to the complexity of this pathway, a recent seminal study showed that lysosomal dysfunction and the dysregulation of autophagic flux were evident in cardiac cells from human patients with AL amyloid-cardiomyopathy, implying that the activation of autophagy would be therapeutically beneficial in AL amyloid-cardiomyopathy [101]. In fact, the data show that the activation of autophagy via rapamycin protects against amyloidogenic light chain-associated cardiac proteotoxicity and prolongs survival in a zebrafish model [101].
Altogether, it is likely that the modulation of autophagy would have both favorable and unfavorable effects when used in patients with AL amyloidosis, thus making it extremely difficult to design safe clinical trials [102].

4. Aggresome

In AL amyloidosis, amyloidogenic, misfolded FLCs are secreted and lead to the formation of amyloid deposits extracellularly. From a biological standpoint, this observation is puzzling because quality control mechanisms do not allow misfolded proteins to transit from the ER to the Golgi. Rather, these unstable protein species are retro-translocated from the ER to the cytosol, ubiquitinated and directed for proteasome-mediated degradation via ERAD [103,104]. Aggresomes represent an alternative pathway for the containment of misfolded proteins in perinuclear inclusion bodies [105]. The formation of aggresomes activates autophagy and ultimately lysosomal degradation, which mediates the disposal of aggregated proteins. While a potential therapeutic role for targeting aggresomes has not been evaluated in AL amyloidosis, studies have been conducted in MM, leading to the development of targeted drugs. Histone deacetylase 6 (HDAC6) is a class I HDAC that is necessary for the proper formation of aggresomes by catalyzing the sequestration of polyUb misfolded proteins in perinuclear areas via the dynein–microtubule axis [106,107,108]. Preclinical studies in MM showed that proteasome inhibition induces aggresome formation, suggesting that aggresomes may represent an escape pathway to survive PI-induced proteotoxic stress. Therefore, HDAC6 represents a promising molecular target to overcome PI resistance [108,109].
Panobinostat, an orally bioavailable, non-selective pan-HDAC inhibitor, is the only FDA-approved HDAC inhibitor in MM. In 2015, panobinostat received accelerated approval by the FDA for its use in combination with bortezomib and dexamethasone in relapsed/refractory MM patients who had received at least two prior regimens, including bortezomib and an immunomodulatory agent [110,111]. However, in December 2021, a request for withdrawal of this approval was submitted, effectively leading to the discontinuation of approved use of panobinostat in MM.
The therapeutic index of panobinostat was narrow due to the frequent and often severe occurrence of gastrointestinal and hematologic side effects. The hypothesis that the efficacy of pan-HDAC inhibitors is mediated by the blockade of HDAC6-mediated trafficking of polyubiquitinated proteins to the aggresome pathway whereas the adverse events are due to non-specific targeting of gene expression led to the development of selective HDAC6 inhibitors, such as ricolinostat (ACY-1215) and citarinostat (ACY-241). A phase Ib trial of ricolinostat in combination with lenalidomide/dexamethasone reported a 55% overall response rate (ORR) in RRMM and a phase I/II on ricolinostat in combination with bortezomib/dexamethasone reported a 29% ORR in RRMM [112,113]. Ricolinostat in combination with pomalidomide/dexamethasone is currently evaluated in clinical trials (NCT01997840 and NCT02189343). Citarinostat is being evaluated in combination with lenalidomide and PVX-410, a cancer vaccine, in smoldering MM in a phase 1 clinical trial (NCT02886065).

5. Unfolded Protein Response (UPR)

The UPR is an evolutionary conserved, tripartite stress response that is triggered by the accumulation of misfolded proteins in the ER and serves the goal of restoring homeostais [114,115]. It does so by decreasing de novo protein synthesis via translation arrest while selectively inducing chaperones to aid in protein transport and folding and enhancing protein degradation via ERAD. If homeostasis cannot be achieved, persistent UPR activation leads to apoptosis [116]. The signal transductors of the UPR are IRE1, ATF6 and PERK. Activated IRE1 functions as an endonuclease, resulting in XBP1 mRNA splicing [117]. Spliced XBP1 (sXBP1) and ATF6 are master transcriptional regulators, leading to lipid biogenesis to sustain ER expansion, ERAD activation, and induction of chaperones and factors regulating redox balance [118]. Alternatively, PERK causes a repression of de novo protein synthesis via the phosphorylation of eIF2α in an attempt to establish homeostasis. However, if the UPR is sustained over time, PERK selectively induces ATF4 expression, which in turns induces the expression of the pro-apoptotic protein CHOP [35].
Among the distinct UPR pathways, only the IRE1-XBP1 branch mediates bortezomib sensitivity [119]. In fact, in MM, a loss of spliced XBP1 (sXBP1) results in the de-differentiation of plasma cells to plasmablasts with decreased Ig production, decreased proteotoxic stress, and reduced sensitivity to PI [119]. Direct inhibitors of the IRE1 endoribonuclease domain have shown promising preclinical activity in MM, and early-phase clinical trials are anticipated soon [120,121].
Our group and others have shown that drugs capable of inhibiting proper protein folding (ER stressors) such as tunicamycin and thapsigargin synergize with PI in vitro, providing proof-of-principle that combinations of PI with ER stressors may be clinically beneficial [11]. However, ER stressors may have a narrow therapeutic index with potential organ toxicity. Recently, a protease inhibitor used in HIV therapy nelfinavir was shown to induce UPR and overcome PI resistance in MM cells in preclinical models [122]. Importantly, the combination of nelfinavir with bortezomib resulted in synergistic clinical activity even in bortezomib refractory MM, with ORR of 50% in patients with bortezomib-refractory and lenalidomide-resistant MM [123].
A cancer-specific isoform of the HSP70 family of ER chaperone GRP78 (or BIP) has been found to be abundantly expressed on the cell surface of solid tumor and MM cells, representing an appealing target for cancer therapy [124]. Naturally occurring IgM antibodies recognizing BIP have been detected in prostate cancer patients, suggesting this represents a highly antigenic target [125]. PAT-SM6 is a fully humanized, IgM monoclonal antibody directed against surface-expressed GRP78 isoform [126]. Preclinical studies have shown that it binds cell-surfaced expressed GRP78 and induces both direct cytotoxicity and complement-dependent cytotoxicity. In a single-agent, phase 1 clinical trial, PAT-SM6 appeared well tolerated, but responses were not observed.
Seminal work from the Kelly and Wiseman groups has recently shown that the selective activation of individual UPR pathways differently impacts the fate of misfolded immunoglobulin free light chains in terms of retention, stabilization or degradation [127]. In particular, the activation of ATF6 resulted in a reduced secretion of misfolded secretory proteins without increased degradation overall, leading to attenuated extracellular aggregation [128,129]. In AL compound 147, an investigational ER proteostasis enhancer based on its ATF6 activating properties was shown to reduce the secretion of amyloidogenic FLC in an ATF6-independent mechanism involving the on target, covalent modification of other proteaostasis factors, including protein disulfide isomerases (PDIs) and LCs independent of ATF6 activation. Instead, 147 reduces amyloidogenic LC secretion through the selective, on-target covalent modification of ER proteostasis factors, including PDIs [130]. This is a prime example of how fundamental basic research can inform the development of novel and potentially impactful therapies in AL amyloidosis.

6. Heat Shock Proteins

The heat shock response (HSR) can be activated by proteotoxicity and serves the purpose of inducing the rapid expression of chaperone proteins, which are known as heat shock proteins (HSPs). These constitute a large family of evolutionary highly conserved proteins that are often classified based on their molecular weight: HSP70, HSP 90, HSP10, etc. Chaperones have emerged as critical players in protein quality control with a variety of roles including inhibiting protein aggregation, maintaining the folding and solubility of proteins and facilitating protein trafficking and localization into correct subcellular compartments [131]. Heat shock factor 1 (HSF1) is the “master regulator” of the HSR, controlling the expression of HSP90 and HSP70, which are the two main chaperones that support the redirection of misfolded proteins for degradation and participate in chaperone-mediated autophagy [132,133]. These two chaperones have been extensively studied in cancer by virtue of their role in supporting the folding of proto-oncogenes and promoting the stabilization of inactive mutant forms of onco-suppressors such as p53 [132,134]. In light of their critical role in proteostasis, HSPs have been regarded as promising targets in plasma cell disorders [135]. In preclinical MM models, inhibition of HSF1, HSP70 or HSP90 leads to activation of the terminal UPR and consequent apoptosis [136,137,138]. HSP90 (e.g., 17-AAG, NVP-HSP990, PU-H71), HSP70 (e.g., PER-16, Ver-155008, MAL3-101) and HSF1 inhibitors (e.g., CCT251236, KRIBB11) showed potent anti-MM activity in preclinical studies [139]. However, phase I/II clinical trials of HSP90 inhibitors luminespib (NVP-AUY922), KW-2478, SNX5422, retaspimycin (IPI-504) and tanespimycin (KOS-953) in RRMM patients revealed a narrow therapeutic index and only modest clinical significance, leading to a discontinuation of further clinical development [140,141,142,143,144].
Extracellular chaperones (EC) stabilize misfolded proteins that escape the intracellular protein quality control and have been implicated in the clearance of unstable protein species via receptor-mediated endocytosis, thereby playing a pivotal role in maintaining extracellular proteostasis [145]. There is significant interest in exploring the role that EC plays in AL amyloidosis and potentially identifying novel therapeutic targets to activate the clearance of misfolded FLC [146].
Human serum amyloid P (SAP) is a plasma protein that is invariably present in all types of amyloid deposits. SAP has shown specific chaperone activity by stabilizing and protecting the fibrillary protein, suggesting a potential role in the pathogenesis of amyloidoses [147,148]. Miridesap, a small molecule investigational compound, triggers the hepatic clearance of circulating SAP but leaves residual SAP in amyloid deposits. This remaining SAP is a specific target for dezamizumab, a fully humanized IgG1 anti-SAP monoclonal antibody, that has been shown to trigger immune-mediated amyloid reabsorption from visceral organs. In a phase 1 dose-escalation study in patients with amyloidosis, including AL, the use of miridesap followed by dezamizumab showed significant organ responses while having a safe profile. However, a phase 2 trial in patients with cardiac disease was terminated early due to toxicity [149,150].

7. Direct Targeting of FLC Secretion

The pathologic mechanism in AL amyloidosis is the deposition of a misfolded FLC. Therefore, the secretion of amyloidogenic FLC is essential to mediate organ damage. There has been interest in developing therapies directly targeting FLC secretion. An antisense oligonucleotide (ASO) targeting the variable region of lambda free light chain was shown to significantly reduce FLC secretion in λ-secreting MM preclinical models [151]. The major limitation with this approach is the need for patient-specific ASO as the λ-light chain variable region is unique in each individual. Nevertheless, this approach provided proof of concept of the feasibility and potential therapeutic utility of targeting FLC secretion.
Work from the Comenzo lab identified a common sequence in the constant region of λ-light chain, and the group developed an siRNA pool targeting this area [152]. The transfection of the anti λ-light chain siRNA pool in λ-light chain-expressing AL amyloidosis and MM PC caused a rapid and substantial decline in FLC secretion and triggered a terminal UPR presumably due to the retention of orphan Ig heavy chain.
Finally, Moscvin et al. have recently shown that the similar induction of a terminal UPR can be achieved by expressing the catalytic domain of several distinct botulinum neurotoxin serotypes and blocking the SNARE-mediated vesicular secretion of FLC [153]. This approach suggests that direct targeting of the secretory pathway of AL PC may be therapeutically effective, leading not only to the suppression of FLC secretion but also triggering rapid apoptosis presumably due to terminal UPR triggered by the retention of unsecreted FLC.

8. Conclusions and Future Directions

AL amyloidosis is the prototypic, malignant disease of proteotoxic stress based on its high synthetic rate and extracellular release of misfolded, aggregation-prone Ig FLCs. The clinical efficacy of PIs in AL amyloidosis has clearly demonstrated the therapeutic potential of targeting this intrinsic vulnerability of AL-PCs. The notion that AL amyloidosis cells release an improperly folded Ig FLC suggest that quality control mechanisms in the ER are faulty. This characteristic makes AL amyloidosis an even more suitable disease target for therapies that impair protein homeostasis as compared to MM. Nevertheless, innate and/or acquired PI resistance remains an unmet clinical challenge, negatively impacting the prognosis of patients.
In this manuscript, we provided an overview of the laboratory rationale for targeting alternative therapeutic targets to the proteasome catalytic subunits within the proteostasis network (Figure 2). In recent years, the efficiency of the UPS in degrading polyUb protein targets has been leveraged to develop the targeted degradation of specific targets by redirecting E3 Ub ligase activity and facilitating proteasome-mediated degradation [73,74,152,153]. Theoretically, this approach allows the unprecedented opportunity to target previously undruggable proteins, such as MYC [153].
We look forward to an exciting era in AL amyloidosis where a deeper understanding of plasma cell biology can provide a foundation for targeting the Achille’s heel of this disorder. We believe that hypothesis-driven, basic science holds the key to develop more effective treatments for AL amyloidosis patients with a potential for cure for most, if not all, our patients. The work and dedication of Professor Merlini in developing novel models for the study of amyloidogenic light chains and in undertaking numerous clinical trials to advance the therapeutic resources available to patients remains the gold standard for physician, investigators and scientists striving in this field.

Funding

This work was supported in part by the NIH/National Institute of Aging grant R21-AG070502-01 (RLC), by NIH/NCI K08CA245100 grant (GB), by the American Italian Cancer Foundation Research Fellowship (MM) and the Donald C. Brockman Memorial Research Grant from the Amyloidosis Foundation (MM).

Acknowledgments

G.B. and M.M. are thankful to the Demarest Lloyd Jr Foundation and the Appleby Cardiac Amyloidosis Fund for their support of the Amyloidosis Program. R.L.C. thanks the Amyloidosis and Myeloma Research Fund, the Sidewater Family Fund, the Amyloidosis Foundation, David and Barbara Levine (in memoriam), and the Demarest Lloyd Jr Foundation.

Conflicts of Interest

Giada Bianchi: Advisory board participation (with personal payment): Pfizer, Karyopharm; Maria Moscvin: nothing to disclose; Raymond Comenzo: Steering committee fees from Janssen Biotech; advisory board fees from Karyopharm Therapeutics; fees for serving on a data and safety monitoring committee from Sanofi-Aventis, and holding patent WO2016187546A1 on anti-CD38 antibodies for treatment of light-chain amyloidosis and other CD38-positive hematologic cancers.

References

  1. Merlini, G.; Dispenzieri, A.; Sanchorawala, V.; Schonland, S.O.; Palladini, G.; Hawkins, P.N.; Gertz, M.A. Systemic immunoglobulin light chain amyloidosis. Nat. Rev. Dis. Primers 2018, 4, 38. [Google Scholar] [CrossRef]
  2. Merlini, G.; Bellotti, V. Molecular mechanisms of amyloidosis. N. Engl. J. Med. 2003, 349, 583–596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Bianchi, G.; Kumar, S. Systemic amyloidosis due to clonal plasma cell diseases. Hematol. Oncol. Clin. N. Am. 2020, 34, 1009–1026. [Google Scholar] [CrossRef] [PubMed]
  4. Rajkumar, S.V.; Dimopoulos, M.A.; Palumbo, A.; Blade, J.; Merlini, G.; Mateos, M.V.; Kumar, S.; Hillengass, J.; Kastritis, E.; Richardson, P.; et al. International myeloma working group updated criteria for the diagnosis of multiple myeloma. Lancet Oncol. 2014, 15, e538–e548. [Google Scholar] [CrossRef]
  5. Hipp, M.S.; Park, S.H.; Hartl, F.U. Proteostasis impairment in protein-misfolding and -aggregation diseases. Trends Cell Biol. 2014, 24, 506–514. [Google Scholar] [CrossRef] [PubMed]
  6. Brehme, M.; Voisine, C.; Rolland, T.; Wachi, S.; Soper, J.H.; Zhu, Y.; Orton, K.; Villella, A.; Garza, D.; Vidal, M.; et al. A chaperome subnetwork safeguards proteostasis in aging and neurodegenerative disease. Cell Rep. 2014, 9, 1135–1150. [Google Scholar] [CrossRef] [Green Version]
  7. Walter, P.; Ron, D. The unfolded protein response: From stress pathway to homeostatic regulation. Science 2011, 334, 1081–1086. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Sontag, E.M.; Vonk, W.I.M.; Frydman, J. Sorting out the trash: The spatial nature of eukaryotic protein quality control. Curr. Opin. Cell Biol. 2014, 26, 139–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Benbrook, D.M.; Long, A. Integration of autophagy, proteasomal degradation, unfolded protein response and apoptosis. Exp. Oncol. 2012, 34, 286–297. [Google Scholar] [PubMed]
  10. Guang, M.H.Z.; Kavanagh, E.L.; Dunne, L.P.; Dowling, P.; Zhang, L.; Lindsay, S.; Bazou, D.; Goh, C.Y.; Hanley, C.; Bianchi, G.; et al. Targeting proteotoxic stress in cancer: A review of the role that protein quality control pathways play in oncogenesis. Cancers 2019, 11, 66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Bianchi, G.; Oliva, L.; Cascio, P.; Pengo, N.; Fontana, F.; Cerruti, F.; Orsi, A.; Pasqualetto, E.; Mezghrani, A.; Calbi, V.; et al. The proteasome load versus capacity balance determines apoptotic sensitivity of multiple myeloma cells to proteasome inhibition. Blood 2009, 113, 3040–3049. [Google Scholar] [CrossRef] [PubMed]
  12. Richardson, P.G.; Sonneveld, P.; Schuster, M.W.; Irwin, D.; Stadtmauer, E.A.; Facon, T.; Harousseau, J.L.; Ben-Yehuda, D.; Lonial, S.; Goldschmidt, H.; et al. Bortezomib or high-dose dexamethasone for relapsed multiple myeloma. N. Engl. J. Med. 2005, 352, 2487–2498. [Google Scholar] [CrossRef] [Green Version]
  13. Driscoll, J.J.; Chowdhury, R.D. Molecular crosstalk between the proteasome, aggresomes and autophagy: Translational potential and clinical implications. Cancer Lett. 2012, 325, 147–154. [Google Scholar] [CrossRef] [PubMed]
  14. Yewdell, J.W.; Anton, L.C.; Bennink, J.R. Defective ribosomal products (drips): A major source of antigenic peptides for mhc class i molecules? J. Immunol. 1996, 157, 1823–1826. [Google Scholar]
  15. Yewdell, J.W.; Schubert, U.; Bennink, J.R. At the crossroads of cell biology and immunology: Drips and other sources of peptide ligands for mhc class i molecules. J. Cell Sci. 2001, 114, 845–851. [Google Scholar] [CrossRef] [PubMed]
  16. Cenci, S.; Mezghrani, A.; Cascio, P.; Bianchi, G.; Cerruti, F.; Fra, A.; Lelouard, H.; Masciarelli, S.; Mattioli, L.; Oliva, L.; et al. Progressively impaired proteasomal capacity during terminal plasma cell differentiation. EMBO J. 2006, 25, 1104–1113. [Google Scholar] [CrossRef] [Green Version]
  17. Obeng, E.A.; Carlson, L.M.; Gutman, D.M.; Harrington, W.J., Jr.; Lee, K.P.; Boise, L.H. Proteasome inhibitors induce a terminal unfolded protein response in multiple myeloma cells. Blood 2006, 107, 4907–4916. [Google Scholar] [CrossRef] [Green Version]
  18. Meister, S.; Schubert, U.; Neubert, K.; Herrmann, K.; Burger, R.; Gramatzki, M.; Hahn, S.; Schreiber, S.; Wilhelm, S.; Herrmann, M.; et al. Extensive immunoglobulin production sensitizes myeloma cells for proteasome inhibition. Cancer Res. 2007, 67, 1783–1792. [Google Scholar] [CrossRef] [Green Version]
  19. Hebert, D.N.; Bernasconi, R.; Molinari, M. Erad substrates: Which way out? Semin. Cell Dev. Biol. 2010, 21, 526–532. [Google Scholar] [CrossRef] [PubMed]
  20. Bochtler, T.; Hegenbart, U.; Kunz, C.; Granzow, M.; Benner, A.; Seckinger, A.; Kimmich, C.; Goldschmidt, H.; Ho, A.D.; Hose, D.; et al. Translocation t(11;14) is associated with adverse outcome in patients with newly diagnosed al amyloidosis when treated with bortezomib-based regimens. J. Clin. Oncol. 2015, 33, 1371–1378. [Google Scholar] [CrossRef]
  21. Neefjes, J.; Jongsma, M.L.; Paul, P.; Bakke, O. Towards a systems understanding of mhc class i and mhc class ii antigen presentation. Nat. Rev. Immunol. 2011, 11, 823–836. [Google Scholar] [CrossRef]
  22. Seifert, U.; Bialy, L.P.; Ebstein, F.; Bech-Otschir, D.; Voigt, A.; Schroter, F.; Prozorovski, T.; Lange, N.; Steffen, J.; Rieger, M.; et al. Immunoproteasomes preserve protein homeostasis upon interferon-induced oxidative stress. Cell 2010, 142, 613–624. [Google Scholar] [CrossRef] [Green Version]
  23. Altun, M.; Galardy, P.J.; Shringarpure, R.; Hideshima, T.; LeBlanc, R.; Anderson, K.C.; Ploegh, H.L.; Kessler, B.M. Effects of ps-341 on the activity and composition of proteasomes in multiple myeloma cells. Cancer Res. 2005, 65, 7896–7901. [Google Scholar] [CrossRef] [Green Version]
  24. Goldberg, A.L. Functions of the proteasome: The lysis at the end of the tunnel. Science 1995, 268, 522–523. [Google Scholar] [CrossRef] [PubMed]
  25. Besche, H.C.; Haas, W.; Gygi, S.P.; Goldberg, A.L. Isolation of mammalian 26s proteasomes and p97/vcp complexes using the ubiquitin-like domain from hhr23b reveals novel proteasome-associated proteins. Biochemistry 2009, 48, 2538–2549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Kisselev, A.F.; Goldberg, A.L. Proteasome inhibitors: From research tools to drug candidates. Chem. Biol. 2001, 8, 739–758. [Google Scholar] [CrossRef] [Green Version]
  27. Adams, J. The development of proteasome inhibitors as anticancer drugs. Cancer Cell 2004, 5, 417–421. [Google Scholar] [CrossRef] [Green Version]
  28. Richardson, P.G.; Sonneveld, P.; Schuster, M.W.; Stadtmauer, E.A.; Facon, T.; Harousseau, J.L.; Ben-Yehuda, D.; Lonial, S.; Goldschmidt, H.; Reece, D.; et al. Reversibility of symptomatic peripheral neuropathy with bortezomib in the phase iii apex trial in relapsed multiple myeloma: Impact of a dose-modification guideline. Br. J. Haematol. 2009, 144, 895–903. [Google Scholar] [CrossRef] [PubMed]
  29. San Miguel, J.F.; Schlag, R.; Khuageva, N.K.; Dimopoulos, M.A.; Shpilberg, O.; Kropff, M.; Spicka, I.; Petrucci, M.T.; Palumbo, A.; Samoilova, O.S.; et al. Persistent overall survival benefit and no increased risk of second malignancies with bortezomib-melphalan-prednisone versus melphalan-prednisone in patients with previously untreated multiple myeloma. J. Clin. Oncol. 2013, 31, 448–455. [Google Scholar] [CrossRef] [Green Version]
  30. Ho, M.; Goh, C.Y.; Patel, A.; Staunton, S.; O’Connor, R.; Godeau, M.; Bianchi, G. Role of the bone marrow milieu in multiple myeloma progression and therapeutic resistance. Clin. Lymphoma Myeloma Leuk. 2020, 20, e752–e768. [Google Scholar] [CrossRef]
  31. Bianchi, G.; Ghobrial, I.M. Molecular mechanisms of effectiveness of novel therapies in multiple myeloma. Leuk. Lymphoma 2013, 54, 229–241. [Google Scholar] [CrossRef] [PubMed]
  32. Hideshima, T.; Richardson, P.; Chauhan, D.; Palombella, V.J.; Elliott, P.J.; Adams, J.; Anderson, K.C. The proteasome inhibitor ps-341 inhibits growth, induces apoptosis, and overcomes drug resistance in human multiple myeloma cells. Cancer Res. 2001, 61, 3071–3076. [Google Scholar] [PubMed]
  33. Hideshima, T.; Ikeda, H.; Chauhan, D.; Okawa, Y.; Raje, N.; Podar, K.; Mitsiades, C.; Munshi, N.C.; Richardson, P.G.; Carrasco, R.D.; et al. Bortezomib induces canonical nuclear factor-kappab activation in multiple myeloma cells. Blood 2009, 114, 1046–1052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Hideshima, T.; Mitsiades, C.; Akiyama, M.; Hayashi, T.; Chauhan, D.; Richardson, P.; Schlossman, R.; Podar, K.; Munshi, N.C.; Mitsiades, N.; et al. Molecular mechanisms mediating antimyeloma activity of proteasome inhibitor ps-341. Blood 2003, 101, 1530–1534. [Google Scholar] [CrossRef] [Green Version]
  35. Cenci, S.; Oliva, L.; Cerruti, F.; Milan, E.; Bianchi, G.; Raule, M.; Mezghrani, A.; Pasqualetto, E.; Sitia, R.; Cascio, P. Pivotal advance: Protein synthesis modulates responsiveness of differentiating and malignant plasma cells to proteasome inhibitors. J. Leukoc. Biol. 2012, 92, 921–931. [Google Scholar] [CrossRef] [PubMed]
  36. Oliva, L.; Orfanelli, U.; Resnati, M.; Raimondi, A.; Orsi, A.; Milan, E.; Palladini, G.; Milani, P.; Cerruti, F.; Cascio, P.; et al. The amyloidogenic light chain is a stressor that sensitizes plasma cells to proteasome inhibitor toxicity. Blood 2017, 129, 2132–2142. [Google Scholar] [CrossRef] [PubMed]
  37. Kastritis, E.; Wechalekar, A.D.; Dimopoulos, M.A.; Merlini, G.; Hawkins, P.N.; Perfetti, V.; Gillmore, J.D.; Palladini, G. Bortezomib with or without dexamethasone in primary systemic (light chain) amyloidosis. J. Clin. Oncol. 2010, 28, 1031–1037. [Google Scholar] [CrossRef]
  38. Reece, D.E.; Hegenbart, U.; Sanchorawala, V.; Merlini, G.; Palladini, G.; Blade, J.; Fermand, J.P.; Hassoun, H.; Heffner, L.; Vescio, R.A.; et al. Efficacy and safety of once-weekly and twice-weekly bortezomib in patients with relapsed systemic al amyloidosis: Results of a phase 1/2 study. Blood 2011, 118, 865–873. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Landau, H.; Hassoun, H.; Rosenzweig, M.A.; Maurer, M.; Liu, J.; Flombaum, C.; Bello, C.; Hoover, E.; Riedel, E.; Giralt, S.; et al. Bortezomib and dexamethasone consolidation following risk-adapted melphalan and stem cell transplantation for patients with newly diagnosed light-chain amyloidosis. Leukemia 2013, 27, 823–828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Palladini, G.; Milani, P.; Foli, A.; Vidus Rosin, M.; Basset, M.; Lavatelli, F.; Nuvolone, M.; Obici, L.; Perlini, S.; Merlini, G. Melphalan and dexamethasone with or without bortezomib in newly diagnosed al amyloidosis: A matched case-control study on 174 patients. Leukemia 2014, 28, 2311–2316. [Google Scholar] [CrossRef] [Green Version]
  41. Venner, C.P.; Lane, T.; Foard, D.; Rannigan, L.; Gibbs, S.D.; Pinney, J.H.; Whelan, C.J.; Lachmann, H.J.; Gillmore, J.D.; Hawkins, P.N.; et al. Cyclophosphamide, bortezomib, and dexamethasone therapy in al amyloidosis is associated with high clonal response rates and prolonged progression-free survival. Blood 2012, 119, 4387–4390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Mikhael, J.R.; Schuster, S.R.; Jimenez-Zepeda, V.H.; Bello, N.; Spong, J.; Reeder, C.B.; Stewart, A.K.; Bergsagel, P.L.; Fonseca, R. Cyclophosphamide-bortezomib-dexamethasone (cybord) produces rapid and complete hematologic response in patients with al amyloidosis. Blood 2012, 119, 4391–4394. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Bianchi, G.; Zhang, Y.; Comenzo, R.L. Al amyloidosis: Current chemotherapy and immune therapy treatment strategies: Jacc: Cardiooncology state-of-the-art review. JACC CardioOncol. 2021, 3, 467–487. [Google Scholar] [CrossRef]
  44. Kastritis, E.; Palladini, G.; Minnema, M.C.; Wechalekar, A.D.; Jaccard, A.; Lee, H.C.; Sanchorawala, V.; Gibbs, S.; Mollee, P.; Venner, C.P.; et al. Daratumumab-based treatment for immunoglobulin light-chain amyloidosis. N. Engl. J. Med. 2021, 385, 46–58. [Google Scholar] [CrossRef]
  45. Bianchi, G.; Anderson, K.C. Understanding biology to tackle the disease: Multiple myeloma from bench to bedside, and back. CA Cancer J. Clin. 2014, 64, 422–444. [Google Scholar] [CrossRef]
  46. Dimopoulos, M.A.; Moreau, P.; Palumbo, A.; Joshua, D.; Pour, L.; Hajek, R.; Facon, T.; Ludwig, H.; Oriol, A.; Goldschmidt, H.; et al. Carfilzomib and dexamethasone versus bortezomib and dexamethasone for patients with relapsed or refractory multiple myeloma (endeavor): A randomised, phase 3, open-label, multicentre study. Lancet Oncol. 2016, 17, 27–38. [Google Scholar] [CrossRef]
  47. Stewart, A.K.; Rajkumar, S.V.; Dimopoulos, M.A.; Masszi, T.; Spicka, I.; Oriol, A.; Hajek, R.; Rosinol, L.; Siegel, D.S.; Mihaylov, G.G.; et al. Carfilzomib, lenalidomide, and dexamethasone for relapsed multiple myeloma. N. Engl. J. Med. 2015, 372, 142–152. [Google Scholar] [CrossRef] [PubMed]
  48. Kuhn, D.J.; Chen, Q.; Voorhees, P.M.; Strader, J.S.; Shenk, K.D.; Sun, C.M.; Demo, S.D.; Bennett, M.K.; van Leeuwen, F.W.; Chanan-Khan, A.A.; et al. Potent activity of carfilzomib, a novel, irreversible inhibitor of the ubiquitin-proteasome pathway, against preclinical models of multiple myeloma. Blood 2007, 110, 3281–3290. [Google Scholar] [CrossRef] [Green Version]
  49. Siegel, D.; Martin, T.; Nooka, A.; Harvey, R.D.; Vij, R.; Niesvizky, R.; Badros, A.Z.; Jagannath, S.; McCulloch, L.; Rajangam, K.; et al. Integrated safety profile of single-agent carfilzomib: Experience from 526 patients enrolled in 4 phase ii clinical studies. Haematologica 2013, 98, 1753–1761. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Waxman, A.J.; Clasen, S.; Hwang, W.T.; Garfall, A.; Vogl, D.T.; Carver, J.; O’Quinn, R.; Cohen, A.D.; Stadtmauer, E.A.; Ky, B.; et al. Carfilzomib-associated cardiovascular adverse events: A systematic review and meta-analysis. JAMA Oncol. 2018, 4, e174519. [Google Scholar] [CrossRef] [PubMed]
  51. Cohen, A.D.; Landau, H.; Scott, E.C.; Liedtke, M.; Kaufman, J.L.; Rosenzweig, M.; Gasparetto, C.; Vesole, D.H.; Sanchorawala, V.; Lentzsch, S.; et al. Safety and efficacy of carfilzomib (cfz) in previously-treated systemic light-chain (al) amyloidosis. Blood 2016, 128, 645. [Google Scholar] [CrossRef]
  52. Garg, M.; Hall, A.; Jenner, M.; Kishore, B.; Lachmann, H.J.; Gillmore, J.D.; Pitchford, A.; Flanagan, L.; Oughton, J.B.; Mahmood, S.; et al. A phase 1 study of carfilzomib-thalidomide-dexamethasone in patients with relapsed/refractory al amyloidosis—Catalyst trial results. Blood 2019, 134, 1890. [Google Scholar] [CrossRef]
  53. Manwani, R.; Mahmood, S.; Sachchithanantham, S.; Lachmann, H.J.; Gillmore, J.D.; Yong, K.; Rabin, N.; Popat, R.; Kyriakou, C.; Worthington, S.; et al. Carfilzomib is an effective upfront treatment in al amyloidosis patients with peripheral and autonomic neuropathy. Br. J. Haematol. 2019, 187, 638–641. [Google Scholar] [CrossRef]
  54. Weyburne, E.S.; Wilkins, O.M.; Sha, Z.; Williams, D.A.; Pletnev, A.A.; de Bruin, G.; Overkleeft, H.S.; Goldberg, A.L.; Cole, M.D.; Kisselev, A.F. Inhibition of the proteasome beta2 site sensitizes triple-negative breast cancer cells to beta5 inhibitors and suppresses nrf1 activation. Cell Chem. Biol. 2017, 24, 218–230. [Google Scholar] [CrossRef] [Green Version]
  55. Chauhan, D.; Catley, L.; Li, G.; Podar, K.; Hideshima, T.; Velankar, M.; Mitsiades, C.; Mitsiades, N.; Yasui, H.; Letai, A.; et al. A novel orally active proteasome inhibitor induces apoptosis in multiple myeloma cells with mechanisms distinct from bortezomib. Cancer Cell 2005, 8, 407–419. [Google Scholar] [CrossRef] [Green Version]
  56. Richardson, P.G.; Zimmerman, T.M.; Hofmeister, C.C.; Talpaz, M.; Chanan-Khan, A.A.; Kaufman, J.L.; Laubach, J.P.; Chauhan, D.; Jakubowiak, A.J.; Reich, S.; et al. Phase 1 study of marizomib in relapsed or relapsed and refractory multiple myeloma: Npi-0052–101 part 1. Blood 2016, 127, 2693–2700. [Google Scholar] [CrossRef] [Green Version]
  57. Koizumi, S.; Irie, T.; Hirayama, S.; Sakurai, Y.; Yashiroda, H.; Naguro, I.; Ichijo, H.; Hamazaki, J.; Murata, S. The aspartyl protease ddi2 activates nrf1 to compensate for proteasome dysfunction. Elife 2016, 5, e18357. [Google Scholar] [CrossRef] [PubMed]
  58. Radhakrishnan, S.K.; den Besten, W.; Deshaies, R.J. P97-dependent retrotranslocation and proteolytic processing govern formation of active nrf1 upon proteasome inhibition. Elife 2014, 3, e01856. [Google Scholar] [CrossRef] [PubMed]
  59. Radhakrishnan, S.K.; Lee, C.S.; Young, P.; Beskow, A.; Chan, J.Y.; Deshaies, R.J. Transcription factor nrf1 mediates the proteasome recovery pathway after proteasome inhibition in mammalian cells. Mol. Cell 2010, 38, 17–28. [Google Scholar] [CrossRef] [Green Version]
  60. Vangala, J.R.; Sotzny, F.; Kruger, E.; Deshaies, R.J.; Radhakrishnan, S.K. Nrf1 can be processed and activated in a proteasome-independent manner. Curr. Biol. 2016, 26, R834–R835. [Google Scholar] [CrossRef] [Green Version]
  61. Tomlin, F.M.; Gerling-Driessen, U.I.M.; Liu, Y.C.; Flynn, R.A.; Vangala, J.R.; Lentz, C.S.; Clauder-Muenster, S.; Jakob, P.; Mueller, W.F.; Ordonez-Rueda, D.; et al. Inhibition of ngly1 inactivates the transcription factor nrf1 and potentiates proteasome inhibitor cytotoxicity. ACS Cent. Sci. 2017, 3, 1143–1155. [Google Scholar] [CrossRef]
  62. Chen, T.; Ho, M.; Briere, J.; Moscvin, M.; Czarnecki, P.G.; Anderson, K.C.; Blackwell, T.K.; Bianchi, G. Multiple myeloma cells depend on the ddi2/nrf1-mediated proteasome stress response for survival. Blood Adv. 2022, 6, 429–440. [Google Scholar] [CrossRef] [PubMed]
  63. Kumar, S.K.; Bensinger, W.I.; Zimmerman, T.M.; Reeder, C.B.; Berenson, J.R.; Berg, D.; Hui, A.M.; Gupta, N.; Di Bacco, A.; Yu, J.; et al. Phase 1 study of weekly dosing with the investigational oral proteasome inhibitor ixazomib in relapsed/refractory multiple myeloma. Blood 2014, 124, 1047–1055. [Google Scholar] [CrossRef]
  64. Kumar, S.K.; Berdeja, J.G.; Niesvizky, R.; Lonial, S.; Laubach, J.P.; Hamadani, M.; Stewart, A.K.; Hari, P.; Roy, V.; Vescio, R.; et al. Safety and tolerability of ixazomib, an oral proteasome inhibitor, in combination with lenalidomide and dexamethasone in patients with previously untreated multiple myeloma: An open-label phase 1/2 study. Lancet Oncol. 2014, 15, 1503–1512. [Google Scholar] [CrossRef]
  65. Dispenzieri, A.; Kastritis, E.; Wechalekar, A.D.; Schonland, S.O.; Kim, K.; Sanchorawala, V.; Landau, H.J.; Kwok, F.; Suzuki, K.; Comenzo, R.L.; et al. A randomized phase 3 study of ixazomib-dexamethasone versus physician’s choice in relapsed or refractory al amyloidosis. Leukemia 2022, 36, 225–235. [Google Scholar] [CrossRef]
  66. Ciechanover, A.; Finley, D.; Varshavsky, A. The ubiquitin-mediated proteolytic pathway and mechanisms of energy-dependent intracellular protein degradation. J. Cell. Biochem. 1984, 24, 27–53. [Google Scholar] [CrossRef] [PubMed]
  67. Ying, J.; Zhang, M.; Qiu, X.; Lu, Y. Targeting the neddylation pathway in cells as a potential therapeutic approach for diseases. Cancer Chemother. Pharmacol. 2018, 81, 797–808. [Google Scholar] [CrossRef]
  68. Enchev, R.I.; Schulman, B.A.; Peter, M. Protein neddylation: Beyond cullin-ring ligases. Nat. Rev. Mol. Cell Biol. 2015, 16, 30–44. [Google Scholar] [CrossRef]
  69. McMillin, D.W.; Jacobs, H.M.; Delmore, J.E.; Buon, L.; Hunter, Z.R.; Monrose, V.; Yu, J.; Smith, P.G.; Richardson, P.G.; Anderson, K.C.; et al. Molecular and cellular effects of nedd8-activating enzyme inhibition in myeloma. Mol. Cancer 2012, 11, 942–951. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Shah, J.J.; Jakubowiak, A.J.; O’Connor, O.A.; Orlowski, R.Z.; Harvey, R.D.; Smith, M.R.; Lebovic, D.; Diefenbach, C.; Kelly, K.; Hua, Z.; et al. Phase i study of the novel investigational nedd8-activating enzyme inhibitor pevonedistat (mln4924) in patients with relapsed/refractory multiple myeloma or lymphoma. Clin. Cancer Res. 2016, 22, 34–43. [Google Scholar] [CrossRef] [Green Version]
  71. Kronke, J.; Udeshi, N.D.; Narla, A.; Grauman, P.; Hurst, S.N.; McConkey, M.; Svinkina, T.; Heckl, D.; Comer, E.; Li, X.; et al. Lenalidomide causes selective degradation of ikzf1 and ikzf3 in multiple myeloma cells. Science 2014, 343, 301–305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Lu, G.; Middleton, R.E.; Sun, H.; Naniong, M.; Ott, C.J.; Mitsiades, C.S.; Wong, K.K.; Bradner, J.E.; Kaelin, W.G., Jr. The myeloma drug lenalidomide promotes the cereblon-dependent destruction of ikaros proteins. Science 2014, 343, 305–309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Schapira, M.; Calabrese, M.F.; Bullock, A.N.; Crews, C.M. Targeted protein degradation: Expanding the toolbox. Nat. Rev. Drug Discov. 2019, 18, 949–963. [Google Scholar] [CrossRef] [PubMed]
  74. Ge, C.; Liao, B.; Zhang, L. Abstract 6367: Kpg-818, a novel cereblon modulator, inhibits hematological malignancies in preclinical models. In Proceedings of the American Association for Cancer Research Annual Meeting, San Diego, CA, USA, 24–29 April 2020; Volume 80, p. 6367. [Google Scholar]
  75. Komander, D.; Clague, M.J.; Urbe, S. Breaking the chains: Structure and function of the deubiquitinases. Nat. Rev. Mol. Cell Biol. 2009, 10, 550–563. [Google Scholar] [CrossRef] [PubMed]
  76. Hanna, J.; Leggett, D.S.; Finley, D. Ubiquitin depletion as a key mediator of toxicity by translational inhibitors. Mol. Cell. Biol. 2003, 23, 9251–9261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. D’Arcy, P.; Brnjic, S.; Olofsson, M.H.; Fryknas, M.; Lindsten, K.; De Cesare, M.; Perego, P.; Sadeghi, B.; Hassan, M.; Larsson, R.; et al. Inhibition of proteasome deubiquitinating activity as a new cancer therapy. Nat. Med. 2011, 17, 1636–1640. [Google Scholar] [CrossRef] [PubMed]
  78. Besche, H.C.; Sha, Z.; Kukushkin, N.V.; Peth, A.; Hock, E.M.; Kim, W.; Gygi, S.; Gutierrez, J.A.; Liao, H.; Dick, L.; et al. Autoubiquitination of the 26s proteasome on rpn13 regulates breakdown of ubiquitin conjugates. EMBO J. 2014, 33, 1159–1176. [Google Scholar] [CrossRef]
  79. Song, Y.; Ray, A.; Li, S.; Das, D.S.; Tai, Y.T.; Carrasco, R.D.; Chauhan, D.; Anderson, K.C. Targeting proteasome ubiquitin receptor rpn13 in multiple myeloma. Leukemia 2016, 30, 1877–1886. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Tian, Z.; D’Arcy, P.; Wang, X.; Ray, A.; Tai, Y.T.; Hu, Y.; Carrasco, R.D.; Richardson, P.; Linder, S.; Chauhan, D.; et al. A novel small molecule inhibitor of deubiquitylating enzyme usp14 and uchl5 induces apoptosis in multiple myeloma and overcomes bortezomib resistance. Blood 2014, 123, 706–716. [Google Scholar] [CrossRef] [PubMed]
  81. Wang, X.; Mazurkiewicz, M.; Hillert, E.K.; Olofsson, M.H.; Pierrou, S.; Hillertz, P.; Gullbo, J.; Selvaraju, K.; Paulus, A.; Akhtar, S.; et al. The proteasome deubiquitinase inhibitor vlx1570 shows selectivity for ubiquitin-specific protease-14 and induces apoptosis of multiple myeloma cells. Sci. Rep. 2016, 6, 26979. [Google Scholar] [CrossRef] [PubMed]
  82. Chauhan, D.; Tian, Z.; Nicholson, B.; Kumar, K.G.; Zhou, B.; Carrasco, R.; McDermott, J.L.; Leach, C.A.; Fulcinniti, M.; Kodrasov, M.P.; et al. A small molecule inhibitor of ubiquitin-specific protease-7 induces apoptosis in multiple myeloma cells and overcomes bortezomib resistance. Cancer Cell 2012, 22, 345–358. [Google Scholar] [CrossRef] [Green Version]
  83. Auner, H.W.; Moody, A.M.; Ward, T.H.; Kraus, M.; Milan, E.; May, P.; Chaidos, A.; Driessen, C.; Cenci, S.; Dazzi, F.; et al. Combined inhibition of p97 and the proteasome causes lethal disruption of the secretory apparatus in multiple myeloma cells. PLoS ONE 2013, 8, e74415. [Google Scholar] [CrossRef] [Green Version]
  84. Le Moigne, R.; Aftab, B.T.; Djakovic, S.; Dhimolea, E.; Valle, E.; Murnane, M.; King, E.M.; Soriano, F.; Menon, M.K.; Wu, Z.Y.; et al. The p97 inhibitor cb-5083 is a unique disrupter of protein homeostasis in models of multiple myeloma. Mol. Cancer Ther. 2017, 16, 2375–2386. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Levine, B.; Kroemer, G. Autophagy in the pathogenesis of disease. Cell 2008, 132, 27–42. [Google Scholar] [CrossRef] [Green Version]
  86. Pengo, N.; Scolari, M.; Oliva, L.; Milan, E.; Mainoldi, F.; Raimondi, A.; Fagioli, C.; Merlini, A.; Mariani, E.; Pasqualetto, E.; et al. Plasma cells require autophagy for sustainable immunoglobulin production. Nat. Immunol. 2013, 14, 298–305. [Google Scholar] [CrossRef] [PubMed]
  87. Milan, E.; Perini, T.; Resnati, M.; Orfanelli, U.; Oliva, L.; Raimondi, A.; Cascio, P.; Bachi, A.; Marcatti, M.; Ciceri, F.; et al. A plastic sqstm1/p62-dependent autophagic reserve maintains proteostasis and determines proteasome inhibitor susceptibility in multiple myeloma cells. Autophagy 2015, 11, 1161–1178. [Google Scholar] [CrossRef]
  88. Moscvin, M.; Ho, M.; Bianchi, G. Overcoming drug resistance by targeting protein homeostasis in multiple myeloma. Cancer Drug Resist. 2021, 4, 1027–1045. [Google Scholar] [CrossRef]
  89. Michallet, A.S.; Mondiere, P.; Taillardet, M.; Leverrier, Y.; Genestier, L.; Defrance, T. Compromising the unfolded protein response induces autophagy-mediated cell death in multiple myeloma cells. PLoS ONE 2011, 6, e25820. [Google Scholar] [CrossRef] [Green Version]
  90. Kawaguchi, T.; Miyazawa, K.; Moriya, S.; Ohtomo, T.; Che, X.F.; Naito, M.; Itoh, M.; Tomoda, A. Combined treatment with bortezomib plus bafilomycin a1 enhances the cytocidal effect and induces endoplasmic reticulum stress in u266 myeloma cells: Crosstalk among proteasome, autophagy-lysosome and er stress. Int. J. Oncol. 2011, 38, 643–654. [Google Scholar]
  91. Mizushima, N.; Komatsu, M. Autophagy: Renovation of cells and tissues. Cell 2011, 147, 728–741. [Google Scholar] [CrossRef] [Green Version]
  92. Denton, D.; Kumar, S. Autophagy-dependent cell death. Cell Death Differ. 2019, 26, 605–616. [Google Scholar] [CrossRef] [Green Version]
  93. Lamy, L.; Ngo, V.N.; Emre, N.C.; Shaffer, A.L., 3rd; Yang, Y.; Tian, E.; Nair, V.; Kruhlak, M.J.; Zingone, A.; Landgren, O.; et al. Control of autophagic cell death by caspase-10 in multiple myeloma. Cancer Cell 2013, 23, 435–449. [Google Scholar] [CrossRef] [Green Version]
  94. Milan, E.; Fabbri, M.; Cenci, S. Autophagy in plasma cell ontogeny and malignancy. J. Clin. Immunol. 2016, 36 (Suppl. 1), 18–24. [Google Scholar] [CrossRef] [Green Version]
  95. Hoang, B.; Benavides, A.; Shi, Y.; Frost, P.; Lichtenstein, A. Effect of autophagy on multiple myeloma cell viability. Mol. Cancer Ther. 2009, 8, 1974–1984. [Google Scholar] [CrossRef] [Green Version]
  96. Yu, L.; Alva, A.; Su, H.; Dutt, P.; Freundt, E.; Welsh, S.; Baehrecke, E.H.; Lenardo, M.J. Regulation of an atg7-beclin 1 program of autophagic cell death by caspase-8. Science 2004, 304, 1500–1502. [Google Scholar] [CrossRef]
  97. Baranowska, K.; Misund, K.; Starheim, K.K.; Holien, T.; Johansson, I.; Darvekar, S.; Buene, G.; Waage, A.; Bjorkoy, G.; Sundan, A. Hydroxychloroquine potentiates carfilzomib toxicity towards myeloma cells. Oncotarget 2016, 7, 70845–70856. [Google Scholar] [CrossRef] [Green Version]
  98. Vogl, D.T.; Stadtmauer, E.A.; Tan, K.S.; Heitjan, D.F.; Davis, L.E.; Pontiggia, L.; Rangwala, R.; Piao, S.; Chang, Y.C.; Scott, E.C.; et al. Combined autophagy and proteasome inhibition: A phase 1 trial of hydroxychloroquine and bortezomib in patients with relapsed/refractory myeloma. Autophagy 2014, 10, 1380–1390. [Google Scholar] [CrossRef] [Green Version]
  99. Guan, J.; Mishra, S.; Qiu, Y.; Shi, J.; Trudeau, K.; Las, G.; Liesa, M.; Shirihai, O.S.; Connors, L.H.; Seldin, D.C.; et al. Lysosomal dysfunction and impaired autophagy underlie the pathogenesis of amyloidogenic light chain-mediated cardiotoxicity. EMBO Mol. Med. 2014, 6, 1493–1507. [Google Scholar] [CrossRef]
  100. Dispenzieri, A. The yin and yang of autophagy in al amyloidosis. Blood 2017, 129, 2044–2045. [Google Scholar] [CrossRef] [Green Version]
  101. Bole, D.G.; Hendershot, L.M.; Kearney, J.F. Posttranslational association of immunoglobulin heavy chain binding protein with nascent heavy chains in nonsecreting and secreting hybridomas. J. Cell Biol. 1986, 102, 1558–1566. [Google Scholar] [CrossRef] [Green Version]
  102. Werner, E.D.; Brodsky, J.L.; McCracken, A.A. Proteasome-dependent endoplasmic reticulum-associated protein degradation: An unconventional route to a familiar fate. Proc. Natl. Acad. Sci. USA 1996, 93, 13797–13801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Kopito, R.R.; Sitia, R. Aggresomes and russell bodies. Symptoms of cellular indigestion? EMBO Rep. 2000, 1, 225–231. [Google Scholar] [CrossRef] [Green Version]
  104. Kawaguchi, Y.; Kovacs, J.J.; McLaurin, A.; Vance, J.M.; Ito, A.; Yao, T.P. The deacetylase hdac6 regulates aggresome formation and cell viability in response to misfolded protein stress. Cell 2003, 115, 727–738. [Google Scholar] [CrossRef] [Green Version]
  105. Hideshima, T.; Qi, J.; Paranal, R.M.; Tang, W.; Greenberg, E.; West, N.; Colling, M.E.; Estiu, G.; Mazitschek, R.; Perry, J.A.; et al. Discovery of selective small-molecule hdac6 inhibitor for overcoming proteasome inhibitor resistance in multiple myeloma. Proc. Natl. Acad. Sci. USA 2016, 113, 13162–13167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Lee, S.W.; Yeon, S.K.; Kim, G.W.; Lee, D.H.; Jeon, Y.H.; Yoo, J.; Kim, S.Y.; Kwon, S.H. Hdac6-selective inhibitor overcomes bortezomib resistance in multiple myeloma. Int. J. Mol. Sci. 2021, 22, 1341. [Google Scholar] [CrossRef] [PubMed]
  107. Santo, L.; Hideshima, T.; Kung, A.L.; Tseng, J.-C.C.; Tamang, D.; Yang, M.; Jarpe, M.; van Duzer, J.H.; Mazitschek, R.; Ogier, W.C.; et al. Preclinical activity, pharmacodynamic, and pharmacokinetic properties of a selective hdac6 inhibitor, acy-1215, in combination with bortezomib in multiple myeloma. Blood 2012, 119, 2579–2589. [Google Scholar] [CrossRef]
  108. Laubach, J.P.; Schjesvold, F.; Mariz, M.; Dimopoulos, M.A.; Lech-Maranda, E.; Spicka, I.; Hungria, V.T.M.; Shelekhova, T.; Abdo, A.; Jacobasch, L.; et al. Efficacy and safety of oral panobinostat plus subcutaneous bortezomib and oral dexamethasone in patients with relapsed or relapsed and refractory multiple myeloma (panorama 3): An open-label, randomised, phase 2 study. Lancet Oncol. 2021, 22, 142–154. [Google Scholar] [CrossRef]
  109. San-Miguel, J.F.; Hungria, V.T.; Yoon, S.S.; Beksac, M.; Dimopoulos, M.A.; Elghandour, A.; Jedrzejczak, W.W.; Gunther, A.; Nakorn, T.N.; Siritanaratkul, N.; et al. Panobinostat plus bortezomib and dexamethasone versus placebo plus bortezomib and dexamethasone in patients with relapsed or relapsed and refractory multiple myeloma: A multicentre, randomised, double-blind phase 3 trial. Lancet Oncol. 2014, 15, 1195–1206. [Google Scholar] [CrossRef]
  110. Yee, A.J.; Bensinger, W.I.; Supko, J.G.; Voorhees, P.M.; Berdeja, J.G.; Richardson, P.G.; Libby, E.N.; Wallace, E.E.; Birrer, N.E.; Burke, J.N.; et al. Ricolinostat plus lenalidomide, and dexamethasone in relapsed or refractory multiple myeloma: A multicentre phase 1b trial. Lancet Oncol. 2016, 17, 1569–1578. [Google Scholar] [CrossRef]
  111. Vogl, D.T.; Raje, N.; Jagannath, S.; Richardson, P.; Hari, P.; Orlowski, R.; Supko, J.G.; Tamang, D.; Yang, M.; Jones, S.S.; et al. Ricolinostat, the first selective histone deacetylase 6 inhibitor, in combination with bortezomib and dexamethasone for relapsed or refractory multiple myeloma. Clin. Cancer Res. 2017, 23, 3307–3315. [Google Scholar] [CrossRef] [Green Version]
  112. Kim, I.; Xu, W.; Reed, J.C. Cell death and endoplasmic reticulum stress: Disease relevance and therapeutic opportunities. Nat. Rev. Drug Discov. 2008, 7, 1013–1030. [Google Scholar] [CrossRef]
  113. Woo, C.W.; Cui, D.; Arellano, J.; Dorweiler, B.; Harding, H.; Fitzgerald, K.A.; Ron, D.; Tabas, I. Adaptive suppression of the atf4–chop branch of the unfolded protein response by toll-like receptor signalling. Nat. Cell Biol. 2009, 11, 1473–1480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Yoshida, H.; Matsui, T.; Yamamoto, A.; Okada, T.; Mori, K. Xbp1 mrna is induced by atf6 and spliced by ire1 in response to er stress to produce a highly active transcription factor. Cell 2001, 107, 881–891. [Google Scholar] [CrossRef] [Green Version]
  115. Lee, A.-H.H.; Iwakoshi, N.N.; Glimcher, L.H. Xbp-1 regulates a subset of endoplasmic reticulum resident chaperone genes in the unfolded protein response. Mol. Cell. Biol. 2003, 23, 7448–7459. [Google Scholar] [CrossRef] [Green Version]
  116. Leung-Hagesteijn, C.; Erdmann, N.; Cheung, G.; Keats, J.J.; Stewart, A.K.; Reece, D.E.; Chung, K.C.; Tiedemann, R.E. Xbp1s-negative tumor b cells and pre-plasmablasts mediate therapeutic proteasome inhibitor resistance in multiple myeloma. Cancer Cell 2013, 24, 289–304. [Google Scholar] [CrossRef] [Green Version]
  117. Papandreou, I.; Denko, N.C.; Olson, M.; Van Melckebeke, H.; Lust, S.; Tam, A.; Solow-Cordero, D.E.; Bouley, D.M.; Offner, F.; Niwa, M.; et al. Identification of an ire1alpha endonuclease specific inhibitor with cytotoxic activity against human multiple myeloma. Blood 2011, 117, 1311–1314. [Google Scholar] [CrossRef] [Green Version]
  118. Ali, M.M.; Bagratuni, T.; Davenport, E.L.; Nowak, P.R.; Silva-Santisteban, M.C.; Hardcastle, A.; McAndrews, C.; Rowlands, M.G.; Morgan, G.J.; Aherne, W.; et al. Structure of the ire1 autophosphorylation complex and implications for the unfolded protein response. EMBO J. 2011, 30, 894–905. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Kraus, M.; Bader, J.; Overkleeft, H.; Driessen, C. Nelfinavir augments proteasome inhibition by bortezomib in myeloma cells and overcomes bortezomib and carfilzomib resistance. Blood Cancer J. 2013, 3, e103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Driessen, C.; Muller, R.; Novak, U.; Cantoni, N.; Betticher, D.; Mach, N.; Rufer, A.; Mey, U.; Samaras, P.; Ribi, K.; et al. Promising activity of nelfinavir-bortezomib-dexamethasone in proteasome inhibitor-refractory multiple myeloma. Blood 2018, 132, 2097–2100. [Google Scholar] [CrossRef] [PubMed]
  121. Li, J.; Lee, A.S. Stress induction of grp78/bip and its role in cancer. Curr. Mol. Med. 2006, 6, 45–54. [Google Scholar] [CrossRef] [PubMed]
  122. Arap, M.A.; Lahdenranta, J.; Mintz, P.J.; Hajitou, A.; Sarkis, A.S.; Arap, W.; Pasqualini, R. Cell surface expression of the stress response chaperone grp78 enables tumor targeting by circulating ligands. Cancer Cell 2004, 6, 275–284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Rasche, L.; Duell, J.; Morgner, C.; Chatterjee, M.; Hensel, F.; Rosenwald, A.; Einsele, H.; Topp, M.S.; Brandlein, S. The natural human igm antibody pat-sm6 induces apoptosis in primary human multiple myeloma cells by targeting heat shock protein grp78. PLoS ONE 2013, 8, e63414. [Google Scholar] [CrossRef] [PubMed]
  124. Shoulders, M.D.; Ryno, L.M.; Genereux, J.C.; Moresco, J.J.; Tu, P.G.; Wu, C.; Yates, J.R., 3rd; Su, A.I.; Kelly, J.W.; Wiseman, R.L. Stress-independent activation of xbp1s and/or atf6 reveals three functionally diverse er proteostasis environments. Cell Rep. 2013, 3, 1279–1292. [Google Scholar] [CrossRef] [Green Version]
  125. Chen, J.J.; Genereux, J.C.; Suh, E.H.; Vartabedian, V.F.; Rius, B.; Qu, S.; Dendle, M.T.A.; Kelly, J.W.; Wiseman, R.L. Endoplasmic reticulum proteostasis influences the oligomeric state of an amyloidogenic protein secreted from mammalian cells. Cell Chem. Biol. 2016, 23, 1282–1293. [Google Scholar] [CrossRef] [Green Version]
  126. Plate, L.; Cooley, C.B.; Chen, J.J.; Paxman, R.J.; Gallagher, C.M.; Madoux, F.; Genereux, J.C.; Dobbs, W.; Garza, D.; Spicer, T.P.; et al. Small molecule proteostasis regulators that reprogram the er to reduce extracellular protein aggregation. Elife 2016, 5, e15550. [Google Scholar] [CrossRef] [PubMed]
  127. Rius, B.; Mesgarzadeh, J.S.; Romine, I.C.; Paxman, R.J.; Kelly, J.W.; Wiseman, R.L. Pharmacologic targeting of plasma cell endoplasmic reticulum proteostasis to reduce amyloidogenic light chain secretion. Blood Adv. 2021, 5, 1037–1049. [Google Scholar] [CrossRef]
  128. Hartl, F.U. Molecular chaperones in cellular protein folding. Nature 1996, 381, 571–579. [Google Scholar] [CrossRef]
  129. Wang, B.; Chen, Z.; Yu, F.; Chen, Q.; Tian, Y.; Ma, S.; Wang, T.; Liu, X. Hsp90 regulates autophagy and plays a role in cancer therapy. Tumour Biol. 2016, 37, 1–6. [Google Scholar] [CrossRef]
  130. Chiang, H.L.; Terlecky, S.R.; Plant, C.P.; Dice, J.F. A role for a 70-kilodalton heat shock protein in lysosomal degradation of intracellular proteins. Science 1989, 246, 382–385. [Google Scholar] [CrossRef] [PubMed]
  131. Boysen, M.; Kityk, R.; Mayer, M.P. Hsp70- and hsp90-mediated regulation of the conformation of p53 DNA binding domain and p53 cancer variants. Mol. Cell 2019, 74, 831–843 e834. [Google Scholar] [CrossRef]
  132. Kimura, H.; Caturegli, P.; Takahashi, M.; Suzuki, K. New insights into the function of the immunoproteasome in immune and nonimmune cells. J. Immunol. Res. 2015, 2015, 541984. [Google Scholar] [CrossRef] [Green Version]
  133. Marcu, M.G.; Doyle, M.; Bertolotti, A.; Ron, D.; Hendershot, L.; Neckers, L. Heat shock protein 90 modulates the unfolded protein response by stabilizing ire1alpha. Mol. Cell Biol. 2002, 22, 8506–8513. [Google Scholar] [CrossRef] [Green Version]
  134. Chatterjee, M.; Andrulis, M.; Stuhmer, T.; Muller, E.; Hofmann, C.; Steinbrunn, T.; Heimberger, T.; Schraud, H.; Kressmann, S.; Einsele, H.; et al. The pi3k/akt signaling pathway regulates the expression of hsp70, which critically contributes to hsp90-chaperone function and tumor cell survival in multiple myeloma. Haematologica 2013, 98, 1132–1141. [Google Scholar] [CrossRef] [PubMed]
  135. Braunstein, M.J.; Scott, S.S.; Scott, C.M.; Behrman, S.; Walter, P.; Wipf, P.; Coplan, J.D.; Chrico, W.; Joseph, D.; Brodsky, J.L.; et al. Antimyeloma effects of the heat shock protein 70 molecular chaperone inhibitor mal3–101. J. Oncol. 2011, 2011, 232037. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Usmani, S.Z.; Chiosis, G. Hsp90 inhibitors as therapy for multiple myeloma. Clin. Lymphoma Myeloma Leuk. 2011, 11 (Suppl. 1), S77–S81. [Google Scholar] [CrossRef]
  137. Seggewiss-Bernhardt, R.; Bargou, R.C.; Goh, Y.T.; Stewart, A.K.; Spencer, A.; Alegre, A.; Blade, J.; Ottmann, O.G.; Fernandez-Ibarra, C.; Lu, H.; et al. Phase 1/1b trial of the heat shock protein 90 inhibitor nvp-auy922 as monotherapy or in combination with bortezomib in patients with relapsed or refractory multiple myeloma. Cancer 2015, 121, 2185–2192. [Google Scholar] [CrossRef]
  138. Cavenagh, J.; Oakervee, H.; Baetiong-Caguioa, P.; Davies, F.; Gharibo, M.; Rabin, N.; Kurman, M.; Novak, B.; Shiraishi, N.; Nakashima, D.; et al. A phase i/ii study of kw-2478, an hsp90 inhibitor, in combination with bortezomib in patients with relapsed/refractory multiple myeloma. Br. J. Cancer 2017, 117, 1295–1302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Reddy, N.; Voorhees, P.M.; Houk, B.E.; Brega, N.; Hinson, J.M., Jr.; Jillela, A. Phase i trial of the hsp90 inhibitor pf-04929113 (snx5422) in adult patients with recurrent, refractory hematologic malignancies. Clin. Lymphoma Myeloma Leuk. 2013, 13, 385–391. [Google Scholar] [CrossRef] [PubMed]
  140. Richardson, P.G.; Badros, A.Z.; Jagannath, S.; Tarantolo, S.; Wolf, J.L.; Albitar, M.; Berman, D.; Messina, M.; Anderson, K.C. Tanespimycin with bortezomib: Activity in relapsed/refractory patients with multiple myeloma. Br. J. Haematol. 2010, 150, 428–437. [Google Scholar] [CrossRef] [Green Version]
  141. Siegel, D.; Jagannath, S.; Vesole, D.H.; Borello, I.; Mazumder, A.; Mitsiades, C.; Goddard, J.; Dunbar, J.; Normant, E.; Adams, J.; et al. A phase 1 study of ipi-504 (retaspimycin hydrochloride) in patients with relapsed or relapsed and refractory multiple myeloma. Leuk. Lymphoma 2011, 52, 2308–2315. [Google Scholar] [CrossRef] [PubMed]
  142. Wyatt, A.R.; Yerbury, J.J.; Ecroyd, H.; Wilson, M.R. Extracellular chaperones and proteostasis. Annu. Rev. Biochem. 2013, 82, 295–322. [Google Scholar] [CrossRef] [Green Version]
  143. Genereux, J.C.; Qu, S.; Zhou, M.; Ryno, L.M.; Wang, S.; Shoulders, M.D.; Kaufman, R.J.; Lasmezas, C.I.; Kelly, J.W.; Wiseman, R.L. Unfolded protein response-induced erdj3 secretion links er stress to extracellular proteostasis. EMBO J. 2015, 34, 4–19. [Google Scholar] [CrossRef] [PubMed]
  144. Coker, A.R.; Purvis, A.; Baker, D.; Pepys, M.B.; Wood, S.P. Molecular chaperone properties of serum amyloid p component. FEBS Lett. 2000, 473, 199–202. [Google Scholar] [CrossRef]
  145. Tennent, G.A.; Lovat, L.B.; Pepys, M.B. Serum amyloid p component prevents proteolysis of the amyloid fibrils of alzheimer disease and systemic amyloidosis. Proc. Natl. Acad. Sci. USA 1995, 92, 4299–4303. [Google Scholar] [CrossRef] [Green Version]
  146. Richards, D.B.; Cookson, L.M.; Berges, A.C.; Barton, S.V.; Lane, T.; Ritter, J.M.; Fontana, M.; Moon, J.C.; Pinzani, M.; Gillmore, J.D.; et al. Therapeutic clearance of amyloid by antibodies to serum amyloid p component. N. Engl. J. Med. 2015, 373, 1106–1114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Richards, D.B.; Cookson, L.M.; Barton, S.V.; Liefaard, L.; Lane, T.; Hutt, D.F.; Ritter, J.M.; Fontana, M.; Moon, J.C.; Gillmore, J.D.; et al. Repeat doses of antibody to serum amyloid p component clear amyloid deposits in patients with systemic amyloidosis. Sci. Transl. Med. 2018, 10, eaan3128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Ohno, S.; Yoshimoto, M.; Honda, S.; Miyachi, S.; Ishida, T.; Itoh, F.; Endo, T.; Chiba, S.; Imai, K. The antisense approach in amyloid light chain amyloidosis: Identification of monoclonal ig and inhibition of its production by antisense oligonucleotides in in vitro and in vivo models. J. Immunol. 2002, 169, 4039–4045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Zhou, P.; Ma, X.; Iyer, L.; Chaulagain, C.; Comenzo, R.L. One sirna pool targeting the lambda constant region stops lambda light-chain production and causes terminal endoplasmic reticulum stress. Blood 2014, 123, 3440–3451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Moscvin, M.; Chen, T.; Czarnecki, P.G.; Gulla, A.; Anderson, K.C.; Bianchi, G. Targeting free light chain secretion via botulinum neurotoxin is a novel therapeutic strategy in al amyloidosis by inducing a terminal unfolded protein response. Blood 2021, 138, 1576. [Google Scholar] [CrossRef]
  151. Winter, G.E.; Buckley, D.L.; Paulk, J.; Roberts, J.M.; Souza, A.; Dhe-Paganon, S.; Bradner, J.E. Drug development. Phthalimide conjugation as a strategy for in vivo target protein degradation. Science 2015, 348, 1376–1381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Nabet, B.; Roberts, J.M.; Buckley, D.L.; Paulk, J.; Dastjerdi, S.; Yang, A.; Leggett, A.L.; Erb, M.A.; Lawlor, M.A.; Souza, A.; et al. The dtag system for immediate and target-specific protein degradation. Nat. Chem. Biol. 2018, 14, 431–441. [Google Scholar] [CrossRef] [PubMed]
  153. Delmore, J.E.; Issa, G.C.; Lemieux, M.E.; Rahl, P.B.; Shi, J.; Jacobs, H.M.; Kastritis, E.; Gilpatrick, T.; Paranal, R.M.; Qi, J.; et al. Bet bromodomain inhibition as a therapeutic strategy to target c-myc. Cell 2011, 146, 904–917. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. The Proteostasis Network. The schema outlines the complex proteostasis network in an AL-PC following the fate of the immunoglobulin (Ig) free light chain (FLC) as an example of highly synthesized protein. For secretory proteins, four proteostasis compartments regulating distinct biological functions can be identified (dashed circles): protein synthesis (1), folding (2), secretion (3) and degradation (4). Critical organelles and proteins participating in the ER proteostasis are outlined in capital letters. Crosstalk with the three ER receptors (IRE1α, ATF6α and PERK) mediating the unfolded protein response (UPR) is also outlined. Abbreviations: mRNA: messenger RNA; ER: endoplasmic reticulum; IgL: immunoglobulin light chain; BIP: Ig binding protein, also known as GRP78, glucose regulated protein 78; FLC: free light chain; UPR: unfolded protein response; ERAD: ER-associated degradation; VCP: valosin-containing protein, also known as p97; Ub: ubiquitin; ROS: reactive oxygen species; HDAC6: histone deacetylase 6; DUB: deubiquitylating enzyme; E1: Ub activating enzyme; E2: Ub conjugating enzyme; E3: Ub ligase enzymes.
Figure 1. The Proteostasis Network. The schema outlines the complex proteostasis network in an AL-PC following the fate of the immunoglobulin (Ig) free light chain (FLC) as an example of highly synthesized protein. For secretory proteins, four proteostasis compartments regulating distinct biological functions can be identified (dashed circles): protein synthesis (1), folding (2), secretion (3) and degradation (4). Critical organelles and proteins participating in the ER proteostasis are outlined in capital letters. Crosstalk with the three ER receptors (IRE1α, ATF6α and PERK) mediating the unfolded protein response (UPR) is also outlined. Abbreviations: mRNA: messenger RNA; ER: endoplasmic reticulum; IgL: immunoglobulin light chain; BIP: Ig binding protein, also known as GRP78, glucose regulated protein 78; FLC: free light chain; UPR: unfolded protein response; ERAD: ER-associated degradation; VCP: valosin-containing protein, also known as p97; Ub: ubiquitin; ROS: reactive oxygen species; HDAC6: histone deacetylase 6; DUB: deubiquitylating enzyme; E1: Ub activating enzyme; E2: Ub conjugating enzyme; E3: Ub ligase enzymes.
Hemato 03 00022 g001
Figure 2. Molecular targets within the proteostasis network for the treatment of MM and AL amyloidosis. In the center of the cartoon is a clonal plasma cells (MM and/or AL amyloidosis plasma cells); also outlined is the nucleus and the endoplasmic reticulum with associated ribosomes (red dots). Elements of the proteostatic network that represent appealing molecular targets for anti-AL amyloidosis/MM therapy are marked by numbers: 1—HSP90 chaperone proteins; 2—ER chaperone GRP78 that is also expressed on the cell surface, representing a target for antibody mediated, complement-induced cytotoxicity (CDC); 3—unfolded protein response sensors (IRE1, PERK and ATF6); 4—the proteasome; 5—aggresome master regulator HDAC6; 6—autolysosome; 7—ubiquitin activating (E1), conjugating (E2) and ligase (E3) enzymes; 8—NEDD8 enzyme (N); 9—cereblon (CRBN)-mediated IKZF1 and IKZF3 proteolysis; 10—deubiquitylating enzymes (DUBs) and Ub receptors; FDA-approved agents are outlined in green while investigational agents are in red. Key components of the proteostasis network are outlined with associated FDA approved (in green) or investigational agents (in red) listed in Table 1. Arrows signify positive activation/induction while T-shaped lines signify inhibition. Dotted lines signify modulation/interaction. Abbreviations: HSP90: heat shock protein 90; GRP78: glucose-regulated protein 78; C1q: Complement Component 1, Q Subcomponent; HDAC6: histone deacetylase 6; CRBN: cereblon; DDB1: DNA damage-binding 1; IKZF1: Ikaros family zinc finger protein 1; IKZf3: Ikaros family zinc finger protein 3; MTOC: microtubule organizing center.
Figure 2. Molecular targets within the proteostasis network for the treatment of MM and AL amyloidosis. In the center of the cartoon is a clonal plasma cells (MM and/or AL amyloidosis plasma cells); also outlined is the nucleus and the endoplasmic reticulum with associated ribosomes (red dots). Elements of the proteostatic network that represent appealing molecular targets for anti-AL amyloidosis/MM therapy are marked by numbers: 1—HSP90 chaperone proteins; 2—ER chaperone GRP78 that is also expressed on the cell surface, representing a target for antibody mediated, complement-induced cytotoxicity (CDC); 3—unfolded protein response sensors (IRE1, PERK and ATF6); 4—the proteasome; 5—aggresome master regulator HDAC6; 6—autolysosome; 7—ubiquitin activating (E1), conjugating (E2) and ligase (E3) enzymes; 8—NEDD8 enzyme (N); 9—cereblon (CRBN)-mediated IKZF1 and IKZF3 proteolysis; 10—deubiquitylating enzymes (DUBs) and Ub receptors; FDA-approved agents are outlined in green while investigational agents are in red. Key components of the proteostasis network are outlined with associated FDA approved (in green) or investigational agents (in red) listed in Table 1. Arrows signify positive activation/induction while T-shaped lines signify inhibition. Dotted lines signify modulation/interaction. Abbreviations: HSP90: heat shock protein 90; GRP78: glucose-regulated protein 78; C1q: Complement Component 1, Q Subcomponent; HDAC6: histone deacetylase 6; CRBN: cereblon; DDB1: DNA damage-binding 1; IKZF1: Ikaros family zinc finger protein 1; IKZf3: Ikaros family zinc finger protein 3; MTOC: microtubule organizing center.
Hemato 03 00022 g002
Table 1. Investigational agents targeting protein homeostasis in plasma cell disorders. This table outlines promising agents targeting protein homeostasis in advanced preclinical or early clinical development in MM.
Table 1. Investigational agents targeting protein homeostasis in plasma cell disorders. This table outlines promising agents targeting protein homeostasis in advanced preclinical or early clinical development in MM.
Drug NameChemical Structure/Mechanisms of ActionStage of DevelopmentNotes
Proteasome Inhibitors
Bortezomib
  • Boronic acid
  • Reversible inhibition of β5 > β1
  • Leads to accumulation of polyUb proteins and proteotoxic death
  • Induces immunogenic cell death
FDA approved as first line in MM and AL amyloidosisPeripheral neuropathy is an impactful side effect
Carfilzomib
  • Epoxyketone
  • Irreversible inhibition of β5
  • Leads to accumulation of polyUb proteins and proteotoxic death
FDA approved as second line in MMCardiovascular adverse events
Ixazomib
  • Orally bioavailable boronic acid
  • Reversible inhibition of β5 > β1
  • Leads to accumulation of polyUb proteins and proteotoxic death
FDA approved as second line in MMGastrointestinal side effects and peripheral neuropathy
Marizomib
  • β-lactone-γ-lactam
  • Irreversible inhibition of β5, β1 and βs
Early phase clinical trials in CNS MM plannedCNS side effects
E3 Ubiquitin ligase modulators
IMiDs (thalidomide, lenalidomide, pomalidomide
  • Cereblon binding and redirection to the selective degradation of IKZF1 and IKZF3
FDA approved across of line of treatment in MM
  • Increased incidence of venous thromboembolic events when used in combination
  • Class X drug in pregnancy
  • Secondary hematologic neoplasms observed post IMiD + alkylator combination
KPG818
  • Small molecule binding to cereblon and modulating E3 ligase activity
Phase 1 clinical trial in hematologic malignancies
TAS4464, MLN4924
  • NEDD8 inhibitors
  • Inhibits neddylation-mediated activation of Cullin-RING E3 Ub ligases
Phase 1 clinical trials in MMTerminated due to liver toxicity (TAS4464) or lack of efficacy (MLN4924)
DUB Inhibitors
VLX-1570
  • Inhibits UCHL5
Phase 1 clinical trial in combination with dex in RRMMTerminated due to grade 5 toxicity(lung toxicity).
P5091, XL177A
  • Inhibits USP7 activity
Preclinical
B-AP15
  • Inhibits USP14 and UCHL5
Preclinical
RA190
  • Inhibits RPN13 and UCHL37
Preclinical
ERAD inhibitors
CB-5083
  • Inhibits p97 inhibition
  • Leads to polyUb protein accumulation
  • Induces terminal UPR
Phase I
terminated
Photophobia and dyschromatopsia reported and probably related to off target effect on PDE6
Autophagy Modulators
ChloroquineAlkalinizes lysosomal pH and inhibits autophagosome–lysosome fusionPhase I/II studies in combination with PI-based backbones in RRMM have been completedInsufficient clinical activity to warrant further development
Aggresome Inhibitors
Ricolinostat
(ACY-1215)
Inhibits HDAC6 and thus aggresome formationPhase I and II studies in combination with IMiDs or bortezomib completed or currently ongoing
Citarinostat
(ACY-241)
Inhibits HDAC6 and thus aggresome formationIn phase I study in combination with PVX-410 vaccine and lenalidomide in smoldering MM
UPR Modulators
Nelfinavir
  • Modulates membrane lipid bilayer composition
  • Induces UPR
Phase I/II studies in combination with lenalidomide or bortezomib in RRMM completedClinical activity shown in lenalidomide or bortezomib refractory MM
PAT-SM6
  • Anti-GRP78 monoclonal IgM antibody
  • Induces UPR
  • Induces complement dependent cytotoxicity
Phase I
Completed in RRMM
Safe with best response SD as single agent
Compound 147
  • Covalent modification of PDIs
  • Reduces secretion of amyloidogenic FLC
Preclinical
HSP Inhibitors
KW-2478
  • HSP90 inhibitor
  • Apoptosis
Phase II
completed
Further clinical development halted due to severe toxicities and/or lack of clinical benefit
NVP-AUY922
  • HSP90 inhibitor
  • Apoptosis
  • Downregulation of survival pathways
Phase I/II
completed
IPI-504
  • HSP90 inhibitor
  • Inhibition of UPR
Phase I
completed
Tanespimycin
(17-AAG; KOS-953)
  • HSP90 inhibitor
  • Inhibition of downstream signaling pathways
  • Induction of UPR
Phase II/III
completed
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Bianchi, G.; Moscvin, M.; Comenzo, R.L. Laboratory-Based Rationale for Targeting the Protein Homeostasis Network in AL Amyloidosis. Hemato 2022, 3, 298-317. https://doi.org/10.3390/hemato3020022

AMA Style

Bianchi G, Moscvin M, Comenzo RL. Laboratory-Based Rationale for Targeting the Protein Homeostasis Network in AL Amyloidosis. Hemato. 2022; 3(2):298-317. https://doi.org/10.3390/hemato3020022

Chicago/Turabian Style

Bianchi, Giada, Maria Moscvin, and Raymond L. Comenzo. 2022. "Laboratory-Based Rationale for Targeting the Protein Homeostasis Network in AL Amyloidosis" Hemato 3, no. 2: 298-317. https://doi.org/10.3390/hemato3020022

Article Metrics

Back to TopTop