Pharmacologic Evaluation of Orally Bioavailable Inhibitors of the 20S Proteasome.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3581-3581
Author(s):  
Christopher J. Kirk ◽  
Monette A. Aujay ◽  
Mark Ho ◽  
Jing Jiang ◽  
Guy J. Laidig ◽  
...  

Abstract Clinical application of proteasome inhibitors in the treatment of hematologic malignancies such as myeloma and lymphoma is restricted in part by the necessity of frequent IV administration and would be improved by oral (PO) administration. Selective inhibitors of the protease subunits of the 20S proteasome can be generated from peptidyl aldehydes, boronates, vinyl sulfones, and epoxyketones. Many of these peptide based proteasome inhibitors are cell permeant and capable of systemic proteasome inhibition upon intravenous (IV) administration to experimental animals such as mice and rats. In the cases of the peptidyl boronate bortezomib (Velcade™) and the epoxyketone PR-171, proteasome inhibition can be achieved in patients with IV administration. However, systemic exposure following PO administration of these inhibitors may be limited by several factors including gastric pH, gastric and intestinal peptidases, efflux pumps, biliary excretion and intestinal and hepatic metabolic activities. We have tested over 80 peptide epoxyketones with potent (IC50 <100 nM) in vitro inhibitory activity against the chymotrypsin-like activity of the 20S proteasome for bioavailability following PO administration in mice. Systemic exposure was monitored by measuring chymotrypsin-like inhibition in a number of tissues and an approximation of the relative bioavailability for selected compounds was measured by comparing the pharmacodynamics of IV and PO administration using a range of doses. These compounds were also tested in vitro for solubility, intestinal cell permeability, intestinal and hepatic metabolism, and sensitivity to the multidrug resistance protein 1 (MDR1) efflux pump in order to determine which properties were associated with oral bioavailability. We have found that oral bioavailability in mice is associated with increased intrinsic solubility and metabolic stability and reduced MDR1 sensitivity. Proteasome inhibition following PO administration is rapid, resulting in maximal proteasome inhibition within 15 minutes. Rapid absorption and clearance of selected compounds was also confirmed in mice and rats by pharmacokinetic analysis. Repeated oral administration was well tolerated at doses that resulted in significant (>80%) proteasome inhibition in most tissues. The anti-tumor efficacy of these orally bioavailable proteasome inhibitors are being assessed in both human tumor xenograft and mouse syngeneic models. The results from these studies will enable further pre-clinical development of potent, orally bioavailable proteasome inhibitors for the treatment of malignant diseases.

2019 ◽  
Vol 39 (10) ◽  
Author(s):  
Jessica Tsalikis ◽  
Mena Abdel-Nour ◽  
Armin Farahvash ◽  
Matthew T. Sorbara ◽  
Stephanie Poon ◽  
...  

ABSTRACTProtein degradation pathways are critical for maintaining proper protein dynamics within the cell, and considerable efforts have been made toward the development of therapeutics targeting these catabolic processes. We report here that isoginkgetin, a naturally derived biflavonoid, sensitized cells undergoing nutrient starvation to apoptosis, induced lysosomal stress, and activated the lysosome biogenesis geneTFEB. Isoginkgetin treatment led to the accumulation of aggregates of polyubiquitinated proteins that colocalized strongly with the adaptor protein p62, the 20S proteasome, and the endoplasmic reticulum-associated degradation (ERAD) protein UFD1L. Isoginkgetin directly inhibited the chymotrypsin-like, trypsin-like, and caspase-like activities of the 20S proteasome and impaired NF-κB signaling, suggesting that the molecule may display its biological activity in part through proteasome inhibition. Importantly, isoginkgetin was effective at killing multiple myeloma (MM) cell linesin vitroand displayed a higher rate of cell death induction than the clinically approved proteasome inhibitor bortezomib. We propose that isoginkgetin disturbs protein homeostasis, leading to an excess of protein cargo that places a burden on the lysosomes/autophagic machinery, eventually leading to cancer cell death.


1998 ◽  
Vol 42 (3) ◽  
pp. 647-653 ◽  
Author(s):  
Weixing Li ◽  
Paul A. Escarpe ◽  
Eugene J. Eisenberg ◽  
Kenneth C. Cundy ◽  
Clive Sweet ◽  
...  

ABSTRACT GS 4071 is a potent carbocyclic transition-state analog inhibitor of influenza virus neuraminidase with activity against both influenza A and B viruses in vitro. GS 4116, the guanidino analog of GS 4071, is a 10-fold more potent inhibitor of influenza virus replication in tissue culture than GS 4071. In this study we determined the oral bioavailabilities of GS 4071, GS 4116, and their respective ethyl ester prodrugs in rats. Both parent compounds and the prodrug of the guanidino analog exhibited poor oral bioavailability (2 to 4%) and low peak concentrations in plasma (C maxs; C max<0.06 μg/ml). In contrast, GS 4104, the ethyl ester prodrug of GS 4071, exhibited good oral bioavailability (35%) as GS 4071 and high C maxs of GS 4071 (Cmax = 0.47 μg/ml) which are 150 times the concentration necessary to inhibit influenza virus neuraminidase activity by 90%. The bioavailability of GS 4104 as GS 4071 was also determined in mice (30%), ferrets (11%), and dogs (73%). The plasma of all four species exhibited high, sustained concentrations of GS 4071 such that at 12 h postdosing the concentrations of GS 4071 in plasma exceeded those necessary to inhibit influenza virus neuraminidase activity by 90%. These results demonstrate that GS 4104 is an orally bioavailable prodrug of GS 4071 in animals and that it has the potential to be an oral agent for the prevention and treatment of influenza A and B virus infections in humans.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2859-2859
Author(s):  
Shirin Arastu-Kapur ◽  
Andrew J. Ball ◽  
Janet L. Anderl ◽  
Mark K Bennett ◽  
Christopher J Kirk

Abstract Abstract 2859 Poster Board II-835 BACKGROUND: The dipeptide boronate proteasome inhibitor bortezomib (BTZ; Velcade®) is approved for the treatment of multiple myeloma and non-Hodgkin's lymphoma. Bortezomib-induced peripheral neuropathy (BIPN, Blood (2008)112:1593-1599) is seen in ∼30% of BTZ-treated patients and can result in dose reductions and discontinuations that may result in suboptimal levels of proteasome inhibition. Carfilzomib (CFZ), a tetrapeptide epoxyketone, is a selective and irreversible proteasome inhibitor that is structurally and mechanistically distinct from bortezomib. Single agent treatment with CFZ has demonstrated strong activity in relapsed and refractory myeloma and a favorable safety profile in Phase 2 trials (ASH2008:864 & 865). Importantly treatment-emergent PN was seen at low levels and did not result in dose modifications or discontinuations. The disparate safety data for these proteasome inhibitors suggest that non-proteasomal mechanisms may underlie BIPN. Using activity-based probes in peripheral blood mononuclear cell (PBMC) lysates, we previously demonstrated inhibition of non-proteasomal proteases by BTZ and other proteasome inhibitors with a boronate pharmacophore (EHA2009:0939). However, the involvement of the proteasome in the peripheral nerve degeneration and BIPN in BTZ-treated myeloma patients remains to be established. AIMS: To establish an in vitro model of peripheral nerve degeneration and to determine the effects of proteasome inhibition by BTZ and CFZ on neurite outgrowth and cell survival. METHODS: SH-SY5Y neuroblastoma cells were differentiated by long term culture in retinoic acid and brain derived nerve growth factor to induce neurite outgrowth. The effects of proteasome inhibitors were measured by high content image analysis of fluorescent images for cell survival (Hoechst nuclear counterstain) and neurite degeneration (FITC-mouse anti-beta-III-tubulin). Phase contrast images were also collected to observe morphological effects and gross cell death. Cell viability and proteasome inhibition was measured in undifferentiated and differentiated cells. The MEROPS (peptidase) database was mined for candidate serine proteases with a P1 selectivity of Leu/Phe/Tyr to identify candidate off-targets CFZ and BTZ and candidate proteases were validated by standard biochemical and cell biology techniques. RESULTS: In differentiated SH-SY5Y cells, the average neurite length decreased by 33% following 24 hr exposure to 10nM BTZ but was unaffected by the same concentration of CFZ. Proteasome inhibition as determined by a fluorescent substrate for the chymotrypsin-like activity was equivalent (∼70%) after a 24 hr exposure for both compounds in differentiated cells, suggesting that neurodegeneration involves non-proteasomal pathways. With 72 hrs continuous exposure, BTZ was 10-fold more potent than CFZ at inducing neurodegeneration. Furthermore, in both undifferentiated and differentiated SH-SY5Y cells, BTZ was 5-fold more cytotoxic than CFZ. Database mining for serine proteases with a selectivity for Leu/Phe/Tyr at P1 was used to identify other potential BTZ targets that might underlie neurotoxicity. One candidate is HtrA2 (also called Omi), an inducible mitochondrial serine protease whose activity protects neurons from stress induced apoptosis (Hum Mol Genet (2005) 14(5):2099-2111). HtrA2 levels increased 2-fold in SH-SY5Y cells treated with either BTZ or CFZ for 6 hrs at 40 nM. Using a gel based assay and purified enzyme preparations, BTZ inhibited HtrA2 activity with an IC50 ∼ 4 nM, equivalent to its activity against the proteasome. In contrast, Carfilzomib did not inhibit HtrA2 at the highest concentration tested (10 mM). CONCLUSIONS: These data demonstrate that BTZ induces neuronal cell death and neurite degeneration in vitro by proteasome-independent mechanisms. We propose that combined inhibition of the proteasome and HtrA2 by BTZ may underlie peripheral nerve toxicities in vitro and may be involved in BIPN in myeloma patients. In this model, CFZ, which mediates equivalent proteasome inhibition to BTZ in neurons, does not induce neurodegeneration due to inactivity against HtrA2. Future profiling of non-proteasomal targets of BTZ, including HtrA2 activity, in patient samples is merited. These results suggest that the favorable safety profile of CFZ in myeloma patients may be a result of its high selectivity for proteasomal proteases. Disclosures: Arastu-Kapur: Proteolix, Inc: Employment. Ball:Millipore Corp: Employment. Anderl:Millipore Corp: Employment. Bennett:Proteolix: Employment. Kirk:Proteolix, Inc: Employment.


Blood ◽  
2012 ◽  
Vol 120 (23) ◽  
pp. 4513-4516 ◽  
Author(s):  
David I. Lichter ◽  
Hadi Danaee ◽  
Michael D. Pickard ◽  
Olga Tayber ◽  
Michael Sintchak ◽  
...  

AbstractVariations within proteasome β (PSMB) genes, which encode the β subunits of the 20S proteasome, may affect proteasome function, assembly, and/or binding of proteasome inhibitors. To investigate the potential association between PSMB gene variants and treatment-emergent resistance to bortezomib and/or long-term outcomes, in the present study, PSMB gene sequence variation was characterized in tumor DNA samples from patients who participated in the phase 3 Assessment of Proteasome Inhibition for Extending Remissions (APEX) study of bortezomib versus high-dose dexamethasone for treatment of relapsed multiple myeloma. Twelve new PSMB variants were identified. No associations were found between PSMB single nucleotide polymorphism genotype frequency and clinical response to bortezomib or dexamethasone treatment or between PSMB single nucleotide polymorphism allelic frequency and pooled overall survival or time to progression. Although specific PSMB5 variants have been identified previously in preclinical models of bortezomib resistance, these variants were not detected in patient tumor samples collected after clinical relapse from bortezomib, which suggests that alternative mechanisms underlie bortezomib insensitivity. This study is registered at www.clinicaltrials.gov as NCT00048230.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 4464-4464 ◽  
Author(s):  
Jane L. Liesveld ◽  
Camille N. Abboud ◽  
Chaohui Lu ◽  
Jeremy Bechelli ◽  
Karen Rosell ◽  
...  

Abstract Proteasome inhibitors block degradation of the NF-κB regulator, IκB, resulting in inhibition of NF-κB nuclear localization. Proteasome inhibitors have been tested in the setting of refractory acute leukemia, with proteasome inhibition demonstrated within leukemic blasts. Arsenic trioxide (AsO3) has also been shown to inhibit NF-κB in leukemia. Since NF-κB is constitutively activated in primitive AML cells and serves as a regulator of many genes which encode proliferative and survival responses, we have begun to explore the effects of AsO3 and bortezomib (PS-341, VelcadeTM ), on AML cell lines and primary cells. The farnesyl transferase inhibitors (FTIs) also inhibit AML proliferation, and their effects in combination with bortezomib have also been explored. Because FTIs may inhibit signal transduction pathways independent of those affected by NF-κB inhibition, and because FTIs may indirectly inhibit NF-κB function via Akt inhibition, the effects of combining these agents with other NF-κB agents on AML cells in vitro have also been explored. Bortezomib, at concentrations of 4nM or greater, inhibited NF-κB in AML cell lines and primary cells as did AsO3 at concentrations of 1 nM or greater. NF-kappa B was measured by ELISA for p65 NF-κB activity. The nonpeptidomimetic FTI, R115777 (J&J), did not inhibit NF-κB at concentrations up to 100 nM, concentrations which effectively inhibit farnesylation of target proteins, whereas the FTI, L-744832 (Merck), was able to inhibit NF-κB expression at 1 μM from 24 to 72 hours of exposure. In the HL60 cell line, inhibited by FTI and bortezomib independently, the combination did not appear to have additive or synergistic effects. Furthermore, the effects of combined exposure to FTI and bortezomib on expression of activated caspase 3 or activated PARP cleavage were no greater than with bortezomib alone. Likewise, combination effects on expression of phosphorylated AKT or ERK were not observed. In contrast, the combination of bortezomib and AsO3 resulted in decreased phospho-ERK expression and increased expression of cleaved PARP, suggesting increased apoptosis. When cytarabine, 100 nM was combined with bortezomib at 1 to 4 nM, no effect on timing of administration was noted, and apoptosis was increased with the combination as evidenced by an increase in cleaved PARP expression. Greater inhibition of proliferation was seen with this combination than with individual agents as demonstrated in MTT assays with combination index calculations suggesting synergism. With this combination, co-culture with an endothelial monolayer did not prevent the increased apoptosis noted with combined cytarabine and bortezomib. These studies suggest that future studies combining proteasome inhibition with standard chemotherapeutic agents or with other inhibitors of NF-κB like Arsenic trioxide may have greater antileukemic activity by inducing apoptosis in AML cells in vivo as well as in vitro, without obvious limitations of other targeted agents and drugs that are extruded by multridrug resistance transporters.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4910-4910
Author(s):  
Tony Muchamuel ◽  
Shirin Kapur ◽  
Christopher J Kirk ◽  
Jing Jiang ◽  
Susan Lee ◽  
...  

Abstract Abstract 4910 Proteasome inhibition is a validated therapeutic strategy for the treatment of multiple myeloma. Carfilzomib (CFZ) is a selective and irreversible proteasome inhibitor that is intravenously administered and an has shown an encouraging safety and efficacy profile in myeloma patients from multiple Phase 2 studies (ASH2008:864&865). CFZ induces prolonged proteasome inhibition in vivo and can be safely administered to patients on dose intensive schedules (e.g. QDx5); however, such schedules lack patient convenience. PR-047 is a novel inhibitor in the same chemical class as carfilzomib that is orally bioavailable Similar to carfilzomib, PR-047 potently targets the chymotrypsin-like activity of the proteasome and induces anti-tumor responses in vitro and in vivo across a range of tumor cell types including myeloma cells. We describe here the proteasome selectivity of PR-047 in vitro and the pharmacodynamics and safety of repeat dose oral administration in experimental animals. We found PR-047, like CFZ, to be highly selective for proteasomal proteases as demonstrated by a lack of inhibitory activity against a panel of non-proteasomal proteases, including serine proteases targeted by dipeptide boronates such as bortezomib, (J. Med. Chem. 2008 51:1068). In rats, well tolerated oral doses resulted in rapid and potent (>80%) inhibition of proteasome activity when the compound was administered either as a solution or in a capsule formulation suitable for clinical use. Similar to carfilzomib, PR-047 was well tolerated when administered on a dose intense schedule. Oral dosing of PR-047 to mice and rats daily for 5 consecutive days resulted in proteasome inhibition following the 5th dose that was equivalent to or greater than inhibition seen after the 1st dose in whole blood and tissues. This suggests that PR-047 promotes prolonged proteasome inhibition in vivo. These doses were well tolerated in GLP-compliant toxicity studies in rats and dogs where PR-047 was administered for 2, 14-day cycles of QDx5 followed by nine days rest. The MTD in rats was ∼6 fold greater than the dose required for >80% proteasome inhibition, suggesting a significant therapeutic window. At the MTD, PR-047 did not affect renal or liver function and was not associated with anemia or leukopenia. In addition, PR-047 administration did not result in behavioral or histologic signs of neurotoxicity. Comparable doses were well tolerated in dogs on the same dose schedule. Similar to CFZ, PR-047 is a selective and irreversible proteasome inhibitors that has potent anti-tumor activity and is well tolerated on dose intensive schedules that result in prolonged proteasome inhibition. These studies support a safe starting dose for Phase 1 clinical trials with PR-047, a novel agent for the treatment of malignant diseases. Disclosures Muchamuel: Proteolix Inc.: Employment. Kapur:Proteolix, Inc.: Employment. Kirk:Proteolix, Inc: Employment. Jiang:Proteolix, Inc.: Employment. Lee:Proteolix, Inc.: Employment. Bennett:Proteolix: Employment. Lewis:Proteolix, Inc.: Employment. Yang:Proteolix, Inc.: Employment. Jumaa:Proteolix, Inc.: Employment. Ring:Proteolix Inc.: Employment. Phiasivongsa:Proteolix Inc.: Employment. Zhou:Proteolix Inc.: Employment. Wang:Proteolix, Inc.: Employment.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2906-2906 ◽  
Author(s):  
Michelle A Hurchla ◽  
Antonio Garcia-Gomez ◽  
Mary C Hornick ◽  
Enrique M Ocio ◽  
Aixiao Li ◽  
...  

Abstract Abstract 2906 Background: Multiple myeloma (MM), a hematological malignancy of terminally differentiated plasma cells, is closely associated with osteolytic bone disease, caused by increased osteoclast (OC) number and resorption and suppressed osteoblast (OB) differentiation and function. Proteasome inhibitors (PIs), such as bortezomib, have become a cornerstone therapy for MM, potently reducing tumor burden and inhibiting pathologic bone destruction. In clinical trials, carfilzomib, a second generation epoxyketone-based PI, has exhibited potent anti-myeloma efficacy and decreased side effects compared to bortezomib. ONX 0912 is an orally bioavailable analog of carfilzomib. However, it is currently not known whether carfilzomib and ONX 0912, in addition to their anti-myeloma activity, may have a beneficial effect on myeloma-associated bone disease similarly to bortezomib. Here, we have conducted in vitro studies to evaluate the ability of each PI to promote osteogenic differentiation and function and to inhibit OC formation and resorption in murine and human cells. We have also studied the effects of orally administered ONX 0912 in both non-myelomatous mice and mice bearing bone marrow (BM) disseminated human myeloma. Patient samples, material and methods: The human MM cell lines MM.1S, U266 and RPMI-8226 were employed. Mesenchymal stem cells from BM samples of healthy donors, MM patients and mice were used for OB studies, whereas PBMCs from healthy volunteers and murine BM macrophages were used to generate OCs. Viability, proliferation and apoptosis were respectively assessed by MTT assay, BrdU incorporation and Annexin V/7-AAD staining. OB differentiation and function were investigated by alkaline phosphatase activity, quantitative mineralization assay and real-time quantitative RT-PCR. Osteoclastogenesis was assessed by enumeration of mutinucleated (≥3) tartrate resistant acid phosphatase-positive cells, whereas OC resorption was assessed on calcium-coated slides. Immunoflurescence analyses and flow cytometry were used to further evaluate OC function. Micro-CT analysis of myelomatous and non-myelomatous bone, bioluminescence imaging of tumor burden, calcein labeling and determination of serum levels of Igλ, CTX and P1NP were used in in vivo models. Results: Carfilzomib and ONX 0912 effectively decreased MM cell viability in vitro following continual or physiologic brief pulse treatment. These PIs were also able to overcome the growth advantage conferred by co-culture with human BM stromal cells or OCs. During their differentiation from mesenchymal stem cell precursors of mice and myeloma patients, physiologic concentrations of carfilzomib and ONX 0912 stimulated osteogenic differentiation and matrix mineralization and enhanced OB activity. In a similar manner, carfilzomib and ONX 0912 inhibited osteoclastogenesis from mouse and human progenitors and inhibited OC resorption. Importantly, the effects of these new PIs were exerted without being cytotoxic to OC/OB precursors. Daily oral administration of ONX 0912 to non-tumor bearing mice increased trabecular bone volume and enhanced bone formation. Finally, in a mouse model of disseminated human MM, ONX 0912 decreased human RPMI-8226 tumor burden and prevented bone loss, with serum markers evidencing both bone anti-catabolic and anabolic effects. Conclusion: In vitro and at physiologic concentrations and dosing, carfilzomib and ONX 0912 not only exert cytotoxic effects on myeloma cells, but also directly enhance OB formation and function and inhibit OC differentiation and resorption. In a disseminated human MM mouse model, orally administered ONX 0912 showed anti-resorptive and bone-anabolic effects in addition to its anti-tumor properties. Our data demonstrate that carfilzomib and ONX 0912 exert combined beneficial effects of anti-myeloma activity and on associated-myeloma bone disease, with favorable pharmacologic and tolerability profiles being reported in patients. This work was supported by funding from the Spanish MICINN-ISCIII (PI081825), Fundación de Investigación Médica Mutua Madrileña 2008, Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, the National Institutes of Health (T32CA113275; P01CA100730; P50CA94056), the St. Louis Mens' Group Against Cancer, and the Holway Myeloma Fund. Disclosures: Kirk: Onyx Pharmaceuticals: Employment. San Miguel:Onyx Pharmaceuticals: Consultancy.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4466-4466 ◽  
Author(s):  
Lenka Besse ◽  
Marianne Kraus ◽  
Andrej Besse ◽  
Juergen Bader ◽  
Thomas Mehrling ◽  
...  

Abstract Background. EDO-S101 is a first-in-class alkylating, histone-deacetylase inhibitor (HDACi) fusion molecule with dual activity that is currently in Phase I. It structurally combines the strong DNA damaging effect of bendamustine with a fully functional pan-HDAC inhibitor, vorinostat. Bendamustine has substantial activity against B-cell malignancies; vorinostat sensitizes the same type of cancers against alkylators or proteasome inhibitors (PI). Bendamustine combined with the PI bortezomib (BTZ) is active against multiple myeloma (MM). Cytotoxicity of PI in MM relies on excess induction of proteotoxic stress and triggering of the unfolded protein response (UPR). Upon proteasome inhibition, HDACi synergize with PI by interfering with the a-tubulin-mediated transport of poly-ubiquitinated proteasome substrates to lysosomal destruction. Indeed, EDO-S101 has strong synergistic cytotoxicity with PI in vitro against hematological malignancies, including MM, mantle cell lymphoma and ABC type diffuse large B-cell lymphoma. The aim of this work is to characterize the molecular mechanism of action of the synergy of EDO-S101 with PI in comparison to its established structurally related drugs, bendamustine and vorinostat. Methods. The cytotoxic and molecular activity of EDO-S101 in combination with BTZ and other types of PI was assessed in vitro using the RPMI-8226 and several other MM cell lines. HDAC-inhibiting activity, accumulation of poly-ubiquitinated proteins and induction of ER stress, apoptotic signaling and autophagy induction were assessed by quantitative PCR and western blotting. Proteasome activity was measured with activity based probes (ABP). Apoptosis was assessed by AnnexinV/FITC staining with flow cytometry. Cell viability was evaluated by MTS assay. Results. EDO-S101 showed substantially stronger cytotoxicity in combination with PI than melphalan, bendamustine, cyclophosphamide or PI combined with equimolar vorinostat. EDO-S101 had higher HDACi-type of activity, compared to vorinostat, as demonstrated judged in particular by increased a-tubulin acetylation, providing a potential mechanistic basis for its superior synergy with PI. Consistent with this, EDO-S101 alone induced moderate cellular accumulation of poly-ubiquitinated proteins already in the absence of proteasome inhibition, which was potentiated when EDO-S101 was combined with BTZ. EDO-S101 induced activation of the UPR-regulators XBP1 and IRE1 known to control BTZ sensitivity of MM, in contrast to vorinostat or bendamustine alone. Co-treatment with BTZ and EDO-S101 or vorinostat resulted in highly synergistic triggering of the UPR (ATF4, CHOP, BIP). Interestingly, EDO-S101 in addition induced the pro-apoptotic machinery via upregulation of NOXA, downregulation of BCL2 and an increase of the BAX/BCL2 ratio, and also activated autophagy, as evidenced by upregulation of LC3A and LC3B. While this pro-apoptotic signaling of EDO-S101 was highly synergistic with BTZ-induced apoptotic signals, co-treatment with BTZ and vorinostat reduced apoptotic signaling compared to BTZ alone. EDO-S101 reduced c-Myc expression by 60%, while vorinostat had no effect on c-Myc levels. The combination BTZ+EDO-S101 decreased c-Myc levels by approx. 90%, while these levels remained unchanged during treatment with BTZ+vorinostat. Conclusion. EDO-S101 is a first-in-class, dual-mechanism, alkylator-HDAC-inhibitor fusion molecule that combines key structural features of bendamustine and vorinostat. The molecular mode of action of EDO-S101 differs from that of its structurally related drugs by a more effective interaction with a-tubulin, which may in part explain superior synergy with PI. Most importantly, EDO-S101 has a direct pro-apoptotic activity via downregulation of c-Myc and BCL2 while upregulating NOXA, features not observed with vorinostat. This results in highly synergistic signaling with the PI-induced pro-apoptotic effects. EDO-S101 is a promising advancement of bendamustine with molecular features clearly different from and superior to a combination of bendamustine with vorinostat. EDO-S101 should be explored in combination with proteasome inhibitors in particular in poor risk B cell neoplasms with c-Myc overexpression such as aggressive MM, Burkitt lymphoma or "double hit" aggressive B cell lymphoma. Disclosures Besse: Mundipharma-EDO: Other: travel support. Mehrling:Mundipharma-EDO: Employment. Driessen:Mundipharma-EDO: Honoraria, Membership on an entity's Board of Directors or advisory committees; celgene: Consultancy; janssen: Consultancy.


2019 ◽  
Author(s):  
Jessica L. Rausch ◽  
Areej A. Ali ◽  
Donna M. Lee ◽  
Yemarshet K. Gebreyohannes ◽  
Keith R. Mehalek ◽  
...  

ABSTRACTThe majority of gastrointestinal stromal tumors (GISTs) are driven by oncogenic KIT signaling and can therefore be effectively treated with the tyrosine kinase inhibitor (TKI) imatinib mesylate. However, most GISTs develop imatinib resistance through secondary KIT mutations. The type of resistance mutation determines sensitivity to approved second-/third-line TKIs but shows high inter- and intratumoral heterogeneity. Therefore, therapeutic strategies that target KIT independently of the mutational status are intriguing. Inhibiting the ubiquitin-proteasome machinery with bortezomib is effective in GIST cells through a dual mechanism of KIT transcriptional downregulation and upregulation of the pro-apoptotic histone H2AX but clinically problematic due to the drug’s adverse effects. We therefore tested second-generation inhibitors of the 20S proteasome (delanzomib, carfilzomib and ixazomib) with better pharmacologic profiles as well as compounds targeting regulators of ubiquitination (b-AP15, MLN4924) for their effectiveness and mechanism of action in GIST. All three 20S proteasome inhibitors were highly effective in vitro and in vivo, including in imatinib-resistant models. In contrast, b-AP15 and MLN4924 were only effective at high concentrations or had mostly cytostatic effects, respectively. Our results confirm 20S proteasome inhibitors as promising strategy to overcome TKI resistance in GIST, while highlighting the complexity of the ubiquitin-proteasome machinery as a therapeutic target.


Blood ◽  
2006 ◽  
Vol 108 (1) ◽  
pp. 286-291 ◽  
Author(s):  
Neil P. Shah ◽  
Francis Y. Lee ◽  
Roger Luo ◽  
Yibin Jiang ◽  
Marjolein Donker ◽  
...  

Mastocytosis is associated with an activating mutation in the KIT oncoprotein (KITD816V) that results in autophosphorylation of the KIT receptor in a ligand-independent manner. This mutation is inherently resistant to imatinib and, to date, there remains no effective curative therapy for systemic mastocytosis associated with KITD816V. Dasatinib (BMS-354825) is a novel orally bioavailable SRC/ABL inhibitor that has activity against multiple imatinib-resistant BCR-ABL isoforms in vitro that is presently showing considerable promise in early-phase clinical trials of chronic myeloid leukemia (CML). Pharmacokinetic analysis suggests that high nanomolar concentrations of dasatinib can be achieved safely in humans. In this study, we demonstrate significant inhibitory activity of dasatinib against both wild-type KIT and the KITD816V mutation in the nanomolar range in in vitro and cell-based kinase assays. Additionally, dasatinib leads to growth inhibition of a KITD816V-harboring human masto-cytosis cell line. Significantly, dasatinib selectively kills primary neoplastic bone marrow mast cells from patients with systemic mastocytosis while sparing other hematopoietic cells. Computer modeling suggests that the KITD816V mutation destabilizes the inactive conformation of the KIT activation loop to which imatinib binds, but it is not predicted to impair binding of KIT by dasatinib. Based upon our results, further evaluation of dasatinib for the treatment of systemic masto-cytosis in clinical trials is warranted. Moreover, dasatinib may be of clinical utility in other disease settings driven by activating KIT mutations. (Blood. 2006;108:286-291)


Sign in / Sign up

Export Citation Format

Share Document