A Novel Non-Competitive Chemical Proteasome Inhibitor Displays Preclinical Activity in Myeloma and Leukemia.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1711-1711
Author(s):  
Xiaoming Li ◽  
Tabitha E Wood ◽  
Remco Sprangers ◽  
Xinliang Mao ◽  
Xiaoming Wang ◽  
...  

Abstract The proteasome is an enzymatic complex that rids cells of excess and misfolded proteins and possesses chymotrypin, trypsin, and caspase-like enzymatic activity. To date, all of the proteasome inhibitors approved for clinical use or in clinical trials inhibit the complex competitively by binding the active sites of the enzymes. Here, we report a novel chemical proteasome inhibitor that binds the alpha subunits of the 20S proteasome and inhibits the complex non-competitively through a dual copper-dependent and independent mechanism. In a screen of a focused chemical library for novel proteasome inhibitors, we identified 5-amino-8-hydroxyquinoline (5AHQ). When added to myeloma or leukemia intact cells or cell extracts, 5AHQ inhibited the enzymatic activity of the proteasome at low micromolar concentrations. In order to obtain further insight into the mechanism of action of 5AHQ, we carried out a kinetic analysis of inhibition of the enzymatic activity of purified T. Acidophilium proteasome. By Lineweaver-Burk plot analysis, 5AHQ inhibited the proteasome non-competitively. Next, we investigated the binding of 5AHQ to the proteasome. By NMR analysis, 5AHQ bound the half-proteasome complex comprised of a pair of α-rings, α7-α7, and clear spectral changes were observed that localized to residues Ile159, Val113, Val87, Val82, Leu112, Val89, Val134, Val24 and Leu136 inside the antechamber. In contrast, the competitive inhibitor MG132 that binds the proteolytic chamber did not produce any changes in spectra of α7-α7, as expected. 5AHQ bound copper in a 2:1 stoichiometry with a logβ′ value of 9.09, and the addition of copper to 5AHQ enhanced 5AHQ-mediated inhibition of the proteasome. However, binding intracellular copper was not sufficient to explain the effects of 5AHQ on the proteasome as analogues of 5AHQ that did not bind copper continued to inhibit the proteasome, copper-binding molecules not structurally related to 5AHQ did not affect the proteasome, and 5AHQ inhibited isolated proteasomes in buffers devoid of copper and other heavy metals. Given the effects of 5AHQ on the proteasome, we examined the effects of this molecule on the viability of leukemia and myeloma cell lines. Leukemia, myeloma and solid tumor cell lines were treated with increasing concentrations of 5AHQ for 72 hours and cell viability was measured by the MTS assay. 5AHQ induced cell death in 9/9 myeloma, 6/10 leukemia, and 3/10 solid tumor cell lines with an LD50 ≤5 uM. Cell death was confirmed by Annexin V staining. Consistent with its mechanism of action as a proteasome inhibitor, the ability of 5AHQ to induce cell death matched its ability to inhibit the proteasome. In addition, 5AHQ-mediated cell death was associated with inhibition of the NF-kappaB signalling pathway. As 5AHQ induced cell death in malignant cells, we evaluated the effects of oral 5AHQ in 3 mouse models of leukemia. Sublethally irradiated NOD-SCID mice were injected subcutaneously with OCI-AML2 or K562 human leukemia cells or intraperitoneally with MDAY-D2 murine leukemia cells. After tumor implantation, mice were treated with 5AHQ (50 mg/kg/day) or buffer control by oral gavage. Oral 5AHQ decreased tumor weight and volume in all 3 mouse models compared to control without causing weight loss or gross organ toxicity. In summary, we have identified a new strategy for inhibition of the proteasome and a lead for a new therapeutic agent for the treatment of hematologic malignancies.

Oncogene ◽  
2002 ◽  
Vol 21 (30) ◽  
pp. 4613-4625 ◽  
Author(s):  
Karuppiah Muthumani ◽  
Donghui Zhang ◽  
Daniel S Hwang ◽  
Sagar Kudchodkar ◽  
Nathanael S Dayes ◽  
...  

2014 ◽  
Vol 20 (2) ◽  
pp. 189-200 ◽  
Author(s):  
Luigi Leanza ◽  
Paul O’Reilly ◽  
Anne Doyle ◽  
Elisa Venturini ◽  
Mario Zoratti ◽  
...  

2011 ◽  
Vol 2 (3) ◽  
pp. 419-422 ◽  
Author(s):  
VESNA BUCAN ◽  
CLAUDIA Y.U. CHOI ◽  
ANDREA LAZARIDIS ◽  
PETER M. VOGT ◽  
KERSTIN REIMERS

1994 ◽  
Vol 42 (7) ◽  
pp. 917-929 ◽  
Author(s):  
E Spiess ◽  
A Brüning ◽  
S Gack ◽  
B Ulbricht ◽  
H Spring ◽  
...  

We investigated the appearance and activity of the cysteine proteinase cathepsin B and its physiological inhibitors, stefins A and B, at the cellular level in human tumor cell lines HS-24, derived from a primary lung tumor (squamous cell), and SB-3, derived from a metastasis (lung adenocarcinoma). In addition to cathepsin B, these tumor cells also expressed the immunologically and functionally related cathepsin L, but not cathepsin H. Stefin A was found in HS-24 but not in SB-3 cells; stefin B was found in both cell types. Using a specific fluorogenic cytochemical assay, the intracellular activity of the enzyme was localized and quantified. Thus, the cellular cathepsin B kinetics for the synthetic substrates Z-Arg-Arg-4M beta NA and Z-Val-Lys-Lys-Arg-4M beta NA, its pH dependence and inhibition by E64, stefins A and B, and cystatin C could be determined. From these measurements it appeared that the enzyme exhibited different cleavage rates for these substrates in the different cell types, showed considerable cleavage activity at neutral pH, which was stable under these conditions for extended time periods, and was highly sensitive to the inhibitors E64 and cystatin C but was considerably less sensitive to stefins, particularly stefin A. By conventional light microscopy, confocal laser scanning microscopy, and electron microscopy the enzymatic activity was localized in lysosomes, as expected, but also in the endoplasmic reticulum, nuclear membrane, and plasma membrane. The endoplasmic reticulum is a site at which only pre-mature enzyme forms exist, which are usually not active. The appearance of enzymatic activity at the plasma membrane confirms earlier biochemical and immunofluorescence microscopic investigations. The different sites of localization within the cells make it likely that different forms of the enzyme are expressed simultaneously, which follow alternate ways of processing and sorting. Taken together, the results support an involvement of the enzyme under extracellular conditions in degradative processes.


2015 ◽  
Vol 29 (5) ◽  
pp. 1026-1033 ◽  
Author(s):  
Giovanna Barbarini Longato ◽  
Giovanna Francisco Fiorito ◽  
Débora Barbosa Vendramini-Costa ◽  
Ilza Maria de Oliveira Sousa ◽  
Sirlene Valério Tinti ◽  
...  

2011 ◽  
Vol 85 (Suppl_1) ◽  
pp. 820-820
Author(s):  
Yvonne A.R. White ◽  
Alan L. Johnson ◽  
Dori C. Woods

2003 ◽  
Vol 17 (4) ◽  
pp. 103-107 ◽  
Author(s):  
Chia-Ni Li ◽  
Hui-Lun Hsu ◽  
Tsu-Lan Wu ◽  
Kuo-Chien Tsao ◽  
Chien-Feng Sun ◽  
...  

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2664-2664
Author(s):  
Alex Hessel ◽  
Malefa Tselanyane ◽  
Fengrong Wang ◽  
Ebenezer David ◽  
Sagar Lonial ◽  
...  

Abstract Patulin (4-Hydroxy-4H-furo 3,2-C-pyran-2(6H)-one) is a first-in-class mycotoxin under development as a novel chemotherapeutic agent. The mechanism of action of Patulin has been reported to include activation of mitogen activated protein kinases (MAPKs) and generation of reactive oxygen species (ROS). We have previously shown Patulin to have activity against myeloma, leukemia, and lymphoma cell lines, as well as primary tumor cells in clinical samples from patients with these diagnoses (Wang BLOOD 2007). Moreover, we have shown that Patulin specifically and potently targets tumor cells over normal cells and effectively killed primary tumor cells of patients with refractory illness. The aim of this study was to determine whether Patulin acts synergistically with the topoisomerase inhibitor doxorubicin or the proteasome inhibitor bortezomib. We hypothesize that drugs with non-cross-reactive modes of action could be complementary. Human tumor cell lines from B-cell lymphoma (DAUDI), T-Cell leukemia and lymphoma (Jurkat and H9, respectively), and myeloid leukemia (HL60) malignancies were tested for their sensitivity to single agents Patulin, bortezomib, and doxorubicin as well as Patulin in combination with the latter two agents (Patulin and bortezomib, P + B; Patulin and doxorubicin, P + D). Cells were treated with a range of concentrations of each single agent and the drugs in combination over 24 hours. Following treatment, cell metabolic activity was assessed using a microculture tetrazolium (MTT) assay and cell viability was assessed by flow cytometry using Annexin V and propidium iodide (PI) staining. Dose-effect curves, median effect plots, and combination index (CI) values were generated in the Compusyn software program for each target cell population. Median-effect doses (IC50s) of individual drugs were interpolated using the y-intercept of median-effect plots. Three dose-effect data points were used to create a range of CI values at different fractions of affect (fa). The lower and upper values of the CI range were used to characterize drug combinations as synergistic, antagonistic, or additive based on Chou’s Symbols for Synergism and Antagonism using CI analysis (Table 1). The role for ROS in the mechanism of action of Patulin was confirmed by flow cytometry showing increased levels of ROS in cell lines following Patulin exposure. Preincubation of cell lines with N-acetyl cysteine (NAC) or concurrent exposure to Patulin and NAC abrogated the cytotoxic activity of the mycotoxin. H9 cells were most sensitive to the effects of Patulin, with an IC50 of 1.2 μM. Combinations of P + B acted synergistically against Jurkat, H9, DAUDI, and HL60 tumor cells; however, P + B also demonstrated moderate antagonism against the Jurkat and H9 cell lines (Table 2). Likewise, combinations of P + D interacted synergistically against Jurkat, H9, DAUDI, and HL60 tumor cell lines while simultaneously demonstrating strong antagonism against the H9 cell line. Patulin kills leukemia and lymphoma cells via generation of intracellular ROS. Synergy of Patulin with either bortezomib or doxorubicin in leukemia and lymphoma cell lines indicates a distinct mechanism of action for the mycotoxin and compared to other chemotherapeutics and supports the rationale for continued development of Patulin as a novel chemotherapeutic mycotoxin. TABLE 1. Chou’s Symbols for Synergism and Antagonism using CI Analysis CI Description < 0.1 Very Strong Synergism 0.1–0.3 Strong Synergism 0.3–0.7 Synergism 0.7–0.85 Moderate Synergism 0.85–0.90 Slight Synergism 0.90–1.10 Nearly Additive 1.10–1.20 Slight Antagonism 1.20–1.45 Moderate Antagonism 1.45–3.3 Antagonism 3.3–10 Strong Antagonism > 10 Very Strong Antagonism TABLE 2. IC50 of single agents patulin, bortezomib, and doxorubicin and CI in hematological cancer cell lines Tumor cell line IC50 of patulin (μM) IC50 of bortezomib (nM) IC50 of doxorubicin (μM) CI: P + B CI: P + D T-Cell Jurkat 1.16 5600 5.2 0.6 -- 1.2 0.1 -- 1.2 H9 1.2 14 0.33 0.4 -- > 1 0.3 -- > 1 B-Cell DAUDI 0.98 1.1 0.19 < 0.1 -- 0.1 < 0.1 -- 0.3 Myeloid HL60 1.7 0.42 0.07 < 0.1 -- 0.1 < 0.1


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4701-4701
Author(s):  
Blake T. Aftab ◽  
Daniel J Anderson ◽  
Ronan Le Moigne ◽  
Stevan Djakovic ◽  
Eugen Dhimolea ◽  
...  

Abstract Hematological malignancies such as multiple myeloma (MM) have an increased reliance on the ubiquitin proteasome system (UPS) presumably as a consequence of their high protein synthetic and secretory burden. Chemical agents that target the proteasome, such as bortezomib and carfilzomib, have been successful in treating multiple myeloma; however patients treated with these drugs ultimately relapse. The AAA-ATPase p97/VCP (p97) facilitates ATP-dependent extraction and degradation of ubiquitinated proteins destined for proteasomal elimination. In addition to ubiquitin-dependent protein degradation, p97 is also closely involved in other aspects of protein homeostasis, including endoplasmic reticulum-associated degradation (ERAD) and autophagy. Pharmacologic inhibition of p97 provides a compelling therapeutic approach for hematological malignancies that rely on tight regulation of protein homeostasis as a component of their survival. CB-5083 is a novel small molecule inhibitor of p97 ATPase activity with nanomolar enzymatic and cellular potency. Treatment of cancer cells with CB-5083 causes a dramatic increase in poly-ubiquitinated proteins as well as an accumulation of substrates of the UPS and ERAD. CB-5083 causes a profound induction of the unfolded protein response (UPR) with consequent activation of the DR5 death receptor, caspase 8, caspase 3/7 and ultimately cell death. Induction of the UPR occurs to a greater magnitude with CB-5083 when compared to the proteasome inhibitor, bortezomib, suggesting the potential for increased efficacy in cancers with sensitivity to UPR-mediated cell death. In addition, activation of apoptosis and cell death occur more rapidly with CB-5083 than with bortezomib. Sequencing of cell lines made resistant to CB-5083 reveals missense mutations mapping to the D2 ATPase site in p97, supporting on-target association with cytotoxicity. In an expanded panel of MM cell lines there is no correlation between the cytotoxic sensitivity to CB-5083 and the cytotoxic sensitivity to proteasome inhibitors, suggesting differential mechanisms of cytotoxicity and potential activity of CB-5083 in proteasome inhibitor resistant settings. Compared to myeloma cell lines, CB-5083 has reduced cytotoxic potency in immortalized stromal cell lines and in patient-derived CD138-negative bone marrow mononuclear cells. Furthermore, unlike the reduced potency demonstrated by carfilzomib in the context of MM cell-bone marrow stromal cell (BMSC) interactions, the cyto-reductive potential of CB-5083 is unaffected in co-cultures of MM cells with patient-derived BMSCs or immortalized BMSCs from healthy donors. In vivo, CB-5083 is orally bioavailable, shows a pharmacodynamic effect in tumor tissue (as measured by poly-ubiquitin accumulation) and demonstrates robust anti-tumor activity across several MM models. CB-5083 treatment of mice bearing subcutaneous xenografts leads to tumor stasis and regression in RPMI8226 and AMO1 MM models, respectively. In advanced models of disseminated, ortho-metastatic disease, intermittent oral administration of CB-5083 demonstrates significant inhibition of myeloma burden and improves survival, with an overall efficacy profile that compares favorably to that of clinically approved proteasome inhibitors. Furthermore, in the Vk*Myc genetically engineered mouse model of MM, treatment with CB-5083 results in a significant reduction in M-spike by 55%. Combination treatment of mice bearing the RPMI8226 subcutaneous xenograft model with CB-5083, dexamethasone and lenalidomide results in tumor regression. Taken together, these data demonstrate that CB-5083 is a potent and selective inhibitor of the p97 ATPase with robust activity in vitro and in vivo in numerous MM models and strongly support clinical evaluation. Based on these observations, a phase 1 dose-escalation trial has recently been initiated and is currently underway in patients with relapsed/refractory multiple myeloma. Disclosures Anderson: Cleave Biosciences: Employment. Le Moigne:Cleave Biosciences: Employment. Djakovic:Cleave Biosciences: Employment. Rice:Cleave Biosciences: Employment. Wong:Cleave Biosciences: Employment. Kumar:Cleave Biosciences: Employment. Valle:Cleave Biosciences: Employment. Menon:Cleave Biosciences: Employment. Kiss von Soly:Cleave Biosciences: Employment. Wang:Cleave Biosciences: Employment. Yao:Cleave Biosciences: Employment. Soriano:Cleave Biosciences: Employment. Bergsagel:ONYX: Consultancy; Janssen: Consultancy; BMS: Consultancy; Novartis: Research Funding. Yakes:Cleave Biosciences: Employment. Zhou:Cleave Biosciences: Employment. Wustrow:Cleave Biosciences: Employment. Rolfe:Cleave Biosciences: Employment.


Sign in / Sign up

Export Citation Format

Share Document