Abstract B154: Co-administration of nicotinic acid with NAMPT small molecule inhibitors results in loss of in vivo efficacy in NAPRT1-deficient tumor models.

Author(s):  
Deepak Sampath ◽  
Jason Oeh ◽  
Yang Xiao ◽  
Xiaorong Liang ◽  
Alexander Vanderbilt ◽  
...  
Neoplasia ◽  
2013 ◽  
Vol 15 (12) ◽  
pp. 1314-IN3 ◽  
Author(s):  
Thomas O'Brien ◽  
Jason Oeh ◽  
Yang Xiao ◽  
Xiaorong Liang ◽  
Alexander Vanderbilt ◽  
...  

2015 ◽  
Vol 76 (2) ◽  
pp. 287-299 ◽  
Author(s):  
Susan M. Christner ◽  
Dana M. Clausen ◽  
Jan H. Beumer ◽  
Robert A. Parise ◽  
Jianxia Guo ◽  
...  

2021 ◽  
Author(s):  
Jana Van Dycke ◽  
Arno Cuvry ◽  
Jan Knickmann ◽  
Annelii Ny ◽  
Sebastian Rakers ◽  
...  

2020 ◽  
Vol 25 (9) ◽  
pp. 985-999
Author(s):  
John Vincent ◽  
Marian Preston ◽  
Elizabeth Mouchet ◽  
Nicolas Laugier ◽  
Adam Corrigan ◽  
...  

Cytoplasmic dynein-1 (hereafter dynein) is a six-subunit motor complex that transports a variety of cellular components and pathogens along microtubules. Dynein’s cellular functions are only partially understood, and potent and specific small-molecule inhibitors and activators of this motor would be valuable for addressing this issue. It has also been hypothesized that an inhibitor of dynein-based transport could be used in antiviral or antimitotic therapy, whereas an activator could alleviate age-related neurodegenerative diseases by enhancing microtubule-based transport in axons. Here, we present the first high-throughput screening (HTS) assay capable of identifying both activators and inhibitors of dynein-based transport. This project is also the first collaborative screening report from the Medical Research Council and AstraZeneca agreement to form the UK Centre for Lead Discovery. A cellular imaging assay was used, involving chemically controlled recruitment of activated dynein complexes to peroxisomes. Such a system has the potential to identify molecules that affect multiple aspects of dynein biology in vivo. Following optimization of key parameters, the assay was developed in a 384-well format with semiautomated liquid handling and image acquisition. Testing of more than 500,000 compounds identified both inhibitors and activators of dynein-based transport in multiple chemical series. Additional analysis indicated that many of the identified compounds do not affect the integrity of the microtubule cytoskeleton and are therefore candidates to directly target the transport machinery.


2006 ◽  
Vol 12 (11) ◽  
pp. 3459-3469 ◽  
Author(s):  
Deepak Sampath ◽  
Lee M. Greenberger ◽  
Carl Beyer ◽  
Malathi Hari ◽  
Hao Liu ◽  
...  

Author(s):  
Sylvia Gruenewald ◽  
Julia Schueler ◽  
Michael Haerter ◽  
Frank Suessmeier ◽  
Kerstin Klingner ◽  
...  

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 306-306
Author(s):  
Meredith W. Miller ◽  
Soni Basra ◽  
Paul C. Billings ◽  
Jamie Gewirtz ◽  
William F. DeGrado ◽  
...  

Abstract Vascular damage due to trauma or disease exposes circulating platelets to collagen in the subendothelial matrix. This is a critical event in the formation of a hemostatic plug or an occluding thrombus because collagen is not only a substrate for platelet adhesion but is also a strong platelet agonist. Platelets possess two physiologic collagen receptors: glycoprotein VI, a member of the immunoglobin superfamily, and the integrin α2β1. To design small molecule inhibitors of the interaction of platelets with collagen, we focused on α2β1 as a target because murine models of α2β1 deficiency display normal bleeding times and only a slight decrease in platelet activation by collagen and because the small number of reported patients with congenital α2β1 deficiency demonstrated only a mild bleeding diathesis. Thus, α2β1 antagonists could be effective anti-thrombotic agents with minimal toxicity, especially when combined with other anti-platelet drugs. We have developed a class of compounds that target the I-like domain of the β1 subunit, an allosteric site that regulates collagen binding to α2β1 by preventing the conversion of α2β1 from an inactive (low affinity) to an active (high affinity) conformation. This class of compounds is based on a proline-substituted 2,3-diaminopropionic acid scaffold. Structure-activity relationship studies of the scaffold have focused on optimization of the proline moiety, the urea functionality, and the sulfonyl group and have resulted in the development of potent inhibitors of α2β1-mediated platelet adhesion to collagen with IC50’s in the high picomolar to low nanomolar range. In particular, optimization of the proline moiety lead to compounds with high potency: transitioning from proline (DB496, IC50 of 29–62 nM) to a thiazolidine (SB68A) improved the IC50 to 2–8 nM; adding a methyl group at the 2 position of the thiazolidine (SB68B) slightly improved the IC50 to 1–12 nM; adding two methyl groups at the 5 position of the thiazolidine (SW4-161) resulted in a lead compound with an IC50 of 0.33–8 nM. As expected, the compounds had no effect on the binding of isolated α2 I-domains to collagen, consistent with their I-like domain mode of activity. Further, they were specific for α2β1-mediated platelet adhesion to collagen because they had no impact on ADP-stimulated platelet aggregation when added at 2 μM, a concentration more than 100-fold greater than the IC50 for inhibition of platelet adhesion to collagen. The compounds were also strong inhibitors of murine platelet adhesion to collagen and when tested in the ferric chloride-initiated murine carotid artery injury model, displayed activity similar to aspirin. Thus, 71% of untreated mice in this thrombosis model developed occlusive thrombi that remained stable for the 30 min duration of the assay, whereas stable thrombi developed in only 32% of mice treated with 1g/kg aspirin orally and in 41% of mice receiving 60 mg/kg CSW4-161intravenously. In summary, we have developed a class of potent inhibitors of the integrin α2β1 that demonstrate both in vitro and in vivo anti-platelet activity. Further development of this class of compounds may result in novel and relatively non-toxic anti-thrombotic agents.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 610-610
Author(s):  
Dharminder Chauhan ◽  
Ze Tian ◽  
Benjamin Nicholson ◽  
Bin Zhou ◽  
Teru Hideshima ◽  
...  

Abstract Abstract 610 Background and Rationale: Ubiquitin-Proteasome Signaling (UPS) pathway is mediated via a large number of components, suggesting many potential sites of pharmacological intervention. Therapeutic targeting of UPS is exemplified by the recent FDA approval of dipeptidyl boronic acid bortezomib first-in-class proteasome inhibitor for the treatment of relapsed/refractory, relapsed, and newly diagnosed multiple myeloma (MM). As with other agents, however, dose-limiting toxicities and the development of resistance limit its long-term utility. Recent efforts have been devoted to discovery and development of small molecule inhibitors of the major components of UPS, such as E1-conjugating enzyme inhibitors, E3 ubiquitin ligase inhibitors (e.g inhibitors of TRAF6, SCF-p27, POSH, MDM2-P53) or Deubiquitylating enzyme (DUB) inhibitor. Therapeutic strategies using these small molecule inhibitors will allow for specific targeting of the UPS and therefore may be less likely than proteasome inhibitors to trigger off-target activities and associated toxicities. In this context, Ubiquitin Specific Protease-7 (USP-7), a deubiquitylating enzyme, is of particular interest since it regulates activity of key biological signaling pathways such as p53, the transcription factor FOXO-4, Cdk inhibitor p27, and PI3K/Akt. In the present study, we examined the anti-tumor activity of a novel USP-7 inhibitor P5091 (Progenra, Inc., Malvern, PA) in MM cells using both in vitro and in vivo model systems. Results: We first confirmed the functional specificity of P5091 using different experimental strategies: 1) Cell-free based experiments using a novel reporter (Ub-PLA2)-based assay showed a potent, specific and selective deubiquitylating activity of P5091 against USP-7 (EC50 = 5.6 mM) versus other DUBs and proteases (EC50 25 to > 50 mM); 2) Treatment of MM.1S MM cells with P5091 blocked USP-7 deubiquitylating activity, without altering proteasome activity; and 3) treatment of HCT-116 (wild type) cells with P5091 markedly decreased viability; conversely, knockout of USP-7 in HCT-116 conferred resistance to P5091 (P < 0.001; n=2). Taken together, these data demonstrate that P5091 targets USP-7. We next examined the effects of P5091 in MM cells. Protein expression analysis showed that USP-7 is highly expressed in MM cells. Treatment of MM cell lines (MM.1S, MM.1R, RPMI-8226, U266, KMS-12BM, and INA-6) and primary patient cells for 72h significantly decreased their viability (IC50 range 5-10 mM) (P < 0.001; n=3) without affecting the viability of normal peripheral blood mononuclear cells, suggesting specific anti-MM activity and a favorable therapeutic index for P5091. P5091-induced apoptosis was confirmed in MM.1S and RPMI-8226 cells, as evidenced by marked increase in Annexin V+ and PI- cell population (P < 0.001, n=3). Importantly, transfection of USP-7 siRNA, but not scrambled (genome control) siRNA, inhibits growth of MM.1S cells, similar to USP-7 inhibitor P5091 (P < 0.001; n=2). In addition, p5091 triggered apoptosis in MM cells even in the presence of MM bone marrow stromal cells. Mechanistic studies further showed that P5091-triggered apoptosis in MM cells is associated with 1) activation of caspase-8, caspase-9, caspase-3, and PARP; 2) activation of Noxa, PUMA, and Bid; 3) downregulation of MM cell growth and survival proteins: Akt, Bclxl, phospho-S6 ribosomal protein, and c-Myc; 4) inhibition of migration of MM cells and angiogenesis; and 5) suppression of USP-7 specific substrate HDM2 with concurrent increased expression of CDK inhibitor p21. Importantly, blockade of HDM2 and p21 using siRNA strategy significantly abrogates P5091-induced MM.1S cell death (P value <0.001; n=2). We next examined the in vivo efficacy of p5091 using human plasmacytoma xenograft mouse model. Treatment of tumor-bearing mice with P5091 (10 mg/kg IV, twice a week for three weeks), but not vehicle alone, significantly (P < 0.001) inhibits MM tumor growth and prolongs survival of these mice (P < 0.001; 13.6 days prologation of survival in mice receiving P5091 vs. vehicle-treated mice). The concentrations of P5091 were well tolerated by mice, without significant weight loss. Conclusions: These preclinical studies will allow for the development of next generation UPS-based therapies and provide the rationale for novel therapeutics targeting USP7 to improve patient outcome in MM. Disclosures: Chauhan: Progenra, Inc: Consultancy. Nicholson:Progenra, Inc: Employment.


Sign in / Sign up

Export Citation Format

Share Document