scholarly journals Discovery and Optimization of Selective Inhibitors of Meprin α (Part II)

Author(s):  
Chao Wang ◽  
Juan Diez ◽  
Hajeung Park ◽  
Christoph Becker-Pauly ◽  
Gregg B. Fields ◽  
...  

Meprin α is a zinc metalloproteinase (metzincin) that has been implicated in multiple diseases, including fibrosis and cancers. It has proven difficult to find small molecules that are capable of selectively inhibiting meprin α, or its close relative meprin β, over numerous other metzincins which, if inhibited, would elicit unwanted effects. We recently identified possible molecular starting points for meprin α-specific inhibition through an HTS effort (see part I, preceding paper). In part II we report the optimization of a potent and selective hydroxamic acid meprin α inhibitor probe which may help define the therapeutic potential for small molecule meprin α inhibition and spur further drug discovery efforts in the area of zinc metalloproteinase inhibition.

2021 ◽  
Vol 14 (3) ◽  
pp. 197
Author(s):  
Chao Wang ◽  
Juan Diez ◽  
Hajeung Park ◽  
Timothy P. Spicer ◽  
Louis D. Scampavia ◽  
...  

Meprin α is a zinc metalloproteinase (metzincin) that has been implicated in multiple diseases, including fibrosis and cancers. It has proven difficult to find small molecules that are capable of selectively inhibiting meprin a, or its close relative meprin b, over numerous other metzincins which, if inhibited, would elicit unwanted effects. We recently identified possible molecular starting points for meprin a-specific inhibition through an HTS effort (see part I, preceding paper). Here, in part II, we report further efforts to optimize potency and selectivity. We hope that a hydroxamic acid meprin α inhibitor probe will help define the therapeutic potential for small molecule meprin a inhibition and spur further drug discovery efforts in the area of zinc metalloproteinase inhibition.


2017 ◽  
Author(s):  
Neel S. Madhukar ◽  
Prashant K. Khade ◽  
Linda Huang ◽  
Kaitlyn Gayvert ◽  
Giuseppe Galletti ◽  
...  

AbstractDrug target identification is one of the most important aspects of pre-clinical development yet it is also among the most complex, labor-intensive, and costly. This represents a major issue, as lack of proper target identification can be detrimental in determining the clinical application of a bioactive small molecule. To improve target identification, we developed BANDIT, a novel paradigm that integrates multiple data types within a Bayesian machine-learning framework to predict the targets and mechanisms for small molecules with unprecedented accuracy and versatility. Using only public data BANDIT achieved an accuracy of approximately 90% over 2000 different small molecules – substantially better than any other published target identification platform. We applied BANDIT to a library of small molecules with no known targets and generated ∼4,000 novel molecule-target predictions. From this set we identified and experimentally validated a set of novel microtubule inhibitors, including three with activity on cancer cells resistant to clinically used anti-microtubule therapies. We next applied BANDIT to ONC201 – an active anti- cancer small molecule in clinical development – whose target has remained elusive since its discovery in 2009. BANDIT identified dopamine receptor 2 as the unexpected target of ONC201, a prediction that we experimentally validated. Not only does this open the door for clinical trials focused on target-based selection of patient populations, but it also represents a novel way to target GPCRs in cancer. Additionally, BANDIT identified previously undocumented connections between approved drugs with disparate indications, shedding light onto previously unexplained clinical observations and suggesting new uses of marketed drugs. Overall, BANDIT represents an efficient and highly accurate platform that can be used as a resource to accelerate drug discovery and direct the clinical application of small molecule therapeutics with improved precision.


2017 ◽  
Author(s):  
Carrow I. Wells ◽  
Nirav R. Kapadia ◽  
Rafael M. Couñago ◽  
David H. Drewry

AbstractPotent, selective, and cell active small molecule kinase inhibitors are useful tools to help unravel the complexities of kinase signaling. As the biological functions of individual kinases become better understood, they can become targets of drug discovery efforts. The small molecules used to shed light on function can also then serve as chemical starting points in these drug discovery efforts. The Nek family of kinases has received very little attention, as judged by number of citations in PubMed, yet they appear to play many key roles and have been implicated in disease. Here we present our work to identify high quality chemical starting points that have emerged due to the increased incidence of broad kinome screening. We anticipate that this analysis will allow the community to progress towards the generation of chemical probes and eventually drugs that target members of the Nek family.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 760-760
Author(s):  
Kimberly A. Hartwell ◽  
Peter G. Miller ◽  
Alison L. Stewart ◽  
Alissa R. Kahn ◽  
David J. Logan ◽  
...  

Abstract Abstract 760 Recent insights into the molecular and cellular processes that drive leukemia have called attention to the limitations intrinsic to traditional drug discovery approaches. To date, the majority of cell-based functional screens have relied on probing cell lines in vitro in isolation to identify compounds that decrease cellular viability. The development of novel therapeutics with greater efficacy and decreased toxicity will require the identification of small molecules that selectively target leukemia stem cells (LSCs) within the context of their microenvironment, while sparing normal cells. We hypothesized that it would be possible to systematically identify LSC susceptibilities by modeling key elements of bone marrow niche interactions in high throughput format. We tested this hypothesis by creating and optimizing an assay in which primary murine stem cell-enriched leukemia cells are plated on bone marrow stromal cells in 384-well format, and examined by a high content image-based readout of cobblestoning, an in vitro morphological surrogate of cell health and self-renewal. AML cells cultured in this way maintained their ability to reinitiate disease in mice with as few as 100 cells. 14,720 small molecule probes across diverse chemical space were screened at 5uM in our assay. Retest screening was performed in the presence of two different bone marrow stromal types in parallel, OP9s and primary mesenchymal stem cells (MSCs). Greater than 60% of primary screen hits positively retested (dose response with IC50 at or below 5 μM) on both types of stroma. Compounds that inhibited leukemic cobblestoning merely by killing the stroma were identified by CellTiter-Glo viability analysis and excluded. Compounds that killed normal primary hematopoietic stem and progenitor cell inputs, as assessed by a related co-culture screen, were also excluded. Selectivity for leukemia over normal hematopoietic cells was additionally examined in vitro by comingling these cells on stroma within the same wells. Primary human CD34+ AML leukemia and normal CD34+ cord blood cells were also tested, by way of the 5 week cobblestone area forming cell (CAFC) assay. Additionally, preliminary studies of human AML cells pulse-treated with small molecules ex vivo, followed by in vivo transplantation, provided further evidence of potent leukemia kill across genotypes. A biologically complex functional approach to drug discovery, such as the novel method described here, has previously been thought impossible, due to presumed incompatibility with high throughput scale. We show that it is possible, and that it bears fruit in a first pilot screen. By these means, we discover small molecule perturbants that act selectively in the context of the microenvironment to kill LSCs while sparing stroma and normal hematopoietic cells. Some hits act cell autonomously, and some do not, as evidenced by observed leukemia kill when only the stromal support cells are treated prior to the plating of leukemia. Some hits are known, such as parthenolide and celastrol, and some are previously underappreciated, such as HMG-CoA reductase inhibition. Others are entirely new, and would not have been revealed by conventional approaches to therapeutic discovery. We therefore present a powerful new approach, and identify drug candidates with the potential to selectively target leukemia stem cells in clinical patients. Disclosures: No relevant conflicts of interest to declare.


2015 ◽  
Vol 2015 ◽  
pp. 1-11 ◽  
Author(s):  
Hui Lin ◽  
Pingfang Song ◽  
Yi Zhao ◽  
Li-Jia Xue ◽  
Yi Liu ◽  
...  

T helper 17 (Th17) cells play a central role in inflammatory and autoimmune diseases via the production of proinflammatory cytokines interleukin- (IL-) 17, IL-17F, and IL-22. Anti-IL-17 monoclonal antibodies show potent efficacy in psoriasis but poor effect in rheumatoid arthritis (RA) and Crohn’s disease. Alternative agents targeting Th17 cells may be a better way to inhibit the development and function of Th17 cells than antibodies of blocking a single effector cytokine. Retinoic acid-related orphan receptor gamma t (RORγt) which acts as the master transcription factor of Th17 differentiation has been an attractive pharmacologic target for the treatment of Th17-mediated autoimmune disease. Recent progress in technology of chemical screen and engineering nucleic acid enable two new classes of therapeutics targeting RORγt. Chemical screen technology identified several small molecule specific inhibitors of RORγt from a small molecule library. Systematic evolution of ligands by exponential enrichment (SELEX) technology enabled target specific aptamers to be isolated from a random sequence oligonucleotide library. In this review, we highlight the development and therapeutic potential of small molecules inhibiting Th17 cells by targeting RORγt and aptamer mediated CD4+T cell specific delivery of small interference RNA against RORγt gene expression to inhibit pathogenic effector functions of Th17 lineage.


Author(s):  
Moses Moustakim ◽  
Thomas Christott ◽  
Octovia P. Monteiro ◽  
James Bennett ◽  
Charline Giroud ◽  
...  

<p>YEATS domain (YD) containing proteins are an emerging</p> <p>class of epigenetic targets in drug discovery. Dysregulation of these modified lysine binding proteins has been linked to the onset and progression of cancers. We herein report the discovery and characterisation of the first small molecule chemical probe, SGC-iMLLT, for the YD of MLLT1 (ENL/YEATS1) and MLLT3 (AF9/YEATS3). SGC-iMLLT is a potent and selective inhibitor of MLLT1/3 -histone interactions. Excellent selectivity over other human YD proteins (YEATS2/4) and bromodomains was observed. Furthermore, our probe displays cellular target engagement of MLLT1 and MLLT3. The first small molecule X-ray co-crystal structures with the MLLT1 YD are also reported. This first in class probe molecule can be used to understand MLLT1/3 associated biology and the therapeutic potential of small molecule YD inhibitors.</p>


2021 ◽  
Vol 4 (1) ◽  
Author(s):  
Akinobu Senoo ◽  
Sho Ito ◽  
Satoru Nagatoishi ◽  
Yutaro Saito ◽  
Go Ueno ◽  
...  

AbstractMany cadherin family proteins are associated with diseases such as cancer. Since cell adhesion requires homodimerization of cadherin molecules, a small-molecule regulator of dimerization would have therapeutic potential. Herein, we describe identification of a P-cadherin-specific chemical fragment that inhibits P-cadherin-mediated cell adhesion. Although the identified molecule is a fragment compound, it binds to a cavity of P-cadherin that has not previously been targeted, indirectly prevents formation of hydrogen bonds necessary for formation of an intermediate called the X dimer and thus modulates the process of X dimerization. Our findings will impact on a strategy for regulation of protein-protein interactions and stepwise assembly of protein complexes using small molecules.


2020 ◽  
Vol 21 (15) ◽  
pp. 5262 ◽  
Author(s):  
Qingxin Li ◽  
CongBao Kang

Small-molecule drugs are organic compounds affecting molecular pathways by targeting important proteins. These compounds have a low molecular weight, making them penetrate cells easily. Small-molecule drugs can be developed from leads derived from rational drug design or isolated from natural resources. A target-based drug discovery project usually includes target identification, target validation, hit identification, hit to lead and lead optimization. Understanding molecular interactions between small molecules and their targets is critical in drug discovery. Although many biophysical and biochemical methods are able to elucidate molecular interactions of small molecules with their targets, structural biology is the most powerful tool to determine the mechanisms of action for both targets and the developed compounds. Herein, we reviewed the application of structural biology to investigate binding modes of orthosteric and allosteric inhibitors. It is exemplified that structural biology provides a clear view of the binding modes of protease inhibitors and phosphatase inhibitors. We also demonstrate that structural biology provides insights into the function of a target and identifies a druggable site for rational drug design.


BioTechniques ◽  
2020 ◽  
Vol 69 (1) ◽  
pp. 70-76
Author(s):  
Xiaoyun Meng ◽  
Lanjun Zhang ◽  
Hong Wei ◽  
Furong Li ◽  
Lihua Hu ◽  
...  

Refolding of human interleukin 17A (IL-17A) has been reported; however, the key refolding protocol was not robust enough to deliver consistent results and to be easily scaled up for crystallization. Here we report an optimized refolding method for IL-17A. Although co-crystal structures of IL-17A with ligands have been obtained with a high-affinity peptide and an anti-IL-17A Fab as stabilizers, neither the production yield nor the characterization of the IL-17A/Fab complex was reported. To facilitate co-crystallization of IL-17A with small-molecule compounds derived from our DNA encoded library, we also describe the method for yield enhancement of anti-IL-17A Fab production and characterize the IL-17A/Fab complex for the first time, providing an essential prerequisite for structure-based drug discovery targeting IL-17A.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 571-571
Author(s):  
Shrikanta Chattopadhyay ◽  
Alison L. Stewart ◽  
Siddhartha Mukherjee ◽  
Cherrie Huang ◽  
Kimberly A. Hartwell ◽  
...  

Abstract Abstract 571 Despite advances in the treatment of multiple myeloma (MM), this disease remains incurable and novel therapeutic strategies are urgently needed. Ideal strategies would overcome resistance factors from the bone-marrow microenvironment (niche) since a variety of inhibitors are rendered less effective by bone-marrow stromal cells (BMSCs) of the MM niche (McMillin et al., Nat Med. 2010 Apr;16(4):483–9). Drug discovery often entails a target-based approach but identifying targets in MM is challenging because of its complex genome and multiple niche interactions. We used a chemical biology approach in which small-molecule inhibitors of MM cells, grown within their niche, are first identified and then used to discover targets within MM or its niche. These compounds also serve as leads for future drug discovery. To model myeloma/niche interactions, we chose an MM cell line MOLP5 that has an obligate dependence on BMSCs to maintain viability. Small-molecule inhibitors were identified by screening ∼25,000 structurally diverse small molecules on GFP-labeled MOLP5 cells co-cultured with primary BMSCs derived from hip replacement samples. MOLP5 growth inhibition was measured by quantifying GFP(+) cells with automated high-throughput microscopy. About 800 hits were counter-screened on BMSCs alone to exclude non-specifically toxic compounds. The remaining 182 MOLP5-selective inhibitors were then tested on 2 other GFP-labeled MM cell-lines, MM1S and INA6, in the presence or absence of BMSCs to exclude compounds that are less effective in the presence of BMSCs. The 64 compounds that overcome BMSC resistance were tested on CD34+ human hematopoietic progenitors to prioritize compounds with selectivity between MM and normal blood cells. The 8 compounds that met these criteria fell into 3 categories: 1) compounds with equal activity in the presence or absence of BMSCs (overcome stromal resistance); 2) compounds with selectivity for BMSC-dependent MOLP5 cells (block stromal viability factors); and 3) compounds with increased activity in the presence of BMSCs (enhance stromal inhibitory factors). Because most efficacious clinical compounds like bortezomib act like compounds in category 1, compound BRD9876 was chosen from this category for mechanistic studies. Gene-expression profiling of BRD9876-treated MM1S cells suggested possible links to mitotic arrest and cell cycle analyses revealed a rapid accumulation of cells in the G2/M phase. Treated cells were stained for the mitotic spindle protein α-tubulin and found to exhibit an aberrant mono-astral mitotic phenotype, reminiscent of the kinesin-5 (Eg5; KIF11) inhibitor monastrol. This was encouraging because a kinesin-5 inhibitor ARRY-520 has shown promising durable responses in multiple myeloma (Shah et al, ASH Annual Meeting 2011; Abstract 1860). To determine if BRD9876 was a kinesin-5 inhibitor, a BRD9876-resistant sub-line of MM1S was developed and the kinesin-5 gene sequenced. BRD9876-resistant cells have a novel kinesin-5 mutation (Y104C) at a site that is distant from the monastrol-binding pocket. Most kinesin-5 inhibitors in clinical development bind the monastrol pocket, and the BRD9876-resistant cells were not cross-resistant to one such inhibitor, ispinesib, suggesting a distinct mode of kinesin-5 inhibition by BRD9876. To identify biomarkers of sensitivity to BRD9876, quantitative dose/response measurements in 98 genetically characterized cell lines (Schreiber & co-workers, submitted) comprising a subset of the Cancer Cell Line Encyclopedia (CCLE) were analyzed. Unbiased analyses correlating genetic features with sensitivity revealed that mutations in the mitotic regulator WEE1 were associated with sensitivity to BRD9876. Validation studies comparing WEE1 mutant to wild-type cell lines confirmed enhanced sensitivity of mutant cells to both BRD9876 and ispinesib suggesting that WEE1 mutations could be a useful biomarker for different kinesin-5 inhibitors. In contrast, co-treatment of WEE1 WT cells with sub-toxic concentrations of the WEE1 inhibitor MK1775 led to marked enhancement of BRD9876 activity but had little effect on ispinesib activity, suggesting a unique synergistic relationship between WEE1 inhibitors and BRD9876. In summary, niche-based screening in multiple myeloma has revealed a novel therapeutic candidate and can complement other drug-discovery approaches against this disease. Disclosures: Ebert: Celgene: Consultancy; Genoptix: Consultancy. Raje:Onyx: Consultancy; Celgene: Consultancy; Millennium: Consultancy; Acetylon: Research Funding; Amgen: Research Funding; Eli-Lilly: Research Funding.


Sign in / Sign up

Export Citation Format

Share Document