Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Sui Xiong Cai is active.

Publication


Featured researches published by Sui Xiong Cai.


Journal of Medicinal Chemistry | 2008

Discovery of 4-aryl-4H-chromenes as a new series of apoptosis inducers using a cell- and caspase-based high throughput screening assay. 4. Structure-activity relationships of N-alkyl substituted pyrrole fused at the 7,8-positions.

William Kemnitzer; John Drewe; Songchun Jiang; Hong Zhang; Candace Crogan-Grundy; Denis Labreque; Monica Bubenick; Giorgio Attardo; Real Denis; Serge Lamothe; Henriette Gourdeau; Ben Tseng; Shailaja Kasibhatla; Sui Xiong Cai

In our continuing effort to discover and develop apoptosis inducing 4-aryl-4H-chromenes as novel anticancer agents, we explored the structure-activity relationship (SAR) of alkyl substituted pyrrole fused at the 7,8-positions. A methyl group substituted at the nitrogen in the 7-position of the pyrrole ring led to a series of potent apoptosis inducers with potency in the low nanomolar range. These compounds were also found to be low nanomolar or subnanomolar inhibitors of cell growth, and they inhibited tubulin polymerization, indicating that methylation of the 7-position nitrogen does not change the mechanism of action of these chromenes. Compound 2d was identified as a highly potent apoptosis inducer with an EC50 value of 2 nM and a highly potent inhibitor of cell growth with a GI50 value of 0.3 nM in T47D cells.


Pain | 1997

Antinociceptive effects of NMDA and non-NMDA receptor antagonists in the tail flick test in mice

Kabirullah Lutfy; Sui Xiong Cai; Richard M. Woodward; Eckard Weber

Abstract Inhibition of spinal glutamate receptors induces antinociceptive effects in numerous animal models of pain. The present study compares the effects of intrathecally administered N‐methyl‐d‐aspartate (NMDA) and non‐NMDA glutamate receptor antagonists on nociceptive responses in the tail flick test. Potency of antagonists at NMDA and &agr;‐amino‐3‐hydroxy‐5‐methyl‐4‐isoxazole‐propionic acid (AMPA) receptors was first measured by electrical assays in Xenopus oocytes expressing rat cerebral cortex poly(A)+ RNA. Subsequently, Swiss Webster mice were injected intrathecally with the antagonists and tested for antinociception. The drugs tested were: NBQX and GYKI‐52466, selective AMPA receptor antagonists, ketamine, MK‐801, R(+) HA‐966 and ACEA‐0762, selective NMDA receptor antagonists, and ACEA‐1031, ACEA‐1328 and ACEA‐0593, NMDA receptor antagonists that also show inhibition of non‐NMDA receptors. Selective NMDA receptor antagonists induced essentially no antinociceptive effects in the tail flick test. Antinociceptive activity generally correlated with inhibition of AMPA receptors. The exception was the non‐competitive AMPA receptor antagonist GYKI‐52466, which was unexpectedly weak. This may be due to inadequate dosing, because the compound has limited solubility, or may be due to differences in the non‐NMDA receptor subtype‐selectivity profile of GYKI‐52466 as compared to competitive antagonists such as NBQX. Overall, our results suggest that inhibition of spinal non‐NMDA receptors is the primary, and necessary, mechanism of antinociception by these drugs in the tail flick test in mice.


British Journal of Pharmacology | 2003

MX1013, a dipeptide caspase inhibitor with potent in vivo antiapoptotic activity

Wu Yang; John Guastella; Jin-Cheng Huang; Yan Wang; Li Zhang; Dong Xue; Minhtam Tran; Richard M. Woodward; Shailaja Kasibhatla; Ben Tseng; John Drewe; Sui Xiong Cai

Caspases play a critical role in apoptosis, and are considered to be key targets for the design of cytoprotective drugs. As part of our antiapoptotic drug‐discovery effort, we have synthesized and characterized Z‐VD‐fmk, MX1013, as a potent, irreversible dipeptide caspase inhibitor. MX1013 inhibits caspases 1, 3, 6, 7, 8, and 9, with IC50 values ranging from 5 to 20 nM. MX1013 is selective for caspases, and is a poor inhibitor of noncaspase proteases, such as cathepsin B, calpain I, or Factor Xa (IC50 values >10 μM). In several cell culture models of apoptosis, including caspase 3 processing, PARP cleavage, and DNA fragmentation, MX1013 is more active than tetrapeptide‐ and tripeptide‐based caspase inhibitors, and blocked apoptosis at concentrations as low as 0.5 μM. MX1013 is more aqueous soluble than tripeptide‐based caspase inhibitors such as Z‐VAD‐fmk. At a dose of 1 mg kg−1 i.v., MX1013 prevented liver damage and the lethality caused by Fas death receptor activation in the anti‐Fas mouse‐liver apoptosis model, a widely used model of liver failure. At a dose of 20 mg kg−1 (i.v. bolus) followed by i.v. infusion for 6 or 12 h, MX1013 reduced cortical damage by approximately 50% in a model of brain ischemia/reperfusion injury. At a dose of 20 mg kg−1 (i.v. bolus) followed by i.v. infusion for 12 h, MX1013 reduced heart damage by approximately 50% in a model of acute myocardial infarction. Based on these studies, we conclude that MX1013, a dipeptide pan‐caspase inhibitor, has a good combination of in vitro and in vivo properties. It has the ability to protect cells from a variety of apoptotic insults, and is systemically active in three animal models of apoptosis, including brain ischemia.


Journal of Medicinal Chemistry | 2009

Discovery of N-(4-Methoxyphenyl)-N,2-dimethylquinazolin-4-amine, a Potent Apoptosis Inducer and Efficacious Anticancer Agent with High Blood Brain Barrier Penetration

Nilantha Sudath Sirisoma; Azra Pervin; Hong Zhang; Songchun Jiang; J. Adam Willardsen; Mark B. Anderson; Gary Mather; Christopher M. Pleiman; Shailaja Kasibhatla; Ben Tseng; John Drewe; Sui Xiong Cai

As a continuation of our structure-activity relationship (SAR) studies on 4-anilinoquinazolines as potent apoptosis inducers and to identify anticancer development candidates, we explored the replacement of the 2-Cl group in our lead compound 2-chloro-N-(4-methoxyphenyl)-N-methylquinazolin-4-amine (6b, EP128265, MPI-0441138) by other functional groups. This SAR study and lead optimization resulted in the identification of N-(4-methoxyphenyl)-N,2-dimethylquinazolin-4-amine (6h, EP128495, MPC-6827) as an anticancer clinical candidate. Compound 6h was found to be a potent apoptosis inducer with EC(50) of 2 nM in our cell-based apoptosis induction assay. It also has excellent blood brain barrier penetration, and is highly efficacious in human MX-1 breast and other mouse xenograft cancer models.


Bioorganic & Medicinal Chemistry Letters | 2008

Discovery of 4-aryl-2-oxo-2H-chromenes as a new series of apoptosis inducers using a cell- and caspase-based high-throughput screening assay

William Kemnitzer; Songchun Jiang; Hong Zhang; Shailaja Kasibhatla; Candace Crogan-Grundy; Charles Blais; Giorgio Attardo; Real Denis; Serge Lamothe; Henriette Gourdeau; Ben Tseng; John Drewe; Sui Xiong Cai

As a continuation of our efforts to discover and develop the apoptosis inducing 4-aryl-4H-chromenes as potential anticancer agents, we explored the removal of the chiral center at the 4-position and prepared a series of 4-aryl-2-oxo-2H-chromenes. It was found that, in general, removal of the chiral center and replacement of the 2-amino group with a 2-oxo group were tolerated and 4-aryl-2-oxo-2H-chromenes exhibited SAR similar to 4-aryl-2-amino-4H-chromenes. The 4-aryl-2-oxo-2H-chromenes with a N-methyl pyrrole fused at the 7,8-positions were highly active with compound 2a having an EC(50) value of 13 nM in T47D cells. It was found that an OMe group was preferred at the 7-position. 7-NMe(2), 7-NH(2), 7-Cl and 7,8 fused pyrido analogs all had low potency. These 4-aryl-2-oxo-2H-chromenes are a series of potent apoptosis inducers with potential advantage over the 4-aryl-2-amino-4H-chromenes series via elimination of the chiral center at the 4-position.


Cancer Research | 2007

MPC-6827: A Small-Molecule Inhibitor of Microtubule Formation That Is Not a Substrate for Multidrug Resistance Pumps

Shailaja Kasibhatla; Vijay Baichwal; Sui Xiong Cai; Bruce J. Roth; Ira Skvortsova; Sergej Skvortsov; Peter Lukas; Nicole Marion English; Nilantha Sudath Sirisoma; John Drewe; Azra Pervin; Ben Tseng; Robert O. Carlson; Christopher M. Pleiman

A novel series of 4-arylaminoquinazolines were identified from a cell-based screening assay as potent apoptosis inducers. Through structure-activity relationship studies, MPC-6827 and its close structural analogue, MPI-0441138, were discovered as proapoptotic molecules and mitotic inhibitors with potencies at low nanomolar concentrations in multiple tumor cell lines. Photoaffinity and radiolabeled analogues of MPC-6827 were found to bind a 55-kDa protein, and this binding was competed by MPC-6827, paclitaxel, and colchicine, but not vinblastine. MPC-6827 effectively inhibited the polymerization of tubulin in vitro, competed with colchicine binding, and disrupted the formation of microtubules in a variety of tumor cell lines, which together showed the molecular target as tubulin. Treatment of MCF-7 breast carcinoma or Jurkat leukemia cells with MPC-6827 led to pronounced G2-M cell cycle arrest followed by apoptosis. Apoptosis, as determined by terminal deoxyribonucleotidyl transferase-mediated dUTP nick end labeling assay, was preceded by loss of mitochondrial membrane potential, cytochrome c translocation from mitochondria to nuclei, activation of caspase-3, and cleavage of poly(ADP-ribose) polymerase. MPC-6827 was equipotent in an in vitro growth inhibition assay in several cancer cell lines regardless of the expression levels of the multidrug resistance ABC transporters MDR-1 (Pgp-1), MRP-1, and BCRP-1. In B16-F1 allografts and in OVCAR-3, MIAPaCa-2, MCF-7, HT-29, MDA-MB-435, and MX-1 xenografts, statistically significant tumor growth inhibition was observed with MPC-6827. These studies show that MPC-6827 is a microtubule-disrupting agent with potent and broad-spectrum in vitro and in vivo cytotoxic activities and, therefore, MPC-6827 is a promising candidate for development as a novel therapeutic for multiple cancer types.


Molecular Cancer Therapeutics | 2005

The discovery and mechanism of action of novel tumor-selective and apoptosis-inducing 3,5-diaryl-1,2,4-oxadiazole series using a chemical genetics approach.

Katayoun Jessen; Nicole M. English; Jean Yu Wang; Sergei Maliartchouk; Shannon P. Archer; Ling Qiu; Regina Brand; Jared Kuemmerle; Han-Zhong Zhang; Kurt R. Gehlsen; John Drewe; Ben Tseng; Sui Xiong Cai; Shailaja Kasibhatla

A novel series of 3,5-diaryl-oxadiazoles was identified as apoptosis-inducing agents through our cell and chemical genetics–based screening assay for compounds that induce apoptosis using a chemical genetics approach. Several analogues from this series including MX-74420 and MX-126374 were further characterized. MX-126374, a lead compound from this series, was shown to induce apoptosis and inhibit cell growth selectively in tumor cells. To elucidate the mechanism(s) by which this class of compounds alters the signal transduction pathway that ultimately leads to apoptosis, expression profiling using the Affymetrix Gene Chip array technology was done along with other molecular and biochemical analyses. Interestingly, we have identified several key genes (cyclin D1, transforming growth factor-β1, p21, and insulin-like growth factor-BP3) that are altered in the presence of this compound, leading to characterization of the pathway for activation of apoptosis. MX-126374 also showed significant inhibition of tumor growth as a single agent and in combination with paclitaxel in murine tumor models. Using photoaffinity labeling, tail-interacting protein 47, an insulin-like growth factor-II receptor binding protein, was identified as the molecular target. Further studies indicated that down-regulation of tail-interacting protein 47 in cancer cells by small interfering RNA shows a similar pathway profile as compound treatment. These data suggest that 3,5-diaryl-oxadiazoles may be a new class of anticancer drugs that are tumor-selective and further support the discovery of novel drugs and drug targets using chemical genetic approaches.


Anti-cancer Agents in Medicinal Chemistry | 2009

Discovery of 4-Aryl-4H-Chromenes as Potent Apoptosis Inducers Using a Cell- and Caspase-Based Anti-Cancer Screening Apoptosis Program (ASAP): SAR Studies and the Identification of Novel Vascular Disrupting Agents

Sui Xiong Cai; John Drewe; William Kemnitzer

Many cancer cells are known to have defects in the apoptosis machinery. Therefore identification of compounds that can activate or promote apoptosis in cancer cells is an attractive approach for targeted therapies. By applying a novel cell- and caspase-based Anti-cancer Screening Apoptosis Program (ASAP) HTS assay, 4-aryl-4H-chromenes were identified as potent apoptosis inducers. 4-Aryl-4H-chromenes were found to induce nuclear fragmentation and PARP cleavage, as well as to arrest cells at the G(2)/M stage followed by apoptosis as determined by the flow cytometry analysis assay in multiple human cell lines (e.g. Jurkat, T47D). These compounds were found to be highly active in the growth inhibition MTT assay, including for paclitaxel resistant, p-glycoprotein overexpressed, MES-SA/DX5 tumor cells. Functionally, they were found to be potent inhibitors of tubulin polymerization and to effectively inhibit the binding of colchicine to tubulin. In addition, several 4-aryl-4H-chromenes were also found to be effective vascular disrupting agents (VDA). One of the lead compounds, EPC2407, is currently in clinical trials as a novel tumor vascular disrupting agent.


Journal of Medicinal Chemistry | 2008

Discovery of 2-Chloro-N-(4-methoxyphenyl)-N-methylquinazolin-4-amine (EP128265, MPI-0441138) as a Potent Inducer of Apoptosis with High In Vivo Activity

Nilantha Sudath Sirisoma; Shailaja Kasibhatla; Azra Pervin; Hong Zhang; Songchun Jiang; J. Adam Willardsen; Mark B. Anderson; Vijay Baichwal; Gary Mather; Kevin Jessing; Raouf A. Hussain; Khanh Hoang; Christopher M. Pleiman; Ben Tseng; John Drewe; Sui Xiong Cai

Using a live cell, high-throughput caspase-3 activator assay, we have identified a novel series of 4-anilinoquinazolines as inducers of apoptosis. In this report, we discuss the discovery of 2-chloro-N-(4-methoxyphenyl)-N-methylquinazolin-4-amine, compound 2b (EP128265, MPI-0441138) as a highly active inducer of apoptosis (EC50 for caspase activation of 2 nM) and as a potent inhibitor of cell proliferation (GI50 of 2 nM) in T47D cells. Compound 2b inhibited tubulin polymerization, was effective in cells overexpressing ABC transporter Pgp-1, and was efficacious in the MX-1 human breast and PC-3 prostate cancer mouse models. In contrast to the SAR of 4-anilinoquinazolines as EGFR kinase inhibitors, the methyl group on the nitrogen linker was essential for the apoptosis-inducing activity of 4-anilinoquinazolines and substitution in the 6- and 7-positions of the quinazoline core structure decreased potency.


Journal of Fluorine Chemistry | 1989

Functionalized Perfluorophenyl Azides: New Reagents for Photoaffinity Labeling

John F. W. Keana; Sui Xiong Cai

Abstract Several substituted perfluorophenyl azides capable of attachment to other molecules by an acylation reaction were synthesized for use as photoaffinity labeling reagents.

Collaboration


Dive into the Sui Xiong Cai's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shailaja Kasibhatla

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar

Eckard Weber

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mingdi Yan

University of Massachusetts Lowell

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge