Network


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

Hotspot


Dive into the research topics where Kaustav Biswas is active.

Publication


Featured researches published by Kaustav Biswas.


Tetrahedron Letters | 2003

Effects of temperature and concentration in some ring closing metathesis reactions

Kana Yamamoto; Kaustav Biswas; Christoph Gaul; Samuel J. Danishefsky

Ring closing metathesis (RCM) has emerged as a powerful tool to construct macrocyclic ring systems. However, the product distribution of monomer and oligomers is often a problem in the formation of medium to large rings. In the course of synthetic studies on the natural product radicicol and its analogs, we have found that the reaction temperature, along with concentration, has significant impact on the outcome of the product ratio. Specifically, carrying out the RCM reaction in refluxing toluene (110°C) at higher dilution affords improved yields of the monomeric macrocycle. Similar observations for another family of macrolactone natural products, the epothilones, are also reported.


Tetrahedron Letters | 2002

Construction of carbohydrate-based antitumor vaccines: synthesis of glycosyl amino acids by olefin cross-metathesis

Kaustav Biswas; Don M. Coltart; Samuel J. Danishefsky

Abstract The synthesis of biologically relevant glycosyl amino acids from corresponding O-allyl glycosides is described. The procedure involves a cross-metathesis reaction with Fmoc- l -allylglycine benzyl ester, followed by reduction of the resulting olefin via catalytic hydrogenation, with the concomitant release of the free acid. This method has also been applied to the breast and prostate cancer antigen Globo-H, to afford a hexasaccharide glycosyl amino acid that has been previously incorporated in a polyvalent antitumor vaccine.


Chemical Communications | 2002

Application of hitherto unexplored macrocyclization strategies in the epothilone series: novel epothilone analogs by total synthesisElectronic supplementary information (ESI) available: experimental details for the synthesis of 14 and 32. See http://www.rsc.org/suppdata/cc/b2/b209941a/

Jon T. Njardarson; Kaustav Biswas; Samuel J. Danishefsky

A total synthesis of Epothilone 490 and a synthesis of 11-hydroxy dEpoB utilizing a vinyl-boronate cross-metathesis followed by a Suzuki macrocyclization. A mild route to reach aldehydes from terminal olefins, anticipating Nozaki-Kishi macrocyclization is described.


Journal of Medicinal Chemistry | 2014

Discovery of clinical candidate 1-(4-(3-(4-(1H-benzo[d]imidazole-2-carbonyl)phenoxy)pyrazin-2-yl)piperidin-1-yl)ethanone (AMG 579), a potent, selective, and efficacious inhibitor of phosphodiesterase 10A (PDE10A).

Essa Hu; Ning Chen; Matthew P. Bourbeau; Paul E. Harrington; Kaustav Biswas; Roxanne Kunz; Kristin L. Andrews; Samer Chmait; Xiaoning Zhao; Carl D. Davis; Ji Ma; Jianxia Shi; Dianna Lester-Zeiner; Jean Danao; Jessica Able; Madelyn Cueva; Santosh Talreja; Thomas Kornecook; Hang Chen; Amy Porter; Randall W. Hungate; James J. S. Treanor; Jennifer R. Allen

We report the identification of a PDE10A clinical candidate by optimizing potency and in vivo efficacy of promising keto-benzimidazole leads 1 and 2. Significant increase in biochemical potency was observed when the saturated rings on morpholine 1 and N-acetyl piperazine 2 were changed by a single atom to tetrahydropyran 3 and N-acetyl piperidine 5. A second single atom modification from pyrazines 3 and 5 to pyridines 4 and 6 improved the inhibitory activity of 4 but not 6. In the in vivo LC-MS/MS target occupancy (TO) study at 10 mg/kg, 3, 5, and 6 achieved 86-91% occupancy of PDE10A in the brain. Furthermore, both CNS TO and efficacy in PCP-LMA behavioral model were observed in a dose dependent manner. With superior in vivo TO, in vivo efficacy and in vivo PK profiles in multiple preclinical species, compound 5 (AMG 579) was advanced as our PDE10A clinical candidate.


Tetrahedron Letters | 2003

Hydroxynorleucine as a glycosyl acceptor is an efficient means for introducing amino acid functionality into complex carbohydrates

Stacy J. Keding; Endo Atsushi; Kaustav Biswas; Andrzej Zatorski; Don M. Coltart; Samuel J. Danishefsky

Abstract A new approach to the synthesis of biologically relevant glycosyl amino acids using a non-natural amino acid as the glycosyl acceptor is described. The procedure involves a glycosylation reaction of a suitable carbohydrate donor with Fmoc- l -hydroxynorleucine benzyl ester. This reaction results in the direct incorporation of the amino acid moiety. The acceptor can be used for the preparation of α- or β- O -linked glycosides depending upon the nature of the glycosyl donor. This method has been applied in the synthesis of six different tumor-associated carbohydrate antigens.


Journal of Medicinal Chemistry | 2016

Single Residue Substitutions That Confer Voltage-Gated Sodium Ion Channel Subtype Selectivity in the NaV1.7 Inhibitory Peptide GpTx-1

Justin K. Murray; Jason Long; Anruo Zou; Joseph Ligutti; Kristin L. Andrews; Leszek Poppe; Kaustav Biswas; Bryan D. Moyer; Les P. Miranda

There is interest in the identification and optimization of new molecular entities selectively targeting ion channels of therapeutic relevance. Peptide toxins represent a rich source of pharmacology for ion channels, and we recently reported GpTx-1 analogs that inhibit NaV1.7, a voltage-gated sodium ion channel that is a compelling target for improved treatment of pain. Here we utilize multi-attribute positional scan (MAPS) analoging, combining high-throughput synthesis and electrophysiology, to interrogate the interaction of GpTx-1 with NaV1.7 and related NaV subtypes. After one round of MAPS analoging, we found novel substitutions at multiple residue positions not previously identified, specifically glutamic acid at positions 10 or 11 or lysine at position 18, that produce peptides with single digit nanomolar potency on NaV1.7 and 500-fold selectivity against off-target sodium channels. Docking studies with a NaV1.7 homology model and peptide NMR structure generated a model consistent with the key potency and selectivity modifications mapped in this work.


Journal of Medicinal Chemistry | 2011

Discovery of potent, orally bioavailable phthalazinone bradykinin B1 receptor antagonists.

Kaustav Biswas; Tanya Peterkin; Marian C. Bryan; Leyla Arik; Sonya G. Lehto; Hong Sun; Feng-Yin Hsieh; Cen Xu; Robert T. Fremeau; Jennifer R. Allen

The bradykinin B1 receptor is rapidly induced upon tissue injury and inflammation, stimulating the production of inflammatory mediators resulting in plasma extravasation, leukocyte trafficking, edema, and pain. We have previously reported on sulfonamide and sulfone-based B1 antagonists containing a privileged bicyclic amine moiety leading to potent series of 2-oxopiperazines. The suboptimal pharmacokinetics and physicochemical properties of the oxopiperazine sulfonamides led us to seek B1 antagonists with improved druglike properties. Using a pharmacophore model containing a bicyclic amine as anchor, we designed a series of amide antagonists with targeted physicochemical properties. This approach led to a novel series of potent phthalazinone B1 antagonists, where we successfully replaced a sulfonamide acceptor with a cyclic carbonyl unit. SAR studies revealed compounds with subnanomolar B1 binding affinity. These compounds demonstrate excellent cross-species PK properties with high oral bioavailability and potent activity in a rabbit biochemical challenge pharmacodynamic study.


ACS Medicinal Chemistry Letters | 2015

Unfolded Protein Response in Cancer: IRE1α Inhibition by Selective Kinase Ligands Does Not Impair Tumor Cell Viability.

Paul Harrington; Kaustav Biswas; David Malwitz; Andrew Tasker; Christopher Mohr; Kristin L. Andrews; Ken Dellamaggiore; Richard Kendall; Holger Beckmann; Peter Jaeckel; Silvia Materna-Reichelt; Jennifer R. Allen; J. Russell Lipford

The kinase/endonuclease inositol requiring enzyme 1 (IRE1α), one of the sensors of unfolded protein accumulation in the endoplasmic reticulum that triggers the unfolded protein response (UPR), has been investigated as an anticancer target. We identified potent allosteric inhibitors of IRE1α endonuclease activity that bound to the kinase site on the enzyme. Structure-activity relationship (SAR) studies led to 16 and 18, which were selective in kinase screens and were potent against recombinant IRE1α endonuclease as well as cellular IRE1α. The first X-ray crystal structure of a kinase inhibitor (16) bound to hIRE1α was obtained. Screening of native tumor cell lines (>300) against selective IRE1α inhibitors failed to demonstrate any effect on cellular viability. These results suggest that IRE1α activity is not essential for viability in most tumor cell lines, in vitro, and that interfering with the survival functions of the UPR may not be an effective strategy to block tumorigenesis.


ACS Chemical Biology | 2017

Engineering Antibody Reactivity for Efficient Derivatization to Generate NaV1.7 Inhibitory GpTx-1 Peptide–Antibody Conjugates

Kaustav Biswas; Thomas Nixey; Justin K. Murray; James Richard Falsey; Li Yin; Hantao Liu; Jacinthe Gingras; Brian E. Hall; Brad Herberich; Jerry Ryan Holder; Hongyan Li; Joseph Ligutti; Min-Hwa Jasmine Lin; Dong Liu; Brian Soriano; Marcus Soto; Linh Tran; Christopher M. Tegley; Anrou Zou; Kannan Gunasekaran; Bryan D. Moyer; Liz Doherty; Les P. Miranda

The voltage-gated sodium channel NaV1.7 is a genetically validated pain target under investigation for the development of analgesics. A therapeutic with a less frequent dosing regimen would be of value for treating chronic pain; however functional NaV1.7 targeting antibodies are not known. In this report, we describe NaV1.7 inhibitory peptide-antibody conjugates as an alternate construct for potential prolonged channel blockade through chemical derivatization of engineered antibodies. We previously identified NaV1.7 inhibitory peptide GpTx-1 from tarantula venom and optimized its potency and selectivity. Tethering GpTx-1 peptides to antibodies bifunctionally couples FcRn-based antibody recycling attributes to the NaV1.7 targeting function of the peptide warhead. Herein, we conjugated a GpTx-1 peptide to specific engineered cysteines in a carrier anti-2,4-dinitrophenol monoclonal antibody using polyethylene glycol linkers. The reactivity of 13 potential cysteine conjugation sites in the antibody scaffold was tuned using a model alkylating agent. Subsequent reactions with the peptide identified cysteine locations with the highest conversion to desired conjugates, which blocked NaV1.7 currents in whole cell electrophysiology. Variations in attachment site, linker, and peptide loading established design parameters for potency optimization. Antibody conjugation led to in vivo half-life extension by 130-fold relative to a nonconjugated GpTx-1 peptide and differential biodistribution to nerve fibers in wild-type but not NaV1.7 knockout mice. This study describes the optimization and application of antibody derivatization technology to functionally inhibit NaV1.7 in engineered and neuronal cells.


PLOS ONE | 2018

Pharmacological characterization of potent and selective NaV1.7 inhibitors engineered from Chilobrachys jingzhao tarantula venom peptide JzTx-V.

Bryan D. Moyer; Justin K. Murray; Joseph Ligutti; Kristin L. Andrews; Philippe Favreau; John B. Jordan; Josie H. Lee; Dong Liu; Jason Long; Kelvin Sham; Licheng Shi; Reto Stöcklin; Bin Wu; Ruoyuan Yin; Violeta Yu; Anruo Zou; Kaustav Biswas; Les P. Miranda

Identification of voltage-gated sodium channel NaV1.7 inhibitors for chronic pain therapeutic development is an area of vigorous pursuit. In an effort to identify more potent leads compared to our previously reported GpTx-1 peptide series, electrophysiology screening of fractionated tarantula venom discovered the NaV1.7 inhibitory peptide JzTx-V from the Chinese earth tiger tarantula Chilobrachys jingzhao. The parent peptide displayed nominal selectivity over the skeletal muscle NaV1.4 channel. Attribute-based positional scan analoging identified a key Ile28Glu mutation that improved NaV1.4 selectivity over 100-fold, and further optimization yielded the potent and selective peptide leads AM-8145 and AM-0422. NMR analyses revealed that the Ile28Glu substitution changed peptide conformation, pointing to a structural rationale for the selectivity gains. AM-8145 and AM-0422 as well as GpTx-1 and HwTx-IV competed for ProTx-II binding in HEK293 cells expressing human NaV1.7, suggesting that these NaV1.7 inhibitory peptides interact with a similar binding site. AM-8145 potently blocked native tetrodotoxin-sensitive (TTX-S) channels in mouse dorsal root ganglia (DRG) neurons, exhibited 30- to 120-fold selectivity over other human TTX-S channels and exhibited over 1,000-fold selectivity over other human tetrodotoxin-resistant (TTX-R) channels. Leveraging NaV1.7-NaV1.5 chimeras containing various voltage-sensor and pore regions, AM-8145 mapped to the second voltage-sensor domain of NaV1.7. AM-0422, but not the inactive peptide analog AM-8374, dose-dependently blocked capsaicin-induced DRG neuron action potential firing using a multi-electrode array readout and mechanically-induced C-fiber spiking in a saphenous skin-nerve preparation. Collectively, AM-8145 and AM-0422 represent potent, new engineered NaV1.7 inhibitory peptides derived from the JzTx-V scaffold with improved NaV selectivity and biological activity in blocking action potential firing in both DRG neurons and C-fibers.

Collaboration


Dive into the Kaustav Biswas's collaboration.

Researchain Logo
Decentralizing Knowledge