Network


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

Hotspot


Dive into the research topics where Neal W. Sach is active.

Publication


Featured researches published by Neal W. Sach.


Nature | 2012

Practical and innate carbon-hydrogen functionalization of heterocycles

Yuta Fujiwara; Janice A. Dixon; Fionn O’Hara; Erik Daa Funder; Darryl D. Dixon; Rodrigo A. Rodriguez; Ryan D. Baxter; Bart Herlé; Neal W. Sach; Michael Raymond Collins; Yoshihiro Ishihara; Phil S. Baran

Nitrogen-rich heterocyclic compounds have had a profound effect on human health because these chemical motifs are found in a large number of drugs used to combat a broad range of diseases and pathophysiological conditions. Advances in transition-metal-mediated cross-coupling have simplified the synthesis of such molecules; however, C–H functionalization of medicinally important heterocycles that does not rely on pre-functionalized starting materials is an underdeveloped area. Unfortunately, the innate properties of heterocycles that make them so desirable for biological applications—such as aqueous solubility and their ability to act as ligands—render them challenging substrates for direct chemical functionalization. Here we report that zinc sulphinate salts can be used to transfer alkyl radicals to heterocycles, allowing for the mild (moderate temperature, 50 °C or less), direct and operationally simple formation of medicinally relevant C–C bonds while reacting in a complementary fashion to other innate C–H functionalization methods (Minisci, borono-Minisci, electrophilic aromatic substitution, transition-metal-mediated C–H insertion and C–H deprotonation). We prepared a toolkit of these reagents and studied their reactivity across a wide range of heterocycles (natural products, drugs and building blocks) without recourse to protecting-group chemistry. The reagents can even be used in tandem fashion in a single pot in the presence of water and air.


Journal of Medicinal Chemistry | 2014

Discovery of (10R)-7-amino-12-fluoro-2,10,16-trimethyl-15-oxo-10,15,16,17-tetrahydro-2H-8,4-(metheno)pyrazolo[4,3-h][2,5,11]-benzoxadiazacyclotetradecine-3-carbonitrile (PF-06463922), a macrocyclic inhibitor of anaplastic lymphoma kinase (ALK) and c-ros oncogene 1 (ROS1) with preclinical brain exposure and broad-spectrum potency against ALK-resistant mutations.

Ted W. Johnson; Paul F. Richardson; Simon Bailey; Alexei Brooun; Benjamin J. Burke; Michael Raymond Collins; J. Jean Cui; Judith Gail Deal; Ya-Li Deng; Dac M. Dinh; Lars D. Engstrom; Mingying He; Jacqui Elizabeth Hoffman; Robert Louis Hoffman; Qinhua Huang; Robert Steven Kania; John Charles Kath; Hieu Lam; Justine L. Lam; Phuong Thi Quy Le; Laura Lingardo; Wei Liu; Michele McTigue; Cynthia Louise Palmer; Neal W. Sach; Tod Smeal; Graham L. Smith; Albert E. Stewart; Sergei Timofeevski; Huichun Zhu

Although crizotinib demonstrates robust efficacy in anaplastic lymphoma kinase (ALK)-positive non-small-cell lung carcinoma patients, progression during treatment eventually develops. Resistant patient samples revealed a variety of point mutations in the kinase domain of ALK, including the L1196M gatekeeper mutation. In addition, some patients progress due to cancer metastasis in the brain. Using structure-based drug design, lipophilic efficiency, and physical-property-based optimization, highly potent macrocyclic ALK inhibitors were prepared with good absorption, distribution, metabolism, and excretion (ADME), low propensity for p-glycoprotein 1-mediated efflux, and good passive permeability. These structurally unusual macrocyclic inhibitors were potent against wild-type ALK and clinically reported ALK kinase domain mutations. Significant synthetic challenges were overcome, utilizing novel transformations to enable the use of these macrocycles in drug discovery paradigms. This work led to the discovery of 8k (PF-06463922), combining broad-spectrum potency, central nervous system ADME, and a high degree of kinase selectivity.


Science | 2016

Strain-release amination

Ryan Gianatassio; Justin M. Lopchuk; Jie Wang; Chung-Mao Pan; Lara R. Malins; Liher Prieto; Thomas A. Brandt; Michael Raymond Collins; Gary M. Gallego; Neal W. Sach; Jillian E. Spangler; Huichin Zhu; Jinjiang Zhu; Phil S. Baran

Opening one ring to tack on another Curious chemists have long sought to learn just how tightly carbon atoms can be bound together. For instance, its possible to form a bond between two opposite corners of an already strained four-membered ring to make an edge-sharing pair of triangles. Gianatassio et al. have now devised a general use for these and related molecular curiosities. They show that appropriately modified nitrogen centers can pop open the most highly strained bond, leaving the more modestly strained ring motif intact. In this way, small rings can emerge as a convenient diversifying element in compounds, including new pharmaceutical candidates. Science, this issue p. 241 Strained rings are appended to compounds of pharmaceutical interest through the use of even more highly strained precursors. To optimize drug candidates, modern medicinal chemists are increasingly turning to an unconventional structural motif: small, strained ring systems. However, the difficulty of introducing substituents such as bicyclo[1.1.1]pentanes, azetidines, or cyclobutanes often outweighs the challenge of synthesizing the parent scaffold itself. Thus, there is an urgent need for general methods to rapidly and directly append such groups onto core scaffolds. Here we report a general strategy to harness the embedded potential energy of effectively spring-loaded C–C and C–N bonds with the most oft-encountered nucleophiles in pharmaceutical chemistry, amines. Strain-release amination can diversify a range of substrates with a multitude of desirable bioisosteres at both the early and late stages of a synthesis. The technique has also been applied to peptide labeling and bioconjugation.


Journal of Medicinal Chemistry | 2014

Design of Potent and Selective Inhibitors to Overcome Clinical Anaplastic Lymphoma Kinase Mutations Resistant to Crizotinib.

Qinhua Huang; Ted W. Johnson; Simon Bailey; Alexei Brooun; Kevin D. Bunker; Benjamin J. Burke; Michael Raymond Collins; Andrew Simon Cook; J. Jean Cui; Kevin Neil Dack; Judith Gail Deal; Ya-Li Deng; Dac M. Dinh; Lars D. Engstrom; Mingying He; Jacqui Elizabeth Hoffman; Robert Louis Hoffman; Patrick Stephen Johnson; Robert Steven Kania; Hieu Lam; Justine L. Lam; Phuong Thi Quy Le; Qiuhua Li; Laura Lingardo; Wei Liu; Melissa West Lu; Michele McTigue; Cynthia Louise Palmer; Paul F. Richardson; Neal W. Sach

Crizotinib (1), an anaplastic lymphoma kinase (ALK) receptor tyrosine kinase inhibitor approved by the U.S. Food and Drug Administration in 2011, is efficacious in ALK and ROS positive patients. Under pressure of crizotinib treatment, point mutations arise in the kinase domain of ALK, resulting in resistance and progressive disease. The successful application of both structure-based and lipophilic-efficiency-focused drug design resulted in aminopyridine 8e, which was potent across a broad panel of engineered ALK mutant cell lines and showed suitable preclinical pharmacokinetics and robust tumor growth inhibition in a crizotinib-resistant cell line (H3122-L1196M).


Organic Letters | 2011

Scalable synthesis of 1-bicyclo[1.1.1]pentylamine via a hydrohydrazination reaction.

Kevin D. Bunker; Neal W. Sach; Qinhua Huang; Paul F. Richardson

The reaction of [1.1.1]propellane with di-tert-butyl azodicarboxylate and phenylsilane in the presence of Mn(dpm)(3) to give di-tert-butyl 1-(bicyclo[1.1.1]pentan-1-yl)hydrazine-1,2-dicarboxylate is described. Subsequent deprotection gives 1-bicyclo[1.1.1]pentylhydrazine followed by reduction to give 1-bicyclo[1.1.1]pentylamine. The reported route marks a significant improvement over the previous syntheses of 1-bicyclo[1.1.1]pentylamine in terms of scalability, yield, safety, and cost.


Organic Letters | 2012

Synthesis of Aryl Ethers via a Sulfonyl Transfer Reaction

Neal W. Sach; Daniel T. Richter; Stephan James Cripps; Michelle Bich Tran-Dube; Huichun Zhu; Buwen Huang; Jean Cui; Scott C. Sutton

A general synthesis of aryl ethers from primary and secondary alcohols and aryl mesylates is presented. The reaction proceeds via a sulfonyl-transfer mechanism. In this paper, we compare the sulfonyl transfer reaction to Mitsunobu ether formation. The reaction can be employed in a multistep synthesis where the aryl mesylate is used as a phenol protecting group and then as an activating group for ether formation. This protecting/activating group strategy is demonstrated using raloxifene as the target.


ACS Medicinal Chemistry Letters | 2011

Highly Selective and Potent Thiophenes as PI3K Inhibitors with Oral Antitumor Activity

Kevin Liu; JinJiang Zhu; Graham L. Smith; Min-Jean Yin; Simon Bailey; Jeffrey H. Chen; Qiyue Hu; Qinhua Huang; Chunze Li; Qing J. Li; Matthew A. Marx; Genevieve Paderes; Paul F. Richardson; Neal W. Sach; Marlena Walls; Peter A. Wells; Aihua Zou

Highly selective PI3K inhibitors with subnanomolar PI3Kα potency and greater than 7000-fold selectivity against mTOR kinase were discovered through structure-based drug design (SBDD). These tetra-substituted thiophenes were also demonstrated to have good in vitro cellular potency and good in vivo oral antitumor activity in a mouse PI3K driven NCI-H1975 xenograft tumor model. Compounds with the desired human PK predictions and good in vitro ADMET properties were also identified. In this communication, we describe the rationale behind the installation of a critical triazole moiety to maintain the intricate H-bonding network within the PI3K receptor leading to both better potency and selectivity. Furthermore, optimization of the C-4 phenyl group was exploited to maximize the compounds mTOR selectivity.


Journal of Medicinal Chemistry | 2017

Discovery of N-((3R,4R)-4-Fluoro-1-(6-((3-methoxy-1-methyl-1H-pyrazol-4-yl)amino)-9-methyl-9H-purin-2-yl)pyrrolidine-3-yl)acrylamide (PF-06747775) through Structure-Based Drug Design: A High Affinity Irreversible Inhibitor Targeting Oncogenic EGFR Mutants with Selectivity over Wild-Type EGFR.

Simon Paul Planken; Douglas Carl Behenna; Sajiv Krishnan Nair; Theodore Otto Johnson; Asako Nagata; Chau Almaden; Simon Bailey; T. Eric Ballard; Louise Bernier; Hengmiao Cheng; Sujin Cho-Schultz; Deepak Dalvie; Judith Gail Deal; Dac M. Dinh; Martin Paul Edwards; Rose Ann Ferre; Ketan S. Gajiwala; Michelle Hemkens; Robert Steven Kania; John Charles Kath; Jean Matthews; Brion W. Murray; Sherry Niessen; Suvi T. M. Orr; Mason Alan Pairish; Neal W. Sach; Hong Shen; Manli Shi; James Solowiej; Khanh Tran

Mutant epidermal growth factor receptor (EGFR) is a major driver of non-small-cell lung cancer (NSCLC). Marketed first generation inhibitors, such as erlotinib, effect a transient beneficial response in EGFR mutant NSCLC patients before resistance mechanisms render these inhibitors ineffective. Secondary oncogenic EGFR mutations account for approximately 50% of relapses, the most common being the gatekeeper T790M substitution that renders existing therapies ineffective. The discovery of PF-06459988 (1), an irreversible pyrrolopyrimidine inhibitor of EGFR T790M mutants, was recently disclosed.1 Herein, we describe our continued efforts to achieve potency across EGFR oncogenic mutations and improved kinome selectivity, resulting in the discovery of clinical candidate PF-06747775 (21), which provides potent EGFR activity against the four common mutants (exon 19 deletion (Del), L858R, and double mutants T790M/L858R and T790M/Del), selectivity over wild-type EGFR, and desirable ADME properties. Compound 21 is currently being evaluated in phase-I clinical trials of mutant EGFR driven NSCLC.


Science | 2018

A platform for automated nanomole-scale reaction screening and micromole-scale synthesis in flow

Damith Perera; Joseph W. Tucker; Shalini Brahmbhatt; Christopher John Helal; Ashley Chong; William Farrell; Paul G. Richardson; Neal W. Sach

A reaction screen in flowing solvent Chemists charged with manufacturing pharmaceuticals have recently been exploring the efficiency advantages of continuous flow techniques. Perera et al. now show that a flow apparatus can also accelerate reaction optimization earlier in the drug discovery process. They modified a high-performance liquid chromatography system to screen a wide variety of solvent, ligand, and base combinations to optimize carbon-carbon bond formation. Injecting stock solution aliquots of the catalyst and reactants into a carrier solvent stream let the authors vary the main solvent efficiently and scale up the optimal conditions for product isolation. Science, this issue p. 429 Chromatographic, flow-based screening of reaction conditions is demonstrated for Suzuki coupling in pharmaceutical research. The scarcity of complex intermediates in pharmaceutical research motivates the pursuit of reaction optimization protocols on submilligram scales. We report here the development of an automated flow-based synthesis platform, designed from commercially available components, that integrates both rapid nanomole-scale reaction screening and micromole-scale synthesis into a single modular unit. This system was validated by exploring a diverse range of reaction variables in a Suzuki-Miyaura coupling on nanomole scale at elevated temperatures, generating liquid chromatography–mass spectrometry data points for 5760 reactions at a rate of >1500 reactions per 24 hours. Through multiple injections of the same segment, the system directly produced micromole quantities of desired material. The optimal conditions were also replicated in traditional flow and batch mode at 50- to 200-milligram scale to provide good to excellent yields.


Journal of the American Chemical Society | 2017

Strain-Release Heteroatom Functionalization: Development, Scope, and Stereospecificity

Justin M. Lopchuk; Kasper Fjelbye; Yu Kawamata; Lara R. Malins; Chung-Mao Pan; Ryan Gianatassio; Jie Wang; Liher Prieto; James Bradow; Thomas A. Brandt; Michael Raymond Collins; Jeff Elleraas; Jason Ewanicki; William Farrell; Olugbeminiyi O. Fadeyi; Gary M. Gallego; James J. Mousseau; Robert Oliver; Neal W. Sach; Jason K. Smith; Jillian E. Spangler; Huichin Zhu; Jinjiang Zhu; Phil S. Baran

Driven by the ever-increasing pace of drug discovery and the need to push the boundaries of unexplored chemical space, medicinal chemists are routinely turning to unusual strained bioisosteres such as bicyclo[1.1.1]pentane, azetidine, and cyclobutane to modify their lead compounds. Too often, however, the difficulty of installing these fragments surpasses the challenges posed even by the construction of the parent drug scaffold. This full account describes the development and application of a general strategy where spring-loaded, strained C–C and C–N bonds react with amines to allow for the “any-stage” installation of small, strained ring systems. In addition to the functionalization of small building blocks and late-stage intermediates, the methodology has been applied to bioconjugation and peptide labeling. For the first time, the stereospecific strain-release “cyclopentylation” of amines, alcohols, thiols, carboxylic acids, and other heteroatoms is introduced. This report describes the development, synthesis, scope of reaction, bioconjugation, and synthetic comparisons of four new chiral “cyclopentylation” reagents.

Collaboration


Dive into the Neal W. Sach's collaboration.

Researchain Logo
Decentralizing Knowledge