Upcoming Webinars

cancel
Showing results for 
Search instead for 
Did you mean: 

Upcoming Webinars

Technology born as a powerful drawing tool is now transforming and empowering a Chemist’s journey.    ChemDraw is the technology that parallels and powers the evolution of this journey. Designed with chemistry in mind, ChemDraw and ChemOffice+ Cloud accelerates countless administrative tasks that impede a Chemist’s ability to spend time on what matters most: Chemistry.    Join Pierre Morieux, a.k.a ChemDraw Wizard, to learn about the newest capabilities that will empower you to create beautiful chemical drawings more efficiently, communicate your research visually, and expedite mundane tasks such as managing molecules and reactions as well as everyday reporting.    Chemists can work more efficiently, communicate their research more clearly, and reach crucial information faster with ChemDraw.    Learn how powerful capabilities in the latest release of ChemDraw and ChemOffice+ Cloud v21 will dramatically increase your productivity and accelerate your chemistry research and discovery by enabling you to:    Improve the efficiency of your research by drawing entire complex reactions schemes in minutes with new hotkeys and improved shortcuts.  Effortlessly and expediently represent 3D organic and organometallic, inorganic, and supramolecular structures.  Make your ChemDraw files come to life with the ability to embed native, animated 3D models in MS PowerPoint directly.  Create chemistry lab reports in a few clicks with the new reporting capability of ChemOffice+, integrated with Signals Notebook.  Who Should Attend: Chemists  Research Chemists  Organic Chemists  Inorganic Chemists  Organometallic Chemists  Polymer Chemists  Ph.D. Students and Post-Docs  Patent Attorneys   Brought to you by: SPEAKERS Pierre Morieux, Ph.D. ChemDraw Global Marketing Manager, ChemDraw Wizard Melissa O'M eara Forensic Science Consultant, C&EN Media Group
PerkinElmer Webinar
Labels (2)
In this Webinar you will learn about the analytical techniques solving today’s challenges in battery component recycling. Learn about robust and practical atomic spectroscopy solutions which detect and mitigate against cross-contamination during the recycling process. See examples of TGA and STA in understanding the degradation and thermal properties of battery materials. Learn how this knowledge allows recyclers to understand the behavior of certain materials during processing. Drive the development of more circular input structures by finding the best laboratory analysis solutions for material reclaim and reuse.   Key Learning Objectives: Understand trends and challenges facing battery recyclers Get an overview of recycling processes and approaches Recognize and prevent cross-contamination of materials during the recycling process Compare ICP-OES vs ICP-MS technologies for materials reclaim Gain insights into pyrometallurgical recycling with TGA and STA analysis   Who Should Attend: Battery and energy storage researchers Laboratory managers Materials scientists Academic researchers in the field Principal investigators on energy-related projects Scientists at battery manufacturers or recycling specialists Brought to you by: Sascha Nowak Head of Analytics & Environment, MEET Battery Research Center Ryan Purcell-Joiner Senior Application Scientist, PerkinElmer Inc. Gerlinde Wita Global Market Leader, PerkinElmer Inc. Kieran Evans Application Scientist, PerkinElmer Inc. Jeff Huber Contributing Editor, C&EN Media Group
PerkinElmer_socialimage_031522.jpg
Labels (2)
Native mass spectrometry (nMS), in which intact protein or nucleoprotein complexes are directly measured, commonly involves exchange of nonvolatile electrolytes (NaCl, PBS, Tris) for volatile electrolytes such as ammonium acetate. This can be time-consuming if offline buffer exchange spin cartridges are used.  Furthermore, some samples may not be stable if left in nonvolatile buffer for more than just a very short period of time. This problem can be avoided if an LC-compatible online buffer exchange column is used to separate protein complex from salt.  In this experiment, no attempt is made to perform size exclusion chromatography to separate protein complexes of different sizes but rather any protein complex that is injected onto the system is simply separated from low mass salts that would otherwise product heavily adducted protein complexes that appear as broad peaks in the mass spectrum.  This webinar will focus on the development of online buffer exchange (OBE) for nMS applications and will describe how the experiment is implemented and how it can be extended by coupling to affinity separation (e.g., IMAC-OBE) to be used to optimize protein overexpression.    Key Learning Objectives: The use of online buffer exchange for high throughput  native mass spectrometry   Online buffer exchange nMS allows to directly screen structural features of large biomolecules even if such biomolecules are not very stable in MS friendly buffers  How the coupling of affinity separation (e.g., IMAC-OBE) to online-buffer exchange can be used to optimize protein overexpression  Who Should Attend: MS practitioners  Researchers/ R&D Managers  Laboratory Managers/ Directors / Supervisors  Laboratory Technicians / Operators  Structural Biologists  Scientists in biopharma engaged in biotherapeutic characterization    Brought to you by:   Vicki Wysocki, Ph.D. Professor, Dept of Chemistry and Biochemistry The Ohio State University Catherine Dold Health & Environment Writer, C&EN Media Group
{e8b86ccd-7c1f-4565-971b-791a7e32af88}_ThermoFisher_032322_bannerimage.jpg
Labels (2)
Catalysts facilitate the creation of almost all synthetic materials we interact with everyday. New materials require new catalysts with enhanced and novel properties. Traditional synthetic approaches for materials discovery are expensive and slow. First-principles simulation has become a reliable tool for the prediction of structures, chemical mechanisms, and reaction energetics for the fundamental steps in homogeneous catalysis. Details of reaction coordinates for competing pathways can provide the fundamental understanding of observed catalytic activity, selectivity, and specificity. Such predictive capability raises the possibility for computational discovery and design of new catalysts with enhanced properties. Unfortunately, this is an arduous process that requires meticulous maintenance, specialized training, and accounting of hundreds of files and properties.   To facilitate the fundamental understanding, design, and discovery of novel catalysts, an automated enterprise solution was designed and developed for collaboration between synthetic and computational chemists on a single web-based platform.    Key Learning Objectives: Discover the predictive capabilities of physics-based modeling in reactivity and catalysis.  See how automated high-throughput screening accelerates synthetic discovery.  Learn how a web-based platform can generate more ideas and drive innovation through the collaboration of computational and synthetic chemists.  Who Should Attend: Synthetic Chemists  Materials Scientists  Chemical Engineers  Digitization Managers  R&D Scientists designing novel materials    Brought to you by:   Thomas Mustard Scientific Lead of Catalysis and Reactivity, Schrödinger Kelly McSweeney Contributing Editor, C&EN Media Group
%7B9d1e6683-5d15-450b-b8a5-3aa64cb8f30c%7D_Schrodinger_033022_bannerimage.png
Labels (2)
The characterization of extractables and leachables (E&L) is an integral part of ensuring biocompatibility for many medical devices and pharmaceutical products. Guidance for E&L has been provided in USP <1663> and <1664> for pharmaceutical products and in ISO 10993-18 for medical devices. The first step in the E&L process involves detection of those compounds which are suspected to be at or above the level of toxicological concern. This process is accomplished through the use of the Analytical Evaluation Threshold (AET) which links the toxicologically relevant concentration to the observed analytical signals. A significant problem in AET evaluation is caused by response factor (RF) variation. It is an unfortunate reality that compounds which are at equivalent concentration do not always or even often give equivalent signal response on various detector systems including mass spectrometers. Recent publications have highlighted these risks for both LCMS (Jordi, et al. J. Pharm. Biomed. **bleep**. 2018, 150, 368–376) and GCMS (Jenke and Odufu, Journal of Chromatographic Science 2012;50:206–212). The prominent and necessary use of surrogate standards for AET evaluation introduces error into the precise estimation of the signal strength which corresponds to the toxicologically relevant concentration. To overcome this problem, an uncertainty factor has been introduced into the AET equation and regulatory agencies have provided recommendations as to values for the UF (GC/MS with UF =4, LC/MS with UF =10). While this approach does account for response variation, it also introduces other difficulties including lower AET values which can be difficult or impractical to achieve which then require additional sample concentration due to limited instrument sensitivity. This has the potential to counteract the perceived benefit resulting in compound loss or degradation and additional regulatory scrutiny of the sample preparation process. This approach also results in a significant potential for false positives (I.e., compounds that are below the AET concentration are determined to have a peak area above the threshold associated with the AET). An alternative approach is therefore desirable. In this webinar, a new case study will be presented using the multidetector approach to AET evaluation contrasted with the use of the recommended UF of 4 and 10 for a model polymer system. The multidetector approach leverages the independence of the response factors for a given compound obtained on different detectors and chromatographic systems to overcome potential weak signals on any one detector and thus reduces the reliance of the method on UF to overcome response variation. The multidetector approach was summarized in two recent publications (Jordi, et al. J. Pharm. Biomed. **bleep**. 2020, 186, 1-14 and Jordi, et al. PDA Journal, vol. 75, No. 2 2021, pg. 289-301). The effectiveness of using a combination of triple detection Liquid Chromatography Mass Spectrometry (LCMS) with Ultraviolet (UV) and Charged aerosol detection (CAD) as well as Gas Chromatography Mass Spectrometry (GCMS) will be presented. Quantitation for a series of reference compounds characteristic of the polymer systems will be used to gauge the potential for false positives using the different AET approaches. Finally, the benefits of this approach for detection of compounds with little to no mass spectrometry response will be highlighted.   Key Learning Objectives: Objective 1: What is the Analytical Evaluation Threshold and how should it be applied?   Objective 2: Review a Case study comparing the use of regulatory agency recommended approaches to AET evaluation and a multidetector approach.   Objective 3: How can using a multidetector approach reduce the need for large UF and by extension the need for additional sample preparation steps?  Who Should Attend: Laboratory managers  Chromatographers  New product developers    Brought to you by:   Dr. Mark Jordi President, Jordi Labs Jeff Huber Contributing Editor, C&EN Media Group
%7Ba6b93935-01d5-4151-9c4d-531c21b453a6%7D_Agilent_033122_bannerimage.jpg
Labels (2)
Continuous manufacturing (flow chemistry) is a key element in the green-chemistry landscape of a pharmaceutical company. In this webinar, Benjamin Martin, PhD from Novartis will use case studies to illustrate the drivers for continuous processing for clinical supplies, the challenges navigated during process R & D, and a summary of how the sustainability metrics, Process-Mass-Intensity (PMI) and Total CO2-Release (TCR), are positively affected.     Hongwei Yang, PhD from WuXi STA will show how industry collaboration between Pharma companies and CDMOs with strong capability to quickly develop or transfer in a flow process and assemble customized lines at plant-scale can accelerate the implementation of continuous manufacturing for drug substance supply with a cost-effective and greener process.    Key Learning Objectives: Sustainability metrics for continuous manufacturing (CM) in pharmaceutical industry  Key drivers for using CM at the clinical stages  How Innovators and CDMOs can work together to speed implementation of CM and bring therapies to patients faster.  Who Should Attend: Pharmaceutical & Fine Chemists  Academic Chemistry Researchers  Process & Development Chemists  Drug Discovery Chemists    Brought to you by: Benjamin Martin, Ph.D. Associate Director Science & Technology Continuous Manufacturing-Upstream Network Leader, Novartis Hongwei Yang, Ph.D. Executive Director and Head of Flow Chemistry, WuXi STA, a subsidiary of WuXi AppTec Melissa O'Meara Forensic Science Consultant, C&EN Media Group
{96bcac5c-e76f-4e01-ae91-fbebd7bf46c4}_STA_040522_bannerimage.png
Labels (2)
There is a trend in the materials industry towards thinner and thinner coatings that create new applications and enable both material and cost savings. However, as coatings become thinner, the traditional methods for coating hardness testing become less accurate or fail entirely.     Although methods are available for measuring the hardness of thin films, up until now, they have tended to be both price-prohibitive and require extremely complex and difficult to use instrumentation.     With the introduction of the Hit 300, accurate hardness testing of thin films is now both accessible and affordable for any lab or production site developing new coating materials.     Anton Paar is proud to present “Accessible and Affordable Hardness Testing of Thin Coatings,” a live webinar discussing the benefits and limitations of traditional hardness testing as well as new developments that enable accurate and affordable hardness measurements of ultrathin coatings.     This webinar will include real-life applications of hardness testing methods, including:     • Hardness testing of cutting tool coatings     • Analysis of thin DLC films     • Alumina-based coatings     • Paints and other polymers    Key Learning Objectives: New developments that have made hardness testing of thin coatings easier to use and more cost-effective  Real-life applications of hardness testing methods that can be done more efficiently with the newest generation of instruments  Who Should Attend: Research Scientists  Laboratory Managers  University Researchers  Quality Engineers    Brought to you by:   Mark Haase Commercial Product Specialist, Anton Paar Kelly McSweeney Contributing Editor, C&EN Media Group
{fa19dcc9-7c86-47bc-bc6b-7910454e17cc}_600x200_Newsletter_Hit_300_02.jpg
Labels (2)
Labels (2)
Multidimensional measurements integrating liquid chromatography, ion mobility spectrometry and mass spectrometry (LC-IMS-MS) provide valuable polarity, structural and mass information simultaneously for lipidomic analyses and show tremendous power for attaining more confident lipid identifications. For all of their advantages, LC-IMS-MS measurements are highly complex and result in huge datasets which are difficult to process in a timely fashion. Thus, developing a data analysis workflow that is capable of accurate and rapid molecular analyses is essential.   The freely available, open-source software Skyline offers targeted processing of lipid data which ultimately allows for confident identification of diverse lipid species. We have developed sample-specific lipid spectral libraries which include over 700 target lipids from multiple lipid categories. Each target lipid is populated with m/z values, normalized retention times, ion mobility collision cross section (CCS) values, and known fragmentation patterns. These values were manually extracted from LC-IMS-MS experimental data and verified using existing literature.   Recently developed aspects of the Skyline small molecule interface are utilized in this workflow including IMS spectrum filtering and retention time prediction (iRT) using a set of ~20 endogenous lipids for gradient correction and LC alignment. Application of lipid CCS value filtering further increased lipid annotation confidence and greatly improved the signal to noise ratio for the target species. These lipid spectral libraries have undergone additional validation studies and have recently been made publicly available through Skyline’s online repository Panorama.   In comparison to previous studies of NIST SRM 1950, this workflow when coupled with an LCIMS-CID-MS platform gave hundreds of confident annotations using a single sample extraction and analysis platform.   Key Learning Objectives: Learn to overcome challenges with large,  highly complex datasets generated from liquid chromatography, ion mobility spectrometry and mass spectrometry lipidomic analyses. Developing sample-specific multidimensional lipid libraries using Skyline. Build a workflow leveraging Skyline automation features such as small-molecule spectral libraries, drift time filtering, iRT retention time prediction, analysis of multiple adducts, and neutral loss fragments. Who Should Attend: Lipidomics researchers Chromatographers/Mass spectrometrists Drug development scientists   Brought to you by:   Kaylie Kirkwood, Ph.D. North Carolina State university Kelly McSweeney Contributing Editor C&EN Media Group
{3f202871-b457-49a5-8f21-f06f3f6d7988}_Agilent_051222_banner.jpg
Labels (2)
Characterizing quality attributes and other characteristics of biologics, vaccines, and gene vectors is essential at each stage of its discovery, development, production, and quality control. Analytical techniques based on light scattering have become powerful tools for characterizing various attributes include molar mass, size, aggregation, physical titer, thermal and colloidal stability.    In this webinar, we highlight robust, reliable, simple, quantitative, and fast ways to use three different light scattering techniques - batch dynamic light scattering (DLS), static light scattering (SLS), and massively parallel phase analysis light scattering (MP-PALS) - to characterize the stability of vaccines and gene therapeutic nanoparticles.    Key Learning Objectives: Basic DLS and MP-PALS theory and instrumentation  How DLS and SLS are applied to study the stability of AAVs and vaccines  How DLS and MP-PALS characterize the stability of lipid nanoparticles  How high-throughput formulation and stability studies are carried out with these techniques  Who Should Attend: Pharmaceutical scientists involved in analytical characterization and formulation of proteins, monoclonal antibodies, and gene therapeutic nanoparticles Scientists and managers in need of robust, reliable, simple, and fast methods for evaluating biophysical properties and stability Managers of academic labs and core facilities developing viral vectors and LNPs   Brought to you by:   Xujun Zhang, Ph.D. Application Scientist, Wyatt Technology Ann Thayer Contributing Editor, C&EN Media Group
{c0bc3828-8fae-48aa-b6eb-bbf7c3c5c505}_Wyatt_052522_banner.jpg
Labels (2)
In the development of biotherapeutics, a thorough understanding of a molecule’s product quality attributes (PQAs), and their effect on various structure-function relationships and long-term stability, is essential for ensuring the safety and efficacy of the product. At present, numerous routine chromatographic and electrophoretic assays are used to characterize and monitor individual PQAs. However, execution of multiple routine methods for batch release, stability time-points, and process/formulation development support becomes time and resource intensive, and often provides an indirect measure of biologically relevant PQAs. Introduced in 2015, the multi-attribute method (MAM), based on LC-MS peptide mapping and automation principles, provides simultaneous and site-specific detection, identification, quantitation, and quality control (monitoring) of PQAs.   A dedicated Pfizer team has been regularly employing MAM on an in-house MAM platform to support biotherapeutic process and product development. In parallel, this team has continually explored and implemented improvements in the Pfizer MAM platform, including sample preparation and data processing automation, to move toward the next generation of MAM. Recently, a pre-commercial demo model of the new Orbitrap Exploris MX mass detector was evaluated in-house by the Pfizer MAM team. Here, the results of the evaluation and an assessment of the Orbitrap Exploris MX mass detector’s suitability as a next generation MAM instrument are presented.   Key Learning Objectives: Pfizer MAM platform milestones for characterization and routine monitoring Automation of sample handling and data processing and reporting Evaluation and optimization of the Orbitrap Exploris MX mass detector for MAM Who Should Attend: Laboratory managers Chromatographers New product developers Brought to you by:   Andrew W. Dawdy, Ph.D. Principal Scientist, BioTherapeutics Pharmaceutical Sciences, Pfizer, Inc. Ann Thayer Contributing Editor, C&EN Media Group
{991319ef-0d17-4f57-a311-1db79697ef84}_ThermoFisher_052522_bannerimage.jpg
Labels (2)
Accurately identifying unknowns by searching a library of GC/MS spectral data is straightforward but often yields ambiguous results. Isomeric compounds that have the same molecular formula, but different structures, can give very similar spectral signatures. In addition, peaks with co-eluting compounds can lead to inconclusive or incorrect results.    Cerno’s GC/ID data processing software utilizes an advanced mixture deconvolution algorithm and then combines information from spectral libraries, retention index (RI) data, and accurate mass formula confirmation to identify unknowns on single quadrupole mass spectrometer systems. The software automatically generates RIs for detected compounds during a single or batch run, without running separate standards. Finally, it visually highlights correct identifications among a long list of possible matches. This post acquisition software works with most commercially available GC/MS systems.    Key Learning Objectives: Learn how to obtain retention index automatically from your unknown sample itself Learn how to obtain accurate mass and spectral accuracy on your single quadrupole GC/MS system Learn how to combine multiple ID metrics and quickly come to a conclusive answer Learn how to make your GC/MS analysis dramatically more productive  Who Should Attend: Analysts and lab managers utilizing GC/MS for qualitative or semi-quantitative analysis. Brought to you by: Don Kuehl, PhD VP of Product Development and Marketing, Cerno Bioscience Ann Thayer Contributing Editor, C&EN Media Group
{8a663ba7-fc36-4b77-8c12-cdcfe08cbe75}_CernoBioScience_062622_bannerimage.png
Labels (2)
In forensic chemistry, the presence of drugs and novel psychoactive substances at low concentrations or in complicated mixtures present challenges to traditional analytical screening methods. At the same time, ensuring timely investigative information, determining appropriate analytical schemes and improving the safety of laboratory personnel, healthcare practitioners and crime scene investigators is crucial.     In this webinar, Colby Ott, PhD, Research Scientist at West Virginia University and Luis E. Arroyo-Mora, PhD, Professor at the Department of Forensic and Investigative Sciences at West Virginia University, will present the fundamentals behind electrochemical-surface-enhanced Raman spectroscopy (EC-SERS) and how the advantages of this technique can be leveraged for on-site screening of drugs of abuse. Our experts will describe how the SPELEC combination potentiostat-Raman spectrometer serves to provide simple, time-resolved data and how EC-SERS can be applied to other applications. Tune in to learn about two EC-SERS methods and their analytical performance as well as an assessment of authentic casework samples which will demonstrate the strength of this approach.    Key Learning Objectives: Develop a basic understanding of time-resolved electrochemical surface enhanced Raman spectroscopy (EC-SERS)  Learn how EC-SERS provides a novel and effective screening platform for drugs of abuse  Discover the advantages of hyphenated EC-SERS instrumentation  Who Should Attend: Anyone interested in learning how spectroelectrochemical and time-resolved techniques can be leveraged to provide reliable and effective analytical platforms  Anyone working in the field of forensic science  Anyone interested in understanding how electrochemistry and Raman spectroscopy can work together to improve detection    Brought to you by:   Speakers: Colby Ott, Ph.D. Research Scientist West Virginia University Luis E. Arroyo-Mora, Ph.D. Associate Professor West Virginia University   Melissa O'Meara Forensic Science Consultant C&EN Media Group
ACS Webinars Hub Placeholder Image.png
Labels (2)
Labels (2)
The identification of new chemical matter represents a critical step in the early stages of a drug discovery project. An increasingly broad collection of platforms are available to perform this task, some more suited to small, focused screening sets and some more adapted to very large screening sets. Recently traditional high-throughput biochemical assays with a functional read-out have been supplemented by a range of assays based on biophysical methods that address the physical interaction between ligand and protein, but are agnostic of the functional consequence (if any).    In this webinar we will focus on those techniques that seek to identify small molecule binders to target proteins using such ‘affinity selection’ methods. We will discuss the current state of the art for screening of fragments, small screening collections, and ultra-large diversity collections using techniques such as NMR, affinity selection mass spectrometry, and DNA-encoded library screens.      Key Learning Objectives: Review techniques that seek to identify small molecule binders to target proteins using such ‘affinity selection’ methods   Explore the current state of the art for screening of fragments, small screening collections, and ultra-large diversity collections using techniques such as NMR, affinity selection mass spectrometry, and DNA-encoded library screens.  Who Should Attend: Drug discovery scientists in academia Drug discovery scientists in biotech and pharmaceutical industry  Chemists  Biologists  Pharmacologists   Bioanalytical Scientists Biophysical Scientists Brought to you by:   Speakers: Dave Madge Vice President WuXi AppTec Alex Satz Senior Director, DEL Strategy WuXi AppTec Grzegorz Popowicz Head, Helmholtz Institute   Penny Jia AS-MS Project Leader WuXi AppTec Ann Thayer Contributing Editor C&EN Media Group
ACS Webinars Hub Placeholder Image.png
Labels (2)
The relative abundance of empty and full AAV capsids in gene therapy vector preparations is a critical quality attribute. A variety of methods are available to perform these measurements including SEC-MALS, which combines separation by size-exclusion chromatography with detection by multi-angle light scattering, UV/Vis, and differential refractometry. However, there are relatively few reports that compare these methods. This webinar compares the outcomes of numerous orthogonal characterization methods on a single vector preparation. We find that SEC-MALS not only provides favorable performance for quality control and product development applications, but that it is uniquely situated to be established as a platform method for the semi-quantitative measurement of empty/full ratios across AAV serotypes, transgenes, and manufacturing processes.   Key Learning Objectives: How SEC-MALS quantifies empty and full AAVs, as well as other attributes Are empty/full readouts from different assays comparable? Other light scattering techniques that quantify AAVs Who Should Attend: CMC teams supporting AAV-based therapies Scientists involved in AAV characterization, analytical method development, process development, and quality control Lab managers that need to select and support an optimal set of AAV characterization methods   Brought to you by: Speakers: Margaret Butko Associate Director, Protein Analytical Characterization at Adverum Prithwijit Sarkar Regional Manager, Northern California, Wyatt Technology Corporation Kelly McSweeney Contributing Editor, C&EN Media Group
ACS Webinars Hub Placeholder Image.png
Labels (2)
The development of rechargeable Li-ion batteries (LIBs) has revolutionized electric vehicles and portable electronic devices. Further advancements are needed to improve the power density, safety, reliability, and lifetime of LIBs. ​​Over the past few decades, atomistic modeling of battery materials has complemented experimental characterization techniques and has become an integral part of the development of new technologies. Reliable atomic scale modeling enables rapid initial evaluation of large chemical and material design space accelerating the development cycle of next-generation battery technologies.    In this webinar, we will demonstrate how Schrödinger’s advanced digital chemistry platform can be leveraged to accelerate the design and discovery of next-generation battery materials with improved properties. We will discuss the application of both physics-based and machine learning techniques for understanding structure-property relationships of different components of batteries including electrodes, electrolytes and electrode-electrolyte interfaces. We also discuss the automated active learning framework for the development of state-of-the-art neural network force fields for modeling liquid electrolytes. The framework allows training the force field using highly accurate range-separated hybrid density functional theory data which enables accurate prediction of critical bulk properties of high-performance liquid electrolytes for application in advanced batteries.    Key Learning Objectives: Understand predictive capabilities of physics-based modeling for battery materials  Learn how automated high throughput simulation workflows enable rapid screening of new battery material candidates  Application of advanced neural network force fields for accurate electrolyte property prediction  Who Should Attend: Synthetic Chemists  Materials Scientists  Digitization Managers  R&D Scientists designing novel battery materials Brought to you by: Speakers: Garvit Agarwal Senior Scientist, Schrödinger Kelly McSweeney Contributing Editor, C&EN Media Group
ACS Webinars Hub Placeholder Image.png
Labels (2)
Labels (2)
Wednesday, October 26, 2022
Over the last two decades, traditional small molecules and antibody drugs have provided tremendous benefits to cancer patients. Due to the complexity of the disease, incomplete response and drug resistance ultimately emerge. New modalities beyond traditional small molecules and antibodies can provide unprecedented opportunities for drugs previously deemed “undruggable” targets. As an example, KRASG12C covalent inhibitors have shown promise for the treatment of Non-small cell lung cancer with KRASG12C mutations. Other KRAS mutants which cannot be inhibited by covalent inhibitors remain “undruggable”. In this webinar, we will discuss the opportunity of new modalities like oligonucleotides and TCR-engineered T cell therapy for treating KRAS mutant cancers, as well as various modality toolboxes to support drug discovery programs.     Key Learning Objectives: Review and highlight the use of various drug modalities in oncology  Dive into specific examples of different modalities for a particular target and learn key takeaways  Who Should Attend: Scientists (biologist, oncologist, chemist, etc.)  Program leaders and strategists  Project managers  Medical doctors  Individuals interested in understanding how a new modalities could expand therapies  Brought to you by: Werngard Czechtizky Executive Director Head Medicinal Chemistry, Chair of AZ Global Chemistry Leadership, Astrazeneca Luping Lin Director Biology, WuXi AppTec Jiafeng Rui Associate Director, Biology, WuXi AppTec Melissa O'Meara Forensic Science Consultant, C&EN Media Group
ACS Webinars Hub Placeholder Image.png
Labels (2)
Computational methods can significantly shorten the drug discovery process, accelerating the identification of hit compounds against a therapeutic target and the progression into promising pre-clinical candidates. When used effectively, computational approaches help predicting the molecules that are most likely to succeed, preventing unnecessary wet chemistry and saving time, money, and resources.    In this webinar, we will show how computational tools help research chemists discover and optimize novel small molecules more efficiently and effectively through the synergistic application of ligand-based and structure-based methods. These include cutting-edge methods such as Free Energy Perturbation (FEP) for accurately predicting the biological activity of new compounds before synthesis, and tools for understanding the structure-activity relationships of ligand series to inform new molecule design.    Key Learning Objectives: Understand how to synergistically apply different computational methods to analyze the information available in a drug discovery project, obtaining valuable information on what to do next.  Learn the benefits and limitations of different methods for predicting the activity of new compounds and find the best molecules to progress.  Who Should Attend: Computational chemists  Medicinal and synthetic chemists  Drug discovery scientists in academia  Drug discovery scientists in biotechnology, pharmaceutical, agrochemical, flavor and fragrance industry  Brought to you by: Giovanna Tedesco Head of Products, Cresset Ann Thayer Contributing Editor, C&EN Media Group
ACS Webinars Hub Placeholder Image.png
Labels (2)
Tuesday, November 1, 2022
Over the last two and a half years, we’ve all had to learn to communicate and work in new ways. With the help of technology, we’ve connected with colleagues across the globe in ways we never thought possible.  The ability to work remotely has ensured continued quality management of products consumed daily. For an analytical chemist or lab manager, quality management may include reviewing and approving data from the safety of your office whether in the lab or at home. No matter where the data is reviewed, completeness, accuracy and traceability must be maintained. This can be accomplished with a secure and efficient software platform.    During this webinar, Kerri-Ann Blake, Product Manager for Titration, and Daniel Schmidt, IT Systems Engineer, will introduce you to the OMNIS data management platform. They will discuss how you can securely implement the software, review and sign data. You’ll learn how to track it all via audit trail from anywhere in the world - giving you peace-of-mind that all your data is being kept safe.    Key Learning Objectives: Introduce OMNIS software’s seamless networking capability and user management system  Demonstrate convenient audit trail features with next level compliance and data integrity  Discuss system security and ease of implementation  Who Should Attend: QC/QA, R&D and Process Development Lab Managers and Users  Anyone performing titrations in a 21 CFR Part 11 Compliant Environment  Those interested in data security and integrity  Brought to you by: Kerri-Ann Blake Titration Product Manager Metrohm USA Daniel Schmidt IT Systems Engineer Metrohm USA Catherine Dold Health & Environment Writer C&EN Media Group
ACS Webinars Hub Placeholder Image.png
Labels (2)