Control tools for flow chemistry processing and their application to the synthesis of bromodomain inhibitors A thesis presented by Richard J. Ingham for the degree of Doctor of Philosophy June 2014 Jesus College Jesus Lane Cambridge CB5 8BL Ley Group Department of Chemistry Lensfield Road Cambridge CB2 1EW Declaration This dissertation is the result of my own work and includes nothing that is the outcome of work done in collaboration except where specifically included in the text and bibliography. This thesis is not substantially the same as any that I have submitted for a degree, diploma or other qualification at any other University. I further state that no part of my thesis has already been or is being concurrently submitted for any such degree, diploma or other qualification. It does not exceed 60 000 words, including tables, footnotes, bibliography and appendices. Richard Ingham, June 2014 i ii Acknowledgments I am hugely grateful to Professor Steven Ley for welcoming me into his research group and making it such a wonderful place to work. I cannot think of anywhere I would have preferred to spend four years! Steve’s infectious enthusiasm for new ideas, support and encouragement make the exciting research that takes place in the Ley group possible. Professor Ian Baxendale was also instrumental during the first half of my time in the group, and in founding the fantastic team ITC. Thanks to Dr Elena Riva for her collaboration in the solid-supported synthesis project, and to Dr Nikzad Nikbin for introducing Elena and I to the “way of the monolith.” Thanks to Dr Claudio Battilocchio for his collaboration with the pyrazine and adamantanone projects (for making the chemistry work, for his frequent displays of musical prowess, and for teaching the ITC the more colourful aspects of the Italian language). Thanks to Daniel Fitzpatrick for construction of the rotating filter prototype, and to Dr Éric S´liwin´ski for creating some fantastic 3D models. Finally, thanks (again) to Nik, to Dr Lucie Guetzoyan, Charlotte Sutherell, Michael Wolling, Julien Rossignol, the members of the SGC and everyone else who took part in the bromodomain project. Thank you to everyone who is in or has passed through the Ley group during my stay, for creating such a special atmosphere, planning the parties, and creating all of the memories. Particular thanks to Richard, Nik, Claudio, Eric, Duncan, James, Charlotte, Gonçalo, Daniel, Lucy and Lucie for taking the time to read this thesis in its varying states of completion, and for their helpful comments and suggestions. The department would not function without the hard work of the technical staff: thanks to Dr Richard Turner, Melvyn Oriss, Keith Parmenter and Matt Pond for keeping the labs working and stocked with reagents and equipment. And thanks to Jude, Jacqui, Jo and Rose for keeping the Ley group stocked with happy people! Finally, a big thank you to all of my friends and family for always being around to provide help and support, for making life both in Cambridge and out so wonderful, and without whom I wouldn’t have made it this far. iii Summary Flow chemistry and continuous processing techniques are now frequently used in synthetic laboratories, taking advantage of the ability to contain reactive or hazardous intermediates and to perform moderate scale-up processes for important compounds. However, only a limited number of methods and tools for connecting flow synthesis steps into a single protocol have been described, and as a result manual interventions are frequently required between consecutive stages. There are two main challenges to overcome. Work-up operations such as solvent extractions and filtrations are invariably needed to ensure high purity of the intermediates. Solutions for achieving this are well established within industrial facilities for continuous production, but adapting such machinery for laboratory use is rarely straightforward. Secondly, the combination of multiple steps tends to result in a more elaborate reactor configuration. The control procedures required to achieve optimum performance may then be beyond the capabilities of a single researcher. Computer control and remote monitoring can help to make such experiments more practical; but commercially-available systems are often highly specialised, and purpose-built at high cost for a particular system, and so are not suitable for laboratory scientists to use routinely. This work describes the development of software tools to enable rapid prototyping of control systems that can integrate multiple instruments and devices (in Chapter 2). These are applied to three multi-step synthesis projects, which also make use of enabling technologies such as heterogeneous reagents and in-line work-up techniques so that material can be passed directly from one stage to the next: In Chapter 1, a series of analogues of a precursor to imatinib, a tyrosine kinase inhibitor used for the treatment of chronic myeloid leukaemia, are prepared. A “catch-react-release” technique for solid-phase synthesis is used, with computer-controlled operation of the reactors. In Chapter 3, a two-step procedure for the synthesis of piperazine-2-carboxamide, a valuable 3D building block, is developed. A computer control system enabled extended running and the integration of several machines to perform optimisation experiments. In Chapter 4, improvements to the continuous synthesis of 2-aminoadamantane-2-carboxylic acid are discussed. This includes an integrated sequence of three reactions and three work- up operations. iv The final chapter describes a project to evaluate the application of control techniques to a medicinal chemistry project. New ligands for BRD9 and CECR2, proteins involved in the recognition of acetylated histone proteins, are produced. A number of triazolopyridazine compounds were synthesised and tested using a number of assay techniques, including a frontal-affinity chromatography system under development within our group. Pleasingly, the qualitative FAC data showed a good correlation with biological assessments made using established assay techniques. Further work using the FAC method is ongoing. Contents Glossary and Abbreviations ix Key to flow reactor symbols xiii Introduction 1 Automation in Synthesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2 Flow Chemistry . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3 Automation in Flow Chemistry . . . . . . . . . . . . . . . . . . . . . . . . . . 5 Objectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6 1 Solid-supported flow synthesis of 2-aminopyrimidines 7 1.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8 1.1.1 Synthesis of 2-aminopyrimidines . . . . . . . . . . . . . . . . . 8 1.1.2 Solid-supported reagents . . . . . . . . . . . . . . . . . . . . . . 9 1.1.3 Monolithic reagents . . . . . . . . . . . . . . . . . . . . . . . . 10 1.1.4 Proposed synthetic route to 2-aminopyrimidines . . . . . . . . . 13 1.2 Development of Chemistry . . . . . . . . . . . . . . . . . . . . . . . . . 14 1.2.1 Generation of the monolithic reagent . . . . . . . . . . . . . . . 14 1.2.2 Monolith characterisation . . . . . . . . . . . . . . . . . . . . . 15 1.2.3 Heterocycle Formation . . . . . . . . . . . . . . . . . . . . . . . 16 1.2.4 Oxidation of monolith M2 . . . . . . . . . . . . . . . . . . . . . 18 1.2.5 Release of 2-aminopyrimidine product . . . . . . . . . . . . . . . 19 1.3 Automation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21 1.3.1 Advantages of automation . . . . . . . . . . . . . . . . . . . . . 21 1.3.2 Solvent switching and stop-flow . . . . . . . . . . . . . . . . . . 24 1.3.3 Multi-step processing . . . . . . . . . . . . . . . . . . . . . . . . 25 1.4 Application to the preparation to derivatives of imatinib amine . . . . . . 27 1.4.1 Reported syntheses of imatinib . . . . . . . . . . . . . . . . . . . 27 1.4.2 Monolith-assisted route to imatinib . . . . . . . . . . . . . . . . 30 1.5 Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32 v vi Contents 2 New control and monitoring software for flow chemistry systems 33 2.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 34 2.1.1 Existing control software . . . . . . . . . . . . . . . . . . . . . . 35 2.1.2 Flow chemistry equipment . . . . . . . . . . . . . . . . . . . . . 38 2.1.3 Interaction with equipment . . . . . . . . . . . . . . . . . . . . . 38 2.1.4 Control computers . . . . . . . . . . . . . . . . . . . . . . . . . 40 2.2 Implementation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 42 2.2.1 Communication . . . . . . . . . . . . . . . . . . . . . . . . . . . 43 2.2.2 Data handling . . . . . . . . . . . . . . . . . . . . . . . . . . . . 47 2.2.3 Command sequences . . . . . . . . . . . . . . . . . . . . . . . . 49 2.2.4 Data manipulation . . . . . . . . . . . . . . . . . . . . . . . . . 50 2.2.5 Pausing experiments . . . . . . . . . . . . . . . . . . . . . . . . 53 2.2.6 Error handling . . . . . . . . . . . . . . . . . . . . . . . . . . . 53 2.2.7 Attached control systems . . . . . . . . . . . . . . . . . . . . . . 53 2.3 Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 54 3 Automated two-step reactions with heterogeneous metal catalysis 57 3.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 58 3.2 Synthesis of primary amides by nitrile hydration . . . . . . . . . . . . . . 58 3.2.1 Hydration under flow conditions . . . . . . . . . . . . . . . . . . 60 3.3 Reaction control and monitoring . . . . . . . . . . . . . . . . . . . . . . 61 3.4 Automation of multiple experiments . . . . . . . . . . . . . . . . . . . . 63 3.5 Integration of two steps . . . . . . . . . . . . . . . . . . . . . . . . . . . 67 3.5.1 Intermediate reservoir . . . . . . . . . . . . . . . . . . . . . . . 67 3.5.2 Combining synthesis and DoE . . . . . . . . . . . . . . . . . . . 70 3.5.3 Extended run . . . . . . . . . . . . . . . . . . . . . . . . . . . . 70 3.6 Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 72 4 An integrated flow synthesis of 2-aminoadamantane-2-carboxylic acid 75 4.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 76 4.1.1 Challenges of telescoped reactions . . . . . . . . . . . . . . . . . 76 4.2 Previous synthetic routes . . . . . . . . . . . . . . . . . . . . . . . . . . 79 4.2.1 Reported batch–mode syntheses . . . . . . . . . . . . . . . . . . 79 4.2.2 First generation flow–mode synthesis . . . . . . . . . . . . . . . 80 4.3 Grignard reaction and aqueous extraction . . . . . . . . . . . . . . . . . 82 4.3.1 Mixing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 82 4.3.2 Contactors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84 4.3.3 Filtration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85 4.3.4 Separation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86 4.3.5 Control of the Grignard reaction . . . . . . . . . . . . . . . . . . 90 4.4 Solvent exchange . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 92 4.5 Ritter reaction and quenching . . . . . . . . . . . . . . . . . . . . . . . . 94 4.5.1 Intermediate reservoir . . . . . . . . . . . . . . . . . . . . . . . 94 4.5.2 Ritter reaction . . . . . . . . . . . . . . . . . . . . . . . . . . . . 95 Contents vii 4.5.3 Base quenching . . . . . . . . . . . . . . . . . . . . . . . . . . . 96 4.5.4 Cyclisation reaction . . . . . . . . . . . . . . . . . . . . . . . . 100 4.6 Ozonolysis and Hydrolysis . . . . . . . . . . . . . . . . . . . . . . . . . 101 4.7 Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 103 4.7.1 Further work . . . . . . . . . . . . . . . . . . . . . . . . . . . . 104 5 Development of inhibitors for bromodomain-containing proteins 105 5.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 106 5.1.1 The bromodomain . . . . . . . . . . . . . . . . . . . . . . . . . 107 5.1.2 Frontal Affinity Chromatography . . . . . . . . . . . . . . . . . 109 5.2 Chemical probes for BRD9 . . . . . . . . . . . . . . . . . . . . . . . . . 112 5.2.1 Literature . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 112 5.2.2 Proposed synthesis . . . . . . . . . . . . . . . . . . . . . . . . . 116 5.2.3 Synthesis of core . . . . . . . . . . . . . . . . . . . . . . . . . . 116 5.2.4 Synthesis of boronic ester coupling partners . . . . . . . . . . . . 124 5.2.5 Cross-coupling . . . . . . . . . . . . . . . . . . . . . . . . . . . 125 5.2.6 Carbamate formation . . . . . . . . . . . . . . . . . . . . . . . . 133 5.2.7 Alternative route . . . . . . . . . . . . . . . . . . . . . . . . . . 134 5.2.8 Biological assessment . . . . . . . . . . . . . . . . . . . . . . . 136 5.2.9 Evaluation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 139 5.3 Chemical probes for CECR2 . . . . . . . . . . . . . . . . . . . . . . . . 140 5.3.1 “Reversed” sulfonamide boronic ester . . . . . . . . . . . . . . . 142 5.3.2 Miyaura borylation . . . . . . . . . . . . . . . . . . . . . . . . . 143 5.3.3 Suzuki cross-coupling . . . . . . . . . . . . . . . . . . . . . . . 147 5.3.4 Biological analysis . . . . . . . . . . . . . . . . . . . . . . . . . 149 5.4 Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 152 Conclusion 155 A Synchronous instrument controller 157 B Control scripts for monolith processing 163 Experimental 169 General Experimental Information . . . . . . . . . . . . . . . . . . . . . . . . 170 Solid-supported synthesis of 2-aminopyrimidines . . . . . . . . . . . . . . . . 172 Piperazine-2-carboxamide synthesis . . . . . . . . . . . . . . . . . . . . . . . 190 2-Aminoadamantane-2-carboxylic acid synthesis . . . . . . . . . . . . . . . . 194 Chemical Probes for BRD9 . . . . . . . . . . . . . . . . . . . . . . . . . . . . 200 Chemical Probes for CECR2 . . . . . . . . . . . . . . . . . . . . . . . . . . . 216 Frontal Affinity Chromatography . . . . . . . . . . . . . . . . . . . . . . . . . 228 References and Notes 228 viii Glossary and Abbreviations  Inner diameter 1D One-dimensional 2D Two-dimensional Ac Acetate ACHC Azobiscyclohexylcarbonitrile (radical initiator) Ar Unspecified aryl substituent BEMP 2-tert-Butylimino-2-diethylamino-1,3-dimethylperhydro-1,3,2-diazaphosphorine BPR Back pressure regulator br. Broad peak (NMR / IR spectroscopy) BRD Bromodomain Bu Butyl CV Column volumes (column chromatography) conv. Conversion COSY 2D 1H correlation spectroscopy (NMR spectroscopy) CPME Cyclopentyl Methyl Ether δ Chemical shift (NMR spectroscopy) d Days, or doublet peak (NMR spectroscopy) DCM Dichloromethane DEPT Distortionless Enhancement by Polarization Transfer (NMR spectroscopy) DIN Deutsches Institut fur Normung E.V. (German Institute for Standardisation) DIPEA Diisopropylethyl amine DMAP 4-Dimethylaminopyridine DME Dimethoxyethane ix x Glossary and Abbreviations DMF N,N-Dimethylformamide DMF-DMA N,N-Dimethylformamide dimethylacetal DMSO Dimethyl sulfoxide DMSO-d6 Deuterated dimethyl sulfoxide DoE Design of Experiments DPE-Phos Bis[(2-diphenylphosphino)phenyl]methane DPPA Diphenoxyphosphoryl azide dppf 1,1′-Bis(diphenylphosphino)ferrocene DtBPF 1,1′-Bis(di-tert-butylphosphino)ferrocene DVB Divinylbenzene EI Electron-Impact Ionisation (mass spectroscopy) EMM Ethoxymethylene(malononitrile) equiv. Equivalents ESI Electrospray Ionisation (mass spectroscopy) Et Ethyl ETFE Ethylene tetrafluoroethylene (polymer) FAC Frontal Affinity Chromatography FEP Fluorinated Ethylene Propylene (polymer) FT-IR Fourier Transform Infrared (spectroscopy) HMBC Heteronuclear Multiple Bond Connectivity (NMR spectroscopy) HMQC Heteronuclear Multiple Quantum Coherence (NMR spectroscopy) HPLC High-Performance Liquid Chromatography HRMS High-Resolution Mass Spectroscopy I2C Inter-Integrated Circuit (serial data communication protocol) IC50 Median inhibition concentration (concentration of a substrate that reduces a protein’s activity by 50 %) ICT Information and Communications Technology ITC Innovative Technology Centre, University of Cambridge ID Inner Diameter IR Infrared isol. Isolated Glossary and Abbreviations xi J Coupling constant (NMR spectroscopy) Kd Dissociation constant LC-MS Liquid Chromatography coupled Mass Spectroscopy m Multiplet (NMR spectroscopy), or medium absorbance (IR spectroscopy) m meta (position around a phenyl ring) M Moles per litre, or unspecified metal atom M+ Molecular ion m-CPBA meta-Chloroperoxybenzoic acid Me Methyl mol% Molar percentage m.p. Melting point Ms Methanesulfonyl MW Microwave m/z Mass to charge ratio (mass spectroscopy) NMR Nuclear Magnetic Resonance (spectroscopy) Nu Unspecified nucleophile o ortho (position around a phenyl ring) OD Outer Diameter p para (position around a phenyl ring) PC Personal Computer PEEK Polyether ether ketone (polymer) PEPPSI Pyridine-enhanced precatalyst preparation, stabilization and initiation PFA Perfluoroether (class of polymer) Ph Phenyl ppm Parts per million PTFE Polytetrafluoroethylene (polymer) QP-BZA QuadraPure benzylamine QP-DMA QuadraPure dimethylamine QP-TU QuadraPure thiourea quant. Quantitative xii Glossary and Abbreviations q Quartet peak (NMR spectroscopy) R Unspecified alkyl substituent RS-232 Recommended Standard 232 (serial data communication standard) s Singlet peak (NMR spectroscopy), or strong absorbance (IR spectroscopy) SGC Structural Genomics Consortium, Oxford University SMS Short Message Service SS Stainless steel t Triplet peak (NMR spectroscopy) TFA Trifluoroacetic acid tR Retention time in minutes (LC-MS) TLC Thin Layer Chromatography THF Tetrahydrofuran USB Universal Serial Bus (serial data communication standard) UV Ultraviolet (spectroscopy) v/v volume / volume w Weak absorbance (IR spectroscopy) w/w weight / weight Key to flow reactor symbols 100 psi RR Pump Mixer Spring-type back-pressure regulator In-line detector Glass column Immobilised reagent Valve Sample loop Heated ow coil Monolithic immobilised reagent xiii xiv Introduction 1 2 Introduction Automation in Synthesis Automation has had a significant impact on many different industries since the mid 20th century. Robots of all sizes and specifications are installed in factories to perform tasks that are routine, or too difficult or strenuous for a person to do unassisted, and allowing 24 hour working with a smaller manpower requirement. In a research laboratory, automation refers to the use of computers or micro-controllers to interact with equipment to perform tasks that the researchers would otherwise have to do manually. There are a number of tasks that already routinely benefit from automation. Repetitive tasks such as injecting HPLC samples or making serial dilutions of samples for biological assays can be performed by laboratory robots, though these are often highly engineered for a particular application and very expensive. In-situ analytical equipment can collect more data about experiments than may be possible with manual observations alone, and digital data collection is often more accurate than manual observations, and can be run indefinitely, without breaks. In general, the use of automation can produce better data, and give researchers more time to do less mundane tasks. In the context of synthetic chemistry, automation is typically associated with combinatorial chemistry.1,2 This involves repetition of simple synthetic steps to produce huge numbers of building blocks or compounds for biological analysis. The problem was that by creating large numbers of compounds using only a small number of reactions, the size of the chemical space that was explored turned out to be relatively limited, and highly dependent on the availability of different starting materials. Combinatorial chemistry went through a phase of popularity in the 1980s and 1990s before falling out of favour having not produced significant results.3,4 However, modern concepts such as diversity-oriented synthesis5 may offer new targets for automated synthesis platforms. There are a number of potential benefits to automating the control of synthetic procedures. The key is that the focus should be on improving the quality of individual reactions rather than the quantity of reactions that are performed. Computers and robots can enable scientists to accomplish more complex tasks more easily than if they were done manually, and by delegating routine work to automated systems skilled chemists can be freed to do more important tasks. There may also be an improvement in the reporting and reproducibility of experiments: by consistent recording of the reaction parameters (such as the amount of reagents used, the timing of their addition, temperatures and heating times), and by accurate measurements and recording of data collected during an experiment, reactions in the lab of Flow Chemistry 3 the future could be made more reproducible and the results even more useful.6 As a result, the quality of the data reported in the literature would only improve.7,8 The use of automated platforms could provide other benefits as well. Disconnecting the scientist from the manual aspects of a reaction means that hazardous materials could be used more safely. Chemists could run reactions from a different room, or even from different institutions or countries, providing global access to key facilities. Flow Chemistry Flow chemistry commonly refers to the continuous processing of chemical reagents within small tubes or channels. In conventional batch-mode chemistry, many addition, mixing and reacting operations for one reaction may be performed in the same vessel. By contrast, in a flow chemistry system the reagents are delivered continuously by pumping devices; multiple streams are mixed and then allowed to react in tube or column reactors which may be heated, cooled or irradiated to provide the desired reaction conditions. Reaction times are determined by the reactor volume and the flow rate, and reaction scale by the duration of operation. The history of flow chemistry is rooted in the continuous processing of petrochemicals. The volume of material handled in processes such as cracking and refining are so large that batch methods would be impractical and economic due to the number of manual operations that would be needed. Early examples of continuous manufacturing of bulk chemicals includes the processes for producing ammonia9 and sulfuric acid,10 where the economies of scale again made continuous processing an ideal tool. The use of continuous methods for fine chemicals manufacture originated as a means for process intensification,11 intended to reduce the footprint and manpower requirements of a manufacturing plant, and also to improve safety, energy efficiency and environmental-friendliness.12 However, the speciality chemicals and pharmaceuticals sectors have been slow to adopt continuous manufacturing, primarily as a result of having much smaller production volumes and a large amount of sunken capital in batch reactor equipment. Work within academic laboratories may lead to the development of new methods and processes that will encourage further uptake of flow technology by enabling new or hazardous reactivity to be exploited under a continuous regime. 4 Introduction Flow reactors are often considered to have a number of advantages over batch-mode reaction vessels. With appropriate static or dynamic mixers, the concentration profile can be controlled predictably; if the mixing is very efficient then remarkable improvements in reaction times can be observed for some transformations.13,14 In-line heterogeneous reagent or catalyst cartridges (containing either inorganic or polymeric reagents) can perform chemical transformations with little or no purification requirement.15 Addition of further reagent streams can enable direct quenching or use of reactive intermediates, leading to improved synthetic efficiency and consistency. The large surface area to volume ratio of a flow reactor can allow precise temperature control, extreme conditions and rapid temperature changes,14 as well as safe containment of exothemic events.16 Factors such as mixing and heat transfer are critical parameters when scaling a reaction; in a flow reactor these are independent of the amount of material that is to be processed. Combined with a small reaction volume (1 – 20 mL) compared to standard batch apparatus (10 – 500 mL), continuous processing can deliver significant improvements in the safety of hazardous transformations.16–19 For example, hydrogenation reactions are increasingly being performed in much smaller, safer continuous flow reactors instead of in batch hydrogenators that had to be housed in concrete bunkers to shield against catastrophic explosions.20,21 There are of course some disadvantages that must be kept in mind when using flow reactors.22 They are by their nature much more complex than a flask, and many of the devices currently available are very susceptible to blockage or disablement by solids that form during the reaction. Flow reactions operate most consistently at steady state, and this is only attained after a period of time that is dependent on the reactor’s volume, so a b c Figure 1 Some of the commercially available flow chemistry systems. (a) Vapourtec R2+/R4. (b) Uniqsis FlowSyn. (c) Syrris Asia. Automation in Flow Chemistry 5 are often inappropriate for running large numbers of small-scale reactions. In this regard, they are much less flexible than batch chemistry apparatus: frequently the reactor must be reconfigured to perform different reactions, meaning that a significant time investment is required for each new procedure. This is another reason why the uptake of continuous manufacturing has been slow in the speciality chemicals and pharmaceuticals sectors. Even so, there are a large number of applications for which continuous processing tech- niques are highly effective. For this reason, flow synthesis has become increasingly popular over the past decade.23 Whilst flow reactions can be performed using any appropriate pumping and heating equipment, many researchers choose to employ commercial reactor platforms such as the Vapourtec R2+/R4, Uniqsis FlowSyn, or Syrris Asia systems which are used within our own laboratory (Figure 1). These systems contain pumps, switching valves and heaters within a conveniently integrated unit, and provide software tools to run computer-controlled experiments. Automation in Flow Chemistry Using these systems, our group and others have reported numerous applications of auto- mated flow chemistry technology to combinatorial chemistry activities.23,24 By utilising tools such as polymer-supported reagents and scavengers,25 the desired compounds can be collected in very high purity with little or no purification. Perhaps as a direct result of these early projects, the control software packages provided with commercial flow chemistry platforms tend to be designed for exactly this kind of a b Figure 2 (a) Fluidic configuration using Vapourtec Flow Commander. (b) Choosing experiments using Vapourtec Flow Commander. 6 Introduction procedure. A fluidic configuration can be programmed in, and then long lists of identical reactions using different starting materials can be run with the aid of a liquid handler (Figure 2). In this regard, commercial flow chemistry equipment offers good tools for starting automation of synthesis within the laboratory. The in-built computer connections mean that the apparatus is ready to automate without significant additional engineering effort. If control and monitoring tools are applied to laboratory platforms then the process of scaling reactions may become even easier. If a synthesis system can be left under computer control then it can be run for a long time without active observation, enabling moderate scale-up operations to be performed within the research laboratory itself. Objectives Until recently, programming skills have tended to be limited to specialists. However, with the increasing pervasiveness of the internet connectivity, and concepts such as the “internet of things”, there is much greater interest in the use of computers to influence the physical world around us.26 In the United Kingdom, the school curriculum is being updated to cultivate software creation as well as more traditional ICT skills.27 This is in part aided by organisations such as the Raspberry Pi foundation,28 whose devices were designed to provide affordable computer systems for everyone. This may result in a greater number of scientists wanting to run experiments under computer control. Flow chemistry can provide access to new reactive windows and easier scalability of synthetic processes. However, most reported flow procedures consist of a single step, and to our knowledge very few true multi-step flow syntheses have been described in the literature.23 One reason for this is that the orchestration of the many devices required for an integrated system is very difficult. Computer control and automation can provide the means to do this, but is currently a relatively unexplored aspect of laboratory flow chemistry, since commercial control system software does not tend to offer a level of flexibility appropriate for discovery workflows. The objective of this project was to apply or develop in-line processing and computer control tools to enable the construction of continuous multi-step flow processes, and to demonstrate these within the research laboratory. Chapter 1 Solid-supported flow synthesis of 2-aminopyrimidines 7 8 Solid-supported flow synthesis of 2-aminopyrimidines 1.1 Introduction* 1.1.1 Synthesis of 2-aminopyrimidines One of the primary goals within the field of organic synthesis is to create cleaner and more efficient methods to assemble functional molecules. 2-Aminopyrimidines are biologically important building blocks, found in a wide range of compounds reported to possess anti- emetic,30 anti-tumour,31–33 anti-malarial,34 pain relief35 and other biological properties (Scheme 1.1a). Besides late-stage formation of the pyrimidine ring from a guanidine moiety, these compounds are commonly prepared by cross-coupling methods31,32,36 or by halogen substitution.30–32,35 However, there is an alternative method for accessing the 2-aminopyrimidine unit involving the displacement of an alkylthiol with a nucleophile such as a secondary amine (Scheme 1.1b).34,37–39 This is potentially very powerful, because the starting materials can be prepared easily and the reaction generally proceeds in good yield. Unfortunately, this protocol has the disadvantage that alkylthiol by-products are released during the reaction, which are malodorous and often toxic. We envisaged that this could be overcome with a solid-supported methodology. N N NH2 NH2 Pr Cl N N H N N HN O N N N N N H CF3 O N S HN O N a 1 2 3 N N H MeS O R3 N R2 R1 N N H O R3NH R2 R1 alcohol solvent 100 - 180 °C O RO O R3 NH NH2MeS b Scheme 1.1 (a) Reported bioactive compounds containing the 2-aminopyrimidine scaf- fold: imatinib (1), a kinase inhibitor;33 2, an anti-malarial compound;34 3, a pain relief compound.35 (b) Method for the preparation of 2-aminopyrimidines by nucleophilic dis- placement of a thiol with an amine.37 * The work described in this chapter is reported in Org. Lett. (Reference 29). 1.1. Introduction 9 1.1.2 Solid-supported reagents Thirty years after the discovery of Bakelite,40 this plastic found an application within chemi- cal synthesis as an ion-exchange resin.41,42 Later, similar ion-exchange resins were used for purification using “catch-and-release” methods.43 Following Merrifield’s seminal reports of a solid-phase technique for peptide synthesis,44 a number of functionalised polymer resins were reported and applied to chemical synthesis, including polymer supported phosphine, halogenation reagents, redox reagents and protecting groups.45 The application of polymer-assisted solution phase synthesis provides a number of ad- vantages over conventional solution-phase chemistry.15 If a reagent is covalently attached to an insoluble polymer support, then it can be removed at the end of the reaction by a Me O N2 P O OMe OMe N3 F Ph OH SO3 NO NMe2.CuI N H S NH2 NMe2 SO3H NH2 Ph N NN F 100 psi MeOH 0.2 mL/min KOtBu MeCN 0.2 mL/min 48 min 100 °C 70 °C A21.CuI A21 PS-Ts-TEMPO A15 QP-BZA Q P-TU 2 equiv. 2 equiv. 1 equiv. 4 5 6 7 Scheme 1.2 A three-step reaction enabled by polymer-supported reagents.51 A sup- ported TEMPO reagent effects an oxidation of alcohol 6 to an aldehyde. Upon heating, this reacts with the Bestmann–Ohira reagent (4) to form an alkyne, which then undergoes an alkyne–azide “click” reaction52 in the presence of copper (immobilised on an Amberlyst-21 resin). A sequence of scavengers (present in excess) then remove any unreacted materi- als: the thiourea cartridge sequesters any copper which is leached from the A21 resin; the benzyl amine cartridge removes any unreacted aldehyde; the sulfonic acid cartridge removes the base and protonates any residues from the phosphoric reagent; finally, the dimethyl amine cartridge removes any remaining acidic material to leave a pure stream of the product. 10 Solid-supported flow synthesis of 2-aminopyrimidines simple filtration. This can reduce the number of labour-intensive workup procedures such as extractions and chromatographic purification that must be performed to obtain materials of high purity. Using these solid-supported reagents, the preparation of complex scaffolds using multi- step pathways can be achieved46–48 with only a small number of intermediate purification steps. A further advantage is that toxic, sensitive or valuable reagents can be confined on the solid material25,46,49 creating a lower exposure hazard than that when working with powders or volatile substances. It is also possible to recycle catalytic reagents easily as they can be separated from the reaction mixture by a simple filtration. For these reasons, polymer-assisted synthesis lends itself particularly well to automation50 to generate large arrays of compounds. Polymer-supported reagents are also well suited to flow chemistry processes, because they can be confined within a cartridge so that the flow stream is not contaminated. They can be used as reagents, catalysts, or scavengers which remove by-products to simplify multi-step synthesis. For example, a three-step synthesis of triazole 7 from alcohol 6 and azide 5 reported by our group51 involved sequential reduction, Bestmann–Ohira reaction and copper-catalysed alkyne–azide cyclisation reactions (Scheme 1.2). The excess unreacted materials were removed by the scavenging columns to afford the product which was collected by concentration and crystallisation. However, the integration of polymer-supported reagents into flow chemistry processes can present some difficulties, depending on the morphology of the polymeric structure. 1.1.3 Monolithic reagents Since the description of Merrifield’s resin,44 by far the most common solid support systems have been “gel-type” polystyrene resins which are prepared as small beads with a diameter of 0.5 – 1 mm. These have a low degree of cross-linking (1 – 2 %), and as a result they tend to swell and contract depending on the solvating power of a particular solvent (Figure 1.3).53 An alternative support method is the use of macroporous beads which have a higher degree of cross-linking and thus greater rigidity, maintaining their structure across a wide range of solvents. However, they still suffer from some of the other problems associated with gel-phase beads. Heterogeneous flow processes involve five conceptual steps for mass transfer between the two phases (Figure 1.4). High molecular-weight solutes diffuse slowly 1.1. Introduction 11 a b Figure 1.3 In a “poor” solvent, which does not effectively solvate the polymer, the polymer beads contract as the solvent is expelled, allowing the fluid stream to channel around them. In a “good” solvent they swell up, a process that can block the flow stream. Whilst this behaviour is not a problem for a batch-mode reaction, in a flow reactor these changes in flow dynamic can impair the reaction. into the stagnant solvent present in the pores of the beads. Although diffusion for low molecular weight solutes is rapid, the large void volume (at least 27 % for spherical beads) provides a low resistance flow path through which convective flow travels preferentially.54 1 2 3 4 5 Figure 1.4 Five steps for reactivity of solutes in a heterogeneous system. (1) External mass transfer through convective flow. (2) Diffusion into stagnant solvent surrounding solid and then into pores. (3) Chemical reaction. (4) Diffusion back into stream. (5) External mass transfer. Monoliths represent another class of immobilised reagent which has been developed to overcome some of these problems. A polymer monolith is a uniformly polymerised porous structure made by precipitation polymerisation of organic or inorganic monomers (as opposed to a system of parallel channels such as found in a catalytic converter). Monoliths were originally developed for protein chromatography,54 for which the slow diffusion was the primary challenge. They were also found to be useful for flow chemistry, since the porous polymer structure contains channels with diameters in a tight distribution around 400 nm, which are large enough to support convective flow54 without containing paths for unimpaired channelling, and keep the flow in close proximity to the pores such that diffusion between the external flow and the stagnant solution can occur rapidly.54 Since monolithic reagents can be prepared in any shape, preparation within a glass column (Figure 1.5a) results in a cylindrical monolith through which a flow stream can pass evenly. 12 Solid-supported flow synthesis of 2-aminopyrimidines 5 μm 10 μm a b c Figure 1.5 (a) Monolith prepared within a glass column. The plugs at either end are replaced with fluidic connections for the subsequent steps. (b, c) Scanning-electron micrographs showing the porous structure of a monolith. Figures 1.5b-c show the structure of the monolithic polymer using scanning-electron micrography. The visible structures constitute the microscale channels that allow convective flow, as well as the nanoscale channels (<100 nm54), which allow reagent ingress only by diffusion. The pore sizes can be tuned by altering the polymerisation mixture to create a robust and reactive monolith. Monoliths have found applications as ion-exchangers,55 chromatographic packings,56 and polymeric reagents and catalysts, for example for Diels-Alder,57 polymerisation,58 cross- coupling,59 or cyclopropanation60 reactions. Many of these involved continuous flow systems due to the benefits of flow-through reactors. In an example of a synthesis involving polymeric reagents, an azide-exchange reactor produced by treatment of a quarternary ammonium monolith with sodium azide was used to generate acyl azides (8) from acyl chlo- rides (9).61 These underwent a Curtius rearrangement to produce an isocyanate (10) product which could be recovered in quantitative yield by removal of the solvent (Scheme 1.6). Monolithic reactors can be produced in the laboratory without requiring any specialist equipment. They are made by co-polymerisation of a monomer or mixture of monomers with a cross-linker and a porogen, usually an alkyl alcohol that is also the solvent for the monomer. As the polymer chain length grows, its solubility drops until it precipitates from the solvent; the pattern of phase separation produces the porous structure. The composition of the solvent system determines the point at which this phase separation occurs and thus affects the porosity. A balance must be struck between the solubility of the components and the desired porosity, because if the porosity is too low then the monolith will create a large back-pressure when placed in a flow stream. In summary, monolithic reagents are a powerful tool for the application of polymer- 1.1. Introduction 13 100 psi MeCN 0.5 mL/min 20 min 120 °C R O N3 R N C O R O Cl NEt3 N3 13 min Azide monolith Drying agent 9 8 10 Scheme 1.6 Use of an azide exchange monolith to generate acyl azides, which were then subjected to high temperature reactor coil to perform a Curtius rearrangement reaction. The drying agent cartridge (sodium sulfate) ensured that any water present in the acetonitrile solvent was removed to prevent undesired reactions with the isocyanate product. supported synthesis methods to flow processes. We proposed to create a monolithic reagent to assist with the synthesis of compounds containing the 2-aminopyrimidine motif. 1.1.4 Proposed synthetic route to 2-aminopyrimidines For the synthesis of 2-aminopyrimidines, we designed a route based on a monolith- supported isothiourea. By attaching this reagent onto a solid support (Scheme 1.7),38,39 release of sulfurous by-products can be prevented, and the potential for contamination with thiourea is reduced. SH2N NH2Cl HS N S N R N N R N R' R'R''-NH isothiourea 2-thiopyrimidine sulfur retained R'' Scheme 1.7 Proposed route to 2-aminopyrimidines involving a solid-supported sulfur moiety. 14 Solid-supported flow synthesis of 2-aminopyrimidines 1.2 Development of Chemistry 1.2.1 Generation of the monolithic reagent Previous work within our group62 had investigated the use of compound 11 as a monomer for the preparation of a monolith which might be used for the synthesis of imidazoles and pyrimidines using a solid-supported technique.38 However, generation of the monolith had proven to be challenging, most likely because the polarity of 11 in its hydrochloride salt form made it poorly soluble in dodecanol, an alcohol used as the porogen. Attempts to solubilise the monomer with a methanol co-solvent led to an amorphous monolith, and this was speculated to be the result of having too much solvent in the polymerisation mixture. Experimentation by Dr Nikzad Nikbin, based on the use of a water/propanol solvent system,63 resulted in a monolith with a much more favourable microstructure, having a rigid structure and a back-pressure profile appropriate for use in a flow reactor (Scheme 1.8). This water/propanol solvent system is important because it allows the polymerisation of charged monomers. This can be beneficial in cases such as this one, where the isothiourea starting material was used in its (easily purified) hydrochloride salt form. Salts are generally very difficult to polymerise using the traditional methods for making polymer beads, because beads tend to be made in an aqueous/organic biphasic system,64 so amphiphilic monomers such as 11 can give poor results. In fact, Masquelin and co-workers38 used the Merrifield resin as a starting material in their synthesis of the same solid-supported isothiouronum salt, rather than attempting to polymerise a functional monomer. However, direct polymerisation of the salt is advantageous because this is likely to result in a solid reagent with consistent functionality across the entire polymer; and if it is made as a cross-linked monolith, then its SH2N NH2Cl ACHC 6.6:1 1-propanol/water 90 °C, 20 h SH2N NH2Cl 11 M1 Scheme 1.8 Preparation of functionalised monolith M1. Polymerisation conditions: 4- (vinylbenzyl)isothiuronium chloride (11; 18.8 % w /w); divinyl benzene (DVB, cross-linker; 12.5 % w /w); 1-propanol (porogen; 57.5 % w /w); water (porogen; 10.9 % w /w); 1,1′- azobis-(cyclohexane carbonitrile) (ACHC, initiator; 1 % w /w relative to 11 + DVB). 1.2. Development of Chemistry 15 morphological properties can be tuned during polymerisation. The monomer (11) was easily prepared by a nucleophilic substitution of the chlorine moiety of 4-vinylbenzyl chloride (12) with thiourea (Scheme 1.9). The presence of an imine functionality was suggested by an absorbance at 1655 cm−1 in the infra-red spectrum, and a 13C resonance at 169 ppm. In the 1H NMR spectrum, the two broad singlet resonances at 9.4 and 9.5 ppm from the NH2 groups are in very similar chemical environments, as expected for this structure. The iminium centre is more deshielded than would be expected for a monoprotonated guanidine moiety,65 possibly due to the greater electronegativity of the sulfur atom compared to an amino group. Following the polymerisation, the new monolith was washed with either ethanol or THF to remove the residual porogen and any unpolymerised material. Very little of the monomer was observed in the efflux stream, indicating a good degree of polymerisation. Having identified an efficient method for producing the functional monoliths, methodology for the solid-supported synthesis of 2-aminopyrimidines could be explored. SH2N NH2Cl Cl H2N S NH2 EtOH reflux, 6 h 13 12 11 Scheme 1.9 Preparation of the functionalised monomer 11 by condensation of thiourea (13) with 4-vinylbenzyl chloride (12). 1.2.2 Monolith characterisation One of the drawbacks of solid-phase synthesis is that analysis of the reaction intermediates is much more challenging than for a solution phase synthesis. Conventional methods such as chromatography or mass spectrometry can only be used for fluid samples, and so it was only possible to use these to characterise the waste stream after a reaction. Elemental microanalysis proved to be a useful technique for following the reactions when on the solid support, particularly as many of the transformations involved a change in the number of nitrogen atoms. Although solid samples might not normally be considered for NMR analysis, solid state- NMR by “magic angle” spinning makes use of a method for overcoming the significant line broadening brought about by anisotropy within the sample. Because the positions and 16 Solid-supported flow synthesis of 2-aminopyrimidines SH2N NH2Cl NC H2N N S N(iPr)2NEtEtOH EtO CN CN 25 °C, 30 min 83%1.01 eq11 14 15 Scheme 1.10 Solution-phase cyclisation of functionalised monomer with EMM. orientations of molecules in a solid are fixed, there is no random tumbling which in the solution phase averages out time-dependent interactions. However, by spinning a sample at 54.7° relative to the magnetic field (the “magic angle”) the influence of anisotropic interactions is reduced, providing greater signal resolution. The resulting spectral peaks are often still quite broad, but in some cases the peak shapes can provide further structural information about the sample.66 It can also be very useful to record the dispersion pattern generated by a solid packed bed such as a monolith, to confirm that the desired porosity has been generated. This could be achieved by injecting a pulse of a dye, a salt solution or a UV-active compound into the column, and then measuring the concentration profile of the output stream using a visible light, conductivity or UV detector.67 Monolith M1 was characterised by infra-red and 13C NMR spectrometry. The iminium resonance at 171 ppm was conserved in the NMR spectrum; in the IR spectrum the absorp- tion at 1647 cm−1, attributed to the imine C−N bond, was also still visible. These data suggested that the key thiouronium structure was preserved during the polymerisation. In some cases reactions were tested in the solution phase using unpolymerised monomers, allowing traditional analytical techniques to be used, before attempting a solid-phase reaction. However, solution- and solid-phase conditions are not always comparable; for instance, more forcing conditions were often needed for the heterogeneous reactions. 1.2.3 Heterocycle Formation Previous reports37,68,69 indicated that an isothiourea would combine with a bifunctional compound such as ethoxymethylene(malononitrile) (14, EMM)38 to form a heterocyclic product. Pleasingly, this was found to be the case; mixing 11 and 14 at room temperature afforded pyrimidine 15 as a precipitate (Scheme 1.10). Hünig’s base (DIPEA) was selected 1.2. Development of Chemistry 17 for these reactions because, being a liquid, it was more amenable to this flow process than the inorganic bases such as K2CO3 used in previous reports. In a mixed-phase system, stirring a crushed monolith overnight with EMM and DIPEA afforded a yellow solid after filtration and washing (Scheme 1.11). SH2N NH2Cl NC H2N N S N (iPr)2NEt EtOH EtO CN CN 25 °C, 18 h M1 14 M2 Scheme 1.11 Solid-phase cyclisation of functionalised monolith with EMM. The formation of the desired pyrimidine M2 was supported by evidence from the elemental analysis, showing 8.8 mmol/g of nitrogen and 2.9 mmol/g sulfur,† suggesting approximately 1.8 mmol/g of the pyrimidine, with 0.8 mmol/g of the isothiourea remaining unreacted. The infrared spectrum showed characteristic signals, such as new absorptions in the regions of 1250 and 1400 cm−1, as well as a strong absorption at 2217 cm−1, indicative of a C−−N functional group. The disappearance of the thiouronium centre at 171 ppm could be observed by solid-state NMR spectroscopy experiments. When performed using a flow reactor (Scheme 1.12), the progress of this reaction could be observed by a change in colour of the monoliths from white to yellow (Figure 1.13). For further information about the functional loading of M1, a known quantity of EMM (14) DIPEA (2.5 equiv.) MeCN 0.1 mL/min (2 equiv.) 1:1 DMF/MeCN 0.1 mL/min CN CN EtO 100 psi unreacted EMM, DIPEA + DIPEA.HCl 14 M1 M2 Scheme 1.12 Flow schematic for the transformation of M1 to M2. EMM (14) and DIPEA are combined in a tee mixer and passed through the monolith at room temperature. The solvent mixture was chosen for maximum solubility of EMM. Only unreacted starting materials were observed by NMR spectroscopy of the reactor output. † The sulfur proportion was inferred from the values measured for carbon, hydrogen and nitrogen, and was consistent with a 1:1.17 ratio of monomer (11) to DVB. 18 Solid-supported flow synthesis of 2-aminopyrimidines Figure 1.13 The monolithic reagents assumed a yellow colour during the cyclisation reaction with EMM (14). The uneven distribution of this colour in the initial stages of the reaction indicates some degree of channeling within the structure. was injected into a monolith. The quantity of EMM remaining in the output stream was measured, indicating that 3 mmol of 14 had reacted with the monolithic reagent (2.0 mmol/g for a monolith weighing approximately 1.5 g). Elemental microanalysis of samples taken from the resulting monolith M2 at the ends, from immediately beneath the edges, and from the core reproducibly gave similar results, indicating a consistent reaction throughout the monolith. 1.2.4 Oxidation of monolith M2 Unlike the methylthiol moiety,34,37 the benzyl sulfur linkage has been reported to be unreactive to substitution without first performing an oxidation of the sulfur atom.39 meta- Chloroperoxybenzoic acid (m-CPBA) is a oxidant which is highly soluble in organic solvents and which has precedence for this oxidation (Scheme 1.14).38 NC H2N N S N m-CPBA CH2Cl2 25 °C, 18 h NC H2N N S N O O 15 16 Scheme 1.14 Solution-phase oxidation using m-CPBA. In a mixed-phase system, oxidation of monolith M2 was performed at room temperature using an excess of m-CPBA to encourage complete oxidation of all the sulfur atoms (Scheme 1.15). The progress of the reaction could be observed in the infrared spectrum, with new strong peaks at 1038 cm−1, 1127 cm−1 and 1325 cm−1 suggesting the presence of 1.2. Development of Chemistry 19 both sulfoxide and sulfone groups. This was puzzling given that an excess of the reagent had been used. However, the exact composition and the effects of this could only be calculated after the nucleophilic release step had been performed. NC H2N N S N m-CPBA CH2Cl2 25 °C, 18 h NC H2N N S N O O NC H2N N S N O M2 M3a M3b Scheme 1.15 Oxidation of monolith M2. This “safety-catch” activation represents an important advantage of the polymer-supported technique, because reactions with other nucleophiles could be performed to access further derivatives of the supported heterocyclic intermediate whilst retaining it on the solid support. The use of latent chemical functionality in this way can provide an advantage by reducing the number of steps needed to reach a complex target. For the synthesis of the 2-aminopyrimidines, no protecting group would be required for the sulfur linkage, so the reactivity of the heterocycle could be exploited without impacting the step economy70 of the overall process. 1.2.5 Release of 2-aminopyrimidine product Following activation, the oxidised thioether could be displaced using an amine nucleophile such as morpholine (17) to provide the desired 2-aminopyrimidine (18). Initial experimen- tation revealed that long residence times were required for a complete reaction even at 100 ◦C. Therefore, we decided to investigate the use of a stop–flow technique that would increase the residence time without requiring a lower flow rate than could be achieved reliably using the pumps. Once approximately one column volume worth of amine solution had been infused, the flow was stopped for a period of time whilst the column was kept at an elevated temperature. This cycle was repeated multiple times to achieve a prolonged residence time, whilst maintaining a high local concentration of the nucleophilic reagent. Although there was no flow of solution within the column, mixing would continue by diffusion allowing the reaction to continue during these periods. 20 Solid-supported flow synthesis of 2-aminopyrimidines SN N H2N O NN N H2N 1,4-dioxane 80 °C, 8 h HN O NC NC O SN N H2N O O NC + M3a M3b 18 35 % Scheme 1.16 Reaction of monolith M3 with morpholine to afford the heterocyclic product 18. The yield is calculated based on the measured loading of isothiourea monolith M1. Following concentration of the output, the pyrimidine product 18 was obtained following a single chromatographic purification. Therefore, a large proportion of the experimental effort was spent operating the flow reactor, and very little reagent and intermediate handling was necessary to obtain the product. We envisaged that an automated system could perform many parts of this synthesis with minimal manual intervention. 1.3. Automation 21 1.3 Automation 1.3.1 Advantages of automation The “catch–react–release” method described is essentially a serial batch procedure, and so the throughput of material is restricted. The size of each batch is limited by the amount of material loaded onto the monolith, and under the thermal polymerisation conditions frequently employed there is currently an upper limit to the size of monolith that can be prepared with a favourable morphology.61 Furthermore, the monoliths were not reusable and to avoid contamination we used new columns for each reaction, so a large number of them had to be prepared. Using the column heater modules for a Vapourtec R2+/R4 reactor unit, we were able to produce batches of four monoliths at a time (Figure 1.17), which was sufficient for the pace of work. However, the protocol for generating the monoliths involved heating the polymerisation mixture for 20 hours. When this procedure was performed manually, preparation of the polymerisation mixture had to be started at about 3 pm to ensure that the monoliths would be ready and the heater could be switched off early the next morning. By implementing a timed shut-off, the preparation could be done at any point during the day, which was more practical. Of course, this simple control task could be achieved with an off-the-shelf plug timer, but a more complete solution was required for the later synthesis steps to enable a programmed sequence of operations to be carried out. In these cases, the software available for control of a b c Figure 1.17 (a) Some of the different sizes of glass column available. From left to right: 3 mm ID × 100 mm length, 6.6 mm ID × 50 mm length, 6.6 mm ID × 100 mm length, 10 mm ID × 100 mm length. (b) Heating jackets installed on a Vapourtec R4 heating unit. (c) Monoliths being prepared in parallel. 22 Solid-supported flow synthesis of 2-aminopyrimidines the R2+/R4 unit was also unsuitable, because it was not possible to add new logic beyond the prescribed applications, and because in some cases we wanted to incorporate additional devices to be controlled simultaneously. Therefore, custom computer software had to be created to control these experiments; the 20 hour heating programme will serve as an introductory example to explain how the solution was created and used. Vapourtec kindly provided details of how their reactors can be remotely controlled. The reactor connects to a PC using an RS-232 serial cable, and then the communication is carried out using a text language (Table 1.18). Communication though a serial port in this way is well within the capabilities of any modern programming language. The Python programming language was chosen because it is quick and easy to read and write, and commonly used for scientific software.71 A sequence to set up the reactor, wait for 20 hours and then turn off the system is shown in Listing 1.19. 1.3. Automation 23 Table 1.18 Typical commands and responses for the Vapourtec R2+/R4 unit. Action Command Response Switch power on "" "OK" Switch power off "" "OK" Set Temperature "  0 100" "OK" Listing 1.19 This programme sets all four heaters to 90 ◦C, runs for 20 hours and then switches off the R4. Note that no data is collected during the experiment and there is no facility for checking that the commands have been received. The \r\n (carriage-return and new line) characters are used to terminate commands. Please note that the serial commands have been redacted in the PDF version of this document. # Python includes modules for serial communication and timing import serial import time # Set up a connection to the R2 connection = serial.Serial( "COM1", baudrate = 19200, timeout = 1 ) # Set all four heaters to 90 degrees for i in range(4): connection.write("  {:d} 90\r\n".format(i)) time.sleep(0.1) # Turn on the heaters connection.write("\r\n") # Wait for 20 hours time.sleep(20 * 60 * 60) # Switch off the heaters connection.write("\r\n") 24 Solid-supported flow synthesis of 2-aminopyrimidines 1.3.2 Solvent switching and stop-flow A second application of automation was that when working with a monolithic reactor, it is important to make gradual solvent switches, much like must be done when changing the solvent within a chromatographic column. This is because a sudden change in solvent composition can block the pores or cause the structure to collapse. Another application was the stop–flow protocol which otherwise required many carefully timed manual operations. Automation of the solvent switching protocol (Listing 1.20), and the stop-flow protocol (Listing B.1 in Appendix B) created a much more efficient working regime, where the bulk of these tasks could be managed by a computer. Initially, we encountered a problem in some cases relating to the under-pressure shut off feature which is hardwired into the Vapourtec machine. Once the reactor is running and the pressure has built up to over 3 bar, if the pressure then drops below this level then the pumps and heaters are automatically switched off after a period of time and the machine reports an error.72 In a normal flow chemistry procedure this is very useful, because a pressure drop may indicate a leak or a sudden rupture in one of the flow tubes. However, when pumping a solution slowly through a monolith this system could be triggered during Listing 1.20 Example of a programme to change the solvent mixture gradually from 100 % solvent A (orange channel) to 100 % solvent B (purple channel) in four increments. This programme makes use of an abstraction object (vapourtec.R2R4) that handles the RS-232 communication (see Appendix A). import transport, vapourtec R2 = vapourtec.R2R4(transport.serial("COM1")) flow_rate = 200 # Total flow rate steps = 4 # Number of increments to make # Calculate change in flow rate for each step step = flow_rate / steps for i in range(steps): change = (i + 1) * step # Adjust the flow rate of both pumps R2.flowrate("orange", flow_rate - change) R2.flowrate("purple", change) # Run each solvent composition for 20 min R2.wait(mins = 20) 1.3. Automation 25 normal operation. Furthermore, if the structure of the monolith was not perfect then large pressure fluctuations could sometimes occur, triggering the safety shut off. Most crucially, it was also triggered when the pump was switched off but the heaters were still running, during the “stop” phase of a stop–flow sequence. The pressure gradually fell to nothing after the pumps were stopped, having the undesired effect that the heaters were switched off as well. To circumvent this problem, the programme performed frequent checks on the status of the machine during each waiting period, as part of the R2.wait() function. If it had stopped due to an under-pressure then the reactor was started again allowing the sequence to continue uninterrupted. 1.3.3 Multi-step processing The combination of solid-phase synthesis and automation means that multiple synthetic steps can be programmed to take place in a sequence, in much the same way as a solid-phase peptide synthesiser is used. The valves on the R2+/R4 reactor can be used to select reagents and solvents for different phases of the synthesis. EtOH DIPEA / MeCN MeCN Reagent / MeCN 100 psi orange waste purple 1 1 5 5 Figure 1.21 Reactor configuration for sequential washing and loading of four monoliths. A multi-position valve was added to take advantage of the four heating manifolds available (Figure 1.21). During stop-flow sequences the reagents could be loaded onto one monolith whilst the others were heating, allowing multiple reactions to be performed simultaneously. All four could be washed and then the cyclisation reaction performed on each in turn. Listing B.2 (Appendix B) shows a programme to wash four new monoliths and then react each in turn with a solution of a reagent. A similar system could be used for performing the nucleophilic release step with four different amines (Figure 1.22, Listing B.3 in Appendix B). In this case the valve was 26 Solid-supported flow synthesis of 2-aminopyrimidines connected to four reservoirs containing the amines, and four collection vessels. A separate wash line allowed the dead volumes to be cleaned out between switching the reagents. NMR measurement of the collected materials confirmed that there was no cross-contamination between the amine reagents. Dioxane Dioxane m-CPBA / DCM DCM Amine 1 Amine 2 Amine 3 Amine 4 100 psi 100 psi 100 psi 100 psiorange load inject waste collect 1 collect 2 collect 3 collect 4purple 1 5 5 1 Figure 1.22 Reactor configuration for nucleophilic displacement from four monoliths with four amines. The additional valve is used to enable thorough rinsing of the system to avoid cross-contamination. Since the two multi-position valves were part of the same unit, Amine 1 was infused into column 1, Amine 2 into column 2, and so on. Multiplexing reactions in this way improves the efficiency of a single piece of apparatus. The complex switching schedule required makes the computer control an important addition. After programming the appropriate sequence of operations, the chemist can be confident that it will be carried out correctly without having to (for example) set lots of stopwatches for himself, or worrying about getting one step wrong. Having developed these automation methods, they were applied to the preparation of analogues of a precursor to the kinase inhibitor imatinib. 1.4. Application to the preparation to derivatives of imatinib amine 27 1.4 Application to the preparation to derivatives of ima- tinib amine Imatinib (1) is a drug marketed by Novartis AG for the treatment of chronic myeloid leukaemia and gastrointestinal stromal tumours.33,73–75 It is marketed in the USA (as Gleevec) and in Europe, Australasia and Latin America (as Glivec), in both cases as its mesylate salt. We envisaged that the polymer-supported methodology for the synthesis of 2-aminopyrimidines could be applied to the synthesis of compound 24 (Scheme 1.23), an important precursor to imatinib. 1.4.1 Reported syntheses of imatinib A number of routes for the synthesis of imatinib have been reported. Many have a number of similarities to the initial discovery route patented by Zimmermann and co-workers.33,73 N N N H N X O N H N N N N H N HN O N N Cl NH2 H2N H N NO2 N O NMe2 NO3 H2N NO2 N O 1. NH2CN, HCl, i-PrOH 80 °C, 3 h 2. HNO3 60 °C 81% (2 steps) NaOH DMF, H2O reflux, 8 h 73% (2 steps) X = NO2 X = NH2 MeO OMe NMe2 neat reflux, 3.5 h Cl + N N N H N HN O Cl Raney Ni N2H4.H2O MeOH 35 °C, 5.5 h 81% neat 130 °C, 1.5 h 96 % 1. K2CO3, THF 20 °C, 1 h 2. H2O 20 °C, 80 min 98% (2 steps) 19 20 21b 22 23 24, 25, 26 1 27 Scheme 1.23 Szczepek’s route to imatinib,76 involving condenstion of enaminone 21b with guanidine 23. This route is similar to the original discovery route.33,73 28 Solid-supported flow synthesis of 2-aminopyrimidines The key 2-aminopyrimidine is constructed by a condensation reaction between an enone (21b) and a guanidine (23) (Scheme 1.23).76–81 One patented route82 involved the Negishi coupling of 3-pyridyl zinc bromide with 2,4-dichloropyrimidine which avoided the high- temperature enone condensation. The key amide bond is often formed by acylation of the aniline revealed by reduc- tion of a nitro group such as that present in compound 24. A number of methods for amide coupling have been reported, including aluminium trichloride mediated reactions with carboxylic acids or esters (Scheme 1.24, Route A),77 as well as using common peptide coupling reagents.83,84 The combination of an acyl chloride with an aniline (Scheme 1.24, Route B)76,77,79,80,85 is a particularly high-yielding method. Construc- tion of the 2-aminopyrimidine centre has also been reported using cross-coupling methods such as by a Buchwald coupling (Scheme 1.25)36,77,86 or by Ullmann coupling.87 N N N H N NH2 N N N H N HN O N N HO O N N AlCl3 (2.5 equiv.) PhMe / MeCN 40 °C, 8 h, 55% Cl O N N Pyridine 50 °C, 4.5 h, 70% Route A Route B 25 28 29 1 Scheme 1.24 Formation of the amide bond from aniline 25. Finally, the synthesis of imatinib has also been performed using some advanced synthe- sis techniques. Our own group developed a flow synthesis procedure36 involving solid- supported reagents and scavengers, and a catch-and-release method for the preparation of the aryl bromide cross-coupling partner 31. This compound was obtained in high purity with no manual purification. Leonetti and co-workers reported a solid-supported synthesis of imatinib (Scheme 1.26),88 although this method required a double protection of the guanidine moiety as well as a number of expensive or toxic reagents such as BEMP and HgCl2 to force the reactions. 1.4. Application to the preparation to derivatives of imatinib amine 29 N N N H N HN O N N N N N NH2 Br HN O N N conditions + 30 31 1 Scheme 1.25 Synthesis of imatinib by cross-coupling methods. Conditions: Pd2(dba)3 · CHCl3, NaOtBu, rac-BINAP, xylene, reflux, 5 h, 72 % after reverse phase HPLC; 77 BrettPhos precatalyst (2 mol %), BrettPhos (2 mol %), K2CO3, t-BuOH, 110 ◦C, 6 h, 84 %;86 BrettPhos precatalyst (2.5 mol %), NaOt-Bu, 2:1 1,4-dioxane / t-BuOH, 150 ◦C, 30 min (continuous flow), 69 % after column chromatography.36 O H O2N NH2 Merrifield Resin O2N NH 1. Ti(OiPr)4, NEt3, THF 25 °C, 18 h 2. NaBH(OAc)3 DCM, 25 °C, 4 h 80% (2 steps) H2N N O N N N H N O N N AllocN AllocHN SMe AllocN AllocHN HgCl2, NEt3, DMF 0 °C, 10 min then 80 °C (MW), 5 min + N H N O N N NH H2N 1. Enaminone PhNO2, BEMP 120 °C (MW) 50 min 2. TFA, CH2Cl2 25 °C, 1 h 90% (2 steps) imatinib Pd(PPh3)4 PhSiH3 CH2Cl2 25 °C, 1 h 98% (2 steps) 32 22 33 3435 36 (37) Scheme 1.26 Solid-phase synthesis of imatinib. The steps were generally high-yielding but some unattractive reagents were required. 30 Solid-supported flow synthesis of 2-aminopyrimidines 1.4.2 Monolith-assisted route to imatinib A collection of enaminones (21) were produced by treatment of an acetylated aryl com- pound with DMF-DMA (20), following Szczepek’s protocol.76 These were reacted with monolith M1 using the flow procedure described earlier in this chapter to afford the different thiopyrimidine monoliths M4. By heating the monolith to 80 ◦C during the heterocycle formation reaction, this transformation proceeded to completion as indicated by elemental analysis. From the oxidised monoliths M5, analogues 38 were prepared (Table 1.28).29 These compounds were isolated in yields from 29 – 72 % (based on a 4.2 mmol loading of monolith M1) over the three steps with only a single chromatographic purification. This represents a powerful technique for the preparation of a number of derivatives of a compound of interest, without requiring a large number of manual purification operations. In order to transform compound 24 into imatinib, we chose to use a coupling with an acyl chloride.87 Before this could be carried out, it was necessary to perform a reduction of the nitro group of 24. A number of different conditions have been reported to be effective for this procedure, including Pd/C and H2, 73 SnCl2, 78 hydrazine and FeCl3, 87 hydrazine and Raney Nickel,76 and sodium borohydride with CoCl2. 76 We chose to employ Pd/C catalysis using an H-Cube reactor (Scheme 1.27), which performs heterogeneous catalysis using hydrogen generated in-situ in an electrolytic cell. No toxic metals like SnCl2 are involved and the use of a heterogeneous catalyst reduces the purification requirement. The resulting aniline (25) was treated with an acyl chloride, and then an SN2 reaction afforded imatinib (1) after purification by column chromatography. N N N H N HN O N N N N N H N NO2 O Cl H2 (15 bar) Pd/C CH2Cl2 H-Cube 40 °C 73% HN N Cl 2. NEt3 (1.15 equiv) THF, 0 °C, 3 h, 57% MeCN, 150 °C (MW) 2 h, 53% N N N H N NH2 1. 24 25 1 Scheme 1.27 Synthesis of imatinib (1) from precursor 24. Yields are unoptimised. 1.4. Application to the preparation to derivatives of imatinib amine 31 Table 1.28 Preparation of 2-aminopyrimidine derivatives 38. SH2N NH2Cl SN N Ar Ar O NMe2 DIPEA EtOH, 80 °C SN N Ar m-CPBA CH2Cl2 O ONR 1R2N N Ar R1R2NH 1,4-dioxane 90−120 °C a Ar = 4-pyridinyl b Ar = 3-pyridinyl c Ar = C6H5 d Ar = 4-(CF3)C6H4 M1 21 M4a–d M5a–d24, 38 Entry Monolith Amine Product Product structure Yielda 1 M5a NH O 38a N N N NO 72 % 2 M5a NH2 OMe 38b N NN H N OMe 29 % 3 M5a NH2 38c N N N H N 50 % 4 M5b NH O 38d N NN N O 71 % 5 M5b NH2 NO2 24 N NN H NO2 N 48 % 6 M5c NH O 38e N NN O 40 % 7 M5d NH O 38f N NN O CF3 62 % a Isolated yield over all three steps, after purification by flash column chromatography, based on an assumed 4.2 mmol loading of monolith M1. 32 Solid-supported flow synthesis of 2-aminopyrimidines 1.5 Conclusion In this chapter an automated, multi-step synthesis for a focused array of substituted 2- aminopyrimidines was described. The method involved the generation of a polymer- supported pyrimidine which was then released to afford an advanced intermediate. A significant advantage of this procedure is that it avoids problems of low solubility which are commonly encountered when working with heterocyclic compounds. Indeed, attempts to directly polymerise intermediate 15 were unsuccessful primarily due to its low solubility (Scheme 1.29). S S N N NC H2N N N NC H2N Polymerisation Conditions 15 M2 Scheme 1.29 Attempts to prepare a monolith using cyclised monomer 15 were unsuc- cessful, primarily due to its poor solubility. Solid phase synthesis is a powerful technique, which can eliminate the need for manual purification methods and thus is very useful for library synthesis. In this case, the oxidative “safety-catch” could enable further derivatisation without the need for protecting groups. However, solid-phase synthesis is strongly associated with combinatorial chemistry and so has generally been overlooked since the latter has fallen out of fashion. Nevertheless, when combined with programmed automation attractive synthetic pathways can be harnessed without the need for specialised combinatorial equipment. The introduction of automation techniques allowed a number of compounds to be rapidly prepared by what would otherwise be a relatively labour-intensive process. The potential applications of automation within synthesis are widespread, and in the next chapter a more generalised system for controlling laboratory devices such as the R2+/R4 reactor is described. Chapter 2 New control and monitoring software for flow chemistry systems 33 34 New control and monitoring software for flow chemistry systems 2.1 Introduction As continuous processing techniques such as flow chemistry become more commonplace in modern laboratories, there is an increasing demand to expand the capabilities of laboratory devices. For instance, the ability to share information between different devices enables seamless co-ordination of an experimental protocol to be achieved. We may wish for the sensor reading from a spectroscopic detector to determine the flow rate of a pump,89 or to trigger a multi-position valve to collect samples at regular intervals throughout the procedure (Figure 2.1). One particular challenge in this regard is communication with different equipment made by different manufacturers. heaterdetector controlled pump supported reagent fractionsdetector Control Figure 2.1 Heterogeneous reagents produce additional dispersion of a reagent slug within the flow system. By connecting a detector and a pump, addition of a reagent stream after the column could be performed at a varying flow rate to match the immediate concentration of the substrate. The same control computer could also interact with other devices, such as valves or fraction collectors, in an integrated fashion. There have been a number of examples of integrated flow systems reported recently, such as for multi-step synthesis,14,89,90 combined synthesis and purification,91 or new work-up technologies,92 which confirms that this is an important aspect of laboratory flow chemistry. Although manufacturers generally provide appropriate control software for use with their particular devices, these are often limited to the applications that the manufacturer had envisaged, and to other devices sold by the same manufacturer. For commercial reasons, the control software is invariably provided in a closed format that cannot be extended to implement control of additional hardware. Fortunately, most manufacturers are willing to share control commands for their products; however, extensive integration work is often necessary in order to connect different devices such that data can be passed between them. In a manufacturing environment, process control systems tend to be custom designed (involving a significant financial and time investment) for each individual setup; the cost 2.1. Introduction 35 and specific nature of the resulting system make this unsuitable for a research laboratory, where a particular reactor configuration may only exist for a few days or weeks. Ideally, a chemist should be able to define a sequence of operations to carry out a control protocol for any new process with minimal up-front effort: it ought to be possible to prototype the control system for an experiment in a few hours at most. In this way, more time can be spent on the engineering and chemistry challenges inherent in the synthesis process, rather than the logistics of control systems and communications interfaces. Furthermore, the control software needs to be sufficiently flexible to enable different pieces of equipment to be swapped in and out of the integrated system without having to make significant changes to the automation protocol. Whilst an industrial process might be designed for hardware that will be dedicated to that system, a process in a research laboratory would have to use whatever devices are available. Flow reactor devices tend to be modular and functionally interchangeable, so they ought to be interchangeable in the control algorithm as well, allowing maximum reuse of programmes from one experiment to another. 2.1.1 Existing control software A large number of software projects can be identified that have been developed to enable laboratory automation. The majority are aimed at running analytical devices, since these are now invariably automated with data stored directly in electronic lab notebooks (ELN,93 such as the IDBS E-Workbook94 as used within our own research group95) or in laboratory information management systems (LIMS)96 such as that sold by Thermo Scientific.97 A significant minority are designed for device automation. Clarity,98 developed between the Evolutionary Biology and Systems Biology departments at Harvard University, is an automation management programme written using the C Sharp (C#) programming language to interface with laboratory robots, and includes a scheduler for executing sequences of operations, in particular allowing each instrument to be involved in many such protocols simultaneously. It also has an interface for starting and monitoring protocols, and an alerting system for monitoring the computer that is running them. Importantly, it has the ability to resume a protocol from a stored log file after a system crash. However, the control protocols are stored simply as lists of method calls, allowing very limited decision making abilities. 36 New control and monitoring software for flow chemistry systems The aim of the project was to provide an alternative to expensive and proprietary commercial integration and task-scheduling solutions such as those provided by Peak Analysis and Automation Ltd.,99 PerkinElmer Inc.100 and infoteam Software AG.101 These systems are generally not suitable for academic use as they tend to be sold on a basis where a consultant from the company will design and implement the controlling software for the particular process that needs to be automated. Furthermore, Clarity and all of the products produced by these companies are primarily aimed at the biochemical sector. Clarity includes interfaces for an incubator, a plate handler and a transfer station, and the case studies described for the commercial products revolve around plate handling and reading. The SiLA (Standardization in Lab Automation) consortium,102 a prominent laboratory automation standards body, has published specifications for interfaces with different classes of device.103 These specifications have been primarily developed to address the require- ments for in vitro screening tasks, and this is confirmed by the operations defined in the specifications (for example: Move Plate, Dispense Liquid Volume, Read Plate, and so on). However, the idea of a specification is very important, because if similar devices have the same interface, then the physical unit can be exchanged without requiring extensive modifications to the control protocol. Flow chemistry applications offer different challenges — most importantly, the control of reactors based on the current state of other devices and instruments. Each process will tend to have a dedicated set of physically and fluidically integrated apparatus for the duration of its operation, so there is no need for the software to allow each device to participate in multiple experiments. Rather, its focus should be on responding to incoming information correctly and rapidly. Flow reactors are modular devices which are frequently repurposed for many different experiments, so each control protocol should represent a low time investment. This is not necessarily possible with the currently available software. Therefore, a new open access and freely-available control solution would be a valuable addition to the flow chemistry toolbox. Some reported projects89,104,105 have designed automation protocols using LabVIEW,106 which is a visual programming tool and control platform developed by National Instruments Corporation. Notably, LabVIEW has also been used to create the control software for commercially available Uniqsis107 and Syrris108 devices. A similar tool is OpenAPC109 which offers a similar visual process development interface, for which new software components can be created using the C++ programming language. Another popular programming language is MATLAB,110 which works in a more conventional textual 2.1. Introduction 37 manner, and has been employed for some automation projects.111 A number of manufacturers provide LabVIEW or MATLAB interfaces for their instruments, and so for simple tasks, the tools provided by these or other programming languages can allow very rapid development. However, without a specification or standard protocol scheduling system, the software for each new flow chemistry process must be created from scratch. This has largely been the case with previous projects:89,92,112–114 for each new project the researchers created a control programme to send the raw commands to the particular machines integrated into the synthesis process. Frequently, the control system is not published with the work; but even if it were, it may be useless without having exactly the same set of equipment (with exactly matching firmware versions). A more powerful solution would be to abstract each machine to its fundamental capabilities, and work to a specification enabling similar reactors and devices to be exchanged easily; each new protocol would be focused on its specific purpose rather than on the implementa- tion of particular instruments. Furthermore, if common requirements such as recording all of the data collected during a protocol and providing an interface to monitor this data in real time are performed automatically, then researchers developing a new process will be free to concentrate on other tasks. Control platforms such as LabVIEW and MATLAB offer a promising ecosystem for running lab automation, but the key disadvantage is the closed-source and proprietary nature of their distribution and data formats. To overcome these disadvantages we selected the Python programming language.115 Its open-source nature encourages collaboration,116 and enables technology to be transferred between institutions without the large initial set-up costs typically involved with commercial packages. There are a number of libraries created for Python which are specifically targeted at scientific and chemical applications.71,117 In common with most programming languages, libraries for communication using a large number of standard protocols are also available, enabling programmes written using Python to communicate with laboratory instruments. The goal of this project was to develop an automation platform that implemented the communication protocols for standard devices, and which provided the ability to define logic behind the commands that need to be sent, and to schedule them to occur at the correct time. 38 New control and monitoring software for flow chemistry systems 2.1.2 Flow chemistry equipment Basic flow chemistry equipment (see page 3) consists of pumps, mixers, switching valves, heaters / coolers and sensors. Broadly speaking, any controllable reactor component will have two modifiable parameters: power (on / off) and some target value (for example: tem- perature or speed), in addition to readouts of current sensor values. Recent developments in reactor technology such as electrochemical,118 photochemical119,120 and microwave121 reactors fall broadly within the same categories, but may of course require different pa- rameters (such as voltage, wavelength or power instead of temperature). Sensors, such as pressure sensors, pH sensors or thermocouples may not have any controllable parameters, instead only providing readings. As mentioned earlier, work has been done already towards defining standards for interfaces for interaction with laboratory instruments: the SiLA specification103 defines interfaces for syringe pumps, liquid pumps, valves, heating/cooling units, as well as generic devices with analogue or digital parameters and outputs. These concepts could be adapted to create a new interface which is initially aimed at comtrolling flow chemistry equipment but which could later be made compatible with the SiLA (or other) standards (Table 2.2). Table 2.2 Parameters for typical flow chemistry equipment. Type Parametera Sensorb HPLC Pump Target flow rate Actual flow rate Syringe Pump Target volume Actual volume Valve Position n/a Heater / Cooler Target Actual value Sensor n/a Sensor value a Generally writeable. b Generally read-only parameters. 2.1.3 Interaction with equipment There is a range of communication mechanisms for laboratory devices, some more common than others. The simplest devices communicate with analogue voltage signals and electronic switches (referred to as contact closures). To use these signals and switches we must physically connect one wire from a device to the controller for each parameter of interest. More recently developed devices use “combined” interfaces such as USB, Ethernet or 2.1. Introduction 39 start / stop power ow rate pressure reading pump head size error messages ... 3V % solvent B 0V Figure 2.3 Analogue connectivity, such as with a hypothetical binary HPLC pump (left) might include a switch to start or stop the pump, and a voltage output to read the solvent composition. For each value to be read or controlled, (for example, the flow rate) an addi- tional physical wire is required. Newer methods such as RS-232 allow many commands and parameters to be conveyed using the same physical connection. RS-232. These are digital interfaces that have larger speeds or bandwidths,* meaning that only one wire is required for each device (Figure 2.3). RS-232 is one of the more common interfaces for laboratory devices, and it works by sending text-based messages along a serial cable. For example, to interrogate a valve manufactured by Valco Instruments Inc.,122 a text command NP (Number of Ports) can be sent, to which the valve responds with 10: the number of available ports. A large majority of instruments support RS-232 communication, and there are many options for adaptors which can transmit RS-232 messages within other protocols such as USB or Ethernet. In these cases, the adaptor hardware is used along with software that creates a “virtual serial port” on the control computer. USB is the successor to RS-232, and has a much higher data rate (typically 4000 × faster).† However, there is limitation in the length of the cables that can be used, and often USB devices require their own software or drivers to perform binary communications, rather than simply using text-based communication. Ethernet is another transport mechanism, which uses a protocol called TCP/IP. It has a long physical range, and is typically 1000 to 10 000 × faster than RS-232.‡ The advantage of using Ethernet communication is that it is used extensively for computer networking, so this form of communication can take advantage of existing building-wide cabling systems. There are some other less commonly encountered protocols, including Modbus and I2C. Modbus can operate over different transport mechanisms such as serial, USB or Ethernet; instead of sending messages, it defines commands to interact with parameters (called * How fast or how much data can be transmitted at the same time. † 480 Mbps for USB 2.0, compared with a maximum of 115 kbps for typical RS-232 devices. To minimise interference, few RS-232 devices operate over 19 kbps. ‡ Modern consumer ethernet equipment operates at 100 to 1000 Mbps. 40 New control and monitoring software for flow chemistry systems registers) stored in the device. I2C is a protocol which operates across simple wires, and is often used internally within instruments to control components such as switching valves. It is also possible to use some of these protocols over a wireless transport layer such as wireless Ethernet; this has the potential to reduce the amount of clutter in the fume cabinet and laboratory. However, when using wireless communication, issues of connection dropouts, interference and security must be considered, introducing significant unwanted complexity to the system. 2.1.4 Control computers Previous work in the laboratory has employed laptop computers outside the fumehood, or desktop computers standing inside the fumehood. These take up space, can be damaged easily by solvents or corrosive substances (requiring expensive replacements) and tend to require a large number of wires to be routed around the fumehood. Of course, without wireless connectivity many of the wires are necessary; nevertheless, the use of smaller and lower-powered computers can reduce clutter and cost, save space and energy, and benefit from increased portability. Additionally, the introduction of graphical interfaces that can be accessed using a portable device such as a tablet computer, rather than being confined to a screen adjacent to the equipment, can provide the chemist with greater flexibility and freedom within the laboratory (Figure 2.4).123 instrument instrument instrument portable device control computer Figure 2.4 The use of wired connections between the instruments and the computer running the control programme ensures reliable communications throughout an experiment. By allowing a user interface to be accessed on portable devices, the operator can work with greater flexibility. Fortunately, the control computer for these systems does not necessarily have to be a standard laptop or desktop machine. Provided that software is written to suit them, there are many compact systems now available (Figure 2.5). Platforms such as Arduino132 and Microsoft .NET Gadgeteer133 contain a small, very low power, programmable microchip 2.1. Introduction 41 a b c d Figure 2.5 (a) Arduino Uno,128 (b) Microsoft .NET Gadgeteer (shown with attached button module),129 (c) Raspberry Pi (model B),130 (d) Intel Galileo board.131 with an array of digital and analogue inputs and outputs, as well as plug-in modules for interacting with motors, sensors, or other devices. Generally these have a small amount of memory for storage, and can run a single looping programme. With their low power consumption they are ideal for developing integrated controllers for new devices. A more powerful device is likely to be needed for controlling multiple machines using easy-to- write programs. Raspberry Pi28 is a small computer with an 800 MHz processor running a Linux operating system, which can run software written in Python, as well as most other Linux software. As ubiquitous computing becomes more commonplace, more miniaturised computer systems are being made available, including the Intel Galileo134 and Edison135 systems, and the Phidgets Single Board Computer (SBC).136 The more flexible and powerful a system is, generally the more expensive it will be both in terms of cost and energy usage: small computers such as the Raspberry Pi provide the capability for performing advanced applications whilst having only a small power requirement. 42 New control and monitoring software for flow chemistry systems 2.2 Implementation As part of this project, a new software tool for computerised control of flow chemistry platforms was developed, under the name Octopus.137 This has a similar structure to that of the Clarity project mentioned earlier,98 but was designed specifically for use with flow chemistry systems. Each device is controlled by a virtual instrument, which is a software component that sends commands and records all of the data produced by the device (Figure 2.6). The virtual instruments are controlled by a scheduler, which executes a control protocol and instructs the virtual instruments to perform the required actions at the correct times. The scheduler and virtual instruments can run on a regular desktop computer, or a miniature computer such as the Raspberry Pi. Multiple protocols can be run independently or on different computers; they all commu- nicate with a user interface server which delivers the experimental data in numerical or graphical formats to client devices using a web site interface. These client devices can be desktop computers or portable devices connected via a wireless network or over the internet. The remainder of this chapter discusses some of the key features of the design. user interface protocol scheduler alerts virtual instrument data storage physical instrument virtual instrument data storage physical instrument additional instruments physical instrument client client client Figure 2.6 The control software system is structured so that each instrument is controlled by a virtual instrument within the software. Experimental protocols are run by a scheduler, which can interface with any number of virtual instrument objects. The state of the instruments and the experiment can be displayed on a web interface which can be accessed by multiple users simultaneously; the user interface can be provided by the same computer or a different computer to the one running the protocol. Events within the protocol can trigger alerts such as emails or SMS messages to be sent to the operator. 2.2. Implementation 43 2.2.1 Communication Concurrency and asynchronous operation A fundamental requirement for the control software was for it to be able to communicate with several different machines and devices simultaneously. Computers perform multi- tasking by rapidly switching between running processes; one simple method for achieving this in software is by using an event-based programming model. Under this system, execution of different parts of the programme is triggered by external messages or events. Commonly, new events wait in a queue and only one event is processed at once — ideally, the part of the programme that processes an event should be very efficient so that new events are dealt with as quickly as they arrive. The event-based model is considered to be conceptually easier and more efficient to run than alternative methods for multi- tasking,138 such as the creation of multiple threads (where the operating system manages task-switching) or by programming for the utilisation of multiple processor cores. Since event-driven programmes can be run on a single processor, and can have a relatively low resource requirement, even quite complex programmes can be run on low-powered devices such as the Raspberry Pi. The capacity for a computer programme to perform multiple tasks simultaneously using the same data is called concurrency.139 It turns out that concurrency represents quite a complex problem and so it is important to consider how it will be achieved. In this case, concurrent operation is beneficial because one of the slowest parts of the system is communication with different devices. Typically, it can take around 200 000 times as long to access a file from the hard drive (magnetic storage) as it does to read a value from memory (RAM; solid-state storage). Even when using the fastest interfaces, communicating over a serial cable or over a network is even slower than accessing data from the hard drive, by a factor of up to a hundred.140 Because reading data stored in the RAM is so much faster, a computer can carry out thousands of calculations or instructions in the time that it takes to receive a response from an external device. A programme that waits for responses from each communication before moving on to the next instruction will waste a lot of time doing nothing, whereas a programme made to run in a concurrent manner can perform its communications asynchronously§ and thus be more efficient by doing other things whilst waiting for responses from remote devices. § Without having to wait for them to complete before moving on to the next instruction. 44 New control and monitoring software for flow chemistry systems “OK” “Start R1” R1 “OK” “Start R2” R2 30 ms 30 ms “OK” “Start R1” R1 “OK” “Start R2” R2 30 ms Synchronous communication Asynchronous communication wait... wait... other instructions message response Figure 2.7 Event-based concurrency enables asynchronous communication so that other tasks can be done while waiting for responses from remote devices such as reactors R1 and R2. (30 ms is a typical time for a message to be delivered to a remote device and then for a response to be received). For example, to start two devices simultaneously, messages must be sent along two different cables with as little time separation as possible (Figure 2.7). Whilst it is possible to send two simultaneous messages using a regular programme, one written with concurrency in mind can achieve this and also listen for and handle the responses, essentially allowing a two-way dialogue with each device. In an event-based system, messages are dispatched to the remote devices asynchronously, and then the arrival of each response triggers an event to which the programme responds, to process the incoming data. Twisted141 is an open-source software package designed to enable the development of soft- ware for running networking tasks using events, using the Python programming language. This package was employed to implement the concurrent operation and communication with flow chemistry instruments. Using the Twisted package as a basis, the virtual instru- ments were developed. These define the commands that are to be sent to each type of machine and how to interpret the responses. Implementation of virtual instruments Python is an object-oriented programming language, which means that it is built around the concept of objects (software representations of physical things). Objects contain properties (data) and define methods (actions that can be performed on the object or its data). With this in mind, the interface to each connected machine was implemented as an object (a virtual instrument), which has properties corresponding to the status, parameters, outputs 2.2. Implementation 45 and sensor readings from the physical instrument.¶ For safety reasons, it is important that the physical control panel on the machine is not disabled (even though there is often the option to do so) so that if necessary it can be controlled manually even while it is ostensibly under computer control. As a result, there may be changes to the state of the physical machine which are not caused by commands sent by the virtual instrument. Therefore, it needs to regularly request this information so that its internal representation of the physical instrument is kept up-to-date. This enables the control programme to make decisions based on the actual state of the machine. To create the virtual instruments, a different class is defined for each type of machine that is to be monitored or controlled. A class is a generator which can create an appropriate object to correspond to a particular machine when given a connection through which to communicate with it. The class is defined with all of the parameters appropriate for that type of machine, and methods for requesting the current state of the physical instrument and updating the object’s parameters accordingly (Figure 2.8). Virtual Pump Object Power on Flow rate 100 µL/min Pressure 7 bar Figure 2.8 A pump object is defined with virtual representations of each property of the pump; for example, pressure, flow rate and power. Some are read-only, such as the pressure; others can be changed to control the device. There is likely to be more than one set of commands being sent to each machine whilst the programme is running (Figure 2.9). In the background, the virtual instrument will be periodically updating its state to match the physical one; at the same time, there may be commands defined by the control sequence for a particular experiment. The ability to manage both foreground and background tasks in the same way is a key advantage of a concurrent system. ¶ The virtual instruments perform a similar function to the R2R4 object which was used in Chapter 1 (see Appendix A). 46 New control and monitoring software for flow chemistry systems Co nt ro l S eq ue nc e data commands updates Command Command virtual Command Command virtual physical physical Figure 2.9 The commands defined by the control sequence cause commands to be sent to the physical instrument. At the same time the virtual instrument object requests updates to keep its internal data up-to-date. The internal data is used for calculations and decision making in the control sequence. Command queues Even though a concurrent programme can run multiple tasks in parallel, in general the physical instruments can only process one command at once (this is true for almost all of the machines encountered during this work). However, multiple commands intended for one machine may be generated within a very short time, and so the programme has to wait for each to have been sent and to have received a response before moving on to the next one. Internally, each machine object contains a connection object to represent the communications link with the physical device. The connection object keeps track of M ac hi ne O bj ec t Queue Connection Object response send Commands from control sequence command handler Parameter update commands report result command update parameters command update parameters command physical instrument comm. loop Figure 2.10 The machine object contains a reference to its connection object, which keeps a queue of waiting commands. When a command gets to the front of the queue, the connection sends it to the machine and waits for a response. When the response has been received an appropriate handler is invoked and then the next command is sent. 2.2. Implementation 47 the queue of commands waiting to be sent, and performs the appropriate action when a response is received from each one (Figure 2.10). If no response is received, this could indicate a temporary or permanent loss of the con- nection to the machine. It is not possible to tell whether the message was received or not so the best course of action is to send the message again. For this reason, most devices’ command interfaces are implemented in the form of a state to end up in, rather than an action to perform. For example, a command should be switch valve to position 1 rather than switch valve to the next position. The latter command would lead to different results depending on how many times it is sent. The continuous monitoring of the current state of each instrument gives the software increased confidence that a machine is in the correct state at any point in the sequence. Logging of all of this data throughout an experiment gives the scientist the ability to check that the reaction had run correctly once it is complete. 2.2.2 Data handling The data collected from the connected machines may be needed to observe the current conditions or to measure trends over time. In order to look for trends, the data must be stored for a period of time to allow calculations to be performed on data collected over a particular time frame. As data are received by the virtual instruments, it is stored in two places: in memory within the property associated with the relevant parameter or sensor, and also to a log file on the hard disk. Over a long experiment, the hard disk log files could then become extremely large, or even fragmented if a maximum file size is enforced. This would make reading data from disk a slow and relatively complex task, and so these log files are used only as an archive for later analysis. All calculations involved in the control sequence are evaluated using the in-memory data. Therefore, calculations can be assumed to be instantaneous with respect to the time taken to communicate with an instrument. However, the Python interpreter requires 16 bytes to store a typical numeric value, so a machine with ten properties being updated ten times per second would use around 100 kB per minute. To avoid a large memory requirement the amount of data stored in memory must be limited. 48 New control and monitoring software for flow chemistry systems D at a O bj ec t Low resolution archive High resolution cache Current value Filter New value single value xed size hard drive archive Figure 2.11 Illustration of data storage within a property object. As new data points are added to the property object, it writes them all to a log on the hard drive, and retains the most recent one for quick access to the current value. All of the data from the last minute are stored for use by calculations that need high time resolution. Old data points are removed from this storage as new ones arrive. A long-term archive of filtered data is also kept so that the overall shape of the graph for any time period can be recalled. For real-time graph presentation and for the purposes of most calculations, a time resolution of even one second is probably far greater than necessary. If there are a few prominent features in a particular data set, such as peaks or oscillations, then the precise detail of any additional noise is generally less important. Therefore, it is possible to filter the data points as they are stored to pick out just enough values to represent the overall shape of the data. Each data object keeps a low-resolution archive of all data that has been collected (Fig- ure 2.11). As new data points are added they are filtered to retain only values that represent changes of more than 5 % of the current maximum spread. Key maxima and minima 0 5 10 15 20 20 25 30 35 40 Ab sor ba nc e Time /min Input Filtered Figure 2.12 A section of filtered data from the UV absorption during an HPLC run. The red crosses indicate the key points that were picked out for storage. In this case, the number of data points was reduced by a factor of 100. 2.2. Implementation 49 are also retained to ensure that a reasonably accurate representation of the data is stored (Figure 2.12). In summary, there is a balance between the requirements to respond to changes as soon as they occur, for any calculations to be based on a full set of current data, and for the programs to operate at as high a speed as possible (minimising any calculation or response delays caused by latency between the computer and the instrument, or the software and the hard drive). This is achieved by maintaining current and historic information about each parameter in the computer’s RAM, so that calculations can be performed instantly without waiting to receive new data from the instrument. 2.2.3 Command sequences To run a sequence of commands, a list of steps to be carried out must be defined, similar to the lines of Python code listed in Chapter 1. However, that code was synchronous: nothing else could happen whilst the programme was waiting for one command to be completed.|| Instead, such a sequence can be implemented using the event-based system. Once each command has finished executing,** it triggers an event that starts the next one. The time taken for a step to complete and become finished depends on the action that it is carrying out. Switching on a machine will only take as long as is takes the machine to respond, whereas waiting for 20 hours will take (not surprisingly) 20 hours. Importantly, since the whole system runs concurrently, multiple sequences can be executed simultaneously, and sequences can have sections where certain parts run in parallel. Control of each of these command sequences is performed as a separate task, alongside the background communication between the virtual and physical instruments. The sequence is constructed from individual instructions, implemented as Step objects, which can be linked together. Each one can perform actions or encapsulate logic, or contain child steps. The step objects keep track of their state, such as ready, running, finished or error (Figure 2.13). A control sequence then consists of a connected list of Step objects. By creating Step implementations for various programming constructs (such as decision making and loops), || In Chapter 1, Python’s time.sleep() command was replaced with a function which checked the current time to decide when to stop whilst monitoring the state of the machine. ** For instance, on receiving a response from the instrument. 50 New control and monitoring software for flow chemistry systems St ep O bj ec t State: reset ready running paused nished error Figure 2.13 Each step keeps track of its state; as soon as it becomes “finished” it triggers an event that starts the following step. arbitrarily complex sequences are made possible (Table 2.14). The fact that the system is built using Python means that any of the large selection of libraries available for the language — for chemical intelligence,142 statistical analysis143 and computer vision,144 among others — can be used for data processing or decision making. Table 2.14 Some of the available step types for building sequences. Command Description set Set a variable or parameter on a machine. wait Wait for a specified period of time. wait_until Wait until a specified calculation evaluates to True. loop_until Run a sequence repeatedly until a specified calculation evaluates to True. do_if Run one sequence if a specified calculation evaluates to True, or another if it evaluates to False. log Store a message in the log file. call Execute a function written in Python. Control scripts are written in Python, defining the machines to be used, the appropriate connections and the sequence of operations to carry out. Once all of this is ready, the control sequence is executed (Listing 2.15). The control sequence may make decisions based on data collected from the machines — to do this, calculations (for example an expression such as r.rate < 0) must be evaluated when their value is needed during execution, rather than when they are defined. 2.2.4 Data manipulation Ensuring that expressions are evaluated when they are required, rather than when they are 2.2. Implementation 51 Listing 2.15 A simple automated protocol using a syringe pump and a temperature sensor, controlling an exothermic reaction. Each instrument is set up with an appropriate serial connection, and then the logic is defined. Here the pump will inject reagent at 50 µl/min until the thermocouple registers a temperature rise over 42 ◦C. The pump is then stopped until the temperature drops to below 38 ◦C. The loop continues until the experiment is stopped using the user interface. For full documentation, refer to Reference 137. # Import the necessary libraries from octopus.runtime import * from octopus.manufacturer import wpi, phidgets from octopus.transport.basic import serial from octopus.transport.phidgets import Phidget # Set up the machines that will be used pump = wpi.Aladdin(serial("/dev/ttyS0", baudrate = 9600)) temp_sensor = phidgets.TemperatureSensor(Phidget(12345), inputs = [{ "index": 0, "type": phidgets.ThermocoupleType.K }]) # Create a reference to the relevant sensor temp = temp_sensor.thermocouples[0].temperature # Define a simple sequence: loop until 1 mL has been added main = loop_while(pump.dispensed < 1000, sequence( wait_until(temp < 38), set(pump.rate, 50), # Only pump while the temperature < 42 C wait_until(temp > 42), set(pump.rate, 0) )) # Start the experiment run(main) defined, is an important consideration when making an asynchronous programme using an imperative language such as Python.145 In Python, the actions of operators like +, * and < can be redefined so that rather than returning the current value, they return an Expression object which has the ability to compute the value when required. The expression object acts like a “magic” variable that keeps itself up-to-date based on the components of the contained expression (Listing 2.16). Similarly to the properties of a virtual instrument (Section 2.2.1), an expression needs to be able to provide trends of its value over a particular time period (because it needs to behave exactly like a variable). However, there is no need for this data to be stored within the expression object because it can easily be constructed from the component variables. 52 New control and monitoring software for flow chemistry systems Listing 2.16 After defining an expression, for example temp < 38, the result can be interrogated at any point, and will depend on the current value of the temp variable. # Define an expression >>> expr = (temp < 38) # The value of the expression depends on the value of the variable >>> temp >>> expr # Later on... >>> temp >>> expr >>> expr.value False Whenever a trend is accessed on an expression, the trends from the component variables over the same time period are obtained and the expression is evaluated at each time point to return the result. This is achieved using the NumPy library146 which can perform efficient arithmetic over arrays of data. For this to be achieved, the data of each trend has to be interpolated so that there are data points at regular time intervals. The NumPy library has a method that can map a data series between different x-values (reversing the data-filtering process in order to run a calculation: Figure 2.17). 0 1 2 3 4 5 0 2 4 6 8 10 12 14 Va lue Time A B A + B Figure 2.17 Arithmetic with interpolated values. A and B are data series (large crosses indicate stored values). To add them, they are interpolated at regular intervals (small dots) and the result is calculated at each point. 2.2. Implementation 53 2.2.5 Pausing experiments Whilst running flow chemistry experiments, it can become necessary to suspend a running sequence in order to make manual interventions, for example in order to fix a leak or clear a blockage. Therefore, a key requirement is the ability to pause a sequence and then resume it without having to start again from the beginning. When instructed to pause, each step propagates this command to any child steps, so that the whole sequence stops running. Crucially, sequences do not run any new steps until a resume command has been issued, and steps that involve a waiting time pause their timers so that the full remaining time must elapse after it is resumed again. The machines that are also involved in an experiment are also sent a pause command, which generally means that pumps and heaters are switched off, but data are still collected from the sensors. 2.2.6 Error handling There are a number of errors that may occur during a running sequence: • Errors in the communication with a machine. • Loss of connection to a machine. • Physical machine faults. • Physical problems with the apparatus. • Logic errors within a command protocol. In some cases automatic recovery is possible; for instance, virtual instrument drivers can include logic to repeat or discard corrupted messages. For more complex problems, handling logic can be incorporated within the control programme, including recovery attempts, notifications (for example via an on-screen message or an SMS notification), or as a last resort, flushing the reactor with solvent and halting the experiment. 2.2.7 Attached control systems A significant simplification in the implementation of complex control algorithms was achieved by adding the ability to connect control loops to individual steps in a sequence. 54 New control and monitoring software for flow chemistry systems For example, when attached to a sequence or wait command, a control loop starts and stops (and pauses and resumes) with this step. The control loops created initially were Bind, StateMonitor and PID. Bind updates a variable based on the value of another, optionally transformed by a function, whenever the master variable changes. StateMonitor executes one sequence and then another as a specified expression transitions from evaluating from True to False and back. This functionality is particularly useful for error handling. PID is a form of feedback controller which adjusts one variable based on changes in another (see Chapter 4).147 The example in Listing 2.15 can alternatively be written using these control loops (List- ing 2.18). The two aspects of the logic have been separated: the StateMonitor controls the temperature-dependent reagent addition, while the wait_until keeps track of the volume that has been added and terminates the StateMonitor at the appropriate time. 2.3 Conclusion An extensible software package was developed to control diverse elements of laboratory flow chemistry apparatus. The use of open-source tools should lead to sharing of control programmes and interfaces for new machines. Furthermore, the relative simplicity of the Python programming language may enable more researchers to create their own automated systems with a minimum amount of training. Whilst this software would not be appropriate for an engineering environment, where commercial software packages with support contracts are favoured for their guaranteed reliability, it is ideal for the research laboratory. Here, it is more important for a software tool to be low cost and have good flexibility than to offer a robust monitoring capability. A key aspect of this software is that the machine interfaces are abstracted, so that the researcher does not have to consider any aspects of communication with the instruments, and algorithms for common operations can be copied between protocols using similar devices. Common tasks such as remote data observation and complete logging are done automatically. By enabling researchers to spend less time on laborious and repetitive tasks, more time can be spent focusing on the chemistry and engineering aspects of a project. Importantly, control programmes can be easily adapted to employ similar equipment that may communicate over a different protocol. 2.3. Conclusion 55 Listing 2.18 The same protocol using an attached control system. This is more powerful than the example in Listing 2.15, because the sequence will terminate immediately once 1 mL has been dispensed, rather than having to wait for an iteration of the loop’s child sequence to complete before the expression is evaluated again. Note: there are ways of accomplishing this without the StateMonitor. # Import the necessary libraries from octopus.runtime import * from octopus.manufacturer import wpi, phidgets from octopus.transport.basic import serial from octopus.transport.phidgets import Phidget from octopus.sequence.control import StateMonitor # Set up the machines that will be used pump = wpi.Aladdin(serial("/dev/ttyS0", baudrate = 9600)) temp_sensor = phidgets.TemperatureSensor(Phidget(12345), inputs = [{ "index": 0, "type": phidgets.ThermocoupleType.K }]) # Create a reference to the relevant sensor temp = temp_sensor.thermocouples[0].temperature # Run until 1 mL has been added wait_for_addition = wait_until(pump.dispensed > 1000) # Add the control loop wait_for_addition.dependents.add(StateMonitor( tests = [temp < 42], trigger_step = set(pump.rate, 0), reset_step = sequence( wait_until(temp < 38), set(pump.rate, 50) ) )) # Start the experiment run(sequence( wait_for_addition, set(pump.rate, 0) )) A key target for future development involves the creation of a graphical interface for designing control sequences. A drag and drop interface would open up the technology to non-programmers with a minimal learning curve. 56 Chapter 3 Automated two-step reactions with heterogeneous metal catalysis 57 58 Automated two-step reactions with heterogeneous metal catalysis 3.1 Introduction* There are two important ways in which software for the rapid prototyping of simple control systems can augment flow chemistry projects. Traditionally, an experimental protocol involving multiple devices might require several people to supervise it to ensure that the required actions, such as starting the different reactors and switching the appropriate valves, occur at the correct times. In contrast, a computerised control system can provide precise control of the experimental timing and parameters, and can be designed to handle incoming information and respond to undesired events in a predictable and consistent manner. (Also, a computer can perform its task without having to contend with the external distractions commonly present in a busy laboratory!) The drawback is that an automated system is only as intelligent as its control programme and therefore lacks the ability of a skilled scientist to respond to unforeseen occurrences. Therefore, whilst it is in the scientist’s interest not to have to continually stay next to the equipment, it is important that they have the ability to observe the state of the system from wherever they are and to receive alerts or hear alarms if there is a problem that cannot be rectified automatically. In this chapter an application of the software control tools described in Chapter 2 is explored, within the context of the development of hydrolysis techniques using a hydrous zirconia catalyst. 3.2 Synthesis of primary amides by nitrile hydration Primary amide moieties are present in a number of biologically-active compounds and their building blocks. Amide formation is a very common transformation within phar- maceutical synthesis,149,150 but the standard protocols for achieving this are hampered by the widespread use of reagents that offer poor atom efficiency.151 One of the most atom-economical and environmentally-friendly methods for the preparation of a primary amide is by the hydration of a nitrile.152 Unfortunately, traditional methods can have low functional group tolerance due to the strong acids or harsh conditions employed. Under base-catalysed conditions over-hydrolysis is commonly observed since the formation of a carboxylic acid is kinetically favourable. * The work described in this chapter is reported in Beilstein J. Org. Chem. (Reference 148). 3.2. Synthesis of primary amides by nitrile hydration 59 Just as with solid-supported reagents (see Chapter 1), heterogeneous catalysis offers a number of benefits, particularly in flow systems where the catalyst can be contained within a cartridge.153 Purification is simplified because the catalyst will not contaminate the flow stream, and it may be possible to use a smaller amount of the catalyst because it can be recycled more easily. In cases where the activity of the catalyst can reduce during an experiment, the conversion can be monitored and the cartridge replaced at an appropriate time. A number of homogeneous metal-catalysed systems for nitrile hydration have been re- ported,152 and recent work within our own group has identified manganese dioxide as a mild heterogeneous catalyst for this transformation.154 Following investigations into the use of zirconium dioxide for Meerwein-Ponndorf-Verley reductions155 and Oppenauer oxidations,156 we were interested to find that zirconia was also an effective catalyst for the hydrolysis of heteroaromatic nitriles. We had identified piperazine-2-carboxamide (39) as an important 3D building block157 and at the time of writing it was costly to purchase even a racemic mixture of this com- pound.158 We envisaged that compound 39 could be obtained by a reduction of pyrazine-2- carboxamide (40), which could itself be prepared by a catalytic hydration of pyrazine-2- carbonitrile (41) (Scheme 3.1). NH2 H N N H O NH2 N N O N N N MOx M/C H2O H2 41 40 39 Scheme 3.1 Two step approach to piperazine-2-carboxamide via metal oxide catalysed hydrolysis followed by metal catalysed reduction. 60 Automated two-step reactions with heterogeneous metal catalysis 3.2.1 Hydration under flow conditions Optimal reaction parameters for the hydration of nitrile 41 (Scheme 3.2) had previously been determined by Dr Claudio Battilocchio. An in-line infrared spectrometer (Mettler-Toledo FlowIR) was used to measure the conversion of the nitrile to the amide. hydrous zirconia 100 ºC 100 psi R1 N N NH2 O N N CN 8:1 EtOH/H2O 0.1 mL/min FlowIR41 39 Scheme 3.2 Optimum reaction parameters for the zirconia hydration. Compound 42 is infused into a heated glass column containing zirconium hydroxide. An in-line IR spectrometer is used to measure the degree of conversion. A sensor with a silicon window (SiComp) was employed so that the nitrile stretching frequency could be observed.† In a normal reaction, only absorptions from the product are observed; characteristic signals are seen at 1685 cm−1 corresponding to the amide C−O 0 0.2 0.4 0.6 0.8 1 1.2 1.4 1.6 0 0.5 1 1.5 2 2.5 3 3.5 4 Ab sor ba nc e / AU Time /h Peak at 1051 cm-1 Peak at 1685 cm-1 Figure 3.3 Infrared absorbance profile of the reactor output during a 3 hour process, indicating the slug of material passing through the reactor column. The response reaches a maximum after approximately one column volume (approx. 1 mL) with a small degree of dispersion. In this case, absolute ethanol without additional water was used as the solvent so the reaction stopped once the water intrinsically contained within the hydrous zirconia was exhausted. This fall-off also occurred over approximately one column volume, indicating a low retention of material by the heterogeneous bed. † Frequencies in the range 1950 – 2250 cm−1 are normally masked by C−C stretching within the standard diamond-windowed (DiComp) sensor. 3.3. Reaction control and monitoring 61 bond stretching frequency, and at 1051 cm−1 in the fingerprint region. These could be used to observe the reacted materials exiting the zirconia column (Figure 3.3). We envisaged that a protocol incorporating automated monitoring and control using data from the IR spectrometer, could assist with the processing material over a long period of time. 3.3 Reaction control and monitoring In a manufacturing plant, the concept of process analytical technologies (PAT) refers to the observation of key parameters by sensors and analytical devices installed into the reactor equipment.159 In some cases a continuous process will have a greater number of more easily observable parameters than a comparable batch process. In addition to chemical analyses of the products there are numerous other properties that can be monitored to characterise the system. A continuous process can be defined by parameters such as the flow rate of the inputs, the temperature of the reactors, and the pressure of the system. Electronic measurements can also be useful; for example, in some continuous processing plants the voltage across a pump can be used as a proxy for the pressure and flow rate. An increased voltage requirement might imply a pressure spike, or a lower voltage a pump failure. Generally, if the reactor conditions stay within acceptable limits (for example, if the flow rates and reactor temperature are consistent, and there are no sudden pressure changes) then the output can be assumed to be of the desired quality. Laboratory processes could potentially benefit from similar systems. The devices depicted in Scheme 3.2 were connected to a computer (Figure 3.4a) and a selection of parameters was monitored using a simple control script that performed two functions. At the start of a run the column heater was powered on, and the reagent injected as soon as the temperature was stable. During the run, the output valve was controlled based on the PAT measurements reported by the Vapourtec R2+/R4 and the FlowIR. The infra-red absorbance represented the conversion at the exit point of the reactor, so readings outside the defined acceptable range were set to trigger immediate changes to the valve. Problems in the pressure (indicating flow rate changes) or the temperature could affect the conversion for one whole column volume and so this was set to stop collection for a longer time (Figure 3.4c). 62 Automated two-step reactions with heterogeneous metal catalysis EtOH / H2O hydrous zirconia 100 oC 20 min waste FlowIR N N NH2 O N N CN ow rate, pressure temperature absorbance collect / waste Python Raspberry Pi 100 psi R2 V1 a b 41 40 c Start Wait for temperature stability Switch to reagent Switch reactor on Set temperatures Set ow rates Wait for absorption to be above a trigger value Wait for absorption to be below a trigger value Switch to collect Switch to waste End Wait until reagent is used up (dened time) Switch to solvent Wash for two column volumes Switch o reactor If there are severe uctuations in temperature or pressure Switch to waste Wait for parameters to become stable Wait one column volume Switch to collect Figure 3.4 (a) Reactor and control setup for the continuous process. (b) Photograph of the apparatus as used. (c) Control sequence: the solid lines connect the commands that are to be executed in sequence. 3.4. Automation of multiple experiments 63 0 20 40 60 80 100 120 140 160 180 200 0 0.5 1 1.5 2 2.5 3 3.5 4 -2 0 2 4 6 8 10 12 °C, µA U ba r Time /h Absorbance /µAU Heater Temp. /°C System Pressure /bar Collecting Figure 3.5 Selected data collected over a three-hour reaction simulating a long process. The infra-red absorbance shown is that at 1685 cm−1, which is indicative of the amide bond in compound 40. The thick bar represents the time during which valve V1 was set to direct the output to the collection vessel. The pressure drop at 1.5 h caused the control system to direct the output to waste until one column volume (a pre-configured waiting time) after the pump had been manually re-primed. To test this behaviour, an air bubble was introduced into the reagent stream. This caused the pump to stop, creating a loss of pressure (Figure 3.5, at 1.5 h). The pressure drop was detected, triggering an interruption in the collection until one column volume after the problem had been rectified by manually priming the pump. Finally, after injection of the reagent had ended, collection stopped automatically by the same mechanism as soon as the output signal fell below a threshold . 3.4 Automation of multiple experiments With pyrazine-2-carboxamide (40) in hand, initial testing showed that reduction of this compound to pyrazine-2-carboxamide (39) could be performed in an H-Cube hydrogenation reactor. When working with a new reaction it is common to investigate the effects of the different variables. Interest is building in the concept of automatic optimisation.104,105,111,113,160 Parameters can be varied gradually using different mathematical methods such as simplex 64 Automated two-step reactions with heterogeneous metal catalysis algorithms161,162 in order to approach a maximum or minimum of some metric (for example: conversion, yield, maximum yield with lowest temperature and time, and so on). Initially, we had hoped to apply such a methodology to these reactions, but unfortunately this turned out to be impractical for two reasons. Firstly, each iteration was very slow (30 to 60 minutes) due to the long stabilisation time required for the H-Cube to reach steady state, the slow reaction rate and the large dead volume within the heterogeneous column. Secondly, many of the available parameters were limited to a set of discrete values (for example, the column temperature could only be set in units of 10 ◦C). In this situation, simple linear optimisation methods are difficult to apply,163 and more advanced machine learning techniques would have been required. Instead, we decided to employ a Design of Experiments method164–166 to investigate which of the available parameters were more important for determining the conversion and selectivity of this reaction. A two-level factorial design with three parameters (temperature: 40 ◦C and 100 ◦C; H2 pressure: 20 bar and full hydrogen mode; and flow rate: 0.1 mL/min and 0.2 mL/min) suggested 16 experiments: two repeats each of eight sets of conditions. waste samples FlowIRH-Cube 100 psi 75 psi V2 P2 N N NH2 O EtOH / H2O sa m pl e / w as te o w ra te te m pe ra tu re , pr es su re s ab so rb an ce a 40 Start Release hydrogen For each condition... Start pump at 0.2 mL/min Set valve to waste Set column temperature Set hydrogen mode Switch on hydrogen if o Wait for temperature stability if temperature changed Wait one column volume if ow rate changed Set ow rate Set valve to correct output Set valve to waste End Wait one column volume Wait 10 min (collection) b Figure 3.6 (a) Reactor setup for the optimisation process. A ten-position valve was included to enable collection of nine different samples. (b) The control sequence ran over an experimental subroutine for each of a pre-programmed list of conditions. 3.4. Automation of multiple experiments 65 This would have been laborious to carry out manually, but multiple similar experiments can be performed easily using an automated protocol. The control of a Knauer HPLC pump, the H-Cube reactor and a ten-port switching valve (V2) (Figure 3.6) was combined into a single programme, allowing up to nine reactions to be performed in sequence. In each iteration, the parameters of the reactors were adjusted to the desired set of conditions, and then a sample was taken once steady state was reached. Two major reduction products were observed: 1,4,5,6-tetrahydropyrazine-2- carboxamide (43; Scheme 3.7) and fully-reduced pyrazine-2-carboxamide (39). The carbonyl stretching frequencies of these two amides were not individually resolvable using the FlowIR (this instrument reported absorption maxima at 1651 and 1695 cm−1 respectively). The secondary frequencies had only low intensities, so unfortunately infrared spectroscopy was not adequate for analysis of the reaction mixture. Therefore, the samples were subsequently analysed by NMR to quantify the results. NH2 H N N H O NH2 N N O Pd/C H2 NH2 H N N H O Pd/C H2 40 43 39 Scheme 3.7 Reduction of the pyrazine ring is likely to occur in at least two steps. Intermediate 43 was also observed in the reaction output under some hydrogenation conditions. To quantify each sample, four parameters were calculated (Table 3.8): the degree of conversion, based on the amount of compound 40 remaining; the amount of the desired product 39 formed; and an estimated amount of undesired compounds, based on the integration of prominent by-product peaks visible in the NMR spectra (a doublet at 3.8 ppm and a triplet at 3.2 ppm, the first corresponding to compound 43 and the other to another, unidentified compound) relative to the integration of compounds 39 and 40. As expected, the hydrogen pressure had the most significant effect on the conversion. The temperature and the flow rate had smaller effects, although the combination of higher temperatures and lower flow rates gave a higher purity output because intermediates such as compound 43 are fully reduced. For maximum efficiency further hydrogenation procedures were carried out at the lower flow rate of 0.1 mL/min and the higher temperature of 100 ◦C. With both steps running smoothly, we turned our attention to the development of an integrated synthetic process. 66 Automated two-step reactions with heterogeneous metal catalysis Table 3.8 R esults for 16 experim ents over the eight sets of conditions specified by the design of experim ents protocol. H 2 pressure had the m ostsignificanteffecton conversion and purity. R un H 2 Pressure /bar Tem perature / ◦C Flow rate /m L /m in C onversion a Product a Side-product1 a Side-product2 a 1 Full 100 0.1 1.00 1.00 0.00 0.00 2 20 bar 100 0.1 0.53 0.35 0.08 0.10 3 20 bar 40 0.2 0.85 0.08 0.07 0.70 4 Full 40 0.1 0.78 0.74 0.04 0.00 5 20 bar 40 0.2 0.52 0.34 0.18 0.00 6 20 bar 40 0.1 0.38 0.26 0.12 0.00 7 20 bar 100 0.2 0.37 0.34 0.03 0.00 8 Full 40 0.2 0.80 0.54 0.26 0.00 9 20 bar 40 0.1 0.46 0.42 0.04 0.00 10 Full 100 0.1 1.00 1.00 0.00 0.00 11 Full 40 0.1 0.73 0.59 0.14 0.00 12 Full 100 0.2 1.00 1.00 0.00 0.00 13 Full 100 0.2 1.00 1.00 0.00 0.00 14 20 bar 100 0.1 0.40 0.37 0.04 0.00 15 Full 40 0.2 0.75 0.52 0.23 0.00 16 20 bar 100 0.2 0.44 0.44 0.00 0.00 a V alues w ere calculated by 1H N M R spectroscopy (400 M H z,M eO H -d 4 ),and are based on the relative integration ofthe peaks atδ 9.24 ppm (starting m aterial),3.13 ppm (product),3.79 ppm (side-product1)and 3.18 ppm (side-product2). 3.5. Integration of two steps 67 3.5 Integration of two steps 3.5.1 Intermediate reservoir When running a batch of experiments such as those performed in the previous section, the problem of resource management is commonly encountered. The amount of starting material required must be calculated based on the anticipated number of experiments. This is an even greater challenge when performing an automated optimisation procedure, as the number of experiments is unlikely to be known in advance. If the starting material has to be synthesised then production of a large excess of material may lead to wasted time and materials. It may be more efficient to make the intermediate in situ and then use it immediately for the automated step under each set of conditions. However, this would normally require that the first and second steps are compatible, for example by having the same flow rate. A make-up pump (Figure 3.9) could be introduced to vary the flow rate in the second step — but this would limit the minimum flow rate for the second step, as well as altering the concentration of the reagents. Step 1 Step 2 make-up pump waste Figure 3.9 A second pump can only add material, which affects the concentration. The lowest possible flow rate for step 2 is determined by the flow rate of step 1. We envisaged that a simple camera-based liquid-level sensor could be used to create a reservoir that would hold a small quantity of the intermediate solution. The start of the following reaction could then be delayed until sufficient material had been collected in the reservoir. Following previous work using visual observation methods to measure a phase boundary for continuous separation92,167 we anticipated that a similar method could be applied to measure the level of any solvent within a reservoir. By adding an air-filled float made from a standard disposable syringe168 the liquid level could be measured using a camera (Figure 3.10). The camera was connected to a computer by USB and integrated into the control system using SimpleCV,144 an open-source computer vision library for the Python programming language. 68 Automated two-step reactions with heterogeneous metal catalysis input output webcam Python SimpleCV a b Figure 3.10 (a) A laboratory flask was converted to a reservoir by adding a coloured float and an inlet tube for the pump. A camera observed the float to measure the liquid level within the flask. (b) Photograph of the apparatus. A sheet of white plastic created contrast between the float and the background, and ensured that no other green objects that could disrupt the reading appeared in the field of view. The programme collected an image from the camera at regular (1 s) intervals and the images were interpreted by a computer-vision algorithm (Figure 3.11). By subtracting the red channel from the green one, the float is highlighted and its position reported. The distance from the bottom of the image represented the volume of liquid in the flask, which was used as part of the control algorithm (see Section 3.5.2). This apparatus has a number of advantages over traditional liquid-level detectors such as ultrasonic, pneumatic or capacitance sensors, optical or air-bubbler detectors, or floats with mechanically or magnetically actuated switches. Since the only part in contact with the liquid is the float, which can be exchanged for different materials, there is less concern about chemical compatibility because none of the electronic components are in contact with the reactive solution. It is also a more universal method because the solvent does not need to be conductive and standard laboratory equipment (flasks) can be easily retrofitted with a float, rather than requiring the design of a bespoke vessel. Furthermore, the introduction of a reservoir disconnects one step from the next, allowing the reactors for different phases to be started and stopped independently. It also enables the introduction of workups and solvent-switches between steps (see Chapter 4), improving the flexibility of optimisation procedures. 3.5. Integration of two steps 69 Height 163 Area 93 Height 166 Area 584 G - R G - B ih g fe dcb a Figure 3.11 Computer-vision image processing. The image (a) is separated into red (b), green (c) and blue (d) components, or channels. There is a much lower intensity of red in the region of the float and this shows up as a dark area. Subtracting the red channel from the green (e) highlights this area whilst filtering out the background (grey and white areas have similar amounts of each colour). In contrast, the result of green – blue (f) does not show any significant features. A threshold filter (g) picks out the most significant areas, and then an erosion filter (h) removes smaller artefacts. The remaining blobs are recognised using the computer vision tool and the vertical position of the centre of the largest (by area) is measured as the height of the float. Note: the lower five images are shown with inverted colours for clarity. 70 Automated two-step reactions with heterogeneous metal catalysis 3.5.2 Combining synthesis and DoE Introduction of the reservoir and the liquid-level sensing logic into the original control programme allowed the two steps to be combined (Figure 3.12, Figure 3.13). The maximum solution required for a single hydrogenation experiment was approximately 6 mL, for which the float height was measured in advance. An experiment was performed only when the liquid level in the reservoir was above this threshold. EtOH / H2O 0.2 mL/min hydrous zirconia 100 oC 20 min R2 V1 V2P2 P1 waste samples FlowIR 100 psi reservoir 100 psi N N NH2 O N N CN webcam ow rate, pressure temperature absorbance collect / waste co lle ct / w as te o w ra te oat height te m pe ra tu re , pr es su re s 41 40 Figure 3.12 Reactor setup for the two-step DoE process. The intermediate was collected in the reservoir, and once the liquid level had risen over a predetermined threshold a hydrogenation experiment was performed. Additional monitoring logic was required so that valve V1 would stop directing material to the reservoir if the liquid level rose too high. This would also trigger an SMS notifica- tion, since it would indicate a problem with pump P2 that was likely to require manual intervention. 3.5.3 Extended run This control programme was extended to perform a long synthetic run using the same reactor configuration (Figure 3.12) by removing the multiple experiment logic and fixing the 3.5. Integration of two steps 71 el se St ar t Po w er o  H -C ub e pu m p Po w er o n H -C ub e pu m p Se t  ow ra te to 0 .2 m L/ m in Se t h ea te r t o 10 0 °C W ai t u nt il he at er > 9 5 °C Se t p re ss ur e lim it to 2 5 ba r Sw itc h R2 in pu t t o re ag en t Sw itc h R2 in pu t t o so lv en t Sw itc h R2 o ut pu t t o co lle ct W ai t u nt il th e liq ui d le ve l i n th e re se rv oi r h as ri se n ag ai n Po w er o  H -C ub e (k ee p hy dr og en ) W ai t f or 4 0 m in (z irc on ia c ol um n an d co nn ec tin g tu be d ea d vo lu m e) A le rt c he m is t i f a ny o f t he pu m ps lo se p re ss ur e W ai t u nt il th e liq ui d le ve l h as be en re du ce d W ai t u nt il IR s ho w s co nv er si on to in te rm ed ia te W ai t u nt il th e re se rv oi r c on ta in s en ou gh m at er ia l t o be gi n If IR re sp on se o f i nt er m ed ia te fa lls to o lo w Sw itc h to w as te u nt il th e IR re sp on se re tu rn s to n or m al If co lu m n te m pe ra tu re fa lls be lo w 9 5 °C o r p um p pr es su re u ct ua te s Sw itc h to w as te u nt il on e co lu m n vo lu m e af te r t he s ys te m ha s re tu rn ed to n or m al W ai t f or re ag en t t o be u se d up (p re co n gu re d vo lu m e) or fo r l oo p to  ni sh If th er e is s u ci en t m at er ia l i n th e re se rv oi r t o pe rf or m o ne ru n at th e re qu ire d o w ra te Sw itc h va lv e to w as te Sw itc h va lv e to w as te If th e liq ui d le ve l i n th e re se rv oi r ris es a bo ve a s et th re sh ol d En d Po w er o  R2 Sw itc h R2 o ut pu t t o w as te W ai t u nt il R2 s w itc he s ba ck to s ol ve nt W ai t u nt il th e st or ed h yd ro ge n ha s be en re le as ed Re le as e hy dr og en Fo r e ac h co nd iti on ... St ar t p um p at 0 .2 m L/ m in Se t v al ve to w as te Se t c ol um n te m pe ra tu re Se t h yd ro ge n m od e Sw itc h on h yd ro ge n if o W ai t f or te m pe ra tu re s ta bi lit y if te m pe ra tu re c ha ng ed W ai t o ne c ol um n vo lu m e if o w ra te c ha ng ed Se t  ow ra te Se t v al ve to c or re ct o ut pu t Se t v al ve to w as te W ai t o ne c ol um n vo lu m e W ai t 1 0 m in (c ol le ct io n) Fi gu re 3. 13 In te gr at io n of co nt ro ll og ic fo r m ul tip le hy dr og en at io n ex pe rim en ts un de r di ffe re nt co nd iti on s, w ith ca m er a- co nt ro lle d m an ag em en to ft he in te rm ed ia te pr od uc ed by th e hy dr at io n st ep . Th e co nt ro ls ys te m s in th e bo x ru n th ro ug ho ut th e pr oc ed ur e. 72 Automated two-step reactions with heterogeneous metal catalysis Start Start pump at 0.1 mL/min Stop pump Set ow rate to 0.2 mL/min Set heater to 100 ˚C Wait until temperature > 95 ˚C Set pressure limit to 25 bar Set input to reagent Set input to solvent Power o reactor Set valve to waste Set valve to collect Switch on column heater Wait for pressure and temperature to build up Switch on hydrogen Wait for hydrogen to be released Switch o H-Cube Wait for 30 minutes (H-Cube dead volume) Alert chemist if any of the pumps lose pressure Wait until IR shows good conversion to intermediate Wait until reservoir contains enough solution to continue Wait until reservoir is almost used up If IR response of intermediate falls too low Switch to waste until the IR response returns to normal If column temperature falls below 95 °C or pump pressure uctuates Wait for 2 hours, or until the reservoir gets close to being full Wait for 40 minutes (dead volume of zirconia column + connecting tube) Wait until input switches back to solvent Collect output If H-Cube temperature falls below 95 °C Stop collecting output until 30 min after temperature returns to normal (H-Cube dead volume) Stop collecting output End Switch to waste until one column volume after the system has returned to normal Figure 3.14 Control sequence for the two-step continuous process. hydrogenation parameters to the optimised values of 0.1 mL/min and 100 ◦C (Figure 3.14). The flow rate of the hydrogenation was limited by the performance of the H-Cube reactor, which would ideally be replaced with a higher-throughput equivalent. Nevertheless, this procedure demonstrates how two steps with disparate flow rates can be joined in sequence. In this case the hydration/hydrogenation sequence was run for 14 hours (Figure 3.15), with the first step running until the intermediate reservoir was full, and then shutting down. However, we envisage that this protocol could be extended to a longer sequence where the first step would run for only half the time, to keep the reservoir topped up. The size of the reservoir could be adjusted to minimise the amount of stored material and the amount of material wasted during startup and shutdown of the first reactor. 3.6 Conclusion The Python software developed in Chapter 2 was applied to a controlled two-step synthesis of piperazine-2-carboxamide (39).148 Over 14 hours, 7.3 g (95 %, 99 % purity) of 39 was produced. We were able to integrate the devices from different manufacturers and define time- or event-triggered transitions between different stages of the experiment, and 3.6. Conclusion 73 -20 0 20 40 60 80 100 120 0 2 4 6 8 10 12 14 Time /h Float Height (arbitrary units) IR Absorbance at 1670 cm-1 /µAU H-Cube column temperature /°C Vapourtec heater temperature /°C Figure 3.15 Selected parameters as observed over a 15 hour reaction. The output from the hydrolysis step is directed into the reservoir as soon as the infra-red absorption at 1670 cm−1 passes a pre-defined threshold. When enough of the intermediate has been collected, the H-Cube is powered on and the hydrogenation is started. The hydrolysis is stopped after the reservoir fills up, while the hydrogenation continues until the intermediate has been used up. responses to events such as pump failures. Data from each device were logged so that the performance of the system could be evaluated after an experiment. The software provided real-time feedback via its web interface so that the system could be left to carry out the experiment unattended. Having established the utility of the software control for this process, we investigated the control of more complex reactor configurations. 74 Chapter 4 An integrated flow synthesis of 2-aminoadamantane-2-carboxylic acid 75 76 An integrated flow synthesis of 2-aminoadamantane-2-carboxylic acid 4.1 Introduction Flow chemistry has the potential to enable moderate scale-up operations within the research laboratory. Greater efficiency can be achieved by creating integrated multi-step processes which are designed such that material produced in one stage can be used directly in the next, reducing the requirement for manual intervention between steps.169 Continuous processing is one good way to carry out such procedures, because by making the appropriate fluidic connections material can move directly from one reaction vessel to the next. However, the continuous processing of a sequence of synthetic steps (often referred to as “telescoping”) creates similar challenges to the design of one-pot reactions in batch mode.170 4.1.1 Challenges of telescoped reactions Chemical compatibility and purification In order to connect two chemical transformations in sequence, either as a one-pot batch or a telescoped flow process, the output of the first reaction must be chemically compatible with the second. This output is likely to contain spent reagents and by-products, which must be removed unless they are compatible with the conditions of the next reaction. For this reason, batch protocols nearly always include work-up or solvent extraction operations, or purification of the desired intermediate by chromatography or crystallisation. A particular challenge for a flow system is that the compatibility of the solvents between the two steps must also be considered. In a batch method it is usually straightforward to change the solvent between steps; in flow this is not trivial, because by its nature the materials must generally be kept in solution. Flow rates and dilution The introduction of flow streams for quenching the reaction output or for reagent addition will lead to an increase in the flow rate within the system, and dilution of the reagents, as more solvent is added (Figure 4.1). The higher flow rate will mean that reactors with larger volumes will be required for later steps, and this effect may make some multi-step processes impractical unless some of the solvent can be removed to concentrate the reagents and reduce the flow rate. 4.1. Introduction 77 0.5 mL/min 0.5 mL/min 1.0 mL/min 0.50 M 1.5 mL/min 0.33 M 2.0 mL/min 0.25 M Figure 4.1 Dilution of reagents and increase in flow rate as additional streams are added. It is also important to start the pumps involved in the downstream processes at the correct times, because otherwise the solvents and reagents for the later stages will be wasted whilst material moves through the earlier steps (Figure 4.2). The addition of an in-line detector to identify when a reaction plug is exiting from one reactor can allow the next reagent to be matched (both temporally and stoichiometrically) so that an appropriate amount of material can be used. No need to start this pump for the rst ~ 20 min (depending on dispersion) 20 min Figure 4.2 Reagents are wasted when downstream pumps are started before the reac- tion plug has passed through previous stages. Solutions The use of solid-supported and heterogeneous reagents has produced a number of elegant multi-step flow chemistry procedures,25,51,171,172 but unless these reagents are used catalyti- cally then the limited quantity of reactive material in any cartridge can make this technique unattractive. A solid reagent cartridge must be replaced when it is exhausted, whereas liquid or gaseous consumables can be added and removed continuously making the process easier to operate. Furthermore, the high cost of some polymer-supported reagents can be prohibitive when large quantities are needed. Continuous workup operations such as solvent extraction,173 filtration,174 distillation175 and evaporation176 are well established when working on a large (manufacturing) scale, but the application of these techniques into a bench-top device presents a number of challenges. These generally result from the formation and handling of solids and slurries within the 78 An integrated flow synthesis of 2-aminoadamantane-2-carboxylic acid system.177–179 The reduced dimensions of the equipment mean that any solid particles that form are much larger in comparison to the pumps and channels. In addition, precipitates can create blockages much more rapidly, and the smaller pumping equipment is often more susceptible to damage or disablement by even very fine suspensions. Continuous solvent extraction is well-developed on the laboratory scale. There are a number of reported methods utilising hydrophobic membranes,180–182 settling tanks,92 selectively wetting channels183 or counter-current flow.184 With two of these devices in succession, it can also be possible to achieve a solvent exchange by passing materials from an organic phase into an aqueous phase, and then back into another organic stream.180 Our group has recently developed an in-line solvent evaporator based on nebulisation of a liquid stream within a heated chamber.185 This may offer one solution to the removal of volatile solvents or in-line concentration of streams to reduce the dilution effect. Tools for continuous (multiplexed) chromatography are commercially available, though expensive; and continuous crystallisation methods have been reported186,187 although the resulting slurry must then be processed. Software control can be used to time reagent addition to minimise waste,89 to ensure that each step is run at the correct time, and to monitor for any problems that arise and either make corrections or alert the operator. In this chapter the application of downstream processing techniques and software control systems to multiple step chemistry involving work-up operations is discussed, using the example of a recently reported synthesis of 2-aminoadamantane-2-carboxylic acid (44, Figure 4.3).188 H2N O OH Figure 4.3 2-aminoadamantane-2-carboxylic acid (44) Our group had originally become interested in the synthesis of this unnatural amino acid during investigations into the synthesis of probes for neurotensin receptors 1 and 2.189 As part of a programme to synthesise large quantities of adamantane-containing compounds,190,191 effective methods to prepare the key component 44 on a large scale were developed. 4.2. Previous synthetic routes 79 4.2 Previous synthetic routes 4.2.1 Reported batch–mode syntheses Previously-reported methods to access 2-aminoadamantane-2-carboxylic acid include a Bucherer–Bergs reaction as employed by Nagasawa and co-workers192 (Scheme 4.4, con- ventional conditions), involving heating with sodium cyanide to form a hydantoin (45) that could be directly hydrolysed to the desired amino acid. However, our group was unable to isolate this compound with reasonable purity either using the original conditions, or modified microwave processing conditions (Scheme 4.4, microwave conditions), due to a high degree of inorganic impurities.188 H2N O OHO N H NH O O NaCN (NH4)2CO3 NaOH a b 46 45 44 Scheme 4.4 Bucherer–Bergs routes to amino acid 44. Conventional conditions: (a) NaCN (2 equiv.), (NH4)2CO3 (1.5 equiv.), EtOH, 170 ◦C, 170 psi, 3 h (99 %); (b) 1.25 N NaOH, 195 ◦C, 250 psi (pressure vessel), 2 h (94 %). Microwave conditions: (a) NaCN (2.2 equiv.), (NH4)2CO3 (1.5 equiv.), 3:1 EtOH/H2O, 170 ◦C, 150 psi (sealed vial), 2 h (94 %); (b) 1.25 N NaOH, 175 ◦C, 3 h (71 %). A second route attempted was based on a Strecker reaction as inspired by Commeyras193 and Edward.194 The nitrile product generated by treatment of the ketone with sodium H2N CO2H O H2N CN HN CN Ph O HN CO2H Ph O NaCN, NH3, NH4Cl EtOH / H2O (68 %) HCl THF / H2O 25 °C, 5 min (93%) HCl AcOH / H2O reflux, 30 h (65%) PhCOCl K2O3 THF / H2O 25 °C, 1 h (52%) 46 47 48 4944 Scheme 4.5 Edward’s Strecker route to amino acid 44. 80 An integrated flow synthesis of 2-aminoadamantane-2-carboxylic acid cyanide was hydrated using a milder method than in Nagasawa’s route. Reaction of nitrile 47 with benzoyl chloride gave an α–amide; under acidic conditions this compound underwent hydrolysis to afford intermediate 49, which could be hydrolysed to give the amino acid (Scheme 4.5). However, a significant drop in reaction yield and purity was noticed when this reaction was performed at scale.188 4.2.2 First generation flow–mode synthesis To overcome these problems, our group reported188 a flow synthesis, involving five separate stages: an initial Grignard addition, followed by a Ritter reaction, cyclisation, ozonolysis and hydrolysis (Scheme 4.6). The Grignard addition of ethynylmagnesium bromide (50) to adamantanone was performed in THF at 40 ◦C, and the product was subjected to an in-line aqueous workup to quench the resulting magnesium salts. Under the continuous conditions the temperature of this exother- mic process was controlled effectively. This was an important consideration because the product 51 had been found to be liable to thermal decomposition at elevated temperatures. Subsequently, compound 51 was subjected to strong acidic conditions in an acetonitrile solvent to effect a Ritter reaction.17,195 The output was quenched by simultaneous addition of ethanolic potassium hydroxide to the collection tank. After filtration, the resulting basic solution could be used directly for the 5-exo-dig cyclisation by pumping though a coil heated to 120 ◦C. Cyclic intermediate 53 was subjected to ozonolysis using a biphasic gas / liquid system, and the resulting ozonide quenched using a polymer-supported thiourea25 to provide azlactone 54. This could either be used directly as a coupling reagent196 or subjected to acidic hydrolysis to afford the amino acid 44. The scope of the chemistry for this flow synthesis had been thoroughly established during the first-generation synthesis.188 However, a number of manual work-up operations were required which prevented the steps from being combined into a single sequence. The aim of this work was to use our knowledge of the process to integrate the chemistry steps to produce a single sequence. We envisaged that recently developed in-line workup techniques, along with appropriate automation and control, could be employed to achieve this. The main obstacles that were identified are highlighted in Scheme 4.6. The Grignard reagent 50 used in Step 1 was only commercially available in tetrahydrofuran (THF), which 4.2. Previous synthetic routes 81 Step 1: Grignard Step 4: Ozonolysis Step 3: Cyclisation Step 5: Hydrolysis Step 2: Ritter 1A 2A 3A 4A 3B 2B 4B 1B 1C 1D Reagent is only available in THF. THF solvent is incompatible with the Ritter step. Aqueous work-up is required to quench and remove magnesium salts. Acid must be quenched. Produces large quantity of insoluble salts. Base must be neutralised before the acid hydrolysis in Step 5. Stoichiometric thiourea to decompose the ozonide. 40 min sonication 40 oC 0.5 M THF NH4Cl (aq)0.20 mL/min 0.5 M THF 0.18 mL/min 40 psi O MgBr HO aqueous extraction remove THF solvent 7 min 30 oC MeCN / AcOH3 M KOH 40:1 EtOH / H2O 3 mL/min 1 mL/min 1 mL/min HO H2SO4, AcOH, Ac2O50 min 120 oC 0.3 mL/min NH O 100 psi lter remove base O3 ozone generator 500 mL/min (7–10% O3 ; 2–3 mmol/min) Ar, O3 Ar gas trap ozonolysis reactor O2 N O N H NH2 S QP-TU CH2Cl2 4 mL/min 0.3 mL/min 18 min 150 oC AcOH 0.4 mL/min 0.4 mL/min HCl, AcOH, H2O N O O H2N OH O 100 psi 50 46 51 51 52 53 54 44 Scheme 4.6 Overview of the first-generation flow route to compound 44.188 The main obstacles to an integrated continuous sequence are highlighted. Isolated yields obtained: Step 1, 106 g, 90 %. Step 2, 22 g, 91 %. Step 3, 8.2 g, 91 %. Step 4, 6.7 g, 95 %. Step 5, 6.3 g, 94 %. 82 An integrated flow synthesis of 2-aminoadamantane-2-carboxylic acid is incompatible with the Ritter reaction in Step 2. Initial experimentation showed that a solvent exchange from THF to acetonitrile was possible using an in-line evaporation device,185 which we considered to be preferable to manually changing the solvent of the Grignard reagent. The Grignard reaction produces magnesium salt by-products that must be removed by an aqueous extraction. Furthermore, each step requires different pH conditions: Step 1 in base, Step 2 in acid, Step 3 in base and Step 5 in acid; so multiple quenching operations are needed. Finally, the first generation synthesis employed a stoichiometric quantity of solid-supported thiourea to decompose the ozonide intermediate, which we hoped to avoid. We turned our attention first to a Grignard reaction and aqueous extraction sequence. 4.3 Grignard reaction and aqueous extraction The output of the Grignard reaction consisted of salt 55 in THF, along with any unreacted reagent 50. To perform an aqueous quench and extraction, this stream must be combined with an aqueous stream, mixed and then separated (Scheme 4.7). Organic 0.3 mL/min phase separatorcontactor HO BrMgO Aqueous 0.5 mL/min aqueous waste mixer55 51 Scheme 4.7 Liquid-liquid extraction involves combination of the aqueous and organic streams at an appropriate mixer (see Section 4.3.4) followed by phase separation. 4.3.1 Mixing The first-generation procedure (Scheme 4.6) involved a standard tee connector ( 0.5 mm, PEEK) immersed in a sonication bath to prevent aggregation of the magnesium salts upon mixing. Indeed, without the sonication bath the tee mixer blocked within 10 minutes. Use 4.3. Grignard reaction and aqueous extraction 83 a mixing zone biphasic streamorganic stream aqueous stream 1/8” OD, 1/16” ID PTFE tubing 1/8” tee union 1/16” OD, 1 mm ID PFA tubing 1/8” OD, 2.4 mm ID ETFE tubing 1/8” OD, 1/16” ID PTFE tubing b c Figure 4.8 (a) Schematic of the tube-in-tube mixer, constructed using a Swagelok tee connector and Swagelok reducers. (b) Photograph of a tube-in-tube mixer. (c) A vibrating micromotor that can be attached to the mixer to reduce precipitate aggregation. of a wider-bore tee mixer (Omnifit,  1.5 mm, PTFE) worked for a longer period (30 – 60 minutes) before becoming blocked. Introduction of a tube-in-tube mixer (Figures 4.8a, b), emerging into a wide-bore tube (R2,  2.4 mm, ETFE) provided a more robust solution that could run for around an hour before precipitates caused a catastrophic pressure increase. Precipitation occurred within the inner tubing as traces of water ingressed against the flow. It was found that a more compact alternative to an ultrasonic bath for preventing aggregation was the attachment of a vibrating micromotor (Figure 4.8c) to the tubing just after the tee mixer; the vibration displaced the precipitates, which were then dissolved by the aqueous stream. With this device in place, the mixer was able to operate continuously for over 6 hours. This mixer was configured so that the inner tubing protruded at least 5 cm into the wide tubing. The aqueous flow around the inner tubing displaced solids adhering to the end of the inner tubing. Slicing the inner tubing at an angle to create a tapered end and thus potentially a larger contact area had no noticeable effect on the traces of precipitate remaining at this point. Without the precipitation, the mixer was found to operate best when the outlet was 84 An integrated flow synthesis of 2-aminoadamantane-2-carboxylic acid Figure 4.9 Orientation of the tube-in-tube mixer. Since the organic solvents in this system (indicated by yellow shading) were less dense than water, the organic phase may have risen towards the tee body if the mixer was oriented downwards. However, if the mixer was oriented upwards then the precipitates could fall towards the tee body. An inverted-U configuration was the most effective, because the organic phase did not move backwards and the solids fell away from the mixer. inclined upwards so that the organic phase would tend to rise away from the tee connector. However, when precipitates were formed they tended to fall back towards the tee body, disrupting the aqueous flow. A better arrangement involved an inverted U-bend in the tubing so that any organic bubbles which moved backwards would travel no further than the apex (Figure 4.9). 4.3.2 Contactors When a biphasic system is formed, it is important to ensure that effective contact occurs between the two phases, for complete extraction.183 Contactors effective in laboratory scale continuous processing include static mixers, dynamic mixers, or slug flow in a capillary. In a static mixer, the flow stream passes through a packed column which disrupts the flow. In a dynamic mixer the flow stream moves past an agitating device such as a magnetic bead contained within the flow tube or column.92,178 Biphasic slug flow creates a mixing effect due to internal circulation within each slug.183,197 Without using wide-bore tubing, we did not have magnetic beads in an appropriate size to create a dynamic mixing effect. Due to the solid particles present, use of a capillary was not suitable, but we anticipated that circulation within the slugs as they passed though R2 would provide a good degree of mixing. Fortunately, we later found that a filtration device was necessary which turned out to provide an additional static mixing effect. 4.3. Grignard reaction and aqueous extraction 85 4.3.3 Filtration Whilst the majority of the salts formed during quenching of the Grignard reaction output were removed by the aqueous stream, a very small quantity of insoluble salts were formed and persisted within the flow stream. These created blockages in the phase separation and evaporation steps, and so a filtration mechanism was included to capture them. Addition of a 50 µm filter into the flow stream was ineffective because even a large filter became blocked by particulates. Instead we envisaged the use of a filtration cartridge with varying degrees of filter, which could accumulate the precipitates without becoming blocked. A number of potential filtration media were identified which could be loaded into a glass column: cotton wool, glass wool, celite, sand, charcoal or glass beads (Figure 4.10). Cotton wool became compacted and then blocked after approximately 4 hours. A glass wool plug was more robust and collected the precipitates without becoming blocked over several runs (up to 10 hours). Although it might have been possible to perform an occasional acidic flush to remove the accumulated particulates, glass wool is hazardous to handle, proved to be difficult to pack into the column, and the plug introduced a large dead volume into the flow system. We were concerned that sand might leach iron into the system, so instead we evaluated the glass wool plug frit small glass beads medium glass beads large glass beads charcoalcelite Figure 4.10 Filter cartridge configurations. (1) A cartridge containing a frit, creating an aqueous chamber through which the organic phase passed. The frit blocked rapidly. (2) A glass wool plug caught larger particles to protect the frit, but there was still a large dead volume. (3) Small glass beads were not stable under the flow conditions and migrated downwards. (4) Celite did not migrate but had undesirable packing within the column. (5) A charcoal powder bed provided effective filtration. The glass beads kept the charcoal powder in place. 86 An integrated flow synthesis of 2-aminoadamantane-2-carboxylic acid use of glass beads of a comparable size to the sand particles (53 – 106 µm). A three-layer column was prepared, with a top layer of the small beads, an intermediate layer of medium beads (0.5 – 0.7 mm) and a bottom layer of large beads (4 mm); under static conditions each layer held the one above in place. However, when the slug-flow stream was applied, the top layer seemed to fluidise, causing mixing of the differently-sized beads such that the smallest ones migrated towards and out of the bottom of the column. We anticipated that smaller particles were more likely to pack than migrate, and so different materials were evaluated in place of the smallest glass beads. Celite powder198 remained in place as desired, but seemed to aggregate upon wetting forming channels through which the majority of the flow stream passed. Charcoal powder199 was much more effective, since it was easy to load into the column, it stayed in place without packing, and it collected the small particulates without blocking. The glass beads collected the larger particles and prevented the charcoal from falling down the column. Although charcoal is more likely to be chemically active than the other options, we did not expect this to be a problem since the adamantane-containing compounds are not aromatic. Pleasingly, this column also provided good mixing, and the biphasic stream emerging from the top of the column was separated into very small slugs. 1H NMR analysis of the aqueous phase after separation could not detect any of the product 51, and removal of the solvent from the organic phase left no inorganic salts, indicating that the quenching and extraction were effective. 4.3.4 Separation We chose a gravity-based separation method,92 whereby the biphasic stream is directed into a reservoir (S1) where the phases separate under gravity. A pump (P4) withdraws the lower aqueous layer, allowing the organic layer to escape from the top (Figure 4.11). When performed in an Omnifit glass column this can operate under pressure,200 and the lack of a membrane means that it is robust to small quantities of solid formation. Unfortunately, under continuous operation, the THF / water solvent mix proved to be highly miscible and the phase separation was not effective: there was no visible meniscus and a significant quantity of the product (51) passed into the aqueous stream. The use of a concentrated salt solution instead of water did not create any significant improvement in the separation, so alternative solvent mixtures for the Grignard sequence were investigated. 4.3. Grignard reaction and aqueous extraction 87 75 psi Sat. NH4Cl (aq) / H2O(3:1 v/v) 0.5 mL/min mixer webcam 40 psi 250 psi organic out aqueous out ( also present in aqueous due to poor separation with THF solvent) HO P3 P4 M2 R2 S1 charcoal lter glass beads 40 min 40 oC FlowIR 0.5 M THF 0.30 mL/min 0.5 M THF 0.27 mL/min 40 psi O MgBr BrMgO P1 P2 R1 M1 a 50 46 55 51 51 15 mm Omnit column xed end tting with O-ring. PTFE; no frit. Drilled and tapped to match Omnit screw thread. Upchurch Scientic pressure-release valve tee piece, bored through to 1/8”. 21G needle inserted into tubing, and trimmed to protrude into glass chamber. 1/16” OD, 0.5 mm ID PTFE tubing. Heavy phase outlet Combined inlet b Figure 4.11 (a) Grignard reaction and liquid-liquid extraction apparatus. For the solvent extraction, the aqueous and organic streams are combined at tee mixer M2, and then pass through a wide-bore tubing (R2) and the filtration cartridge into the phase separator (S1). The high-pressure BPR on the lower exit prevents material being forced through pump P4. The low-pressure BPR on the upper exit acts as a check valve to prevent back-flow. (b) Construction of the tee fitting for the bottom port of the phase separation chamber. 88 An integrated flow synthesis of 2-aminoadamantane-2-carboxylic acid Use of diethyl ether as a solvent for the stream of adamantanone (46) led to very good separation, but low solubility of compound 51 in a THF / Et2O solvent mixture resulted in increased precipitation in the reactor and aqueous mixer (Table 4.12). Addition of toluene led to improved solvation of the product, and also a less compressible solvent system which could be pumped more reliably. A 3 : 2 (v/v) mixture of toluene / Et2O was found to provide a good balance of solvation, immiscibility with water when combined with the THF stream, and sufficient volatility to allow the solvent-switch into acetonitrile. Table 4.12 Solvent conditions for the adamantanone stream in the Grignard reaction. Both THF and toluene are readily removed under the nebulising conditions.185 Solvent mixture Outcome THF High miscibility with water. Et2O Poor pump operation due to high compressibility. Low solubility of the starting material. Toluene Low solubility of reaction products. Toluene / Et2O (1 : 1 v/v) Some precipitation of starting material. Toluene / Et2O (3 : 2 v/v) Good solubility. Although the use of Et2O is undesirable, 201 it was necessary when using this equipment configuration because volatile solvents were required for a practical solvent exchange operation. Extractor control As in the originally reported phase separation system,92 the separation tank was observed using a camera and the position of the phase boundary measured using computer-vision enabled monitoring of a plastic float167 (see Section 3.5.1). This enabled the system to compensate for flow rate fluctuations: if the two aqueous pumps did not operate at exactly the same flow rate (for example, in the event of a pump failure or mis-calibration) then over time the phase boundary would drift up or down. By controlling the flow rate of the pump that removes the lower phase based on the position of the interface, this was avoided (Figure 4.13*). * For the Python control protocol, refer to Reference 202. 4.3. Grignard reaction and aqueous extraction 89 Solvent Extractor Feedback Control max P4 ow rate: V1 collecting: (P1 + P2 + P3) total rate V1 waste: (P3 + P5) total rate Run control systems until end of experiment Reservoir S1 oat height Aqueous out pump P4 rate > 0  on < 0  oOrganic stream ow rate Aqueous in pump P3 power Aqueous out pump P4 power Acetonitrile pump P6 power Reservoir S1 oat too high or too low Alert chemist (pump failure) Figure 4.13 Control protocol for the solvent extractor. The flow rate of pump P4 is adjusted based on the liquid level in S1. If the float moves out of bounds, an alert is triggered. If the organic stream is flowing (either valve V4 is collecting or pump P5 is powered on) then the three extractor pumps are powered on. The original system used a proportional-gain controller to control the output pump.200 A similar algorithm was implemented for this system, but in this case incorporating a predictive extrapolation to slow down the rate of change of the response as it approaches the set point, to reduce the effect of the lag time between the flow rate adjustment and the response which otherwise tended to lead to oscillations in the system. This is based on a proportional-differential controller; the sample interval was set to 20 seconds to reduce the effect of noise on the derivative term. A threshold was added so that the flow rate was kept constant when the liquid level was near the desired point (Figure 4.14). The feedback control was performed according to: errort = heightt− target di f f erential = errort− error(t−interval) change = (0.25×|errort |)× (errort + interval×di f f erential) response = responset + change where target is the desired float height (in pixels as observed by the camera), heightt is the float height at the current time t, and response is the new flow rate of the pump. 90 An integrated flow synthesis of 2-aminoadamantane-2-carboxylic acid 200 250 300 350 400 450 0 0.2 0.4 0.6 0.8 1 1.2 1.4 1.6 0.2 0.4 0.6 0.8 1.0 Flo at he igh t /p ixe ls Flo w r ate /m L/m in Time /h Float height Flow rate Figure 4.14 Response of the pump rate to the float height during part of an experiment. The liquid level is maintained within a small window (8 mm). The plateaus indicate when the threshold filter was taking effect. 4.3.5 Control of the Grignard reaction Control systems were also added to the Grignard reaction step (Scheme 4.15). The output from the reactor was monitored by an IR spectrometer. The absorption at 1120 cm−1 was indicative of the product, and by passing the output through valve V4 it could be directed to waste until the product was observed. An additional pump (P5) was added to flush solvent 40 min 40 oC FlowIR solvent waste 0.30 mL/min 0.5 M THF THF MeOH 0.28 mL/min 0.5 M 3:2 Toluene/Et2O 40 psi 100 psiO MgBr BrMgO P1 P2 R1 M1 V4 V1 V2 V3 3:2 Toluene/Et2O 0.5 mL/min P5 50 46 55 Scheme 4.15 Grignard reactor configuration with four valves and an additional pump P5 to enable flushing of the pumps with solvent, and flushing of mixer M1 to prevent blockages by precipitation at times when the output of R1 was being directed to waste. 4.3. Grignard reaction and aqueous extraction 91 through valve V4 at 0.5 mL/min whenever the reactor output stream was being directed to waste. This ensured that there was no standing solution within the mixer, preventing precipitation and blockage. The system was also adapted to reduce the incidence of problems with pump P1 caused by the reagent stream. Some batches of ethynylmagnesium bromide contained small quantities of particulates from water ingress, which could disable the pump. A 2 µm in-line filter was installed before the pump in order to filter the solution prior to entering the pump, and a rigorous drying protocol was used before pumping the organometallic reagent. Nevertheless, pumping performance was occasionally degraded. The control software monitored the pressure sensor readings on the pump outputs to identify the characteristic oscillations caused by reduced effectiveness of either pump’s check valves. If the oscillations persisted for over a minute, then the input streams were switched to the solvents (valves V1 and V3) and the pump flushed at a higher flow rate (1 mL/min) for ten seconds. If this did not rectify the problem, the solvent was switched to methanol (valve V2) and the flushing process repeated before reverting to THF. If this was also unsuccessful then the operator was alerted, otherwise normal operation was resumed. Using these control systems reliable pumping was achieved for several hours at a time with only minimal intervention. An overview of the control protocol is shown in Figure 4.16. Grignard Reaction Experiment Complete Switch P1, P2 to solvent Set pressure limit to 30 bar Set ow rates for pumps P1 – 5 Power on FlowSyn Wait for R1 temperature Switch P1, P2 to reagent Run control system until reagent is exhausted (pre-congured duration) Wait for product IR peak to decrease below threshold Wait for product IR peak to rise above threshold Wait 20 min Power o FlowSynStart collecting output Stop collecting output IR output below threshold Monitor pump P1, P2 pressure Valve V4 position Pump P5 power Switch valve V4 to waste waste  on / collect  o Figure 4.16 Control protocol for the Grignard reaction. Once the system is running, two control loops are started that switch P5 on and off, and control the position of V4 based on the IR sensor output. 92 An integrated flow synthesis of 2-aminoadamantane-2-carboxylic acid 4.4 Solvent exchange The organic output from the separator was combined at mixer M3 ( 0.5 mm, PEEK) with a stream of acetonitrile delivered by pump P6 and directed into the solvent evaporator. The evaporation device developed within our group185 consists of a narrow-bore tube ( 0.1 mm) projecting into a glass chamber. A jet of gas, usually nitrogen, is emitted from a slightly larger tube surrounding the first one (Figure 4.17). The gas causes the liquid stream to break up into droplets, and their larger surface area promotes evaporation. Solvents with high evaporation rates (DIN 53170 standard203) are readily removed by this system, enabling a solvent exchange from a mixture of THF, diethyl ether and toluene into acetonitrile, which was used as both the solvent and a reagent for the subsequent Ritter reaction step. MeCN 1.2 mL/min THF / Et2O / Toluene N22 L/min 100 psi HO organic out (MeCN) vapours out (condenser) HO a bP6 M3 P7 peristaltic 51 51 Figure 4.17 Solvent extractor apparatus. The organic stream from S1 is combined with a stream of acetonitrile and directed into the evaporation chamber. A jet of N2 nebulises this stream and vapourises the THF, Et2O and toluene solvents to leave a solution of 51 in acetonitrile, which is removed by peristaltic pump P7. The combined gases pass out from the top of the chamber, whilst the liquids are withdrawn from the bottom by a peristaltic pump (P7). This pump also functions as a back-pressure regulator, preventing the liquid from being forced out by the gas pressure before it has had time to concentrate. A peristaltic pump also has the advantage that it can operate just as effectively when there is no liquid feedstock present, unlike normal HPLC pumps. Under optimised conditions (Table 4.18, entry 15) the THF content of the stream was reduced to around 5 % which was found to be sufficiently low to prevent extensive poly- merisation under the acidic Ritter reaction conditions. The incoming spray of acetonitrile 4.4. Solvent exchange 93 Ta bl e 4. 18 S ol ve nt ev ap or at io n co nd iti on s to m ax im is e TH F re m ov al an d av oi d so lid or so lv en ta cc um ul at io n w ith in th e co lu m n. Th e ou tle tp um p (P 7) w as tu ne d su ch th at th e liq ui d w as no tw ith dr aw n to o qu ic kl y; th e pr es en ce of so lv en ti n th e co lu m n pr ev en te d th e de po si tio n of so lid s. E nt ry G as pr es su re E va po ra to r P6 flo w ra te P7 flo w ra te So lid L iq ui d M eC N :T H F :T ol ue ne :E t 2 O a /b ar Te m p. /◦ C /m L m in −1 /m L m in −1 de po si tio n pr es en t 1 1. 0 23 1. 2 0. 6 L ot s A cc um ul at io n N ot de te rm in ed 2 1. 2 23 1. 2 1. 0 – – 1 :0 .0 5 :0 .0 3 :0 3 1. 2 25 1. 2 1. 3 L ot s – 1 :0 .1 1 :0 .0 7 :0 .0 1 4 1. 2 25 1. 2 1. 0 L ot s Sl ow A cc um . 1 :0 .0 9 :0 .0 7 :0 .0 1 5 1. 2 25 1. 2 1. 6 L ot s Sl ow A cc um . 1 :0 .1 0 :0 .0 7 :0 .0 1 6 1. 2 25 1. 2 1. 6 L ot s A cc um ul at io n 1 :0 .0 8 :0 .0 7 :0 7 1. 4 23 1. 1 0. 7 So m e – 1 :0 .0 8 :0 .0 7 :0 .0 1 8 1. 4 21 1. 1 0. 7 – – 1 :0 .0 6 :0 .0 6 :0 9 1. 4 21 1. 1 0. 6 So m e Pr es en t 1 :0 .0 7 :0 .0 7 :0 10 1. 4 23 1. 2 1. 0 – – 1 :0 .0 8 :0 .0 6 :0 .0 1 11 1. 4 23 1. 2 0. 8 – Pr es en t 1 :0 .0 7 :0 .0 7 :0 12 1. 4 23 1. 2 0. 6 So m e Pr es en t 1 :0 .0 5 :0 .0 6 :0 13 1. 4 21 1. 2 0. 8 – Pr es en t 1 :0 .0 8 :0 .0 7 :0 .0 1 14 1. 4 21 1. 2 0. 6 – – 1 :0 .0 6 :0 .0 7 :0 15 1. 4 22 1. 2 0. 6 – Pr es en t 1 :0 .0 4 :0 .0 4 :0 a D et er m in ed by 1 H N M R sp ec tr os co pi c an al ys is . 94 An integrated flow synthesis of 2-aminoadamantane-2-carboxylic acid washes any solids from the walls of the chamber, and the resulting solution is directed into the next phase of the process. This represents an important development for continuous processing, since combining reactions with differing solvent requirements often poses a significant challenge to multi- step procedures. The possibility to change the solvent of a reagent stream in a continuous fashion will enable a larger spectrum of chemistries to be connected together in flow processes. 4.5 Ritter reaction and quenching 4.5.1 Intermediate reservoir The output of the solvent-switch operation was collected in a reservoir (see Section 3.5.1), which in this case was made from a solid-phase extraction (SPE) cartridge (S2). These are equipped with a Luer connection at the bottom from which the solution could be withdrawn (Figure 4.19, left), and a 20 µm frit which provides protection for the subsequent pump (P8). The use of this reservoir was necessary because the output from the peristaltic pump (P7) did not have a constant flow rate, and more importantly this pump cannot generate the pressure required to run a flow reaction at elevated temperature with a BPR. The reservoir also evens out any variation in concentration created by the evaporator. Figure 4.19 Three of the work-up devices, mounted such that the reservoir and the separator could be observed by one camera. Left: Intermediate reservoir consisting of an SPE cartridge with a 20 µm frit and plastic float. Centre: Liquid extraction settling tank; the float indicates the phase interface. Right: Charcoal/glass bead filter cartridge. 4.5. Ritter reaction and quenching 95 Ritter Reaction PI Control max 1.4 mL/min min 0.15 mL/min Wait for 20 min Run control systems until end of experiment Switch pump P10 to reagent Wait until reservoir contains enough material to start Reservoir S2 oat height Reagent pump P8 rate o / onAcid pump P9 power Base pump P10 power Reservoir S2 oat too low Reservoir S2 oat too high Reservoir S2 oat no longer too low Pause PI control Stop reagent pump P8 Wait 1h in case reservoir rells Wait for reservoir to ll up Resume PI control Switch valve V5 to waste Reservoir S2 oat level dropped Switch valve V5 to collect Flush with solvent for 40 min Stop pump P8 Figure 4.20 Control protocol for the Ritter reaction step and control loops for reservoir S2. The flow rate of pump P8 is adjusted based on the liquid level; if this falls too low then the system waits for one hour before flushing the system with solvent. Pump P10 is switched on whenever acid is being injected. Additionally, the reservoir could be used to start the following step only when sufficient material was available, and also to stop it if the material ran out, preventing disablement of pump P8 by air bubbles (Figure 4.20). This allowed the control of the different steps to be kept separate, leading to a less complex control system. 4.5.2 Ritter reaction The reagent stream was withdrawn from the reservoir and mixed with an acidic stream at a tee mixer (M4,  0.5 mm, ETFE) before passing through a flow coil (R3, 10 mL, PFA) which provided approximately 15 minutes residence time at 25 ◦C. The flow rate of the reagent stream was controlled by a second feedback loop such that the quantity of reagent in the reservoir remained relatively constant, to allow buffering of the variable flow rate of pump P7. A valve (V5) was set to discard the output from the evaporator if the reservoir was close to overflowing (Scheme 4.21). The acid stream was initially set at 0.2 mL/min to provide an excess of acid for the reaction. By using an acid-resistant pump and removing all PEEK components from the fluidic path, we were able to use a stream of neat sulfuric acid at a lower flow rate of 0.05 mL/min, rather than as a solution in acetic acid and acetic anhydride. This reduced the dilution and 96 An integrated flow synthesis of 2-aminoadamantane-2-carboxylic acid N22 L/min HO vapours out waste peristaltic P7 S2 M4 R3 P8 V5 webcam 0.15 - 1.0 mL/min H2SO4 0.05 mL/min 1:2:1 H2SO4 / Ac2O / AcOH 0.2 mL/min 30 min 25 oC P9 or NHO 51 52 Scheme 4.21 The output of the evaporation chamber is directed into reservoir S2. The reservoir is used to buffer the material passed into the Ritter reaction step. Valve V5 is triggered to switch to waste if the reservoir is in danger of overflowing. Pump P9 is switched on only when pump P8 is running. created an overall lower flow rate, and more importantly reduced the quantity of acid that had to be quenched in the following stage. 4.5.3 Base quenching The chemistry of the 5-exo-dig cyclisation reaction (Step 3) had been extensively explored during the development of the first generation synthesis.188 An acid-catalysed cyclisation of compound 52 had been reported204 but this reaction was very slow, requiring up to 6 hours and was thus unsuitable for a flow process. The base-catalysed reaction was much more efficient, being complete in 40 – 60 minutes at 120 ◦C. The key consideration was to avoid the introduction of significant quantities of water, because this had been found to result in hydration of the residual acetonitrile, competing with the desired reaction and reducing the overall rate of cyclisation. Thus, a solution of potassium hydroxide in 40 : 1 ethanol / water (v/v)188 was initially used to quench the acidic stream. Removal of the salts afforded a primarily ethanolic stream which could be used directly in the cyclisation reaction. It was very important to remove all of the solid material generated by the quenching reaction, since it had the potential to damage the low-pressure reagent selection valves or block the check valves in the pump, causing it to fail. Therefore, a number of strategies for removing the salts formed during the quenching reaction were explored. 4.5. Ritter reaction and quenching 97 Filtration method Initial attempts to perform this filtration continuously were promising, as combination of the two streams within a second tube-in-tube tee mixer (M5) generated a slurry which passed through a wide-bore residence tubing (R4;  2.4 mm, 30 cm, ETFE) and was dropped onto a sintered glass filter funnel (Figure 4.22). In dilute cases this worked well, but at the full concentration, precipitation was not complete by the time it passed though the filter, resulting in large quantities of solid settling in the reservoir. The ethanolic slurry had a high viscosity, and R4 eventually became blocked with solid. The slurry was too viscous for a vibrating motor (such as that used for the previous extraction process) to keep it moving effectively. KOH (3 M) 40:1 EtOH / H2O 2-3 mL/min MeCN, H2SO4 0.4 - 0.7 mL/min NHO wide-bore residence tubing to reservoir (S3) a b M5 R4 P10 sand glass sinter funnel52 Figure 4.22 Removal of salts by filtration using a glass sinter funnel. A sand bed prevents the sinter from becoming blocked by the salt. In this case, further dilution of the stream was not practical because the combined flow rate was already high in order to provide sufficient base to completely neutralise the acid. Since a relatively long residence time (up to an hour) was required for the cyclisation step there was a practical limit on the flow rate for this step without needing a reactor with a very large volume. Extraction method With this restriction on the flow rate in mind, we considered an alternative system involving a second extraction. It was possible to use a biphasic stream of Hünig’s Base (DIPEA) in cyclopentyl methyl ether (CPME, a high boiling point solvent which has a low miscibility 98 An integrated flow synthesis of 2-aminoadamantane-2-carboxylic acid with water) and aqueous KOH to neutralise the acid and remove the generated acetate salts in the aqueous phase. Under these conditions, the resulting slurry after mixing had a significantly lower viscosity compared to the case where ethanolic KOH was used. Aided by a second vibrating micromotor, blockages at mixer M5 were avoided. The slurry was directed into a commercially available phase separation device182 containing a replaceable expanded macroporous PTFE membrane, which allows only the organic phase to pass through (Figure 4.23). A 5 M aqueous solution of KOH could be used at a lower flow rate, ensuring complete quenching of the acid stream whilst reducing the volume of solvent required. KOH (4 M) H2O1 mL/min DIPEA (0.5 M) CPME 0.5 mL/min MeCN, H2SO4 0.4 - 0.7 mL/min NHO wide-boreresidence tubing hydrophobic membrane separator to reservoir (S3) aqueous and salts out peristaltic a b M5 R4 P10 P11 P12 52 Figure 4.23 (a) Hydrophobic membrane separation apparatus. The acidic stream is mixed with a biphasic base stream and then directed into the membrane separator. The organic phase passes through the membrane and drops into the reservoir; the lower aqueous phase is removed by peristaltic pump P12. (b) The separator consists of a chamber and PTFE membrane. A custom glass chamber was made such that the liquid level might be monitored by a camera and modulated with a pump such that none of the organic phase was removed by the peristaltic pump. The sulfate salts were not soluble in aqueous or organic solvents, but we envisaged that a peristaltic pump could remove the remaining aqueous phase and sulfate salts from the bottom of the separator. The organic solution passed through the membrane and dripped into the reservoir for use in the following step. We were pleased to find that a good extraction was achieved in this case, and that heating of compound 52 with DIPEA in CPME led to the desired cyclisation. Unfortunately, the peristaltic pump was not able to remove the solids from the phase separator efficiently, and its inlet tube blocked rapidly (the peristaltic tubing did not 4.5. Ritter reaction and quenching 99 withstand immersion in CPME, so a fitting was required to connect a PTFE tubing which would extend into the phase separator. The blockage occurred at this fitting). Therefore, we turned our attention to further development of the filtration method to remove this salt. Continuous filtration method An effective neutralisation method was developed by using a KOH stream with a mixture of ethanol and water as solvent. By adjusting the amount of water in this solution, the viscosity of the resulting mixture was minimised to prevent blockages in the mixer, whilst still retaining a high conversion in the subsequent step. The salt that was produced was removed by a filter allowing the remaining solution to pass through into reservoir S3. We envisaged that some of the water would be retained by the salt so that only a small amount of the water would pass into the cyclisation step. Using a glass sinter attached to a motor to allow it to rotate, a continuous filtration platform was constructed (Scheme 4.24, Figure 4.25a).205 The slurry passing through R4 was dripped onto the rotating disc; the rotation allowed the liquid to pass through before the residual solid was scraped off. A replaceable paper filter caught the solid, whilst allowing the liquid to percolate through. A hole in the centre of this filter allowed most of the solution that passed through the sinter to drip straight through it. A final thin layer of cotton wool was added to stop any large solids that were not caught by the paper filter. This system was found to operate successfully for periods of up to six hours, with periodic replacement of the filters. KOH (4 M) 4:1 EtOH / H2O2 mL/min MeCN, H2SO4 0.4 - 0.7 mL/min NHO wide-bore residence tubing to reservoir (S3) cotton wool replaceable lter solids removed rotating sinter liquids pass through M5 R4 P10 52 Scheme 4.24 Continuous filtration apparatus. The slurry stream was dropped onto a sintered glass plate that was attached to a rotating motor. The liquid passed through the sinter into the reservoir, and excess solids were pushed off by a fixed bar onto a paper filter, from where any remaining solution could percolate through. 100 An integrated flow synthesis of 2-aminoadamantane-2-carboxylic acid a b Figure 4.25 (a) Continuous filtration apparatus. (b) Reservoir S3. The plastic surround provides a blank background for the camera, and the dots drawn on the reservoir trigger the camera’s autofocus on the optimal focal plane. 4.5.4 Cyclisation reaction The 5-exo-dig cyclisation was performed by passing the basic stream collected in reservoir S3 (Figure 4.25b) through a flow coil heated to 120 ◦C (Scheme 4.26). Another 20 µm frit in the bottom of the reservoir protected the pump from any solids that managed to pass through the main filtration device. The flow rate was controlled by another feedback loop based on the volume present in the reservoir (Figure 4.27). Although this resulted in a fluctuating flow rate for the subsequent step, a large 50 mL reactor coil (R5,  2.5 mm, SS) provided sufficient residence volume for the flow rate to be varied between 1 to 3 mL/min without affecting the degree of conversion of the reaction. KOH, EtOH NHO S3 P13 webcam 1.0 – 3.0 mL/min R5 25 min (average) 120 oC 5 bar gas-pressure BPR N O N2 gas out 52 53 Scheme 4.26 Cyclisation step apparatus. Intermediate 52 was collected in reservoir S3 and then pumped through reactor R5 at 120 ◦C to effect the cyclisation. Feedback control based on the liquid level of S3 attempted to match the flow rate of P13 with the rate of the solution passing through the filter into S3. 4.6. Ozonolysis and Hydrolysis 101 Problems with blockages in the BPR were encountered when using a standard spring-based BPR after R5. This was avoided by using a prototype gas pressure BPR developed within our group.190,206 This device consists of a sealed vessel pressurised using a N2 supply which can be connected at the end of a flow system. The product 53 was periodically drained from the vessel using a manual valve at the bottom. Cyclisation Reaction PI Control max 3 mL/min min 1 mL/min Set heaters to 120 °C Wait for heaters to heat up Set pump P13 to pump solvent at 0.1 mL/min Set pump P13 to pump reagent at 1 mL/min Run control system until end of experiment Wait until reservoir S3 contains enough material to start Wait for reservoir S3 to start to ll up Reservoir S3 oat height Reagent pump P13 rate Figure 4.27 Control protocol for the cyclisation step. The reaction started when sufficient material has been collected in reservoir S3, and then the flow rate of pump P13 was controlled based on the liquid level. Unfortunately, this device could not then be connected to feed into the next step. Further- more, the base had to be removed before the final hydrolysis step to prevent salt formation which might complicate the purification of the final compound. We were also concerned that if this basic stream was mixed with a gaseous stream in the ozonolysis reaction, deposi- tion of salts within the mixer could block the reactor. Therefore, the synthesis was broken at this point to allow manual solvent evaporation and aqueous extraction to be performed. 4.6 Ozonolysis and Hydrolysis To improve on the first generation flow synthesis, we envisaged that reported ozonolysis conditions using an acetone/water solvent207 could be used to decompose the carbonyl oxide intermediate in situ, avoiding the formation of an ozonide and thus avoiding the requirement for the solid-supported thiourea reagent. Pleasingly, these conditions proved to be very effective, generating a mixture for which the NMR spectra were compatible with a 1 : 1 ratio of azlactone 54 and uncyclised 56,188 along with formaldehyde and hydrogen peroxide by-products. The peroxide was removed by a bed of manganese dioxide,208 through which the liquid output of the ozonolysis reaction was allowed to percolate. The output was directly combined with an acidic stream to perform the hydrolysis of both compounds to the desired amino acid 44. The product was collected from the solvent 102 An integrated flow synthesis of 2-aminoadamantane-2-carboxylic acid O3 ozone generator O2 + O3 1:5:8 HCl, AcOH, H2O 0.25 mL/min MnO2 5:1 acetone / H2O 2 mL/min 0.25 mL/min ozonolysis reactor (solvent evaporator) 18 min 150 oC O2 N O N O O H2N O OH HCl NHO OH O 100 psi + P14 P15 P16 R6 R7 53 54 56 44 Scheme 4.28 Ozonolysis and hydrolysis sequence. A stream of compound 53 in acetone / water was combined with a gaseous stream of O3/O2 from the ozone generator. These were mixed and allowed to react in R6, and then directed into column R7 from which the gases could escape and a plug of MnO2 decomposed the hydrogen peroxide by-product. The output was combined with an acidic stream and heated to effect hydrolysis before being collected using a second solvent evaporator device. stream by evaporation of the acids and solvents (Scheme 4.28). The heated output of reactor R7 was passed directly into a solvent evaporator without cooling. The stream at 150 ◦C was vapourised very efficiently, so all solvents and acids were removed to leave a solid precipitate of compound 44. 4.7. Conclusion 103 4.7 Conclusion The three chemistry steps and three work-up steps to prepare the cyclised compound 53 were combined into an integrated sequence which could operate semi-autonomously for at around six hours to produce 9 g of 53 (92 %, 9.0 mmol/h). The integration of flow chemistry steps represents a significant challenge, as when multiple devices are used there are many parameters which must be managed simultaneously. There is an even greater challenge when suspensions are generated or used, because key components of flow chemistry apparatus that are currently standard are not suitable for handling solids (in particular: most pumps, mixers and connections, and BPR assemblies). Technologies adapted from those used for larger scale systems may help to enable such processes. The combination of a tube-in-tube mixer and a vibrating micromotor was used to avoid blockages whilst performing the quenching reactions, during which which precipitates were formed. A prototype continuous filtration device was used to remove the salts formed during a continuous quenching reaction, allowing two reactions with different pH conditions — a Ritter reaction and a base-catalysed cyclisation — to be performed in succession. A nebulising evaporator was used to perform an in-line solvent exchange, allowing the Grignard and Ritter steps — with incompatible solvent systems — to be performed in sequence. Finally, the ozonolysis reaction was improved by the use of an aqueous solvent system and manganese dioxide catalysed decomposition of the hydrogen peroxide by- product instead of a solid-supported thiourea. Software control helped to make the management of multiple devices feasible. A camera- enabled system controlled the rate at which the aqueous phase was withdrawn from the separation tank, to compensate for flow rate fluctuations in the input streams. Similarly, the application of camera-enabled reservoirs meant that the steps could be started automatically as soon as the reagent solutions became available, and stopped if upstream problems caused the input stream to stop. Feedback control loops were used to match the flow rates between these steps so that they did not stop and start more frequently than necessary. The inclusion of these reservoirs meant that the software control could be designed in a modular fashion, with each step being mostly independent, resulting in a simpler control programme. 104 An integrated flow synthesis of 2-aminoadamantane-2-carboxylic acid 4.7.1 Further work A number of potential improvements were identified, to advance this project or to enable similar projects in the future. Further development of the control software could allow the system could be left running unattended overnight. The key to this would be a safe automatic shut-down in the case of a problem, in a manner that left the system in a state that it could re-start immediately as soon as the problem was rectified. The second half of the reaction sequence could also be fully automated by the inclusion of additional reservoirs to connect the steps. Improvements to the filtration device might result in a device which is less susceptible to clogging by the solid material, and from which removal of the collected salts is easier. We envisage that this might be a device based on a centrifugal principle, or a device including a pressure differential across the filter, to force the liquid through and leave a dry solid residue to be removed. The addition of an internal liquid level sensor and a controllable valve at the bottom could enable the gas pressure BPR to be fully automated, by periodically releasing the collected material into a reservoir feeding another reactor. It might be possible to develop a self- regulating solvent evaporator by the application of computer vision to measure the quantity of solids and liquid inside the evaporation chamber, and adjust the temperature and flow rates accordingly. With further development of these devices, techniques and software, it is possible that in the future integrated processes such as this one might be can be constructed and used as a matter of course, rather than having to be developed as a dedicated project. Chapter 5 Development of inhibitors for bromodomain-containing proteins 105 106 Development of inhibitors for bromodomain-containing proteins 5.1 Introduction A typical process for the discovery of a biologically-active molecule, whether for phar- maceutical or for research purposes, often includes hit identification, lead optimisation and then a structural optimisation stage based on the results of biological testing. “Hit” compounds, which are compounds with a low but measurable activity against the target, are often identified by either physical or virtual high-throughput screening (HTS) of a library of compounds against a biological target. The efficiency of HTS methods has increas- ingly been called into question209 and more recently, fragment-based discovery210–212 has become more common. This entails the measurement of weak activities of small “frag- ments”,* followed by the design of more active compounds by connecting these fragments together to form larger hit molecules. By whichever method they are identified, the compounds that are from the first stage will give an idea of the structural features that may be important for binding, and thus demarcate a region of “chemical space” for further investigation. Subsequently, chemical synthesis of a number of similar compounds will allow the biological activity of these chemotypic regions to be described in more detail, and provide information about how the structure influences the activity: the structure–activity relationship (SAR). Repeated cycles of design and experimentation may eventually provide a potent and selective lead compound. There are a number of companies and academic research groups investigating how this cycle (or parts of it) can be automated to speed up the development process, and to what extent this can augment the traditional discovery pattern. The reports thus far have often focused on increased automation: to achieve 24/7 working and produce greater numbers of test compounds. Some have implemented machine learning or intelligent search protocols to improve the automated compound design process so as to reduce the number of trials,213 in much the same way as optimisation protocols for individual reactions harness numerical and linear programming methods for increased efficiency.160,164 The question of whether an automated process can contribute to a molecular design project by performing intelligent ligand design with efficient use of the structure–activity data has not yet been answered. A well-designed algorithm can make decisions in well-defined and reproducible ways, and investigation into the most effective way to incorporate this into a discovery programme is still ongoing. * Compounds with fewer than 16 heavy atoms and a high proportion of hydrogen-bond donors and acceptors. 5.1. Introduction 107 The generation of realistic chemical complexity may well be beyond the scope of current compound-design algorithms,214 particularly if a route for manual or automated synthesis of each novel structure must be suggested as well. Fortunately, a great deal of research is ongoing into these areas.215 Finally, given the huge challenge of predicting the activity of a certain ligand when com- bined with a protein, the SAR method is a key technique for compound development. An individual novel compound may have no detectable activity, but an array of 20 similar compounds might contain one or two active molecules.216 Alongside work to further develop machine-assisted techniques for producing arrays of functional compounds,24,50,112 there is also ongoing work within our group to apply in- telligent automation to close the loop of lead optimisation by integrating chemistry and biology.217 To understand the challenges involved in a typical discovery programme we undertook a synthesis assignment in collaboration with the Structural Genomics Consortium (SGC). 5.1.1 The bromodomain A key research area in which the SGC is involved is in the study of human proteins, especially those containing bromodomain (BRD) modules, and ligands which modify or inhibit their function. Bromodomains constitute one part of a large family of protein modules which assist in gene regulation by recognising or performing post-transcriptional modifications of chromatins. The bromodomain is a protein domain which recognises –N–acetylated lysine residues on a histone protein.218 Histones form a scaffold that DNA winds around to form nucleosomes, which condense to form a chromosome. One effect of acetylation is to alter the charge on the histone; this is thought to weaken the association between the nucleosomes, allowing the genes in that particular region of DNA to become more accessible to the transcription factors that start the process of protein synthesis (Figure 5.1). Therefore, bromodomains are a key instrument in gene regulation within organisms and thus an important area for study, in particular in relation to diseases such as cancers.218 Over 40 proteins in the human body have been identified which contain bromodomains.218 Despite significant differences in the amino acid sequences of the BRD modules, they have a conserved structure formed of four α–helices which surround a hydrophobic acetylated 108 Development of inhibitors for bromodomain-containing proteins N H O Transcription factor Inaccessible gene Acetylated histone DNA Histone Histone tail Accessible gene Figure 5.1 Histones are proteins around which DNA is wrapped within a chromosome. Acetylation of the histone tails is an epigenetic modification which (amongst other effects) is thought to weaken the association between adjacent histones, allowing greater access of the genes to transcription factors, thus affecting gene expression. lysine binding site.218 The role of BRDs is poorly understood, and hence there is a need for selective ligands. Investigation by the SGC had bromosporine (57) as a potent inhibitor of many BRDs.219 Figure 5.2 shows a co-crystal structure of bromosporine with the BRD4 bromodomain unit. The key interactions are those between ASN-140 and the northern triazole and its pendant carbamate NH. The southern tolyl moiety displays a pi–pi interaction with TRP-81, and the sulfonamide unit with ASP-88. Two regions of this molecule were identified for further exploration: the heterocycle amine and the sulfonamide. Depending on the observed activity of the analogous compounds, the objective would be to develop either (a) a more potent pan-bromodomain inhibitor, or (b) a more selective inhibitor for one or more BRDs. a b Figure 5.2 (a) Crystal structure and ligand interaction diagram of the pan-bromodomain inhibitor bromosporine (57) bound into the BRD4A(1) bromodomain. (b) The key interaction between compound 57 and BRD4A(1) is from the triazine nitrogen atoms to an asparagine residue and a water molecule. 5.1. Introduction 109 5.1.2 Frontal Affinity Chromatography Recently, the technique of Frontal Affinity Chromatography (FAC)220 on a miniaturised scale has been developed.217 This method allows an assessment of the binding strength of a test compound to be made using chromatographic equipment and an immobilised protein. This technique was originally used for studying the interaction between carbohydrates and lectins,220,221 but when used in combination with mass spectrometry can be applied to a wider range of biological targets.222–225 Miniaturisation of the column containing the immobilised protein reduces the amount of protein that is required: the high cost of some proteins means that this is a very important consideration, and smaller columns offer the ability to measure affinities with smaller quantities of ligand (which may also be highly valuable). Proteins can be immobilised using a number of methods, such as by covalent attachment223 or more commonly by a streptavidin–biotin H–bonded linkage.217 Since a protein can be biotinylated at either the C or the N terminus, the latter is more likely to lead to a site-selective immobilisation (i.e. where all of the protein molecules are attached from the same position on the protein) so that the interaction of substrates with protein is consistent. Biotin units can be attached to the terminus of a protein by expressing an avidin tag and then performing a biotinylation;226 the protein can then be immobilised in a column containing solid-supported streptavidin units (Figure 5.3). The interaction of biotin with the streptavidin protein is the strongest known biological non-covalent interaction, having a Kd of the order of 4×10−14 M;227 this strong attachment is crucial because it prevents the protein from being washed off under a continuous solvent stream. Biotinylation Enzyme Immobilisation Supported Streptavidin Figure 5.3 A protein (blue) with an avidin tag expressed onto the protein terminus is biotinylated (red) and immobilised by being captured by a solid-supported (green) streptavidin unit (brown). The streptavidin complex has four units and thus four binding sites, but due to the size of the protein they may not all be filled. Under a FAC protocol, a relatively large quantity of a compound of interest is infused into the protein-containing column. Whilst an inactive compound will pass through in one column volume, an active compound will bind to the protein and be retained. The “front” of 110 Development of inhibitors for bromodomain-containing proteins Co nc en tr at io n Void Marker Compound Volume Δv Immobilised Protein No anity = No retention Anity = Retention Figure 5.4 In a FAC analysis, the compound of interest is continuously infused into a column containing an immobilised protein. Compounds are retained according to the strength of their affinity for the enzyme. The breakthrough volume is (arbitrarily) chosen to be the mid-point between the lowest absorption and the plateau, and is recorded relative to that of a void marker that has no affinity for the protein. the compound will emerge only after all of the binding sites have filled up (Figure 5.4). The relative binding affinities of two compounds can be compared by measuring the respective breakthrough volumes (which are proportional to the breakthrough times). The FAC platform consists of an HPLC system running an isocratic buffered aqueous solvent system, which is important for maintaining the stability of the immobilised protein. Downstream of the column there are standard analytical detectors, such as a UV diode array detector or a mass spectrometer (Figure 5.5). By injecting a series of compounds of the same concentration, an order of affinity can be es- Compact mass spectrometer HPLC Pump 15 µL column Make-up pump Dynamic mixer (removes pulsing) Buer Solutions UV/Vis Detector Column Oven Figure 5.5 The Frontal Affinity Chromatography apparatus consists of an HPLC system, with a column (right) containing the immobilised enzyme. The pump delivers an isocratic stream of an aqueous buffer, and 600 µL of the sample solution is infused (large compared to the 15 µL volume of the column) into the FAC column that is maintained at 20 ◦C by the column oven. The output is detected by a UV/Vis diode array detector and/or a mass spectrometer. 5.1. Introduction 111 tablished. Alternatively, by injecting each compound as a series of different concentrations, the binding coefficient Kd can be calculated from the breakthrough volumes. As part of this project we hoped that the FAC technique could be used to discriminate between molecules and give results which were consistent with mainstream assay techniques. Processing FAC data If a solution of substrate A with concentration [A0] is continuously infused into a column on which there are Bt immobilised protein units, then some of the substrate molecules will bind to the proteins and be retained on the column. The number of molecules of substrate removed from the stream corresponds to the retention volume: Bocc = ∆V [A0] = (V −V0)[A0] (5.1) where V is the substrate’s breakthrough volume, and V0 is that of a void marker. The ratio of occupied (Bocc) to unoccupied (Bunocc) sites will depend on the binding affinity Kd . They are related by the equilibrium: A+Bunocc k+1−−⇀↽− k−1 ABocc (5.2) and assuming that the activity quotient of the equilibrium is constant: Kd = k−1 k+1 = [Bunocc][A] [Bocc] (5.3) This assumption is reasonable in this case because the analysis is taking place in a buffered aqueous solution, which has a high ionic potential.228 Since the protein is immobilised, [B] = B Vcolumn (5.4) where Vcolumn is the volume of the column. Since Bt = Bocc+Bunocc, Kd = (Bt−Bocc)[A] Bocc (5.5) = Bt [A] Bocc − [A] (5.6) 112 Development of inhibitors for bromodomain-containing proteins Therefore, using equation (5.1): Kd = Bt [A] (V −V0)[A0] − [A] (5.7) Under the conditions of constant infusion, [A] = [A0] and so Kd +[A0] = Bt (V −V0) (5.8) (V −V0) = BtKd +Bt 1 [A0] (5.9) This is analogous to a Michaelis-Menten system, and so a Lineweaver-Burk linearisation229 can be used to calculate Kd and Bt by plotting (V −V0) against 1[A0] . 5.2 Chemical probes for BRD9† 5.2.1 Literature Previously reported approaches to the triazolopyridazine core such as seen in bromo- sporine (57, Figure 5.6) have generally proceeded by generation of the triazole 60 from a dichloropyridazine precursor 58, using for example acetyl hydrazide231,232 or in two steps using hydrazine and then either triethyl methanetricarboxylate,233 acetic acid234–236 or acetic anhydride237,238 (Scheme 5.7). These reports suggest that after an SNAr reaction N N N N N H S OO H NEtO O Figure 5.6 Bromosporine (57). † The work described in this section is reported in MedChemComm (Reference 230). 5.2. Chemical probes for BRD9 113 onto one of the C−Cl positions, the other bond becomes deactivated and so only single substitutions are observed. N N Cl Cl R N N HN Cl RN2H4.H2O NH2 N N Cl Rconditions NN AcNHNH2 58 59 60 Scheme 5.7 Reported preparations of the triazolopyridazine core, by either hydrazination followed by cyclisation, or by direct cyclisation using a hydrazide. R = H, Me,239 NH2 235 or CO2H. 236 A standard method for the preparation of the pyridazine starting material 58 is by treatment of furan-2,5-dione (61) with hydrazine followed by chlorination of the pyridazine-3,6-diol intermediate 62240 (Scheme 5.8). However, many 3,6-dichloropyridazine derivatives are now commercially available. O O O N2H4.2HCl NH NH O O N N OH OH N N Cl Cl POCl3 61 62a 62b 63 Scheme 5.8 Synthesis of 3,6-dichloropyridazine (63) from furan Cross-coupling of triazolopyridazine 60 (R = H) is well represented in the literature, with examples of Suzuki,241 Stille,242 Negishi243 and other metal-mediated reactions. However, to the best of our knowledge, there have been no reports of cross-coupling reactions onto substituted triazolopyridazines. Cross coupling of an analogous imidazopyridazine has been established244 using palladium acetate and S-Phos (66; Scheme 5.9a), and more recently245 using Pd2(dba)3 and X-Phos (69; Scheme 5.9b). Finally, nucleophilic substitution at the same position has been reported using secondary amines, although low yields were reported (Scheme 5.9c).236 114 Development of inhibitors for bromodomain-containing proteins N N N Cl H2N N N N Ar H2N ArBPin (1.5 equiv.) Pd(OAc)2 (10 mol%) S-Phos (20 mol%) K3PO4 (2 equiv.) 5:2 n-butanol / water 100 °C, 48 h 23% PCy3 OMeMeO S-Phos a 64 65 66 N N N Cl H NR N N NH NR R = 6-(piperidin-1-yl)pyridin-2-yl PhB(OH)2 (1.5 equiv.) Pd2(dba)3 (10 mol%) X-Phos (40 mol%) K2CO3 (3 equiv.) 10:1 dioxane / water 100 °C, 4 h 58% PCy3 i-Pri-Pr i-Pr X-Phos b 67 68 69 N N NN Cl MeO2C N N NN N DMSO 130 °C (MW) 30 min 15% Ph H N MeO2C Ph c 70 71 72 Scheme 5.9 (a) Cross-coupling of imidazopyridazine 64 with an arylboronic ester using Pd(II) and S-Phos.244 (b) Cross-coupling of imidazopyridazine 67 with phenylboronic acid using Pd(0) and X-Phos.245 (c) SNAr reaction of triazolopyridazine 70 with a secondary amine.236 SGC synthesis The synthesis of 57 as reported by the SGC219 began with a sequence to form the tria- zolopyridazine 76 from 3,4,6-trichloropyridazine (73). An SNAr reaction using ammonia N N Cl Cl Cl N N Cl Cl H2NNH3 EtOH 51% N N HN Cl H2N 40% N2H4.H2O NH2 N N Cl H2NAcOH NN 82% 73 74 75 76 Scheme 5.10 Synthesis of precursor 76 starting from 3,4,6-trichloropyridazine (73). 5.2. Chemical probes for BRD9 115 N N Cl H2N NN Boc2O DMAP THF 0 °C to r.t. N N Cl BocHN NN Boronic acid___ Pd(PPh3)4 K2CO3 Dioxane / H2O 120 °C N NBocHN NN NO2 N NH2N NN NO2 TFA, CH2Cl2 25 °C EtOCOCl NEt3 CH2Cl2 25 °C N N H N NN NO2 O EtO SnCl2.H2O EtOH reflux N N H N NN NH2 O EtO N N H N NN N H O EtO MsCl, Pyridine 25 °C 78%87% 23% 100% 44%40% S OO NO2 B(OH)2 76 77 78 79 8081 82 57 Boronic acid 78 Scheme 5.11 Synthesis of bromosporine from precursor 76. afforded 4-amino-3,6-dichloropyridazine (74), before hydrazination and cyclisation with acetic acid (Scheme 5.10). Attempts to perform cross-coupling reactions with 76 gave only traces of the desired products.246 To overcome this problem, a protection strategy was adopted whereby the free amine was carbamoylated. The cross-coupling reaction then proceeded in acceptable yield to give an arylated product that was deprotected with trifluoroacetic acid (TFA). Following deprotection, the desired carbamate moiety was installed (81) before the pendant amine was revealed by a reduction with tin chloride. The methanesulfonyl group was then installed to provide the desired compound 57 (Scheme 5.11). 116 Development of inhibitors for bromodomain-containing proteins 5.2.2 Proposed synthesis In order to produce analogues of 57 a more efficient synthetic protocol than the SGC protocol previously described was attempted. We hypothesised that electron-rich phos- phine ligands for palladium such as S-Phos or X-Phos247 could make the cross-coupling more efficient, perhaps without the need for a protecting group; and that use of the com- pleted sulfonamide as cross-coupling partner would lead to a more convergent synthesis (Scheme 5.12). N N Cl H2N NN Pd / Ligand N NH2N NN RR B OO 76 83 84 Scheme 5.12 Proposed cross-coupling between 76 and a boronic ester. In addition, the original starting material 3,4,6-trichloropyridazine (73, see Scheme 5.10) was prohibitively expensive at the time, and so a route based on the more readily available carboxylic acid derivative 85 was used. There were a number of different options to explore for this synthesis, depending on the order in which the synthetic steps were to be carried out (Scheme 5.13). 5.2.3 Synthesis of core Carboxylic acid route (Route I) Previous work by Dr Nikzad Nikbin248 had found that protecting the carboxylic acid group of compound 85 as an ester was difficult and only possible using diazomethane, providing the methyl ester in low yield. Other protection strategies had been considered, but each possibility was considered to add considerable complexity to the reaction procedure. Nevertheless, Route I was very attractive: retention of the pendant carboxyl group would enable late-stage installation of the carbamate via a Curtius rearrangement (perhaps even after the cross-coupling reaction had been carried out) for efficient derivatisation at this position. Unfortunately, although compound 89 was detectable by LC-MS the subsequent 5.2. Chemical probes for BRD9 117 N N Cl Cl OCN N N HN Cl H2N NH2 N N Cl H2N NN N N Cl Cl HO2C Curtius N N Cl Cl H2N N N Cl Cl H NRO O ROH N2H4 N N Cl H N NN Cyclisation RO O N N HN Cl HO2C NH2 N N Cl HO2C NN N2H4 Cyclisation N N Cl OCN NN Curtius Route I Route II Route III Carbamate Formation N N HN Cl H NRO O NH2 N2H4 Cyclisation Hydrolysis Carbamate Formation Route IV Hydrolysis Hydrolysis 85 86 R = t-Bu, 87 88 89 R = t-Bu, 90 91 92 R = t-Bu, 77 76 93 Scheme 5.13 Proposed routes to access key intermediate 76 from pyridazine 85. 118 Development of inhibitors for bromodomain-containing proteins cyclisation procedure proved to be very challenging in the presence of a free carboxylic acid, particularly with respect to isolation, and no product (92) could be isolated. Carbamate and amine routes (Routes II and III) Initial experimentation using microwave irradiation suggested that the Curtius rearrange- ment to give 86 was an effective reaction; in situ hydrolysis (Route II) was not successful but reaction with tert-butanol (Route III) to give the Boc product 87 worked well. Unfortunately, the hydrazine derivative 90 was unreactive to the cyclisation conditions and so hydrolysis of 87 to 88 had to be carried out before the cyclisation could be performed (Route IV). Pleasingly, the cyclisation of compound 91 was straightforward, as both intermediate 91 and the product 76 could be isolated in reasonable yield by simple filtration. Early attempts at cross-coupling reactions with compound 77 indicated that considerable optimisation would be required, and that the carbamate group was unlikely to survive the conditions. Thus, more material was to be prepared using Routes III and IV to provide enough of the key intermediate 76 to test a number of cross-coupling conditions. Azide formation and Curtius rearrangement in continuous flow The Curtius rearrangement involves the thermal decomposition of an acyl azide to form an isocyanate.249 Carboxylic acid starting materials 94 can be converted directly to acyl azides 95 using diphenyl phosphoryl azide (DPPA, 96) as the azide source (Scheme 5.14).250 R OH O R N O N N R N O N N R N C Oheat - N2 azide source R H NNuH Nu O 94 95 97 98 Scheme 5.14 The Curtius rearrangement transforms an acyl azide (95) to an isocyanate (97). This can subsequently react with a nucleophile such as water or an alcohol to form an amide (98). There are a number of hazards associated with this reaction. DPPA is toxic and explosive, nitrogen gas is released, and the isocyanate products are also toxic and very reactive. Most of these problems can be avoided by performing the reaction in continuous flow:19 the small volume of a flow reactor means that only a small quantity of the DPPA is heated at any one 5.2. Chemical probes for BRD9 119 time; a back-pressure regulator (BPR) can be used to contain the gas that is released; and the isocyanate can be reacted in situ to avoid holding stocks of it. When an alcohol such as tert-butanol is added to the reaction mixture this combines with the isocyanate intermediate in situ to produce a carbamate. In the flow reactor, a stream of DPPA was combined with a stream of the carboxylic acid, triethylamine and tert-butanol at a tee mixer (M1;  0.5 mm, ETFE) and then passed into a series of four heated flow coils (R1;  1 mm, PFA, total volume 40 mL). The efflux passed through a BPR (100 psi) and then a collection valve V1. Initial runs encountered problems related to precipitation of a gummy solid during the reaction. This caused blockages in a number of places, notably in the heating coils, and at the BPR and the collection valve. Optimisation of the solvent system and flow rates led to Table 5.15 Development of conditions for the Curtius reactions in flow. 100 psi 120 °C time waste M1 R1/R2 V1 n Toluene / MeCN Toluene / MeCN N N Cl Cl H Nt-BuO O (0.25 M) NEt3 (1.0 M) t-BuOH (1.5 M) DPPA (0.375 M) 85 87 Entry Solvent ratio Scale Reactor Flow Rate Residence Yield (Tol. / MeCN) /mmol volume /mL /mL/mina time /min 1248 100 : 0 2b — — 240 0.4 g (77 %) 2248 50 : 50 4 20 0.20 100 0.3 g (36 %) 3 0 : 100 2 10 0.10 100 0.1 g (15 %) 4248 70 : 30 6 20 0.17 120 0.5 g (32 %) 5 90 : 10 20 20 0.17 120 — c 6248 80 : 20 10 20 0.17 120 1.1 g (45 %) 7 80 : 20 40 30 0.17 180 3.2 g (30 %)d 8 70 : 30 20 20 0.17 120 2.1 g (40 %) 9 70 : 30 20e 20 0.17 120 2.6 g (50 %) 10 70 : 30 40e 50 0.36 140 — f 11 70 : 30 40e 50 0.36 140 8.5 g (39 %)g a Flow rate of each stream. b Performed in batch, in a sealed vial under microwave irradiation. DPPA was added in three portions. c Rapid blockage in the BPR. d Blockage after 10 hours. e Reactor R2 used; valve V1 not present. f Blockage after 17 hours. g Combined yield from two 40 mmol batches. 120 Development of inhibitors for bromodomain-containing proteins improvements in the performance of the flow system (Table 5.15, entries 2 – 8), but the reactor would frequently become blocked after long periods of operation. To minimise the risks of such blockages, the flow coils R1 were swapped for two larger- bore alternatives R2 ( 2.5 mm, SS, total volume 50 mL) and valve V1 was removed (Figure 5.16). The proportion of acetonitrile in the solvent was increased to 30 % which improved solvation of the precipitates without compromising the yield (Table 5.15, entries 9 – 11). Although the BPR was a frequent location for blockages, it was essential for preventing the solvent from boiling during the reaction and for containing the evolved N2 gas. We wanted to run the reaction for long periods of time in order to obtain a large quantity of 100 psi 120 °C, 2.3 h 2 x 25 mL steel reactor T-mixer 21.5g 21.3g M1 R2 computer residual mass pressure temperature remote access (0.25 M) NEt3 (1.0 M) t-BuOH (1.5 M) 7:3 PhMe / MeCN DPPA (0.375 M) 7:3 PhMe / MeCN N N Cl Cl H Nt-BuO O a 85 87 b Figure 5.16 Curtius reaction with remote monitoring and attempted mass monitoring, using wide-bore stainless steel heated flow coils. (a) Reactor schematic, showing data connections to the control computer. (b) Photograph of system. 5.2. Chemical probes for BRD9 121 the carbamate intermediate 87. Under standard control, the increase in pressure created by a blockage caused the reactor to stop immediately; often the contents of R2 — 40 mL of reagents — would be lost. Immediate notification in the case of a blockage would mean that the material already inside the reactor could be collected and cycled again later, conserving the valuable carboxylic acid starting material 85. Using the software described in Chapter 2 we created a control sequence to run this reaction and allow live monitoring of the reactors. Alerts were set to be triggered by elevated pressures or if the reactor stopped due to an over- or under-pressure. The ability of the software to stop a procedure mid-sequence and then resume it from the same point enabled the reactor to be cleaned out when blocked and for the contents to be recovered, before re-starting the reaction. Incorporation of balances in flow synthesis After the reaction was nominally complete it was discovered that a significant quantity of the solution of pyridazine 85 was remaining. Despite both pumps working as expected when tested independently before the reaction, the observation of excess reagent suggested that this pump was not delivering the correct flow rate during the reaction. Digital balances were used to record the mass of liquid within the reservoirs (Figure 5.16). The rate of change of the mass remaining in each reservoir could be calculated, and would remain constant with a consistent flow rate through the pump. The control software would also be able to make decisions based on the masses (Figure 5.17), such as switching the pump input valves to solvent when either reagent was about to be used up to avoid infusing air into the pump or wasting reagent. The software could also make real-time adjustment of the flow rates to achieve a steady flow rate. 55.2g mass derivative ow rate controlled Figure 5.17 The mass of the reagent was measured by a digital balance. The first derivative of the measurement should be zero to indicate a constant flow rate. Deviations in the first derivative could be fed back to the pump to adjust its flow rate. 122 Development of inhibitors for bromodomain-containing proteins -60 -40 -20 0 20 40 60 0 1 2 3 4 5 6 7 8 Ma ss /g Time /h Balance 1 Balance 2 a -6 -5 -4 -3 -2 -1 0 1 0 1 2 3 4 5 6 7 8 M as s c ha ng e /1 0- 3 Time /h Balance 1 b 0 5 10 15 20 25 30 0 1 2 3 4 5 6 7 8 Pr es su re /b ar Time /h Pump 1 Pump 2 c 0 4 8 12 16 20 24 5.4 5.5 5.6 5.7 5.8 Pr es su re /b ar Time /h Pump 1 Pump 2 d Figure 5.18 (a) Change in mass of two balances over the course of a reaction. The difference in zero indicates that they were not equally tared at the start. The slow decrease after the end of the experiment may suggest evaporation of the residual solvent. (b) The derivative of the mass has too much noise to be useful. (c) There was a problem with the pressure of pump B near the end of the reaction. (d) Close-up of pump B’s pressure loss. Unfortunately, in this system the flow rate was too low to be reliably calculated in real-time based on the change in mass. The available balances were only accurate to 0.1 g, and so the differential of the reading produced was very noisy even after being passed through a smoothing filter (Figure 5.18). The recorded data could be used to calculate an approximate flow rate over the entire reaction, but were not adequate for real-time decision making.‡ Purification The carbamate product 87 was found to be retained on silica giving poor recovery. It was postulated that the reason for this poor recovery may be that the carbamate is lost, leaving the basic amine, as it is known to be removed under the acidic conditions. However, the same poor recovery was again observed even using a basic solvent system previously known to inhibit carbamate deprotection. KP-NH amine-functionalised silica produced by ‡ Pleasingly, later testing with a more accurate balance confirmed that it was possible to visualise a mass change with sufficient accuracy to be a useful measure at low flow rates. 5.2. Chemical probes for BRD9 123 Biotage AB251 was found to offer a good alternative. KP-NH is a proprietary functionalised silica, described as acting like a mixture of primary and secondary amines, suitable for chromatography of amine functionalised materials. This modified silica was found to be effective for the purification of carbamate 87 from the diphenyl phosphate and other by- products of the Curtius rearrangement. The Curtius reaction was performed up to 40 mmol scale over 8 hours to provide 8 g of the product, in 30 – 50 % yield after chromatography, which was sufficient for the following step. The introduction of a batch chromatographic purification meant that it would be difficult to combine this reaction with the following steps without development of an alternative methodology to trap out the by-products. However, there is still a benefit to performing the azide formation and Curtius rearrangement reactions in flow, from the reduced hazards of handling the azide reagent on large scale. Cyclisation The desired amine 88 could be unmasked by removing the carbamate from 87 using aqueous HCl. However, the high solubility of the product 88 in water meant that the greatest isolated yield was achieved when this reaction was performed with an anhydrous solution of HCl in dioxane. A precipitate of compound 88 hydrochloride slowly formed; this was collected by filtration and then re-dissolved in NH3 / MeOH solution. A precipitate of free amine 88 formed, which was collected and washed with a minimum quantity of methanol (Scheme 5.19). N N Cl Cl H2N N N Cl Cl H Nt-BuO O 1. HCl/dioxane (30 equiv.) 25 °C, 24 h 2. NH3, MeOH, 25 °C, 30 min quant. (6.4 g) 87 88 Scheme 5.19 Deprotection of carbamate 87 using acid, and isolation by precipitation from ammonia in methanol. Hydrazine 91 was prepared by heating compound 88 with an excess of hydrazine hydrate. The resulting precipitate provided compound 91 without further purification (Scheme 5.20). Compound 91 was heated in excess glacial acetic acid to give product 76 as a precipitate, which was collected by filtration and purified by recrystallization from water (Scheme 5.21). 124 Development of inhibitors for bromodomain-containing proteins N N HN Cl H2N N N Cl Cl H2N NH2NH2.H2O (30 equiv.) H2O 100 °C (MW), 3 h NH2 80% (combined)(4 × 1.5 g) 88 91 Scheme 5.20 Reaction of aminopyridazine 88 with hydrazine hydrate. N N Cl H2N N N HN Cl H2N AcOH (30 equiv.) 120 °C (MW), 5 h NH2 NN 50% (after recrystallisation) (4 × 1.0 g batches) 91 76 Scheme 5.21 Cyclisation of compound 91 with acetic acid to afford intermediate 76. Development of a flow procedure for these steps was not attempted, because the starting material was relatively valuable and many of the steps up to 76 involved precipitation purification which has not yet been effectively developed for small-scale flow reactions. Attempts to improve solvation of the intermediates by introduction of additional solvents were found to result in lower conversions and increased difficulty of isolation. In summary, all of the reactions from 87 to 76 gave acceptable yields at moderate scale using batch methods, with yields limited mainly by the filtration processes. 5.2.4 Synthesis of boronic ester coupling partners A small collection of boronic esters were prepared in order to make the desired analogues of bromosporine (57). Boronic esters are reactive equivalents to boronic acids for Suzuki couplings, which have the advantage of being crystalline and thus easier to purify and store than the equivalent boronic acids.252 Boronic esters, like other boronic acid equivalents, are converted to boronic acids in the basic conditions of the Suzuki coupling.252 Treatment of (3-amino-4-methyl)phenylboronic acid 99 with a slight excess of pinacol gave complete conversion to the boronic ester (100). Following the removal of the solvent this could be directly combined with a benzenesulfonyl chloride derivative (101) to form the 5.2. Chemical probes for BRD9 125 Table 5.22 Synthesis of boronic ester coupling partners 102. B(OH)2 NH2 B NH2 OO B N H OO S OO Cl Pinacol (1.1 equiv.) MeCN 25 °C, 18 h Pyridine (2 equiv.) EtOH 25 °C, 18 h Cl S OO Cl 99 100 101 102 101 Product Yielda 2-Cl 102a 68 % 3-Cl 102b 55 % 4-Cl 102c 77 % 3,4-diCl 102d 63 % a Isolated yield after double recrystalli- sation from ethanol. coupling partners (102; Table 5.22). These were purified by recrystallization from ethanol in reasonable yield over the two steps. 5.2.5 Cross-coupling The Suzuki reaction The carbon-carbon bond forming cross coupling reaction has been one of the most important recent developments in synthetic chemistry.253,254 The ability to join molecules without using traditional functional group methods created the opportunity to synthesise complex targets more easily, leading to an explosion in the adoption of this strategy.255 Cross-coupling generally occurs between a molecule bearing an electron-poor carbon atom covalently bonded to a halogen or an oxygen atom, and a second molecule with an electron-rich carbon centre bonded to a metal. Of the numerous types of such partnerships that are possible, the palladium catalysed Suzuki cross-coupling reaction254 between an organo halide and an aryl boronic acid is one of the most reliable, having a large substrate scope.255 Importantly, the key boronic acid intermediates have an improved safety profile when compared to equivalent organotin reagents used in Stille coupling:256 boronic acids also have a much lower environmental impact and toxicity.256 Furthermore, aryl boronic 126 Development of inhibitors for bromodomain-containing proteins acids and their derivatives, such as boronic esters,252 boron-MIDA complexes257 and trifluoroborate salts258 are thermally stable and chemically inert, enabling them to be safely stored under standard laboratory conditions whilst being suitable for use in further chemical processing. Novel ligands for the Suzuki reaction have been developed and many are commercially available pre-complexed to palladium. Many of these ligands are much less sensitive to air and moisture than simpler phosphine reagents, thus reducing the requirement for glove-box manipulation. Their better handling characteristics have led to the development of Suzuki reaction methodologies that can be performed in air, and even in the presence of water. The mechanism of the Suzuki reaction (Scheme 5.23)254 is very similar to that of other metal-catalysed cross-coupling reactions,259 being facilitated by a transition metal moving between two oxidation states. The electron-rich Pd(0) catalyst is relatively nucleophilic and can react with an electron poor carbon such as that bonded to a halogen. In this oxidative addition process, electrons move from the Pd to form a Pd−C bond. The resulting Pd(II) complex is relatively electrophilic and liable to be attacked by a carbon nucleophile; in LnPd(0) Cl Pd(II) Ln Cl Pd(II) Ln R'O MOR' R B(OH)2 R B(OH)2 OR' MOR' R'O B(OH)2 OR' Pd(II) L(n-1) R R MCl oxidative addition reductive elimination base organoborate transmetallation organoboronic acid aryl halide metathesis L + L Scheme 5.23 Mechanism of the Suzuki cross-coupling reaction.254 5.2. Chemical probes for BRD9 127 this case, a carbon–boron bond. This transmetallation process leaves two carbon atoms associated to the Pd; their close proximity encourages the formation of a C−C bond. This is the reductive elimination part of the cycle, and the two electrons released by the bond formation reduce the Pd centre to Pd(0), ready to participate in another oxidative addition reaction. The palladium ligand-bound reagents are often added in an oxidised Pd(II) form, which must be converted into the active catalyst Pd(0) before being able to participate in the oxidative addition reaction. This reduction reaction can be performed in situ by the base or the boronic acid260 (the latter process uses up an equivalent of the boronic acid coupling partner). Therefore, minimised catalyst loadings are preferred, being more efficient for cost, for environmental reasons and to avoid increased consumption of the boronic acid. Heteroaromatic cross-coupling The coupling system for the desired reaction (Scheme 5.24) was expected to be challenging. In any aryl ring system, aryl chlorides are the least reactive of the aryl halides towards oxidative addition, because of the lower polarisation of the C−Cl bond compared to C−Br or C−I bonds, which reduces the activity of the oxidative addition reaction.256 Free amine groups compromise Suzuki reactions and are normally protected, because they are thought to bind to the Pd(II) and shut down the catalytic cycle.261 Finally there is a regioselectivity issue with a chloride functionality present on both of the coupling partners. However, we envisaged that the electron-deficient chlorotriazolopyridazine (76) would be active towards oxidative addition, and that this electron deficiency would provide sufficient regioselectivity between it and the aryl chloride coupling partner (102). Dialkylbiarylphos- phine ligands such as S-Phos and X-Phos262 are reported to work well in systems with N H S OO Cl N N NN H2N Cl B N H OO S OO Cl N N NN H2N Cross-coupling 76 102 103 Scheme 5.24 Proposed cross-coupling reaction. 128 Development of inhibitors for bromodomain-containing proteins heteroaryl chlorides,247 and even those with free amine groups.261 The cyclohexyl groups present in these ligands make the phosphine phosphorus electron rich, which has the effect of making the Pd(0) more active to oxidative addition. They are reported to be sufficiently bulky to promote reductive elimination, and also to increase the proportion of L1Pd species (palladium atoms with only one ligand), which are more open to approach by the elec- trophilic carbon species.262 Finally, the presence of ortho-substituents on the lower ring of both of these ligands increases the stability and activity of the palladium by preventing the formation of palladacycles through C−H insertion into the ligand.263 Initial experimentation248 failed to reproduce the cross coupling conditions reported219 for the synthesis of 57 (see Scheme 5.11). Investigation of the reaction components and optimisation discovered that a Pd2(dba)3 / X-Phos combination gave positive results when conducted in an n–butanol / water solvent system.248 Alcoholic solvents are thought to be particularly effective in this reaction because the presence of alkoxide base accelerates the transmetallation.264 Of the common bases used for Suzuki couplings,262 K3PO4 was found to be the most promising. It was hypothesised that the increased conversion was due to the greater basicity of PO4 3– compared to other bases such as CO3 2–. As suggested earlier, the carbamate protecting group was found not to survive the Suzuki conditions; in the presence of water at high temperature it is likely to be removed.265 However, some conversion was observed under these conditions and thus investigation of this reaction was continued with the free amine (Table 5.25). The temperature proved to be optimal at 100 ◦C; lower temperatures had been found to give very low conversions, and higher temperatures led to decomposition of the starting material by protodeboronation to compound 104 (entry 2). The conversion was observed to be highly dependent on catalyst loading (entries 1, 3 and 4), although the loading should ideally be kept as low as possible. It was found that the amount of boronic ester 102 had only a small effect on the conversion; since the boronic ester was difficult to remove by chromatography, the quantity was reduced to only a 10 % excess for future reactions. The addition of stronger bases such as KOH or a phosphazene base (BEMP) gave no improvement to the conversion. Deactivation of catalyst Previous work had shown that 24 – 44 % of the cross-coupling product was obtained when the amine substituent was not present in coupling partner 76.230 We postulated that the poor 5.2. Chemical probes for BRD9 129 Table 5.25 Investigation of the effect of temperature, reaction time and catalyst loading on the Suzuki cross-coupling reaction. N H S R OO Pd2(dba)3 X-Phos K3PO4 (2 equiv.) 7:3 n-BuOH / H2O MW N N NN H2N Cl B N H OO S R OO N N NN H2N N H S R OO 76 102 103 104 Entry R Equiv. Equiv. Equiv. Temp. Time Product Conv. Conv.102 Pd2(dba)3 ligand / ◦C /h to 103a to 104b 1 2-ClC6H4 1.5 10 % 20 % 100 4 103b 60 % 25 % 2 2-ClC6H4 1.5 10 % 20 % 120 4 103b 65 % 45 % 3 2-ClC6H4 1.5 20 % 40 % 100 4 103b 35 % 20 % 4 2-ClC6H4 1.5 1 % 2 % 100 8 103b 80 % 30 % 5 4-ClC6H4 1.5 10 % 20 % 100 4 103d 60 % 30 % 6 4-ClC6H4 1.1 10 % 20 % 100 4 103d 65 % 30 % 7248 Me 1.5 50 %c 100 % 105 3 103a 100 %d n/ae a Conversion measured by LC-MS; relative peak areas for compounds 103 and 76. b Conversion measured by LC-MS; relative peak areas for compounds 104 and 102. c 4 equiv. K3PO4 used. d 10 % isolated by column chromatography on KP-NH silica. e The majority of the remaining boronic ester was deborylated. conversions seen in Table 5.25 were due to deactivation of the palladium catalyst by one of the reaction components, which was supported by the formation of palladium precipitates during the reaction. Indeed, when the reaction was performed using a stoichiometric quantity of the palladium catalyst full conversion was observed, but the product was very difficult to isolate (Table 5.25, entry 7). Catalyst deactivation is likely to be caused by poisoning or sequestration, possibly by the primary amine261 or by a bidentate interaction with the amine and the triazole. The monophosphine ligand X-Phos is reported to give good results even with heteroaryl chlo- rides which have free amine substituents,247 which would suggest that the X-Phos ligand has a particularly strong interaction with the palladium centre. It was noticed that a large amount of material was retained by the silica gel during chromato- graphic purification, and this might indicate that polar complexes between the palladium 130 Development of inhibitors for bromodomain-containing proteins and either the starting material or product were forming. However, attempts to break any such complexes by stirring in acid conditions prior to chromatography were unsuccessful. Alternative ligands Therefore, despite the positive reports of using X-Phos in this kind of system,245 we postulated that a bidentate ligand might be held more strongly to the metal centre and resist catalyst deactivation. Since the substrates 76 and 102 are relatively low in steric bulk (they do not have any substituents in an ortho position) we anticipated that a bidentate ligand would not have a negative effect on the rate of oxidative addition. The reaction conditions were optimised using the methyl-derivative boronic ester (102e) which was not expected to give cross-coupling side reactions. During these investigations (Table 5.27) only starting materials, product 103a and homocoupled product 105 were observed. Improved degassing of the solvent prior to the reaction appeared to give superior results (Table 5.27, entries 2 and 4). Slightly increased conversion and lower homocoupling was observed when using DPE-Phos (Figure 5.26) as a ligand (entry 3), and even further improvement when using DtBPF (entry 5), which is likely to have higher activity due to the greater electron density on the phosphorus atoms. The homocoupling product was very difficult to remove from the reaction mixture, and so attempts were made to reduce its formation. A more rapid reaction turnover may disfavour the competing homocoupling process. Reducing the catalyst loading compromised the conversion (entry 6), and increasing the equivalence of boronic ester present had minimal effect (entry 7). However, increasing the proportion of n-BuOH in the solvent mixture gave lower homocoupling (entry 8). P P Fe PhPh PhPh Pd(II)Cl2 PdCl2.dppf P P Fe t-But-Bu t-But-Bu Pd(II)Cl2 PdCl2.DtBPF O P P Pd(II)Cl2 Ph Ph Ph Ph PdCl2.DPEPhos Figure 5.26 Palladium complexes with bidentate ligands employed in the Suzuki reaction. 5.2. Chemical probes for BRD9 131 Table 5.27 Investigation of the use of different bidentate ligands for the Suzuki cross- coupling reaction. N H S OO Catalyst K3PO4 (4 equiv.) x:y n-BuOH / H2O 100 °C, time N N NN H2N Cl B N H OO S OO N N NN H2N N H S OO H N S OO 76 102e 103a 105 Entry Equiv. Catalyst (equiv.) a Solv. Ratio Time Conversionb Homoc.b 102e (x:y) /h to 39 (105) 1 1.1 Pd/dppf (0.1) 7 : 3 4 20 % 100 % 2 1.1 Pd/dppf (0.1)c 7 : 3 4 60 % 70 % 3 1.1 Pd/DPE-Phos (0.1) 7 : 3 2 80 % 60 % 4 1.1 Pd/DPE-Phos (0.1)c 7 : 3 2 90 % 50 % 5 1.1 Pd/DtBPF (0.10)c 7 : 3 4 90 % (64 %)d 40 % 6 1.1 Pd/DtBPF (0.05)c 7 : 3 4 80 % 50 % 7 2 Pd/DtBPF (0.10)c 7 : 3 4 60 % 30 % 8 1.1 Pd/DtBPF (0.10)c 9 : 1 4 60 % 20 % 9 1.1 Pd/DtBPF (0.10)c 7 : 3 2 50 % 50 % 10 1.1 Pd/DtBPF (0.10)c 1 : 0 2 0 % n/a 11 1.1 Pd/DtBPF (0.10)c dioxane 2 0 % n/a a Precatalysts: Pd/dppf = PdCl2 · dppf ·CH2Cl2; Pd/DPE-Phos = PdCl2 ·DPE−Phos; Pd/DtBPF = PdCl2 ·DtBPF. b Approximate consumption of compound 76 / generation of homocoupled compound 102; as determined using LC-MS peak areas. c Freeze-dry degassing technique used (instead of standard nitrogen-bubbling technique). d Isolated by preparative HPLC. The optimisation studies indicated that, PdCl2 ·DtBPF was the most successful cata- lyst/ligand combination, and that a reduction in the amount of water present was beneficial. Although the presence of water is known to be important in Suzuki processes — it can be involved in in the hydrolysis of the boronic ester and formation of the reactive boronate species — DtBPF was reported to have been used in anhydrous dioxane, specifically in order to avoid side-reactions caused by water.266 However, no reaction was observed when anhydrous n-butanol (entry 10) or dioxane (entry 11) were used as solvents. 132 Development of inhibitors for bromodomain-containing proteins Purification The optimised cross-coupling conditions using PdCl2 ·DtBPF in n-butanol / water were used to obtain compounds 103 in moderate yields. The original hypothesis that the Suzuki cross-coupling could be performed selectively in the presence of aryl chlorides appeared to be mostly correct, but although the desired product tended to be the major one there were many by-products that made isolation very challenging. Table 5.28 Suzuki cross-coupling reactions to provide intermediates 103. N H S R OO PdCl2.DtBPF (0.1 equiv.) K3PO4 (4 equiv.) 9:1 n-BuOH / H2O 100 °C, 4 h N N NN H2N Cl B N H OO S R OO N N NN H2N (1.5 equiv.) 76 102 103 Entry R Compound Yielda 1 Me 103a 64 % 2 (2-Cl)C6H4 103b 49 % 3 (3-Cl)C6H4 103c 19 % 4 (4-Cl)C6H4 103d 30 % 5 (3,4-Cl)C6H3 103e 13 % a Isolated yield after preparative HPLC. Column chromatography using KP-NH silica had previously been found to consistently enable separation of compounds with a primary amine substituent when normal-phase silica failed. However, although isolation of compounds 103 by column chromatography was possible, it was found to be more efficient to separate the whole mixture using prepar- ative HPLC, which provided material of high purity for the final step to be investigated. Compound 103e was notably difficult to purify, as a result of the many minor by-products present in the mixture. These appeared to be the products of unwanted secondary Suzuki coupling reactions, as evidenced by 1H NMR and LC-MS. 5.2. Chemical probes for BRD9 133 5.2.6 Carbamate formation Combination of the Suzuki products 103b–e with 2 – 4 equivalents of ethyl chloroformate in the presence of DMAP gave good conversion to one major product, as observed by LC-MS (Table 5.29). Nevertheless, further purification by preparative HPLC was required to give sufficiently pure samples for NMR analysis. Interestingly, when THF was used as the solvent, product 108 was obtained; whereas when the reaction was performed in DCM, double addition of the carbamate moiety occurred to provide compound 109. Table 5.29 Installation of carbamate moieties onto compounds 103b–e. The desired product (107), with the carbamate present only on the heterocyclic amine, was not ob- served in any case. Ethyl chloroformate____ DMAP NEt3 Solvent 25 °CN H S OO N N NN H2N R Product N H S OO N N NNH N R O EtO N S OO N N NNH N R O EtO N S OO N N NN H2N R OEtO OEtO 103 (106) 107 108 109 R 103 Solvent Equiv. base Equiv. 106 Product Mass (Yield) 2-Cl 103b THF 10 2 108b <1 mg a,b 3-Cl 103c THF 10 2 108c <1 mg a,b 4-Cl 103d THF 2 2 108d 11 mg (95 %)c 3,4-Cl 103e DCM 2 4 109e 2 mg (38 %)a a Isolated yield following preparative HPLC. b Full conversion was observed by LC-MS, but the reaction scale was too small to record an accurate yield. c Isolated by solvent extraction. 134 Development of inhibitors for bromodomain-containing proteins This result was surprising: in particular, we had expected the sulfonamide NH to be deactivated with respect to the aromatic amine, and indeed the relative chemical shifts of these two environments in the NMR spectra of compound 103b–e suggested that this was the case. However, NMR analysis of the products of this reaction (Figure 5.30 shows 103c as an example) indicated that it was the sulfonamide that had in fact reacted first. 5.2.7 Alternative route The alternative route, as reported for bromosporine (57),219 whereby the carbamate group is installed whilst the aryl amine is masked as a nitro moiety, before reduction and sulfonamide formation, turned out to be even less efficient (Scheme 5.31). The amine seemed to be non-nucleophilic and almost completely deactivated, and only very low conversions were observed upon treatment with ethyl chloroformate. The starting material had to be cycled multiple times through this process in order to obtain useful quantities of the desired carbamate. NO2 NEt3 (10 equiv.) DMAP (1 equiv.) THF, 25 °C, 30 min N N NNH N (1.5 equiv.) NO2 N N NN H2N Cl O OEt O EtO trace 80 106 81 Scheme 5.31 Carbamate formation from nitro analogue 80 was unsuccessful. The pendant amine seemed to be very deactivated in the presence of the nitro group. Experiments conducted using the methylsulfonamide derivative 103a suggested that carba- mate formation took place on the sulfonamide nitrogen rather than on the heteroaryl amine, even when multiple equivalents of a strong base such as sodium hydride were added to attempt to activate this position.267 The carbamoylation provided four additional compounds which were suitable subjects for assessing their performance using the Frontal Affinity Chromatography (FAC) and other biological assay techniques. 5.2. Chemical probes for BRD9 135 0. 5 1. 0 1. 5 2. 0 2. 5 3. 0 3. 5 4. 0 4. 5 5. 0 5. 5 6. 0 6. 5 7. 0 7. 5 8. 0 8. 5 f1 (p pm ) 2.67 2.63 1.95 1.93 0.95 0.96 0.87 1.75 0.89 0.88 0.86 2.91 2.88 2.02 1.02 0.97 1.09 1.19 1.03 0.89 0.84 0.96 3.05 1.05 0.99 0.97 3.81 0.94 1H 1H 1H (H et A r- H ) 2H 2H 2H (r ot am er s) B N H SO O O O C l NNN N H 2N N H SO O C l NNN N H 2N N SO O O O C l Tr ac e 1 Tr ac e 2 Tr ac e 3 10 2b 10 3c 10 8c Fi gu re 5. 30 1 H N M R sp ec tra of 10 2b (tr ac e 1) ,1 03 c (tr ac e 2) an d 10 8c (tr ac e 3) sh ow in g ch an ge s in th e ch em ic al sh ift s of th e N H pr ot on s th at su gg es tt ha tt he ca rb am at e m oi et y w as in st al le d on to th e su lfo na m id e am in e. 136 Development of inhibitors for bromodomain-containing proteins 5.2.8 Biological assessment Compounds 103, 108b–d and 109e were assessed for binding activity with bromodomain proteins, including a FAC assay using BRD9. The FAC column was prepared by filling a 15 µL guard cartridge with biotinylated BRD9 protein immobilised onto streptavidin-coated beads. Column performance was validated by injecting samples of bromosporine (57) and analogue 110 at different concentrations.230 The quantity of BRD9 units available for binding was determined to be approximately 2 nmol, and the trend and magnitude of the affinity constants of 57 and 110 corresponded well with reported IC50 data. 219 N N NNH N N H S OO EtO O N N NNH N O t-BuO 57 110 Figure 5.32 Compounds used for validating the BRD9 FAC column. A disadvantage of a chromatographic method is that there is the possibility of binding by non-specific interactions, such as those involving the streptavidin units or the polymer support onto which the BRD9 is loaded. In particular, streptavidin exists as a tetramer, with four biotin-binding positions per unit. The bulk of the BRD9 protein bound in one site is likely to block the other sites from being occupied, so there are spare binding sites available to interact with the test compounds. To determine the effect of non-specific interactions, bromosporine (57) and the void marker 111 were run down a column loaded with streptavidin-coated beads. No significant dif- ference in the retention times were observed between compounds 57 and 111, confirming that the retention times observed during the initial validation were caused primarily by interactions with BRD9. Void compound 111 is not UV active, but can be be detected using a mass spectrometer. The use of a mass spectrometer enables a wider range of compounds to be detected and thus tested, and has greater sensitivity compared to a UV detector; however, it also imposes some restrictions on the analysis method. In particular, a volatile buffer had to be used to 5.2. Chemical probes for BRD9 137 avoid filling the nebulisation chamber with salt deposits. Fortunately, the BRD9 column was found to be stable and functional in the presence of a volatile ammonium acetate buffer. HO2C N H O N N NN CF3 N N NN CF3 OMe N N NNH N N H S OO EtO O 111 57 112 113 Figure 5.33 Additional compounds used during the FAC experiments. Compounds 103b,c,e, 108b–d and 109e, along with bromosporine (57) and analogues 112268 and 113,268 were injected separately into the BRD9 FAC-MS system. By performing single injections with each compound at the same concentration (Figure 5.35), a ranking of affinities was constructed by observing the relative retention times within the functionalised column. These results were compared with thermal shift assays performed against a number of other proteins spread across the BRD phylogenetic tree (Table 5.34).230,246 Table 5.34 Tm shift data collected230 for compounds across a suite of bromodomain- containing proteins. Protein 57 112 113 103b 103e 108b 108c 108d 108e 109e BRD9 (∆Tm,obs) +8.8 -0.6 +0.6 +2.4 +4.3 +4.4 n.d. +2.7 +1.7 +1.1 BRD1 (∆Tm,obs) +2.5 -1.6 +0.0 +2.3 +3.7 -1.1 +1.8 +2.0 +2.0 +1.5 BRD4A (∆Tm,obs) +6.6 -3.5 -1.3 +6.5 +6.7 +6.2 +5.6 +4.3 +1.8 +3.2 BRPF1A (∆Tm,obs) +0.6 n.d. n.d. +1.0 +5.4 n.d. n.d. +1.7 +1.8 -0.3 BRPF3 (∆Tm,obs) +2.1 n.d. n.d. +1.1 +3.1 n.d. n.d. +1.6 +0.4 +0.0 CECR2 (∆Tm,obs) +8.3 -7.4 -0.1 +3.0 +5.6 n.d. n.d. -10.9 +1.7 +1.6 CREBBP (∆Tm,obs) +3.8 -2.1 -0.9 +4.1 +4.3 +3.5 +4.7 +4.6 +2.0 +3.6 EP300A (∆Tm,obs) n.d. +1.3 +1.3 +3.5 +8.0 +4.2 +4.1 +4.3 +2.5 +3.7 TIF1 (∆Tm,obs) +0.9 -1.7 -0.5 +1.3 +1.8 n.d. +0.2 +1.2 +0.9 +0.5 ∆Tm,obs = Difference in temperature of denaturation of the protein with and without the ligand (at 10 µM), data in ◦C; n.d. = not determined. 138 Development of inhibitors for bromodomain-containing proteins 0.0 0.5 1.0 1.5 2.0 2.5 0 10 20 30 40 50 60 Ion Count /106 Tim e /m in N N H 2 N N N NH S O O C l C l N N H 2 N N N NH S O O C l N N H 2 N N N NH S O O C l N N N N N S O O E tO O C l H 2 N < < < N N H 2 N N N N S O O C l E tO O N H A c C O O H N N H 2 N N NCl A B C D E F G A B C D E F G 111 76 108b 103b 103c 108d 103e Figure 5.35 FA C -M S analysis ofcom pounds 76,103 and 108 using a B R D 9-functionalised colum n.C om pounds 112 and 113 eluted betw een com pounds 111 and 76,and are notshow n. B rom osporine (57) did notelute during the 120 m inute run,due to its strong affinity forB R D 9. 5.2. Chemical probes for BRD9 139 5.2.9 Evaluation The test compounds were generally found to have reduced activity for BRD9. Interestingly, compound 103e had increased selectivity for BRPF1A, CECR2 and EP300A, and com- pounds 103b and 108b retained activity for BRD4 whilst losing their activity for BRD9 and CECR2. Most importantly, the observed trend in the FAC retention times of the test compound and standard correlated very well with the measured Tm shift of the same com- pounds with BRD9 (Table 5.36), which gave us additional confidence in the use of this technique for rapid assessment of new compounds. Table 5.36 Comparison of FAC results with Tm shift data. Compound ∆Tm,obs (BRD9) FAC rankinga 57 +8.8 1 103e +4.3 2 108d +2.7 3 103c n.d. 4 103b +2.4 5 108b +4.4 6 108e +1.7 n.d. 109e +1.1 n.d. 113 +0.6 7 112 -0.6 8 a The FAC ranking was obtained by visual iden- tification of the curve fronts of the FAC-MS traces (Figure 5.35) and ordering them from the latest to the earliest. By tuning the column size it should be possible to analyse compounds with different ranges of affinities within a reasonable time. The 15 µL column with 2 nmol binding capacity was found to be able to distinguish between low-to-medium affinity substrates (equivalent to ∆Tm,obs between 0 and 5 ◦C), so it may be useful as a technique for fragment-based drug discovery programmes. 140 Development of inhibitors for bromodomain-containing proteins 5.3 Chemical probes for CECR2 Analogous compounds prepared as part of the research programme were included in a screening assay against a range of bromodomain proteins.246 Compounds with a “reversed” sulfonamide moiety (compounds 115–118, Table 5.38) exhibited increased selectivity for CECR2, another bromodomain protein (Figure 5.37). The identification of these compounds as being selective for CECR2 was used to inspire the design and synthesis of a small array of similar compounds. Importantly, the pendant amine or carbamate chain appeared to be less important for CECR2 binding (compounds 115–119 have increased potency for CECR2 compared to BRD4A), and so we decided to a b c Figure 5.37 (a) Crystal structure of bromosporine (57) bound into BRD4A(1). (b) Bromo- sporine docked into CECR2, calculated using Schrödinger Glide.248 (c) Calculated binding interactions between bromosporine (57) and CECR2. Bromosporine is calculated to make similar interactions with CECR2 as with BRD4A(1), including between the northern triazole and an asparagine residue, and between the pendant sulfonamide and an aspartic acid residue. 5.3. Chemical probes for CECR2 141 Table 5.38 Structure-activity relationship data for a number of compounds as determined by SGC experiments. A red background indicates strong binding, green indicates weak binding. Compound PotencyBRD9 CREBBP CECR2 BRD4A BRD2 57 N N NNH N N H S OO EtO O 0.12 a 0.16 b 0.02 a 0.20 a 0.41 a 114 N N NNH N NO2 EtO O 1.18 a 10000 b 0.04 a 35.9 a n.d. 115 N N NN S N O O 7.14 c 170.6 a 0.05 b 10000 a n.d. 116 N N NN S N O O n.d. 10000 a 0.36 a 8.91 c n.d. 117 N N NN S N O O 22.9 c 10000 a 0.50 b 10000 a n.d. 118 N N NN S N O O 17.8 c 10000 a 15.9 b n.d. n.d. 119 N N NN N H S OO 13.6 c 10000 a 69.5 b 10000 a n.d. Data are given as IC50, measured or estimated by: a AlphaScreen; b AlphaScreen (single concentration); c Tm shift (estimated). 142 Development of inhibitors for bromodomain-containing proteins focus initially on deaminated analogues of 57. Since this amine group was thought to be the main source of problems with the cross-coupling step, we anticipated that this strategy might enable more rapid access to the compounds. This targeted array would test how well the trend in the binding affinities of the deaminated analogues correlated with that of the aminated analogues, providing further confirmation of whether FAC is an effective technique for providing data on fragments as well as final compounds. 5.3.1 “Reversed” sulfonamide boronic ester Previous work had investigated chlorosulfonylation of aminotolueneboronic ester 114269,270 or tolueneboronic ester 114271,272 (Scheme 5.39).273 Unfortunately, these attempts to install the sulfonyl group onto a phenyl boronic ester proved to be unfruitful, thus a different strategy for their preparation was investigated whereby the boronic acid group was installed onto a phenylsulfonamide. BPin X BPin S Cl O O BPin S NR 1R2 O O HNR1R21. Sulfonylation 2. Chlorination 114a; X = NH2 114b; X = H 115 116 Scheme 5.39 Unsuccessful chlorosulfonylation of phenylboronic esters. An effective method was the Miyaura borylation274 of bromobenzylsulfonamides 118 prepared by the treatment of sulfonyl chlorides 117 with cyclic amines (Scheme 5.40). S Cl OO HNR1R2 Br S NR1R2 OO Br Borylation S NR1R2 OO PinB 117 118 119 Scheme 5.40 Access to boronic esters 119 by Miyaura borylation of sulfonamides 118. The sulfonamides 118 were generated smoothly by treatment of a sulfonyl chloride 117 with a secondary amine at room temperature. This reaction was found to have a good solvent tolerance, with the highest solubility of the starting material 117 observed in CPME, 5.3. Chemical probes for CECR2 143 DME or n-butanol. Compounds 117 were treated with polymer-supported amine base (QP- DMA) before addition of the amine to sequester any traces of hydrolysed sulfonyl chloride which would be unreactive under these conditions. With this route in hand, borylation of the sulfonamides 118 was investigated. 5.3.2 Miyaura borylation In 1995, Miyaura and coworkers reported274 that tetra(alkoxo)diborons such as bis(pina- colato)diborane 120 can act as a boron nucleophile for cross-coupling with organic elec- trophiles in the presence of a base. LnPd(0) Pd(II) R Ln Cl Pd(II) R Ln AcO KOAc Pd(II) R L(n-1) PinB BPin KCl oxidative addition reductive elimination base transmetallation aryl halide metathesis L B B O O O O AcO B O O Pd(II)Cl R ClR (PPh3)n-1 PPh2R BPin L = PPh3 + L 120 121 Scheme 5.41 Mechanism of the Miyaura borylation reaction.274 The accepted mechanism (Scheme 5.41) is analogous to the Suzuki-Miyaura cross coupling reaction,274 and includes the critical presence of a base such as potassium acetate to generate an activated intermediate which undergoes transmetallation with the diboron. The use of KOAc is postulated to disfavour homocoupling products; stronger bases such as K2CO3 or K3PO4 lead to the formation of biaryls by further coupling of the transmetallated palladium boronate intermediate with the aryl halide. In cases where electron-poor or electron-rich aryl halides are used, slow transmetallation can permit a competing pathway involving aryl exchange with the phosphine ligand to 144 Development of inhibitors for bromodomain-containing proteins afford small quantities of a phenylboronate by-product (121).275 Borylation is reported to be accelerated by polar solvents such as DMSO or DMF, with slower reactions in dioxane or toluene.274 Considering the difficulty of removing reaction products from such polar solvents, alternative solvents were considered. CPME was selected as the first solvent to be evaluated, because it had been found to be useful in the previous step, a beneficial feature potentially allowing telescoping of steps. CPME is more polar than other commonly used solvents such as dioxane (with a molecular dipole moment of 1.27 D compared to 0.45 D for 1,4-dioxane276), and thus functionalisation using this solvent was investigated first (Table 5.42). Under the first set of conditions attempted, the reaction was found to proceed with good conversion to the desired product (123a) (entry 1). However, the formation of an inorganic Table 5.42 Investigation of ligand systems for Miyaura borylation. S N OO O B2Pin2 Catalyst KOAc (3 equiv.) Solvent Temperature, Time B S N OO O SN O O O O O 2 Br 122a 123a 124 Entry Catalysta Equiv. Solvent (v/v) Temp. Time Conv. b Homoc.b B2Pin2 /◦C /h to 123a (124) 1 Pd/dppf 3 CPME 110 3 100 %c 0 % 2 Pd/XP 3 CPME 110 3 100 %c 0 % 3 Pd/XP 1.5 CPME 110 3 100 %c 0 % 4 Pd/XP 1.5 CPME 110 1 100 %c 0 % 5 Pd/XP 1.5 8 : 1 CPME / H2O 110 1 25 % 0 % 6 Pd/XP 1.5 n-BuOH 110 1 100 % 15 % 7 Pd/XP 1.5 9 : 1 n-BuOH / H2O 110 1 100 % 25 % 8 Pd/PEPPSI 1.1 n-BuOH 110 1 0 % – 9 Pd/PEPPSId 1.1 n-BuOH 110 1 100 % 65 % 10 Pd/XP 1.1 n-BuOH 150 1 95 %e 20 % a Catalysts: Pd/XP = Pd2(dba)3 ·CHCl3 (2 mol%), X-Phos (4 mol%); Pd/dppf = dppf ·PdCl2 · CH2Cl2 (4 mol%); PD/PEPPSI = PEPPSI−IPr ·PdCl2 (3 mol%). b As determined using LC-MS peak areas. c Formation of precipitate observed. d 5% catalyst loading. e 12% isolated yield (column chromatography). 5.3. Chemical probes for CECR2 145 precipitate, likely to be from the palladium or the KOAc, was also observed. Similar results were observed using X-Phos as a ligand (entry 2); in this case HPLC analysis suggested a cleaner reaction. Reduction of the amount of bis(pinacolato)diborane (120) added to the reaction had no significant effect on the conversion (entry 3). A shorter time of 1 hour was equally effective (entry 4). We wanted to investigate the possibility for performing the three-step borylation process in continuous flow as this would enable the use of automation to generate an array of coupling partners. For this reason, we investigated further conditions to attempt to reduce the processing time and also avoid the generation of a precipitate, since this would be highly undesirable within a heated flow reactor coil. The precipitate observed in entries 1 – 4 was found to be water soluble; unfortunately, inclusion of water in the solvent mixture to give a CPME / water ratio of 8/,:/,1 (v/v) did not promote the reaction, possibly due to the low miscibility of these solvents leading to sequestration of the base in the aqueous phase (entry 5). n-Butanol is more polar (1.66 D) than CPME, and is often recommended for Suzuki-type reactions; this solvent was good in both the sulfonamide formation and Suzuki steps, allowing the possibility for reaction telescoping. However, in this case the use of n-butanol tended to lead to the generation of the homocoupled product (124) (entries 6 – 7). Oxygen is postulated to be involved in homocoupling processes.277 However, oxgyen was unlikely to be present under the reaction conditions because the same degassing protocol was followed as for the Suzuki reactions in Section 5.2.5, which had been found to remove almost all of the oxygen. It was likely therefore that the homocoupling product was formed due to a slow transmetallation step in the mechanism. In a limited catalyst and ligand screen, PEPPSI-IPr (Figure 5.43) was included as it is reported to be an effective ligand for borylation,278 and is in general a highly active P P Fe PhPh PhPh Pd(II)Cl2 Pd NN iPr iPr iPr iPr N ClCl Cl PdCl2.dppf PEPPSI-IPr Figure 5.43 Palladium complexes employed in the Miyaura borylation reaction. 146 Development of inhibitors for bromodomain-containing proteins ligand for Suzuki-type processes.279 N-Heterocyclic carbene ligands are in general stronger electron donors than phosphine ligands;280 however, the use of PEPPSI-IPr showed no advantage for this system (entries 8 – 9). In a similar, related system, a limited screen of solvents and bases was conducted (Ta- ble 5.42), using dppf (Figure 5.43) as a ligand because it had been observed to keep the palladium complex to in solution throughout the reaction (Table 5.42, entry 1). Using an n-BuOH / MeCN solvent system, the sulfonamide (125a) remained in solution. Full conversion was obtained with low degree of homocoupling (Figure 5.43, entry 1); however, the isolated yield of compound 126a was only 53 %. Considering the bases, triethylamine has been reported as an effective base for the Miyaura borylation281 and since it is a liquid there is no concern of solubility. However, the use of NEt3 led to a reduced conversion as measured by LC-MS even though there was little effect on the quantity of 126a isolated (entry 2). Furthermore, although the use of NEt3 enabled the use of different solvent systems whilst maintaining a homogeneous reaction mixture, there was no significant improvement in the isolated yield (entries 3 – 5). Table 5.44 Investigation of Miyaura borylation parameters using PdCl2(dppf) ·CH2Cl2 catalyst complex. Br S N OO O B2Pin2 (1.1 eqiv.) PdCl2(dppf).CH2Cl2 Base (3 equiv.) Solvent Temperature, Time B S N OO O SN O O O O O 2 125a 126a 127 Entry Base Solvent (v/v) Temp. Time Conv.a Homoc. a Yieldb /◦C /min (127) of 126a 1 KOAc 2:1 n-BuOH / MeCN 150 60 100 % 20 % 53 % 2 NEt3 2:1 n-BuOH / MeCN 150 60 50 % 0 % 54 % 3 NEt3 MeCN 150 60 35 % 0 % 50 % 4 NEt3 1:1 CPME / MeCN 150 60 40 % 0 % 58 % 5 NEt3 8:2 DME / H2O 150 60 100 % 25 % 53 % 6 KOAc 2:2:1 n-BuOH / MeCN / H2O 150 30 100 % 15 % 52 % 7 KOAc 1:1 EtOH / H2O 150 10 100 % 15 % 58 % 8 KOAc 1:1 MeCN / H2O 150 10 100 % 45 % 24 % a Approximate consumption of 125a / generation of 127; peak areas measured by LC-MS. b Isolated yield, following column chromatography. 5.3. Chemical probes for CECR2 147 The use of water as an additive to the n-BuOH / MeCN solvent system was evaluated as it allowed better solvation of the inorganic base. The results indicate that there was no detrimental effect on the conversion or yield, and in this case the reaction was complete within 30 minutes as evidenced by LC-MS (entry 6). Monophasic aqueous solvent systems were also investigated, because these may have led to more favourable continuous flow conditions. Both ethanol / water and acetonitrile / water solvent systems led to full consumption of the starting material 125a within 10 minutes as observed by LC-MS, but without significant improvement in the isolated yield. The homocoupling product (127) was difficult to separate from the desired product by column chromatography, leading to the low isolated yields. Therefore, in the interests of time, we decided to progress with a batch-mode synthesis method using the original KOAc / CPME conditions, since these had been found to produce the smallest quantity of by-products. 5.3.3 Suzuki cross-coupling Initial screening of reaction conditions using the procedure previously investigated for the preparation of the BRD9 probes (see Section 5.2.5) indicated very high and clean conversion as observed by LC-MS, but only a trace of compound 128a was isolated after purification by preparative HPLC (Scheme 5.45). The poor recovery and isolation of compound 128a indicated that purification was the most significant problem for these compounds. Pd2(dba)3 (10 mol%) X-Phos (20 mol%) K3PO4 (4 equiv.) 4:1 n-BuOH / H2O 100 °C, 2 h B S N OO O S N OO OO O N N NN Cl NN N N (< 1 mg) 129 126a 128a Scheme 5.45 Initial attempt at cross-coupling between 129 and boronic ester 126a. The mass of the product recovered after preparative HPLC was too low for accurate measurement. One-pot borylation and cross-coupling reactions are precedented in the literature,282 so an attempt at telescoping the clean Miyaura borylation reaction with the Suzuki cross-coupling reaction was attempted. Pleasingly, this methodology allowed rapid access to an array 148 Development of inhibitors for bromodomain-containing proteins Br S NR 1R2 O O N N NN 3 steps S Cl O O 4-substituted, 130 3-substituted, 131 4-substituted, 128 3-substituted, 132 Scheme 5.46 One-pot synthesis of compounds 128 and 132. Conditions: Step 1. HNR1R2 (1.1 equiv.), 8 : 1 n-BuOH / water, 25 ◦C, 5 min. Step 2. B2Pin2 (1.1 equiv.), Pd2(dba)3 (0.02 equiv.), X-Phos (0.04 equiv.), KOAc (3 equiv.), CPME, 110 ◦C, 1 h. Step 3. 129 (1 equiv.), Pd2(dba)3 (0.2 equiv.), X-Phos (0.4 equiv.), K3PO4 (3 equiv.), 4 : 1 n-BuOH / water, 100 ◦C, 2 h. of compounds; after each step the solvents were removed and new reagents were added without purification of the intermediates and the final products were isolated by column chromatography (Scheme 5.46, Table 5.47). Table 5.47 Compounds synthesised for biological testing against CECR2. Entry Boronic Ester R Compound Yielda 1 BPin S R O O N O 128a 2 % 2 N 128b 2 % 3 N 128c 4 % 4 N N 128d 3 % 5 S R O O BPin N 132a 12 % 6 N 132b 4 % 7 N 132c 1 % a Isolated yield (column chromatography) over three steps. 5.3. Chemical probes for CECR2 149 5.3.4 Biological analysis The affinities of compounds 128a,c,d and 132a,b, as well as compounds 133248 and 134248 for BRD proteins were determined using four different methods. Firstly, using the FAC method described in Section 5.2.8, solutions of each compound (12.5 µM in ethanol) were injected into a column functionalised with CECR2 protein. All of the compounds were UV-active, so the output was detected by UV spectroscopy, using DMSO as the co-solvent and void marker. This assay method allowed an approximate ranking of their affinities to be contructed (Figure 5.49). The results were found to be highly dependent on concentration; compound 132b was very poorly soluble in the ethanol / DMSO system making assessment of its binding ambiguous, with a large margin of error. In a second FAC assay method, injections of each substrate at three different concentrations (6.25, 12.5 and 25 µM) were performed, to obtain a trend of ∆v against concentration. To calculate ∆v for each compound, a sigmoid curve was fitted to the “front” of the absorbance signal pattern. These data were used to calculate a value for Kd for each compound by performing a nonlinear curve fitting of ∆v against concentration (Figure 5.48). 0 100 200 300 400 500 0 5 10 15 20 25 30 Ab sor ba nc e Time /min a 0.2 0.4 0.6 0.8 1.0 1.2 1.4 1.6 1.8 2.0 0 5 10 15 20 25 30 (V - V 0 ) [ A 0 ] / 10 -9 n m ol [A0] /µM Non-linear t Points b Figure 5.48 (a) FAC curves for compound 128c at 6.25, 12.5 and 25 µM. (b) Nonlinear regression of (V −V0)[A0] = (Bt [A])/(Kd +[A]) to obtain a value for Kd . In a third (virtual) method, the structures were docked into a model of the CECR2 protein in silico using Schrödinger Glide,283 to estimate their relative binding affinities. The affinities that were calculated by Glide§ followed a similar trend to the FAC results, suggesting that the same binding site is active in the protein in vitro and in the FAC column. The involvement of water atoms, which are found to be conserved within the binding sites of many bromodomain proteins, may affect compound binding.284 The effect of water § Glide generates numerical binding scores; more negative scores indicate stronger bonding. 150 Development of inhibitors for bromodomain-containing proteins                  N N NN S N N O O N N NN S N O O O B r S N O O NN N NS N O O NN N N S N O O NN N N S N HN E tO O O O NN N N S N HN E tO O O O NN N N S N O O A A B B C C D D E E F F H G G H < < < < 132b 128a 128d 118a 128c 132a 133 134 Figure 5.49 Injections ofa num berofcom pounds ata single concentration (12.5 µM )to the C E C R 2 FA C colum n,show ing consistent retention for each substrate over atleastthree experim ents. T he results give an approxim ate ranking ofthe com pounds (top). A n artefactis visible in each trace at16.5 m inutes;this is thoughtto have been created by the end ofthe 600 µL injection loop. 5.3. Chemical probes for CECR2 151 N N NN N O O S N N N N S N O O O N N N N S N N O O N N N N S N O O N N NN S N O O N N NN S N O O H NEtO O N N NN S O O H NEtO O 57 132b 128a 128d 128c 132d 134 Figure 5.50 Depiction of the test compounds docked into CECR2, showing the most favourable binding orientations as calculated by Glide. 152 Development of inhibitors for bromodomain-containing proteins molecules in compound binding may be used to explain the high binding results returned for compounds 128a and 132b which were found to have a low affinity in the FAC assay. In the fourth method, the compounds were analysed246 against a set of BRD proteins using Tm shift measurements (Table 5.51). Table 5.51 Tm shift data collected for compounds 128, 132, 133 and 134 across a suite of bromodomain-containing proteins.246 Compound ∆Tm,obsBRD1 BRD4A BRD9 BRPF1B CECR2 CREBBP BRPF3 128a −0.92 0.17 2.64 −0.63 0.66 −1.34 −0.01 128c −0.27 1.30 2.35 0.35 0.48 0.48 −0.11 128d 0.14 1.20 1.40 0.30 0.61 −0.81 0.19 132a 0.89 4.79 1.15 0.78 0.74 0.79 0.43 132b 0.30 1.14 1.90 0.12 0.83 0.36 0.30 133248 0.82 7.39 6.44 2.57 8.08 2.21 0.91 134248 0.49 4.86 7.03 2.41 9.80 1.71 0.12 ∆Tm,obs = Difference in temperature of denaturation of the protein with and without the ligand, data in ◦C. Compounds 133 and 134, which have a pendant carbamate moiety similar to bromosporine, showed a greater affinity for the proteins. This indicates that the presence of this side-chain significantly affects their affinity for CECR2. Furthermore, the assay methods show the ability to distinguish between substrates that have weak binding affinities. This result is very promising, since fragments and intermediates such as 131a could be tested at early stages during a library synthesis providing additional data with which to direct a discovery project. 5.4 Conclusion An array of compounds was generated and tested against a set of bromodomain compounds using both conventional and FAC biological assay methods. The data produced by the FAC technique was found to correlate well with that produced by the conventional methods (Table 5.52). With an appropriately-sized column, FAC is able to distinguish between compounds with relatively low affinities for the protein under analysis, suggesting that it could be an effective assay method for fragment-based discovery programmes. 5.4. Conclusion 153 Table 5.52 Comparison of data from different assay techniques for compounds 132, 128, 133 and 134. (A more negative Glide score indicates stronger calculated affinity). Compound FAC rankinga FAC Kd /µM Glide score ∆Tm,obs (CECR2) 134 1 n.d. −8.3 9.80 133 2 54±6 n.d. 8.08 132b n.d. n.d. −7.6 0.83 132a 3 45±7 −7.8 0.74 128c 4 41±8 −7.4 0.48 128d 5 62±30 −6.4 0.61 128a 6 208±156 −7.3 0.66 a Determined by visual inspection of absorption curves (Figure 5.49) to identify the breakthrough volume, then ordering from the latest to the earliest. Since FAC is fundamentally a chromatographic technique it is possible that with further development FAC analysis could be integrated into a flow synthesis platform to provide rapid feedback on compounds as they are produced. Alternatively, with additional equip- ment such as a column-switching device sometimes included in laboratory HPLC setup, compounds could be tested against a suite of proteins within a matter of hours using a bench-top system. Flow synthesis was found to provide benefits in the early stages of this synthesis to enable relatively large-scale reactions to be performed using hazardous reagents. Future develop- ment of flow chemistry systems285 or alternative automated batch-synthesis platforms286 may lead to improvements in the later stages of a discovery programme where materials are being handled in smaller quantities and under a greater variety of conditions. 154 Conclusion The goal of this work was to investigate how automation could contribute to flow synthesis programmes, and to develop tools to make computer-enabled processes easier to implement so that the use of this technology might become more commonplace in the future. “Solid-phase” synthesis is an important application of immobilised reagents, a key compo- nent in the flow chemistry toolbox. Automation methods were found to enable the more repetitive synthetic tasks to be performed under complete computer control so that long tasks, or operations requiring complex sequences, could be carried out overnight and with greater consistency between experiments. The software packages currently supplied with commercially available flow chemistry platforms were found to be incapable of the more complex operations that might be required. The available laboratory automation software packages are primarily targeted at biochemical assaying applications. A new control and monitoring system (Octopus) was developed using the Python programming language, and the source code released under an open-source license so that publications using this software might be more readily reproducible. This system was applied to a two-step hydration/reduction sequence to obtain piperazine- 2-carbonitrile, a valuable 3D building block. This involved the integration of a Vapourtec R2+/R4 system, a Knauer HPLC pump, a FlowIR spectrometer, an H-Cube hydrogenation reactor and a multi-position valve; this would have been much more challenging using pre- vious automation technologies. Application of the system for running multiple experiments, such as for a Design of Experiments procedure, was demonstrated. Automation techniques were also found to be invaluable in the multi-step flow synthesis of 2-aminoadamantane-2-carboxylic acid, which required intermediate work-up operations between different stages. We were able to implement multiple independent steps with 155 156 Conclusion control of several different devices and operations, which would have been very difficult to achieve manually. Finally, flow chemistry and remote control and monitoring methods were applied where appropriate to a discovery programme for inhibitors of bromodomain proteins. The Frontal- Affinity Chromatography (FAC) technique was shown to provide results that were consistent with conventional assay methods. Since FAC could conceivably be integrated into a flow synthesis system it has the potential to form part of a machine-assisted, “closed-loop” discovery programme where biological data are fed directly into an automated synthesis platform for iterative development of biologically-active molecules. In summary, automation technology has allowed more complex flow synthesis platforms to be operated under precise control. A number of other research programmes within our laboratory have started to use these automation systems. Future development of the software control package should make this technology accessible to non-programmers. Another important area for future development will be for the intelligent automation of analysis and purification procedures, since the running of reactions is probably one of the easier aspects of synthetic chemistry to automate! Nevertheless, routine automation of experiments could remove some of the variation inherent in synthesis, and lead to much more reproducible and better documented procedures. Appendix A Synchronous instrument controller 157 158 Synchronous instrument controller Listing A.1 vapourtec.py Class for creating objects to control Vapourtec R2+/R4 machines using a Python script. Commands are synchronous, i.e. the programme freezes until a response is received from the machine. Please note that the commands have been redacted in the PDF version of this document. import time import math import sys class R2R4 (object): transport = None def __init__ (self, transport): """ Initialise the controller with a transport object. """ transport.setTerminator("\r\n") transport.open() self.transport = transport def _send (self, line): return self.transport.send(line) def _press (self, key): """ Emulate a button press on the machine. """ return self._send(" " + str(key)) def status (self): """ Get the current status. returns: 0 = off, 1 = on, >1 = error """ return self._send("").split()[0] def on (self): """ Power on the machine. """ return self._send("") def off (self): """ Power off the machine. """ return self._send("") def waste (self): """ Direct output to waste. """ return self._press(8) def collect (self): """ Collect output. """ return self._press(9) vapourtec.py 159 def temp (self, heater, target): """ Set heater temperature. heater = 0, 1, 2 or 3. target = -1000 or 20 to 250 """ return self._send("  {:d} {:d}".format(heater, target)) def valve (self, channel, position): """ Switch a valve. channel = "orange", "purple" or "both" position = "solvent", "reagent", "load" or "inject" """ positions = ["solvent", "reagent", "load", "inject"] orange = [0, 1, 4, 5] purple = [2, 3, 6, 7] if channel == "orange": return self._press(orange[positions.index(position)]) if channel == "purple": return self._press(purple[positions.index(position)]) if channel == "both": return ( self._press(orange[positions.index(position)]), self._press(purple[positions.index(position)]) ) def flowrate (self, channel, rate): """ Set the pump flowrate. channel = "orange", "purple" or "both" rate = flowrate in uL/min """ if channel == "both": return ( self._send(" 0 {:d}".format(rate)), self._send(" 1 {:d}".format(rate)) ) if channel == "orange": channel = 0 if channel == "purple": channel = 1 return self._send(" {:d} {:d}".format(channel, rate)) def pressures (self): """ Read the current pressures """ return self._send("").split("&").pop().split(',')[1:] # Continued... 160 Synchronous instrument controller def wait (self, hours = 0, mins = 0, seconds = 0, keepalive = False): """ Wait for specified time, printing out current pressure every minute. If keepalive = True, reactor is switched back on if it stops """ # Caclulate the time to stop end_time = time.time() + (hours * 60 * 60) + (mins * 60) + seconds while 1: now = time.time() # Stop at the end time if (now > end_time): break; left = math.ceil((end_time - now) / 60) m_left = left % 60 h_left = left / 60 p = self._pressures() # Print the current pressure readings sys.stdout.write( "\rWaiting: %dh %dm Pressures S:%d A:%d B:%d " % ( h_left, m_left, int(p[0]), int(p[1]), int(p[2]) ) ) sys.stdout.flush() # If the R2 is not running, start it. if keepalive: for j in range(60): if self.status() != 1: self.on() time.sleep(1) # Check once per second. else: time.sleep(60) print "\n" vici.py 161 Listing A.2 vici.py Class for creating objects to control Valco VICI microelectric multiposition valves. import time import math import sys class MultiValve (object): transport = None positions = 0 def __init__ (self, transport): transport.setTerminator("\r") transport.open() self.transport = transport self.positions = self.num_positions() def _send (self, line): return self.transport.send(line) def num_positions (self): """ Read the current number of positions """ return int(self._send("NP").split('=')[1]) def position (self): """ Read the current valve position """ return int(self._send("CP").split('=')[1]) def advance (self, number): """ Move by [number] positions """ return self.move(self.position() + int(number)) def move (self, position, direction = 'fastest'): """ Move to [position]. direction = "clockwise", "counterclockwise" or "fastest" """ if (direction in ('f', 'fastest')): command = 'GO' elif (direction in ('c', 'cw', 'clockwise')): command = 'CW' elif (direction in ('a', 'cc', 'counterclockwise')): command = 'CC' # Make sure position is between 1 and NP position = ((int(position) - 1) % self.positions) + 1 # Trying to move to current position would give an error if self.position() == position: return 'OK' # Continued... 162 Synchronous instrument controller # Move, then get new pos immediately. (The valve will not respond to # GO/CC/CW commands, so the CP request is needed so that transport.send # doesn't wait forever, and is useful as CP only responds once the move # has been completed.) new_pos = int(self._send(command + str(position) + "\rCP").split('=')[1]) if new_pos == position: return 'OK' else: return 'Error' Listing A.3 transport.py Class for creating objects to communicate over a serial connection. Messages are sent with a terminating sequence, and then the response is read until the terminating sequence is received. class serial (object): ser = None _terminator = None _terminator_len = 0 def __init__ (self, port): import serial self.ser = serial.Serial() self.ser.port = port self.ser.baudrate = 19200 self.ser.timeout = 1 def setTerminator (self, terminator): self._terminator = terminator self._terminator_len = len(terminator) def open (self): self.ser.open() def close (self): self.ser.close() def send (self, line): buff = "" self.socket.send(line + self._terminator) while len(buff) < self._terminator_len \ or buff[-self._terminator_len:] != self._terminator: buff = buff + self.ser.read() return buff[:-self._terminator_len] Appendix B Control scripts for monolith processing 163 164 Control scripts for monolith processing Listing B.1 Example of a programme to perform a stop-flow sequence over a number of cycles. import transport, vapourtec R2 = vapourtec.R2R4(transport.serial("COM1")) flow_rate = 200 # Flow rate in uL/min column_volume = 3000 # Column volume in uL cycles = 4 # Number of stop-flow cycles to run temp = 100 # Heating temperature wait_mins = 120 # Heating time per cycle # Set up reactor R2.valve("orange", "reagent") R2.temp(1, temp) R2.on() # Wait for the heater (~80 C/min) time.sleep(temp / 80.) for i in range(cycles): print "Running cycle {:d}".format(i) # Pump reagent in R2.flowrate("orange", flow_rate) R2.wait(mins = column_volume / float(flow_rate)) # Adjust the flow rate of both pumps R2.flowrate("orange", 0) R2.wait(mins = wait_mins, keepalive = True) # Final solvent wash print "Washing" R2.valve("orange", "solvent") R2.flowrate("orange", flow_rate) R2.wait(mins = column_volume / float(flow_rate)) R2.off() 165 Listing B.2 Programme to wash four new monoliths and then perform a cyclisation reaction on each one. import transport, vapourtec, vici R2 = vapourtec.R2R4(transport.serial("COM1")) V = vici.MultiValve(transport.serial("COM2")) # Function to run a solvent switch def solvent_switch (old_channel, new_channel, flow_rate, steps): for i in range(steps): change = (i + 1) * flow_rate / steps R2.flowrate(old_channel, flow_rate - change) R2.flowrate(new_channel, change) R2.wait(mins = 20) # Set up reactor R2.valve("both", "solvent") R2.flowrate("purple", 0) R2.flowrate("orange", 100) for i in range(4): R2.temp(i, 25) R2.on() # Run 4 columns for i in range(4): print "Column {:d}".format(i + 1) V.move(i + 2) print "Ethanol Wash" R2.wait(hours = 2) print "Solvent Switch to MeCN" solvent_switch("orange", "purple", 200, 4) print "Cyclisation" R2.valve("both", "reagent") R2.flowrate("both", 100) R2.wait(hours = 5) print "Rinse\n" R2.temp(i, -1000) R2.valve("both", "solvent") R2.wait(mins = 20) R2.flowrate("purple", 0) R2.off() 166 Control scripts for monolith processing Listing B.3 Programme to perform oxidative activation on four columns, and then run interleaved stop/flow reactions using a different amine for each column. The load/inject valve on the orange channel is used to isolate the multi-position valve during position changes to avoid contaminating columns with the wrong reagents; careful washing of the dead volume is required. import transport, vapourtec, vici R2 = vapourtec.R2R4(transport.serial("COM1")) V = vici.MultiValve(transport.serial("COM2")) cycles = 4 # Number of stop-flow cycles column_volume = 3000 # Monolith dead volume # Function to run a solvent switch def solvent_switch (old_channel, new_channel, flow_rate, steps): for i in range(steps): change = (i + 1) * flow_rate / steps R2.flowrate(old_channel, flow_rate - change) R2.flowrate(new_channel, change) R2.wait(mins = 20) # Set up reactor - initially pump DCM R2.valve("both", "solvent") R2.flowrate("purple", 100) R2.flowrate("orange", 0) V.move(1) for i in range(4): R2.temp(i, 25) R2.on() # Continued... 167 # Run oxidation + solvent switch on 4 columns for i in range(4): print "Column {:d}".format(i + 1) R2.valve("orange", "reagent") R2.flowrate("purple", 0) R2.flowrate("orange", 90) R2.wait(mins = 2) # Flush out any dioxane print "Pre-wash" R2.valve("orange", "load") V.move(i + 2) R2.valve("orange", "inject") print "Oxidation" R2.wait(hours = 2) print "Solvent Switch to Dioxane" R2.valve("both", "solvent") R2.flowrate("purple", 200) R2.flowrate("orange", 0) solvent_switch("purple", "orange", 200, 4) print "Post-wash" R2.valve("orange", "load") V.move(1) R2.valve("orange", "inject") R2.flowrate("purple", 500) R2.flowrate("orange", 0) R2.wait(mins = 1) # Wash through with Dioxane R2.valve("purple", "reagent") R2.valve("orange", "solvent") R2.flowrate("both", 500) R2.wait(mins = 2) # Continued... 168 Control scripts for monolith processing # Run stop-flow release cycles for cycle in range(cycles + 1): # Once for each column, per cycle... for i in range(4): print "Column {:d}".format(i + 1) # Wash through to second valve with dioxane R2.flowrate("orange", 500) R2.flowrate("purple", 0) R2.wait(mins = 1) if cycle < cycles: R2.temp(i, 100) # Wash through to first valve with amine R2.valve("orange", "load") V.move(i + 2) R2.flowrate("purple", 200) R2.wait(mins = 2) # Inject amine into column R2.flowrate("orange", 0) R2.flowrate("purple", 100) R2.valve("orange", "inject") R2.wait(mins = column_volume / 100.) R2.flowrate("purple", 0) # Rinse with dioxane after last cycle else: R2.temp(i, -1000) R2.valve("orange", "load") V.move(i + 2) R2.flowrate("orange", 100) R2.valve("orange", "inject") R2.wait(mins = column_volume / 100.) # Set up to wash through to second valve # with dioxane (on next cycle) R2.valve("orange", "load") V.move(1) R2.valve("orange", "inject") R2.off() Experimental 169 170 Experimental General Experimental Information Ethyl acetate, hexane, dichloromethane and toluene were obtained from Fisher Scientific and distilled before use. Absolute ethanol was obtained from Sigma-Aldrich, and HPLC grade methanol, isopropanol and acetonitrile were obtained from Fisher Scientific and used without further purification. Unless otherwise specified, all other reagents and solvents were obtained from commercial sources and used without further purification. All reactions were performed under an atmosphere of air unless otherwise specified. Silica gel chromatography was performed on a Biotage SP1 purification system with pre- packed SiliCycle silica cartridges (4, 12, 25 or 40 g), or similar cartridges manually packed with Biotage KP-NH silica. Analytical thin-layer chromatography (TLC) was performed using pre-coated glass-backed plates (Merck Kieselgel 60 F254) and visualized under ultra- violet light (short and long-wave) or using potassium permanganate (KMnO4). LC-MS analysis was performed on an Agilent HP 1100 series chromatography (Mercury Luna 3u C18 (2) column) attached to a Waters ZQ2000 mass spectrometer with ESCi ionisation source in ESI mode. Elution was carried out at a flow rate of 0.6 mL/min using a reverse phase gradient of MeCN and water containing 0.1 % formic acid: Time /min % MeCN 0.0 5 1.0 5 4.0 95 5.0 95 7.0 5 8.0 5 1H NMR spectra were recorded in CDCl3, DMSO-d6, or MeOH-d4 on a Bruker Avance DPX-400, DRX-500 Cryo or DRX-600 spectrometer with residual CHCl3, DMSO or MeOH as the internal reference (δH = 7.26, 2.50 or 4.87 ppm, respectively). 1H resonances are reported to the nearest 0.01 ppm. 13C NMR spectra were recorded in CDCl3, DMSO-d6 or MeOH-d4 on the same spectrometers with the central peak of CHCl3, DMSO or MeOH as the internal reference (δC = 77.0, 39.5 or 49.1 ppm, respectively). 13C resonances are reported to the nearest 0.01 ppm. DEPT-135, COSY, HMQC, and HMBC experiments were used to aid structural determination and spectral assignment. The multiplicity of the 1H and 13C signals are indicated as: s = singlet, d = doublet, t = triplet, q = quartet, m = multiplet, Experimental 171 br = broad, dd = doublet of doublets, etc. Coupling constants (J) are quoted in Hz and reported to the nearest 0.1 Hz. Infrared spectra were recorded on a Perkin-Elmer Spectrum One FT-IR spectrometer using universal ATR sampling accessories. Letters in parentheses refer to relative absorbance with respect to the most intense peak: w = weak (< 40 %), m = medium (40 – 75 %), s = strong (> 75 %). Melting points were performed on a Stanford Research Systems MPA100 (OptiMelt) automated melting point system and are uncorrected. High resolution mass spectrometry (HRMS) within ±5 ppm was carried out either on a Waters Micromass LCT Premier spectrometer using time of flight with positive electrospray ionisation, or by Mr Paul Skelton on a Bruker BioApex 47e FTICR spectrometer using positive ESI or EI at 70 eV, to within a tolerance of 5 ppm of the theoretically calculated value. Elemental composition microanalysis was performed by the Microanalytical Laboratories at the Department of Chemistry, University of Cambridge and results are reported to two decimal places. 172 Experimental Solid-supported synthesis of 2-aminopyrimidines General Experimental Information Monolithic reagents were formed and used in glass Omnifit columns, sealed for polymeri- sation with custom made PTFE end pieces and heated using a Vapourtec R4 convection heating system. Magic Angle Spinning NMR spectra were recorded by Dr David Reid on a Bruker DRX- 400 spectrometer at room temperature using a Bruker HR-MAS probe at the Cambridge University Chemical Laboratory. Divinylbenzene (80 % technical grade) was obtained from Sigma-Aldrich. Amino[(4-ethenylbenzyl)sulfanyl]methanaminium chloride (11)287 3 S1H2N NH2 2' 3' 5' 6' Cl 4-Vinylbenzyl chloride (60.0 g, 0.394 mmol) and thiourea (25.00 g, 0.329 mmol) in ethanol (165 mL) were heated to reflux for 6 h, until all of the thiourea had been consumed as observed by TLC (mobile phase: 1:1 EtOAc / hexane, v/v). The ethanol was removed under reduced pressure and the resulting yellow solid was washed with ether (3×20 mL). Drying in vacuo gave the title compound (67.1 g, 0.29 mmol, 90 %) as an off-white solid, m.p. >250 ◦C (decomp.). 1H NMR (600 MHz, DMSO-d6): δ /ppm = 9.49 (br. s, 2 H, NH2), 9.37 (br. s, 2 H, NH2), 7.46 (d, J = 8.1 Hz, 2 H, H-2′), 7.39 (d, J = 8.1 Hz, 2 H, H-3′), 6.71 (dd, J = 10.9 Hz and 6.7 Hz, 1 H, H-5′), 5.83 (d, J = 6.7 Hz, 1 H, H-6′cis), 5.26 (d, J = 10.9 Hz, 1 H, H-6′trans), 4.52 (s, 2 H, H-3). 13C NMR (100 MHz, DMSO-d6): δ /ppm = 169.29 (C-1), 136.75 (C-4′), 136.06 (CH, C-5′), 134.93 (C-1′), 129.33 (CH, C-2′), 126.46 (CH, C-3′), 114.83 (CH2, C-6′), 33.97 (CH2, C-3). IR (neat): ν /cm−1 = 3270 (w, br.), 3179 (w, br.), 3043 (m, br.), 2722 (w), 1655 (s), 1629 (w), 1508 (w), 1437 (m), 1405 (m), 1306 (w), 1256 (w), 1206 (w), 1164 (w), 1113 (w), 1093 (w), 1017 (w), 987 (m), 915 (m), 908 (m), 832 (m), 675 (s, br). HRMS: m/z calc. for [C10H13S1N2] + ([M – Cl]+) 193.0794, found 193.0800, ∆ = 3.1 ppm. Experimental 173 Thiouronium chloride monolith (M1) SH2N NH2Cl The reaction scale specified below forms a monolith that fills a 7.845 mL glass Omnifit column (10 mm ID × 100 mm length). Multiple monoliths, or monoliths of other volumes, can be made from the same stock solution by multiplying the values below as appro- priate. Divinylbenzene (DVB) (0.77 g, 5.9 mmol) was added to amino[(4-ethenylbenz- yl)sulfanyl]methanaminium chloride (1.16 g, 5.0 mmol) suspended in 1-propanol (3.59 g, 81.5 mmol) and water (0.65 g, 36.1 mmol) and the mixture was heated to 50 ◦C. Once complete dissolution had been achieved, 1,1′-azobis(cyclohexanecarbonitrile) (ACHC) (19.4 mg, 0.08 mmol, 0.3 mol% with respect to the monomer + crosslinker) was added and the mixture stirred for another 5 minutes without further heating. A 10 mm ID × 100 mm length glass column was filled to a height of 7 cm with this solution, and both ends of the column were sealed with custom-made PTFE end plugs. The column was heated at 90 ◦C for 20 hours using a Vapourtec R4 convection heater, resulting in a rigid white monolith (the start of precipitation polymerisation was observed after 30 minutes at temperature). Following polymerisation, the monolith was allowed to cool to room temperature, and the end seals were exchanged for standard tubing connectors. To remove the porogen and any residual nonpolymeric material, the monolith was then heated to 60 ◦C and washed with ethanol at 0.2 mL/min, without a back-pressure regulator, for 1 hour using a Vapourtec R2+ flow system. A 100 psi back-pressure regulator was added to the system and the monolith was then flushed at 0.2 mL/min for another hour. The flow rate was then slowly increased to 1.5 mL/min over the next hour, the monolith inverted and then the process repeated. The monolith was finally flushed at 0.2 mL/min for 30 minutes while being allowed to cool to room temperature to give the title compound (dry weight 1.6 g) as a white monolith. 13C NMR (100 MHz, solid state): δ /ppm = 171.38, 144.98, 128.23, 40.35. IR (neat): ν /cm−1 = 2160 (w, br.), 1647 (m), 1438 (m). Microanalysis: found C 67.47%, H 6.71%, N 7.34%, Cl 9.25% (Loading = 2.6 mmol/g, based on proportions of N and Cl). 174 Experimental Linear pressure response of monolith M1: 2 4 6 8 10 12 14 16 0.04 0.06 0.08 0.10 0.12 0.14 0.16 0.18 0.20 Pre ssu re (ba r) Flow rate (mL/min) 4-Amino-2-((4-vinylbenzyl)thio)pyrimidine-5-carbonitrile (15) NC H2N 4 N 2 S 2' 3' 5' 6' N 6 To a mixture of amino[(4-ethenylbenzyl)sulfanyl]methanaminium chloride (11, 1.6 g, 7.0 mmol) and ethoxymethylene(malononitrile) (620 mg) in ethanol (40 mL) was added (i−Pr)2NEt (1.3 mL) in one portion at 25 ◦C with stirring. The mixture was stirred for 1 hour, during which time a yellow precipitate formed. The solvent was removed, and the residue partitioned between ethyl acetate (30 mL) and water (30 mL). The organic phase was washed with water (2×30 mL), brine (2×30 mL), dried (MgSO4) and the solvent removed to afford the title compound (1.6 g, 5.8 mmol, 83 %) as a yellow solid. A sample was purified by column chromatography for analysis (eluent: ethyl acetate / hexane; 0 : 1 to 3 : 7 over 35 CV, 3 : 7 to 9 : 1 over 5 CV, then 9 : 1 for 20 CV), m.p. 163 – 164 ◦C. 1H NMR (400 MHz, CDCl3): δ /ppm = 8.34 (s, 1 H, H-6), 7.35 (br. s, 4 H, H-2′ and H-3′), 6.69 (dd, J = 17.6 Hz and 10.9 Hz, 1 H, H-6′trans), 5.73 (dd, J = 17.6 Hz and 0.9 Hz, 1 H, H-6′cis), 5.60 (br. s, 2 H, NH2), 5.24 (dd, J = 10.9 Hz and 0.9 Hz, 1 H, H-5 ′), 4.35 (s, 2 H, CH2). 13C NMR (100 MHz, CDCl3): δ /ppm = 175.73, 161.55, 159.61 (CH), 136.84, 136.30 (CH), 129.18 (CH), 126.41 (CH), 114.94, 114.04 (CH2), 86.57, 35.22 (CH2). IR (neat): ν /cm−1 = 3804 (w), 3751 (w), 3711 (w), 3630 (w), 3600 (w), 3399 (w), 3328 (w), Experimental 175 3217 (w), 2383 (w), 2359 (w), 2343 (w), 2326 (w), 2310 (w), 2225 (w), 2126 (w), 2036 (w), 1908 (w), 1647 (s), 1587 (s), 1538 (s), 1511 (m), 1474 (m), 1432 (w), 1405 (w), 1381 (s), 1326 (w), 1236 (m), 1159 (m), 1100 (w), 1048 (w), 1018 (w), 986 (w), 971 (w), 951 (m), 899 (m), 844 (m), 773 (m), 754 (w), 733 (w), 710 (w), 682 (w). HRMS: m/z calc. for [C14H13N4S] + ([M + H]+) 269.0861, found 269.0857, ∆ = −1.5 ppm. Monolith M2 N S N NC H2N A solution of ethoxymethylene(malononitrile) (15 mmol, 3 equiv.) in DMF / MeCN (1 : 1, v/v, 20 mL) and (i−Pr)2NEt (5.21 mL, 30 mmol, 6 equiv.) in MeCN (20 mL) were each pumped at 0.1 mL/min, to combine in a tee mixer ( 0.5 mm, PEEK). The resulting stream passed into the monolith M1, which was maintained at 25 ◦C. The system was pressurised using a 100 psi back-pressure regulator placed in-line after the monolith. During the loading process, the monolith gradually assumed a dark yellow colour. After loading, the system was flushed for 1 hour with EtOH at 0.2 mL/min, whilst maintaining the temperature at 25 ◦C. The obtained monolith M2 was used directly in the following step. IR (neat): ν /cm−1 = 3361 (w), 2919 (w), 2218 (w), 1618 (m), 1574 (s), 1535 (m), 1511 (m), 1485 (m), 1419 (w), 1363 (s), 1315 (w), 1221 (m), 1020 (w), 950 (w), 779 (m), 709 (m). Dry weight: 1.47 g. Microanalysis: found C 72.43%, H 5.82%, N 12.39%. Oxidised monolith M3 N S N NC H2N O O Monolith M2 was washed for 30 minutes with CH2Cl2 pumped at 0.5 mL/min. A solution of m-CPBA (15 mmol, 3 equiv.) in CH2Cl2 (20 mL) was pumped, from a reservoir stored at 0 ◦C (ice/water bath), at 0.1 mL/min through the monolith, which was maintained at 25 ◦C. 176 Experimental The monolith assumed a pale yellow colour after the oxidation reaction. At the end of the reaction, the system was flushed with CH2Cl2 for 2 hours at room temperature with a flow rate of 0.1 mL/min. The obtained monolith M3 was used directly in the following step. IR (neat): ν /cm−1 = 2919 (w), 2231 (w), 2162 (w), 2023 (w), 2012 (w), 1955 (w), 1642 (m), 1576 (m), 1545 (m), 1511 (m), 1486 (w), 1441 (m), 1382 (m), 1325 (m), 1218 (m), 1168 (m), 1127 (s), 1038 (s), 1020 (m), 961 (w), 881 (w), 829 (m), 787 (m), 710 (s). Dry weight: 1.63 g. Microanalysis: found C 67.32%, H 5.47%, N 8.27%. 4-Amino-2-morpholin-4-ylpyrimidine-5-carbonitrile (18)288 4 N 2 N 6 N O 2' 3' N H2N Monolith M3 was washed for 30 minutes with dioxane pumped at 0.5 mL/min. A solution of morpholine (30 mmol, 6 equiv.) and (i−Pr)2NEt (5.21 mL, 30 mmol, 6 equiv.) in dioxane (20 mL) was infused with 5 mL of amine solution, pumping at 0.1 mL/min. After 50 minutes, the pump was stopped and monolith was heated at a temperature of 80 ◦C. After 2 hours, the reaction solution was flushed off with a new portion of the reaction solution (flow rate 0.1 mL/min) and the output collected in a flask. The flushing operation was repeated four times to afford 20 mL of reaction solution. After the last loading process, the monolith was washed with EtOH at 0.2 mL/min for 30 minutes. The combined output was concentrated under vacuum, and the resulting solid suspended in CH2Cl2 (50 mL) and washed with water (3×50 mL). The organic phase was dried (MgSO4) and the solvent removed under reduced pressure. The crude material was purified by flash column chromatography (eluent: CH2Cl2 / MeOH from 10 : 0 to 9.8 : 0.2 over 40 CV) to afford the title compound (297 mg, 1.45 mmol, 35 % over 3 steps) as a white solid, m.p. 227 – 229 ◦C (dichloromethane / methanol) [lit: 229 – 231 ◦C (ethanol)289]. 1H NMR (500 MHz, DMSO-d6): δ /ppm = 8.26 (s, 1 H, H-6), 7.28 (br. s, 2 H, NH2), 3.70 (t, J = 4.8 Hz, 4 H, H-3′), 3.59 (t, J = 4.8 Hz, 4 H, H-2′). 13C NMR (126 MHz, DMSO-d6): δ /ppm = 162.87 (C-4), 162.03 (CH, C-6), 160.79 (C-2), 117.29 (CN), 78.66 (C-5), 65.99 (CH2, C-2 ′), 43.76 (CH2, C-3′). IR (neat): ν /cm−1 = 3418 (w), 3335 (m), 3231 (m), 2960 (w), 2906 (w), 2852 (w), 2222 (m), 1643 (s), 1590 (s), 1540 (s), 1516 (s), 1477 (s), 1443 Experimental 177 (s), 1349 (s), 1311 (m), 1275 (s), 1251 (s), 1223 (m), 1193 (m), 1117 (m), 1103 (s), 1066 (m), 1036 (m), 970 (s), 918 (m), 905 (s), 851 (m), 838 (m), 793 (m), 784 (s), 756 (m), 732 (w), 698 (w). HRMS: m/z calc. for [C9H12N5O] + ([M + H]+) 206.1042, found 206.1051, ∆ = 4.4 ppm. General Procedure A for the preparation of enaminones 21 A mixture of the ketone (50 mmol) and 1,1-dimethoxy-N,N-dimethylmethanamine (7.30 mL, 1.1 equiv.) was heated at 100 ◦C for 12 hours. The reaction mixture was concentrated under vacuum and crystallized from diethyl ether. The crystals were collected by filtration, washed with diethyl ether (2×20 mL) and dried under high vacuum to give crystalline solid. (E)-3-(Dimethylamino)-1-(pyridin-4-yl)prop-2-en-1-one (21a)290,291 2' N 3' 2 O 3 N Prepared according to General Procedure A, to give the title compound (7.2 g, 41 mmol, 82 %) as yellow crystals, m.p. 115 – 116 ◦C (diethyl ether) [lit: 115 – 117 ◦C (ethyl acetate / hexane)291]. 1H NMR (400 MHz, CDCl3): δ /ppm = 8.65 (dd, J = 4.4 Hz and 1.7 Hz, 2 H, H-2′), 7.78 (d, J = 12.3 Hz, 1 H, H-3), 7.63 (dd, J = 4.4 Hz and 1.7 Hz, 2 H, H-3′), 5.60 (d, J = 12.3 Hz, 1 H, H-2), 3.13 (s, 3 H, Me), 2.90 (s, 3 H, Me). 13C NMR (100 MHz, CDCl3): δ /ppm = 186.56 (C-1), 155.21 (CH, C-3), 150.21 (CH, C-2′), 147.25 (C-4′), 121.22 (CH, C-3′), 91.75 (CH, C-2), 45.32 (CH3), 37.45 (CH3). IR (neat): ν /cm−1 = 3032 (w), 1637 (m), 1594 (w), 1562 (m), 1523 (m), 1485 (w), 1432 (m), 1408 (m), 1367 (m), 1320 (w), 1274 (m), 1247 (m), 1130 (m), 1070 (m), 1058 (m), 1013 (m), 992 (m), 904 (m), 848 (m), 774 (s), 698 (s), 660 (s). HRMS: m/z calc. for [C10H13N2O] + ([M + H]+) 177.1028, found 177.1032, ∆ = 2.3 ppm. Microanalysis: calc. (found) for C10H12N2O C 68.16% (68.03%), H 6.86% (6.88%), N 15.90% (15.79%). 178 Experimental (E)-3-(Dimethylamino)-1-(pyridin-3-yl)prop-2-en-1-one (21b)291 5' 6' N 2' 4' 2 O 3 N Prepared according to General Procedure A, to give the title compound (6.6 g, 37 mmol, 75 %) as orange crystals, m.p. 78 – 79 ◦C (diethyl ether) [lit: 71 – 73 ◦C (ethyl acetate / hexane)291]. 1H NMR (400 MHz, CDCl3): δ /ppm = 9.03 (d, J = 2.2 Hz, 1 H, H-2′), 8.61 (dd, J = 4.8 Hz and 1.8 Hz, 1 H, H-6′), 8.13 (dt, J = 7.9 Hz and 2.0 Hz, 1 H, H-4′), 7.78 (d, J = 12.3 Hz, 1 H, H-3), 7.30 (dd, J = 7.9 Hz and 4.8 Hz, 1 H, H-5′), 5.63 (d, J = 12.3 Hz, 1 H, H-2), 3.12 (s, 3 H, Me), 2.89 (s, 3 H, Me). 13C NMR (100 MHz, CDCl3): δ /ppm = 186.35 (C-1), 154.68 (CH, C-3), 151.47 (CH, C-2′), 148.93 (CH, C-6′), 135.67 (C-3′), 135.05 (CH, C-4′), 123.29 (CH, C-5′), 91.86 (CH, C-2), 45.27 (CH3), 37.43 (CH3). IR (neat): ν /cm−1 = 3372 (w), 3054 (w), 2918 (w), 2805 (w), 1637 (s), 1575 (s), 1533 (s), 1439 (m), 1411 (s), 1364 (s), 1328 (s), 1275 (s), 1249 (s), 1186 (m), 1125 (m), 1107 (m), 1065 (s), 1035 (m), 1023 (s), 1001 (m), 965 (m), 902 (s), 813 (m), 771 (s), 723 (s), 701 (s), 684 (s). HRMS: m/z calc. for [C10H13N2O] + ([M + H]+) 177.1028, found 177.1028, ∆ = 0 ppm. Microanalysis: calc. (found) for C10H12N2O C 68.16% (67.75%), H 6.86% (6.83%), N 15.90% (15.67%). (E)-3-(Dimethylamino)-1-phenylprop-2-en-1-one (21c)291 3' 4' 2' 2 O 3 N Prepared according to General Procedure A, to give the title compound (6.9 g, 39 mmol, 79 %) as yellow crystals, m.p. 90 – 91 ◦C (diethyl ether) [lit: 89 – 92 ◦C (diethyl ether)292]. 1H NMR (400 MHz, CDCl3): δ /ppm = 7.91 – 7.83 (m, 2 H, H-2′), 7.76 (d, J = 12.3 Hz, 1 H, H-3), 7.46 – 7.32 (m, 3 H, H-4′ and H-3′), 5.68 (d, J = 12.3 Hz, 1 H, H-2), 3.06 (s, 3 H, Me), 2.87 (s, 3 H, Me). 13C NMR (100 MHz, CDCl3): δ /ppm = 188.56 (C-1), 154.22 (CH, C-3), 140.54 (C-1′), 130.84 (CH, C-4′), 128.10 (CH, C-2′), 127.46 (CH, C-3′), 92.18 (CH, C-2), 44.98 (CH3), 37.20 (CH3). IR (neat): ν /cm−1 = 3024 (w), 2910 (w), 2808 (w), 1634 Experimental 179 (m), 1596 (w), 1582 (m), 1537 (s), 1483 (m), 1446 (w), 1429 (m), 1410 (m), 1363 (m), 1310 (m), 1273 (s), 1232 (m), 1205 (m), 1122 (m), 1079 (w), 1052 (s), 1025 (m), 1014 (m), 1000 (m), 925 (w), 899 (m), 807 (w), 754 (m), 739 (s), 696 (s), 659 (s). HRMS: m/z calc. for [C11H14NO] + ([M + H]+) 176.1075, found 176.1067, ∆ = −4.5 ppm. Microanalysis: calc. (found) for C11H13NO C 75.40% (75.33%), H 7.48% (7.49%), N 7.99% (7.98%). (E)-3-(Dimethylamino)-1-(4-(trifluoromethyl)phenyl)prop-2-en-1-one (21d)291 3' 2' 2 O 3 N F3C Prepared according to General Procedure A, to give the title compound (8.6 g, 35 mmol, 71 %) as yellow crystals, m.p. 96 – 97 ◦C (diethyl ether) [lit: 99 – 100 ◦C (ethyl acetate / hexane)291]. 1H NMR (400 MHz, CDCl3): δ /ppm = 7.95 (d, J = 8.0 Hz, 2 H, H-2′), 7.80 (d, J = 12.3 Hz, 1 H, H-3), 7.63 (d, J = 8.0 Hz, 2 H, H-3′), 5.65 (d, J = 12.3 Hz, 1 H, H-2), 3.13 (s, 3 H, Me), 2.90 (s, 3 H, Me). 13C NMR (100 MHz, CDCl3): δ /ppm = 187.02 (C-1), 154.80 (CH, C-3), 143.69 (q, 5JC−F = 1.1 Hz, C-2′), 132.17 (q, 2JC−F = 32.3 Hz, C-4′), 127.70 (CH, C-1′), 125.06 (q, 3JC−F = 3.8 Hz, CH, C-3′), 123.97 (q, 1JC−F = 273.3 Hz, CF3), 91.91 (CH, C-2), 45.12 (CH3), 37.29 (CH3). IR (neat): ν /cm−1 = 1638 (m), 1586 (m), 1547 (s), 1489 (w), 1427 (m), 1365 (w), 1319 (m), 1279 (m), 1237 (m), 1157 (s), 1106 (s), 1063 (s), 1011 (m), 982 (m), 960 (m), 902 (m), 856 (s), 843 (m), 819 (w), 777 (s), 764 (m), 750 (m), 695 (s), 680 (m). HRMS: m/z calc. for [C12H13NO 19F3] + ([M + H]+) 244.0949, found 244.0956, ∆ = 2.9 ppm. Microanalysis: calc. (found) for C12H13NOF3 C 59.26% (59.39%), H 4.97% (4.98%), N 5.76% (5.74%). 180 Experimental General Procedure B for the preparation of monolith M4 N S N N N S N N N S N M4a M4b M4c N S N CF3 N S N S N S N OMe M4d M4e M4f Solutions (20 mL each) of enaminone (15 mmol, 3 equiv.) and (i−Pr)2NEt (5.21 mL, 30 mmol, 6 equiv.) in EtOH were each pumped at 0.1 mL/min, to combine in a tee mixer ( 0.5 mm, PEEK). The resulting reaction stream passed into the monolith M1, which was maintained at 80 ◦C. The system was pressurised using a 100 psi back-pressure regulator placed in-line after the monolith. During the loading process, the monolith gradually assumed a pale yellow colour. After loading, the system was flushed for 1 hour with EtOH at 0.2 mL/min, whilst maintaining the temperature at 80 ◦C. The obtained monoliths M4 were used directly in the following step. For monolith M4a: IR (neat): ν /cm−1 = 3663 (m), 2988 – 2901 (s), 1406 – 1382 (s), 1250 – 1230 (s), 1066 (s), 892 – 879 (m). 13C NMR (100 MHz, solid state): δ /ppm = 144.22, 127.70, 39.82. Dry weight: 1.6 g. Microanalysis: found C 78.90%, H 6.2%, N 8.49%, Cl 0%. Experimental 181 General Procedure C for the preparation of monolith M5 N S N N O O N S N N O O N S N O O M5a M5b M5c N S N CF3 O O N S N S O O N S N OMe O O M5d M5e M5f The monolith was washed for 30 minutes with CH2Cl2 pumped at 0.5 mL/min. A solution of m-CPBA (15 mmol, 3 equiv.) in CH2Cl2 (20 mL) was pumped, from a reservoir stored at 0 ◦C using an ice-water bath, at 0.1 mL/min through the monolith M4, which was maintained at room temperature. The monolith retained its pale yellow colour after the oxidation reaction. At the end of the reaction, the system was flushed with CH2Cl2 for 2 hours at room temperature with a flow rate of 0.1 mL/min. The obtained monoliths M5 were used directly in the following step. The enaminone starting materials could be recovered by removing the solvent from the collected waste stream, partitioning the concentrated mixture between ethyl acetate and water, and then collecting and drying the organic phase. For monolith M5a: IR (neat): ν /cm−1 = 3663 (m), 2988 – 2901 (s), 1715 (w), 1406 – 1381 (s), 1250 – 1229 (s), 1066 (s), 892 (m). Dry weight: 1.6 g. Microanalysis: found C 75.20%, H 5.7%, N 8.2%, Cl 0%. 182 Experimental General Procedure D for the preparation of 2-amino pyrimidines (38) The monolith was washed for 30 minutes with dioxane pumped at 0.5 mL/min. A solution of amine (30 mmol, 6 equiv.) and (i−Pr)2NEt (5.21 mmol, 30 mmol, 6 equiv.) in dioxane (20 mL) was prepared. The monolith M5 was infused with 5 mL of amine solution, pumping at 0.1 mL/min. After 50 minutes, the pump was stopped and monolith was heated at a temperature of 90 ◦C (except for Entry 4: 120 ◦C). After 2 hours, the reaction solution was flushed off with a new portion of the reaction solution (flow rate 0.1 mL/min) and the output collected in a flask. The flushing operation was repeated four times to afford 20 mL of reaction solution. After the last loading process, the monolith was washed with EtOH at 0.2 mL/min for 30 minutes. The combined output was concentrated under vacuum, and the resulting oil was suspended in CH2Cl2 (50 mL) and washed with water (3×50 mL). The organic phase was dried (MgSO4) and the solvent removed under reduced pressure. The crude material was purified by flash column chromatography to afford the desired pyrimidine compound. 4-(4-(Pyridin-4-yl)pyrimidin-2-yl)morpholine (38a) N 1 N 3 2 N 1' 2' O 6 N 7 Prepared from M5a and morpholine using General Procedure D. Purification by flash column chromatography (eluent: CH2Cl2 / MeOH from 10 : 0 to 9.8 : 0.2 over 40 CV) afforded the title compound (726 mg, 3 mmol, 73 % over 3 steps) as a pale yellow solid. 1H NMR (500 MHz, CDCl3): δ /ppm = 8.74 (dd, J = 5.0 Hz and 1.6 Hz, 2 H, H-7), 8.46 (d, J = 5.0 Hz, 1 H, H-2), 7.90 (dd, J = 5.0 Hz and 1.6 Hz, 2 H, H-6), 7.00 (d, J = 5.0 Hz, 1 H, H-3), 3.91 – 3.89 (m, 4 H, H-2′), 3.81 – 3.79 (m, 4 H, H-1′). 13C NMR (126 MHz, CDCl3): δ /ppm = 161.99 (C-1 or C-4), 161.81 (C-1 or C-4), 159.13 (CH, C-2), 150.27 (CH, C-7), 145.07 (C-5), 121.01 (CH, C-6), 106.18 (CH, C-3), 66.84 (CH3, C-2 ′), 44.22 (CH3, C-1 ′). IR (neat): ν /cm−1 = 2538 (w, br.), 2159 (s), 2031 (s), 1977 (s), 1583 (s), 1561 – 1543 (s, br.), 1487 (s), 1340 (s), 1248 (s), 985 – 962 (s), 800 (s). HRMS: m/z calc. for [C13H15O1N4] + ([M + H]+) 243.1240, found 243.1232, ∆ = −3.5 ppm. Experimental 183 N-(4-Methoxybenzyl)-4-(pyridin-4-yl)pyrimidin-2-amine (38b) N 1 N 3 2 N H 1'3' 4' MeO N 7 6 Prepared from M5a and (4-methoxyphenyl)methanamine using General Procedure D. Purification by flash column chromatography (eluent: CH2Cl2 / MeOH from 10 : 0 to 9.7 : 0.3 over 40 CV) afforded the title compound (320 mg, 1.2 mmol, 29 % over 3 steps) as a pale yellow solid. 1H NMR (500 MHz, CDCl3): δ /ppm = 8.77 (d, J = 5.5 Hz, 2 H, H-7), 8.46 (d, J = 5.1 Hz, 1 H, H-2), 7.92 (d, J = 5.5 Hz, 2 H, H-6), 7.35 (d, J = 8.4 Hz, 2 H, H-4′), 7.07 (d, J = 5.1 Hz, 1 H, H-3), 6.92 (d, J = 8.4 Hz, 2 H, H-3′), 5.61 (br. s, 1 H, NH), 4.69 (d, J = 5.5 Hz, 2 H, H-1′), 3.83 (s, 3 H, H-6′). 13C NMR (126 MHz, CDCl3): δ /ppm = 162.61 (C-1 or C-4), 162.46 (C-1 or C-4), 159.43 (CH, C-2), 159.09 (C-5′), 150.46 (CH, C-7), 145.04 (C-5), 131.15 (C-2′), 129.04 (CH, C-3′), 121.18 (CH, C-6), 114.18 (CH, C-4′), 107.04 (CH, C-3), 55.46 (CH3, C-6 ′), 45.23 (CH2, C-1′). IR (neat): ν /cm−1 = 2161 (w, br), 1977 (w, br), 1570 (s), 1548 (s), 1510 (s), 1454 (s), 1346 (s), 1230 (s), 1175 (s), 1030 (s), 795 (s). HRMS: m/z calc. for [C17H17O1N4] + ([M + H]+) 293.1402, found 293.1410, ∆ = 2.7 ppm. N-Cyclohexyl-4-(pyridin-4-yl)pyrimidin-2-amine (38c) N 1 N 3 2 N H 1' 2' 3' 4' N 7 6 Prepared from M5a and (4-methoxyphenyl)methanamine using General Procedure D. Purification by flash column chromatography (eluent: CH2Cl2 / MeOH from 10 : 0 to 9.8 : 0.2 over 40 CV) afforded the title compound (533 mg, 2.1 mmol, 51 % over 3 steps) as a white solid. 1H NMR (400 MHz, CDCl3): δ /ppm = 8.74 (d, J = 4.5 Hz, 2 H, H-7), 8.40 (d, J = 5.1 Hz, 1 H, H-2), 7.86 (d, J = 4.5 Hz, 2 H, H-6), 6.96 (d, J = 5.1 Hz, 1 H, H-3), 5.18 (d, J = 8.0 Hz, 1 H, H-1′), 2.13 – 1.22 (m, 11 H, NH and cyclohexyl). 13C NMR (100 MHz, CDCl3): 184 Experimental δ /ppm = 162.09 (C-1 or C-4), 159.31 (CH, C-2), 150.48 (CH, C-7), 144.87 (C-5), 120.90 (CH, C-6), 106.25 (CH, C-3), 49.80 (CH, C-1′), 33.13 (CH2, C-2′), 25.74 (CH2, C-4′), 24.86 (CH2, C-3 ′), (C-1 or C-4 was not visible). IR (neat): ν /cm−1 = 2921 (w, br), 2852 (s), 1581 (s), 1548 (s), 1583 (s), 1528 (s), 1421 (s), 1368 (s), 796 (s). HRMS: m/z calc. for [C15H19N4] + ([M + H]+) 255.1610, found 255.1611, ∆ = 0.4 ppm. 4-(4-(Pyridin-3-yl)pyrimidin-2-yl)morpholine (38d) N 1 N 3 2 N 1' 2' O 6 10 9 8 N Prepared from M5b and morpholine using General Procedure D. Purification by flash column chromatography (eluent: CH2Cl2 / MeOH from 10 : 0 to 9.7 : 0.3 over 40 CV) afforded the title compound (713 mg, 2.9 mmol, 70 % over 3 steps) as a white solid. 1H NMR (500 MHz, CDCl3): δ /ppm = 9.24 (d, J = 2.3 Hz, 1 H, H-6), 8.69 (dd, J = 4.8 Hz and 1.7 Hz, 1 H, H-8), 8.42 (d, J = 5.1 Hz, 1 H, H-2), 8.33 (ddd, J = 8.0 Hz and 2.3 Hz and 1.7 Hz, 1 H, H-10), 7.45 – 7.38 (m, 1 H, H-9), 6.98 (d, J = 5.1 Hz, 1 H, H-3), 3.90 (dd, J = 5.7 Hz and 4.0 Hz, 4 H, H-2′), 3.80 (dd, J = 5.7 Hz and 4.0 Hz, 4 H, H-1′). 13C NMR (126 MHz, CDCl3): δ /ppm = 161.90 (C-1 or C-4), 161.86 (C-1 or C-4), 158.76 (CH, C-2), 150.95 (CH, C-6), 148.26 (CH, C-8), 134.58 (CH, C-10), 133.24 (C-5), 123.59 (CH, C-9), 105.90 (CH, C-3), 66.85 (CH2, C-2 ′), 44.24 (CH2, C-1′). IR (neat): ν /cm−1 = 2868 (w, br), 1586, (s), 1565-1556 (s), 1444-1412 (s), 1346 (s), 1253 (s), 1110 (s), 984 (s), 962 (s), 790 (s), 800 (s). HRMS: m/z calc. for [C13H15O1N4] + ([M + H]+) 243.1246, found 243.1252, ∆ = 2.5 ppm. Experimental 185 N-(2-Methyl-5-nitrophenyl)-4-(pyridin-3-yl)pyrimidin-2-amine (24)293,294 N 1 N 3 2 N H 2'4' 5' 7' NO2 6 10 9 8 N Prepared from M5b and 2-methyl-5-nitroaniline using General Procedure D. Purification by flash column chromatography (eluent: CH2Cl2 / MeOH from 10 : 0 to 9.7 : 0.3 over 40 CV) afforded the title compound (619 mg, 2.01 mmol, 49 % over 3 steps) as a pale yellow solid. 1H NMR (400 MHz, DMSO-d6): δ /ppm = 9.31 (s, 1 H, H-6), 9.18 (s, 1 H, NH), 8.81 (s, 1 H, H-2′), 8.70 (d, J = 4.7 Hz, 1 H, H-8), 8.61 (d, J = 5.2 Hz, 1 H, H-2), 8.46 (d, J = 8.2 Hz, 1 H, H-10), 7.87 (dd, J = 8.2 Hz and 2.5 Hz, 1 H, H-4′), 7.59 – 7.45 (m, 3 H, H-3, H-9 and H-5′), 2.42 (s, 3 H, H-7′). 13C NMR (100 MHz, DMSO-d6): δ /ppm = 161.56 (C-1), 160.25 (C-4), 159.60 (CH, C-2), 151.57 (CH, C-6), 148.13 (CH, C-8), 145.80 (C-3′), 138.85 (C-6′), 138.54 (C-1′), 134.24 (CH, C-10), 131.84 (C-5), 131.15 (CH, C-5′), 123.80 (CH, C-9), 117.82 (CH, C-4′), 117.33 (CH, C-2′), 108.85 (CH, C-3), 18.33 (CH3, C-7′). IR (neat): ν /cm−1 = 3250 (w), 3099 (w), 3066 (w), 1602 (w), 1580 (m), 1557 (m), 1531 (s), 1515 (s), 1477 (m), 1456 (m), 1440 (m), 1420 (m), 1406 (s), 1345 (s), 1305 (m), 1284 (m), 1270 (m), 1248 (w), 1208 (w), 1196 (w), 1136 (w), 1108 (w), 1085 (w), 1027 (m), 991 (w), 961 (w), 930 (w), 882 (w), 830 (w), 817 (w), 810 (w), 791 (s), 754 (w), 736 (m), 698 (m), 686 (m). HRMS: m/z calc. for [C16H14N5O2] + ([M + H]+) 308.1148, found 308.1145, ∆ = −1.0 ppm. 4-(4-Phenylpyrimidin-2-yl)morpholine (38e) N 1 N 3 2 N 1' 2' O 8 7 6 Prepared from M5c and morpholine using General Procedure D. Purification by flash column chromatography (eluent: CH2Cl2 / MeOH from 10 : 0 to 9.8 : 0.2 over 40 CV) afforded the title compound (482 mg, 2.0 mmol, 48 % over 3 steps) as a pale yellow solid. 186 Experimental 1H NMR (500 MHz, CDCl3): δ /ppm = 8.38 (d, J = 5.1 Hz, 1 H, H-2), 8.05–8.01 (m, 2 H, H-6), 7.48–7.45 (m, 3 H, H-7 and H-8), 6.97 (d, J = 5.1 Hz, 1 H, H-3), 3.92–3.88 (m, 4 H, H-2′), 3.82–3.78 (m, 4 H, H-1′). 13C NMR (126 MHz, CDCl3): δ /ppm = 164.34 (C-1), 161.98 (C-4), 158.30 (CH, C-2), 137.53 (C-5), 130.52 (CH, C-8), 128.66 (CH, C-7), 126.98 (CH, C-6), 106.11 (CH, C-3), 66.92 (CH2, C-2 ′), 44.28 (CH2, C-1′). IR (neat): ν /cm−1 = 2969 (w, br.), 2850 (s), 1588 (s), 1564 (s), 1547 (s), 1475 (s), 1441-1426 (m, br), 1340 (s), 1248 (s), 982 (s), 787 (s), 694 (s). HRMS: m/z calc. for [C14H16N3O] + ([M + H]+) 242.1293, found 242.1294, ∆ = 0.4 ppm. 4-(4-(4-(Trifluoromethyl)phenyl)pyrimidin-2-yl)morpholine (38f) N 1 N 3 2 N 1' 2' O 6 7 CF3 Prepared from M5d and morpholine using General Procedure D. Purification by flash column chromatography (eluent: CH2Cl2 / MeOH from 10 : 0 to 9.9 : 0.1 over 40 CV) afforded the title compound (803 mg, 2.6 mmol, 63 % over 3 steps) as a pale yellow solid. 1H NMR (500 MHz, CDCl3): δ /ppm = 8.43 (d, J = 5.1 Hz, 1 H, H-2), 8.13 (d, J = 8.1 Hz, 2 H, H-7), 7.71 (d, J = 8.1 Hz, 2 H, H-6), 6.98 (d, J = 5.1 Hz, 1 H, H-3), 3.93 – 3.89 (m, 4 H, H-2′), 3.85 – 3.79 (m, 4 H, H-1′). 13C NMR (126 MHz, CDCl3): δ /ppm = 163.04 (C-1), 162.12 (C-4), 158.94 (CH, C-2), 141.14 (C-6), 127.48 (C-5), 125.77 (q, 3JC−F = 3.8 Hz, CH, C-7), 124.12 (q, 1JC−F = 272.6 Hz, C-9), 106.42 (CH, C-3), 67.03 (CH2, C-2′), 44.42 (CH2, C-1 ′), (C-8 was not visible). IR (neat): ν /cm−1 = 2960 (w, br.), 2861 (s), 1592 (s), 1556 (s), 1490 (s), 1440 (s), 1325 (s), 1252 (s), 1164 (s), 1100 (s), 1068 (s), 983 (s), 852 (s), 800 (s). HRMS: m/z calc. for [C15H15N3OF3] + ([M + H]+) 310.1167, found 310.1166, ∆ = −0.3 ppm. Experimental 187 6-Methyl-N1-(4-(pyridin-3-yl)pyrimidin-2-yl)benzene-1,3-diamine (25)293 10 9 8 N 6 N 1 N2 3 H N 2' 4' 5' NH2 7' A solution of compound 24 (105 mg, 0.34 mmol) in CH2Cl2 (15 mL, approx. 0.02 mM) was prepared. A H-Cube hydrogenation reactor equipped with a 5 % Pd/C cartridge was prepared as follows: The reactor and column were washed with CH2Cl2 at 1 mL/min for 5 minutes. For the reaction, the system pressure regulator was set to 20 bar and the hydrogen pressure to 35 bar in “controlled” mode. The column temperature was set to 40 ◦C, and the solution of 24 infused at 0.5 mL/min. The product was cycled through the reactor once more, and then the collected material concentrated under reduced pressure to afford the title compound (69 mg, 73 %) as a yellow oil. 1H NMR (400 MHz, CDCl3): δ /ppm = 9.26 (d, J = 2.2 Hz, 1 H, H-6), 8.71 (dd, J = 4.8 Hz and 1.7 Hz, 1 H, H-8), 8.48 (d, J = 5.1 Hz, 1 H, H-2), 8.33 (dt, J = 8.0 Hz and 2.2 Hz, 1 H, H-10), 7.59 (d, J = 2.4 Hz, 1 H, H-2′), 7.41 (dd, J = 8.0 Hz and 4.8 Hz, 1 H, H-9), 7.13 (d, J = 5.1 Hz, 1 H, H-3), 6.94 – 7.03 (m, 2 H, NH and H-5′), 6.41 (dd, J = 8.0 Hz and 2.4 Hz, 1 H, H-4′), 3.65 (br. s, 2 H, NH2), 2.24 (s, 3 H, H-7 ′). 13C NMR (100 MHz, CDCl3): δ /ppm = 162.53 (C-1), 160.70 (C-4), 158.99 (CH, C-2), 151.40 (CH, C-6), 148.54 (CH, C-8), 145.12 (C-3′), 137.90 (C-1′), 134.43 (CH, C-10), 132.75 (C-5), 131.03 (CH, C-5′), 123.60 (CH, C-9), 118.16 (C-6′), 110.66 (CH, C-2′ or C-4′), 108.47 (CH, C-2′ or C-4′), 108.02 (CH, C-3), 17.21 (CH3, C-7 ′). HRMS: m/z calc. for [C16H16N5] + ([M + H]+) 278.1406, found 278.1404, ∆ = −0.7 ppm. 188 Experimental 4-(Chloromethyl)-N-(4-methyl-3-((4-(pyridin-3-yl)pyrimidin-2-yl) amino)phenyl)benzamide (27)295 10 9 8 N 6 N 1 N2 3 H N 2' 5' HN 8' O 10' 11' 13' Cl 7' Compound 25 (69 mg, 0.25 mmol) was suspended in distilled THF (4 mL). Triethylamine (0.07 mL, 1.15 equiv.) was added, and the mixture cooled to 0 ◦C and stirred for 10 minutes. A solution of 4-(chloromethyl)benzoyl chloride (54 mg) in distilled THF (2 mL) was added dropwise over 3 minutes. During the addition a darkening in colour of the solution was observed. The mixture was stirred at 0 ◦C for 3 hours. After this time, LC-MS analysis indicated complete conversion. Water (5 mL) was added dropwise over 5 minutes, and the mixture allowed to warm to room temperature and then stirred for 18 hours, during which time a yellow precipitate formed. Filtration afforded the title compound (61 mg, 57 %) as a yellow crystalline solid. 1H NMR (500 MHz, DMSO-d6): δ /ppm = 10.22 (br. s, 1 H, NHamide), 9.26 (d, J = 2.1 Hz, 1 H, H-6), 8.97 (br. s, 1 H, NH), 8.67 (dd, J = 4.8 Hz and 1.7 Hz, 1 H, H-8), 8.50 (dd, J = 5.2 Hz and 1.7 Hz, 1 H, H-2), 8.47 (dq, J = 8.0 Hz and 2.2 Hz, 1 H, H-10), 8.08 (dt, J = 10.1 Hz and 2.9 Hz, 1 H, H-2′), 7.94 (d, J = 8.3 Hz, 2 H, H-10′), 7.57 (d, J = 8.3 Hz, 2 H, H-11′), 7.51 (dd, J = 8.0 Hz and 4.8 Hz, 1 H, H-9), 7.43 – 7.50 (m, 1 H, H-4′), 7.42 (dd, J = 5.2 Hz and 2.1 Hz, 1 H, H-3), 7.20 (d, J = 8.3 Hz, 1 H, H-5′), 4.83 (s, 2 H, H-13′), 2.21 (s, 3 H, H-7′). 13C NMR (126 MHz, DMSO-d6): δ /ppm = 164.89 (C-8′), 161.64 (C-4), 161.22 (C-1), 159.55 (CH, C-2), 151.42 (CH, C-8), 148.23 (CH, C-6), 141.00 (C-12′), 137.88 (C-6′), 137.04 (C-1′), 134.95 (C-9′), 134.54 (CH, C-10), 132.28 (C-5), 130.13 (CH, C-5′), 128.84 (CH, C-10′), 128.07 (CH, C-11′), 123.88 (CH, C-9), 117.14 (CH, C-2′), 116.67 (CH, C-4′), 107.63 (CH, C-3), 45.52 (CH2, C-13′), 17.75 (CH3, C-7′). LC-MS: tR = 4.80 min, m/z 430.18 ([M + H]+). HRMS: m/z calc. for [C24H21N5O35Cl] + ([M + H]+) 430.1429, found 430.1415, ∆ = −3.2 ppm. Experimental 189 N-(4-Methyl-3-((4-(pyridin-3-yl)pyrimidin-2-yl)amino)phenyl)-4-((4- methylpiperazin-1-yl)methyl)benzamide (Imatinib, 1)33 10 9 8 N 6 N 1 N2 3 H N 2' 4' 5' HN 8' O 10' 11' 13' N 14' 15'N 16' 7' Compound 27 (11.5 mg, 27 µmol), N-methylpiperazine (0.05 mL) and acetonitrile (2 mL) were combined and heated in a sealed vial under microwave irradiation at 150 ◦C for 2 hours. The solvent was removed and the resulting solid purified by flash column chromatography to afford the title compound (7 mg, 53 %) as a yellow solid. 1H NMR (400 MHz, DMSO-d6): δ /ppm = 10.17 (s, 1 H, NHamide), 9.27 (d, J = 2.1 Hz, 1 H, H-6), 8.98 (s, 1 H, NH), 8.68 (dd, J = 4.8 Hz and 1.7 Hz, 1 H, H-8), 8.51 (d, J = 5.1 Hz, 1 H, H-2), 8.48 (dt, J = 7.9 Hz and 2.0 Hz, 1 H, H-10), 8.08 (d, J = 2.2 Hz, 1 H, H-2′), 7.90 (d, J = 8.2 Hz, 2 H, H-10′), 7.52 (dd, J = 7.9 Hz and 4.7 Hz, 1 H, H-9), 7.48 (dd, J = 8.2 Hz and 2.2 Hz, 1 H, H-4′), 7.41 – 7.46 (m, 3 H, H-3 and H-11′), 7.20 (d, J = 8.5 Hz, 1 H, H-5′), 3.52 (s, 2 H, H-13′), 2.34 (m, 8 H, H-14′ and H-15′), 2.22 (s, 3 H, H-7′), 2.14 (s, 3 H, H-16′). 13C NMR (100 MHz, DMSO-d6): δ /ppm = 165.23, 161.57, 161.15, 159.46 (CH), 151.37 (CH), 148.18 (CH), 142.09, 137.76, 137.18, 134.40 (CH), 133.74, 132.19, 130.00 (CH), 128.60 (CH), 127.56 (CH), 123.77 (2 × CH), 117.17 (CH), 116.69 (CH), 107.49 (CH), 61.61 (CH2), 54.70 (CH2), 52.58 (CH2), 45.74 (CH3), 17.66 (CH3). LC-MS: tR = 4.26 min, m/z 494.48 ([M + H]+). HRMS: m/z calc. for [C29H32N7O] + ([M + H]+) 494.2668, found 494.2690, ∆ = −4.5 ppm. 190 Experimental Piperazine-2-carboxamide synthesis General Experimental Information Hydrous zirconia was kindly donated by MEL Chemicals (cod. XZO 631/01). FlowIR spectrometers from Mettler Toledo, with silicon or diamond windows, were used for in-line analyses. The flow hydration reaction was performed using a Vapourtec R2+/R4 flow platform. A Knauer K-120 HPLC pump was used for the hydrogenation step, in combination with a ThalesNano H-Cube reactor. Unless otherwise specified, flow tubing is PFA  1 mm with ETFE connections. Pyrazine-2-carboxamide (40)296 100 ºC 100 psi R2 N N NH2 O N N CN 1´ 35 6 FlowIR A solution of pyrazine-2-carbonitrile (41; 7.4 g, 70 mmol) in EtOH / H2O (0.6 M, 8 : 1 v/v) was pumped at 0.2 mL/min through column reactor R2 (100 mm × 10 mm, 5 g hydrous zirconia) heated at 100 ◦C (residence time approximately 20 minutes). Pressure was provided by a 100 psi back-pressure regulator. Concentration of the reactor output afforded the title compound (8.5 g, 69 mmol, >98 %) as a colourless solid, m.p. 189.4 – 189.8 ◦C (ethanol) [lit. 190 ◦C (H2O) 296]. For the control programme, see: https://gist.github.com/richardingham/0a58a291bad2e3b9009f 1H NMR (400 MHz, DMSO-d6): δ /ppm = 9.18 (d, J = 1.5 Hz, 1 H, H-3), 8.84 (d, J = 2.5 Hz, 1 H, H-6), 8.70 (dd, J = 2.5 Hz and 1.5 Hz, 1 H, H-5), 8.25 (br. s, 1 H, NH), 7.86 (br. s, 1 H, NH). 13C NMR (100 MHz, DMSO-d6): δ /ppm = 165.09 (C-1′), 147.38 (CH, C-5), 145.08 (C-2), 143.63 (CH, C-6), 143.36 (CH, C-3). IR (neat): ν /cm−1 = 3410 (m), 3148 (m), 1704 (s), 1608 (m), 1579 (m), 1524 (m), 1478 (w), 1436 (m), 1376 (s), 1182 (m), 1164 (m), 1086 (m), 1053 (m), 1022 (s), 869 (m), 785 (s), 669 (s). LC-MS: tR = 0.28 min, Experimental 191 m/z 124.19 ([M + H]+). HRMS: m/z calc. for [C5H6ON3] + ([M + H]+) 124.0505, found 124.0504, ∆ = −0.8 ppm. Microanalysis: calc. (found) for C5H5ON3 C 48.78% (48.69%), H 4.09% (4.19%), N 34.13% (33.70%). (R,S)-Piperazine-2-carboxamide (39)297 FlowIRH-Cube 100 psi 75 psi N N NH2 O N H N H NH2 O 1´ 2 35 6 A solution of pyrazine-2-carboxamide (40; 0.62 mg, 5.0 mmol) in EtOH / H2O (0.6 M, 8 : 1 v/v) was pumped at 0.1 mL/min using a Knauer K-120 HPLC pump (through tubing  0.5 mm, PTFE then SS) into the H-Cube reactor, which was equipped with a 10 % Pd/C catalyst cartridge, heated to 100 ◦C. The reactor was held at pressure by a 100 psi and 75 psi BPR in series (connected to the reactor by tubing  0.5 mm, PTFE). Concentration of the reactor output afforded the title compound (0.61 g, 4.8 mmol, 95 %) as a colourless solid, m.p. 143 – 145 ◦C (ethanol) [lit. 144 – 145 ◦C (ethanol)297]. 1H NMR (400 MHz, MeOH-d4): δ /ppm = 3.35 (dd, J = 9.9 Hz and 3.3 Hz, 1 H, H-2), 3.09 (dd, J = 12.4 Hz and 3.4 Hz, 1 H, H-3cis), 2.96 (dt, J = 12.2 Hz and 2.8 Hz, 1 H, H-6), 2.90 – 2.77 (m, 1 H, H-5), 2.75 (dd, J = 12.4 Hz and 2.6 Hz, 1 H, H-6), 2.73 – 2.61 (m, 2 H, H-3trans and H-5). 13C NMR (100 MHz, MeOH-d4): δ /ppm = 176.50 (C-1′), 59.61 (CH, C-2), 49.69 (CH2, C-3), 46.19 (CH2, C-2), 45.64 (CH2, C-1). IR (neat): ν /cm−1 = 3333 (m), 3308 (m), 3194 (w), 2949 (m), 2904 (m), 2832 (m), 1674 (m), 1611 (m), 1488 (w), 1438 (m), 1409 (m), 1368 (w), 1355 (m), 1306 (s), 1245 (m), 1187 (w), 1137 (m), 1117 (s), 1071 (w), 1057 (m), 1003 (w), 960 (w), 909 (m), 823 (s), 723 (m), 666 (s). LC-MS: tR = 0.26 min, m/z 130.14 ([M + H]+). HRMS: m/z calc. for [C5H12ON3] + ([M + H]+) 130.0975, found 130.0979, ∆ = 3.0 ppm. Microanalysis: calc. (found) for C5H11ON3 C 46.50% (46.49%), H 8.58% (8.50%), N 32.53% (32.30%). 192 Experimental DoE Reduction Experiments To the reactor setup above was appended a Valco VICI 10-position switching valve, such that samples could be collected from the different experiments. See Figure 3.12, page 70. Representative NMR spectra: 2.53.03.54.05.56.06.57.07.58.08.59.0 ppm Experiment 15 Experiment 13 Experiment 7 For the control programme, see: https://gist.github.com/richardingham/83401127622036c6afd0 Experimental 193 Telescoped flow synthesis (See Figure 3.12, page 70). A solution of pyrazine-2-carbonitrile 41 in EtOH / H2O (0.6 M, 8 : 1 v/v) was passed through a zirconia column reactor as described above in the experimental section for compound 40. The intermediate solution was used directly without purification in the second step. This could be performed either by matching the flow rates of the two steps, or using a reservoir arrangement as described in Section 3.5.1. The intermediate solution was delivered to the H-Cube apparatus (flow rate 0.1 mL/min) using a Knauer K-120 pump as described in the experimental section for compound 39. Over a total of 14 h of operation, 7.3 g of compound 39 was generated in 95 % yield and 99 % purity. For control programme, see: https://gist.github.com/richardingham/31f6f8efa47771c2ed02 194 Experimental 2-Aminoadamantane-2-carboxylic acid synthesis General Experimental Information THF, toluene and methanol were obtained from Fisher Scientific and distilled before use. Diethyl ether was obtained from Sigma-Aldrich and distilled before use. Integrated flow synthesis Reagent Stream A A solution of 2-adamantanone (7.5 g, 50 mmol) in toluene / diethyl ether (3 : 2 v/v, 100 mL), stored in an oven-dried 250 mL, one-necked, pear-shapedflask equipped with a rubber septum and under a N2 atmosphere. Reagent Stream B A solution of ethynyl magnesium bromide (0.5 M), in a Sigma- Aldrich SureSeal bottle equipped with an inert gas inlet connected to a nitrogen manifold. Solvent Stream A THF stored under a nitrogen atmosphere. Solvent Stream B Toluene / diethyl ether (3 : 2 v/v) stored under a nitrogen atmosphere. This can be switched to a stream of distilled methanol using valve V2. Stage 1 Reagent Stream A was delivered at a flow rate of 0.18 mL/min (P2; Uniqsis FlowSyn) and combined at tee mixer M1 (Uniqsis FlowSyn integrated tee mixer) with Reagent Stream B delivered at a flow rate of 0.20 mL/min (P1; Uniqsis FlowSyn) and then heated to 40 ◦C in reactor R1 ( 1 mm, PFA, 14 mL: 37 minutes residence time). A 100 psi back pressure regulator was employed to regulate the pressure of the system. After exiting the coil the flow stream passed through a FlowIR spectrometer in order to detect the presence of the Grignard product 51. The detector was linked to valve V4 (Uniqsis FlowSyn integrated loop-switching valve) by software control (see Section 4.3.5, page 90) and the output stream was directed to the next stage only when this product was detected. Pump P5 (Knauer WellChrom K120) delivered Solvent Stream A at 0.5 mL/min through valve V4 when the reactor output was being discarded. Experimental 195 Quenching Stream C A saturated solution of NH4Cl combined in a ratio of 3 : 1 (v/v) with distilled water (250 mL). Stage 2 The reaction solution was combined with Quenching Stream C, delivered at a flow rate of 0.5 mL/min, (P3, Knauer Smartline S100) using a tube-in-tube mixer (M2, see Section 4.3.4); the output was directed into reactor R2 ( 2.4 mm, 600 mm length, ETFE). A micro vibration motor was attached to R2, reducing the occurrence of blockages at M2. The biphasic plug flow system formed was delivered to a glass column ( 6.6 mm, 100 mm length, glass) packed with charcoal and glass beads in order to remove magnesium aggregates by filtration (see Section 4.3.3, page 85). The resulting clear biphasic system was then delivered to liquid/liquid separating column S1 ( 10 mm, 100 mm length, glass). The aqueous phase was withdrawn continuously by pump P4 (Knauer Smartline S100) at a flow rate based on the position of the interface (see Section 4.3.4, page 88). The organic layer was forced out of the top of the column into the next stage. A pressure differential created by the two back-pressure regulators prevented undesired siphoning of liquid through pump P4. Stage 3 The organic output from S1 was combined at a tee mixer M3 ( 0.5 mm, PEEK) with a stream of MeCN delivered at 1.2 mL/min by pump P6 (Knauer WellChrom K120). The mixture was introduced into a nebulising evaporation device at 20 ◦C (see Section 4.4, page 92) along with a stream of N2 gas (1.4 bar). The remaining solution was withdrawn from the bottom of the evaporation chamber using a peristaltic pump (P7) at a flow rate of 1 mL/min. The solution was collected in reservoir S2. The volume of material within S2 was measured by a digital camera (see Section 4.5.1, page 94) and this data was used to control the flow rate of pump P8 (Vapourtec R2+) in order to match the input and output flow rates. Stage 4 This solution was pumped at a flow rate of 0.1 to 1.4 mL/min by pump P8 and combined at mixer M4 ( 0.5 mm, ETFE) with a stream of concentrated sulfuric acid that was delivered at 0.05 mL/min by pump P9 (Vapourtec R2+ with acid-resistant fittings). The mixture was passed through reactor R3 ( 1 mm, PFA, 10 mL: minimum 7 minutes residence time). Quenching Stream D A 4 M solution of KOH in EtOH / H2O (4 : 1 v/v). 196 Experimental Stage 5 The output of reactor R3 was combined with Quenching Stream D in a second tube-in-tube mixer (M5). Quenching Stream D was delivered at 2 mL/min by pump P10 (Vapourtec R2+); the output was directed into reactor R4 ( 2.4 mm, 300 mm length, ETFE). Another vibrating device was attached to R4, reducing the occurrence of blockages at M5. The stream exiting this reactor was directed onto the continuous filtration device (see Section 4.5.3, page 99) that removed the salts formed by the quenching of the acid to leave a basic ethanolic solution of compound 52 which was collected in reservoir S3. Stage 6 The solution collected in reservoir S3 was pumped (P13, Vapourtec R2+) to reactor R5 ( 2.5 mm, stainless steel, 50 mL: minimum 25 minutes residence time) heated to 120 ◦C. The reactor output was collected and the solvent removed. The residue was redissolved in ethyl acetate and washed with water, dried (MgSO4), and then the solvent removed. Stage 7 A solution of 2-methyl-4-(adamantane-2′-spiro)-5-methylidene oxazoline (53) in acetone / H2O (5 : 1 v/v, 0.25 M) was delivered at 2 mL/min by pump P14 (Knauer WellChrom K120) to meet a stream of ozone (0.5 bar, flow rate 500 mL/min) and these were combined in a Y-mixer ( 0.5 mm, PEEK) and directed into a 1 mL tube reactor R6 ( 2.5 mm, 180 mm length, PFA, 20 s residence time). The solution was directed to column R7 ( 10 mm, 100 mm length, glass) from which the gases can escape, containing amorphous MnO2 (2 g) through which the liquids percolate and the solution was collected. This solution was pumped at 0.25 mL/min P15 (Vapourtec R2) and combined at a tee mixer ( 0.5 mm, ETFE) with a stream of HCl / AcOH / H2O (1 : 5 : 8 v/v/v) delivered at 0.25 mL/min by pump P16 (Vapourtec R2) and directed into reactor R8 ( 1 mm, 10 mL, PFA) heated to 150 ◦C. The output of the reactor was finally directed into a solvent evaporator device. The solid recovered from the evaporator was washed with hot acetonitrile (2×50 mL per gram of product) and diethyl ether (2×50 mL per gram of product), before being recrystallized from MeOH (0 ◦C) to give 2-aminoadamantane-2-carboxylic acid (135). Experimental 197 2-Ethynyladamantan-2-ol (51)298,299 HO Collected as an off-white solid, by concentration of the solution in acetonitrile m.p. 102 ◦C (acetonitrile) [lit: 100 – 102 ◦C (ether)300]. An analytical sample was recrystallised from ethanol / water. 1H NMR (400 MHz, CDCl3): δ /ppm = 2.56 (s, 1 H), 2.21 – 2.16 (m, 4 H), 1.96 – 1.96 (m, 3 H), 1.82 – 1.77 (m, 4 H), 1.70 (br. s, 2 H), 1.55 – 1.58 (m, 3 H). 13C NMR (100 MHz, CDCl3): δ /ppm = 88.51, 72.70 (CH), 72.50, 39.31 (CH), 37.60 (CH2), 35.37 (CH2), 31.54 (CH2), 26.40 (CH), 26.23 (CH). IR (neat): ν /cm−1 = 3477 (w), 3363 (w), 3312 (m), 3179 (m), 2928 (m), 2898 (s), 2858 (m), 2091 (w), 1466 (w), 1449 (w), 1407 (w), 1349 (w), 1326 (w), 1309 (w), 1284 (w), 1244 (w), 1194 (w), 1118 (w), 1101 (m), 1061 (s), 1042 (w), 1023 (s), 998 (s), 943 (w), 925 (m), 879 (w), 835 (w), 802 (w), 755 (m), 693 (w). LC-MS: tR = 4.63 min, m/z 159.00 ([M – H2O] +). HRMS: m/z calc. for [C12H17O] + ([M + H]+) 177.2550, found 177.2558, ∆ = 4.5 ppm. Microanalysis: calc. (found) for C12H16O C 81.77% (81.76%), H 9.15% (9.10%). The structure was confirmed by X-ray crystallographic analysis, and deposited at Cambridge University with the unique reference SL1406. 198 Experimental 2-Ethynyl-2-acetamidoadamantane (52)299 NHO Collected by concentration of the reaction solution, partitioning between ethyl acetate and water, then drying and concentration of the organic phase. The title compound was collected as a white solid, m.p. 147 ◦C (ethyl acetate) [lit: 138 – 139 ◦C (ethanol)299]. 1H NMR (400 MHz, CDCl3): δ /ppm = 5.41 (br. s, 1 H), 2.46 (s, 1 H), 2.40 (br. s, 2 H), 2.35 (br. s, 1 H), 2.31 (br. s, 1 H), 2.01 (s, 3 H), 1.82 – 1.77 (m, 4 H), 1.70 (br. s, 2 H), 1.55 – 1.58 (m, 3 H). 13C NMR (100 MHz, CDCl3): δ /ppm = 169.12, 86.18, 71.73 (CH), 56.47, 37.64 (CH2), 35.06 (CH), 34.37 (CH2), 32.18 (CH2), 27.24 (CH), 26.76 (CH), 24.23 (CH3). IR (neat): ν /cm−1 = 3287 (m), 2903 (m), 2166 (w), 1645 (s), 1537 (s), 670 (m); LC-MS: tR = 4.39 min, m/z 218.14 ([M + H]+). HRMS: m/z calc. for [C14H20NO] + ([M + H]+) 218.1545, found 218.1548, ∆ = 1.4 ppm. Microanalysis: calc. (found) for C14H19NO C 77.78% (77.75%), H 8.81% (8.75%), N 6.45% (6.40%). 2-Methyl-4-(adamantane-2′-spiro)-5-methylidene oxazoline (53)301 N O Collected as a yellow solid after Stage 6, m.p. 62 ◦C (ethyl acetate). 1H NMR (400 MHz, CDCl3): δ /ppm = 4.79 (d, 1 H, J = 2.4 Hz), 4.52 (d, 1 H, J = 2.4 Hz), 2.44 (d, 2 H, J = 12.4 Hz), 2.21 (d, 2 H, J = 12.4 Hz), 2.03 (s, 3 H), 1.86 (br. s, 3 H), 1.74 (s, 2 H), 1.69 – 1.55 (m, 5 H). 13C NMR (100 MHz, CDCl3): δ /ppm = 166.00, 158.67, 87.92 (CH2), 76.03, 38.70 (CH2), 37.08 (CH), 34.06 (CH2), 31.40 (CH2), 27.67 (CH), 26.41 (CH), 14.11 (CH3). IR (neat): ν /cm−1 = 3342 (w), 3031 (w), 2938 (w), 2801 (w), 1643 (w), 1495 (w), 1453 (w), 1363 (w), 1337 (w), 1282 (w), 1239 (w), 1212 (w), 1167 (w), 1139 (w), 1062 (s), 1042 (m), 1017 (m), 979 (w), 953 (w), 911 (w), 849 (w), 789 (m), 737 Experimental 199 (s), 698 (s), 662 (m). LC-MS: tR = 4.49 min, m/z 218.07 ([M + H]+). HRMS: m/z calc. for [C14H20NO] + ([M + H]+) 218.1545, found 218.1541, ∆ = −1.8 ppm. Microanalysis: calc. (found) for C14H19NO C 77.78% (77.80%), H 8.81% (8.77%), N 6.45% (6.44%). 2-Aminoadamantane-2-carboxylic acid hydrochloride (44)194 H2N OH O HCl Collected as an off-white solid after recrystallisation from methanol, m.p. >258 ◦C (de- comp.) [lit: 312 – 314 ◦C (decomp.)192]. 1H NMR (400 MHz, CDCl3): δ /ppm = 8.70 (br. s, 3 H), 2.23 (s, 2 H), 2.16 (d, 2 H, J = 13.2 Hz), 1.93 (d, 2 H, J = 13.2 Hz), 1.73 – 1.90 (m, 4 H), 1.62 – 1.66 (m, 2 H). 13C NMR (100 MHz, CDCl3): δ /ppm = 171.59, 63.92, 37.54 (CH2), 34.41 (CH), 32.25 (CH2), 31.22 (CH), 26.27 (CH). IR (neat): ν /cm−1 = 3223 (w), 2930 (m), 2874 (m), 2775 (m), 1729 (s), 1589 (m), 1507 (s), 1476 (m), 1373 (m), 1282 (w), 1239 (m), 1221 (m), 1146 (s), 1119 (w), 1100 (s), 1087 (m), 1066 (w), 1006 (w), 966 (w), 931 (w), 904 (w), 857 (m), 837 (m), 752 (w), 717 (s), 676 (w). LC-MS: tR = 0.31 min, m/z 196.10 ([M + H]+). HRMS: m/z calc. for [C11H18NO2] + ([M – Cl]+) 196.1338, found 196.1336, ∆ = −1.0 ppm. Microanalysis: calc. (found) for C11H18NO2Cl: C 57.02% (56.90%), H 7.83% (7.97%), N 6.04% (6.06%). 200 Experimental Chemical Probes for BRD9 tert-Butyl (3,6-dichloropyridazin-4-yl)carbamate (87)302 N N Cl H Nt-BuO O Cl For reactor scheme, see Figure 5.16, page 120. Reagent Stream A: 3,6-Dichloropyridazine-4-carboxylic acid (8.0 g, 40 mmol), tri- ethylamine (11.6 mL, 80 mmol, distilled over CaCl2 before use) and tert-butanol (18.5 g, 250 mmol) in toluene / acetonitrile (7 : 3 v/v, 125 mL). Reagent Stream B: Diphenyl phosphoryl azide (13.5 mL, 75 mmol) in toluene / ace- tonitrile (7 : 3 v/v, 125 mL). Method: The reagents were stored in sealed flasks, pressurised to 1 bar under a nitrogen atmosphere, and each stream was drawn through a pump at 0.179 mL/min. The streams are combined ( 1 mm PFA tubing) in a tee mixer ( 0.5 mm, ETFE) and then heated at 120 ◦C in a continuous flow coil ( 2.5 mm, 2×25 mL volume, SS) before passing through a 100 psi back-pressure regulator. The output was collected and the solvent removed. The residue was extracted into toluene (2×250 mL) from 0.1 M aqueous citric acid (50 mL). The combined organic extracts were dried (MgSO4) and concentrated. The resultant mixture was purified by column chromatography (50 g KP-NH silica), using a solvent system of ethyl acetate / hexane (0 : 1 to 2 : 8 over 20 CV, then 2 : 8 for 5 CV) to afford the title compound (8.5 g, 32 mmol, 39 %) as a white solid, m.p. 110 – 111 ◦C (ethyl acetate) [lit: 81 – 83 ◦C302]. 1H NMR (400 MHz, CDCl3): δ /ppm = 8.36 (s, 1 H), 7.20 (br. s, 1 H, NH), 1.56 (s, 9 H). 13C NMR (100 MHz, CDCl3): δ /ppm = 156.51, 150.88, 144.86, 137.28, 114.58 (CH), 84.11, 28.00 (CH3). IR (neat): ν /cm−1 = 3286 (w), 3186 (w), 3146 (w), 3086 (w), 2978 (w), 2933 (w), 2216 (w), 2168 (w), 2030 (w), 1994 (w), 1979 (w), 1760 (w), 1732 (s), 1693 Experimental 201 (w), 1638 (w), 1590 (w), 1547 (s), 1489 (s), 1457 (m), 1411 (w), 1393 (w), 1367 (m), 1341 (m), 1277 (m), 1265 (w), 1237 (s), 1188 (m), 1151 (s), 1131 (s), 1118 (s), 1056 (m), 1026 (m), 1010 (m), 959 (s), 882 (m), 860 (s), 788 (m), 753 (m), 740 (m), 688 (m). HRMS: m/z calc. for C9H12O2N3[ 35Cl]2 ([M + H] +) 264.0301, found 264.0292, ∆ = −3.3 ppm. 3,6-Dichloropyridazin-4-amine (88)302 N N Cl H2N Cl tert-Butyl (3,6-dichloropyridazin-4-yl)carbamate (87) (10 g, 37.8 mmol) is dissolved in dichloromethane (40 mL). 4 M HCl in dioxane (125 mL) was added, with rapid formation of a precipitate. The reaction mixture was allowed to stir at 25 ◦C for 1 day, and then the solid was collected by filtration and resuspended in DCM (100 mL). 2 M NH3 in methanol (50 mL) was added and stirred at 25 ◦C for 30 minutes. The solid product was collected by filtration, washing with DCM and then dried in vacuo. The title compound (6.2 g, quant.) was collected as a white solid, m.p. 204.6 – 204.8 ◦C (dichloromethane) [lit: 194 – 196 ◦C (ethyl acetate)302]. 1H NMR (400 MHz, DMSO-d6): δ /ppm = 7.1 (br. s, 2 H, NH2), 6.8 (s, 1 H). 13C NMR (100 MHz, DMSO-d6): δ /ppm = 154.53, 146.26, 143.72, 108.49 (CH). IR (neat): ν /cm−1 = 3459 (w), 3290 (w), 3209 (w), 3094 (w), 3049 (m), 2795 (w), 2694 (w), 1635 (s), 1563 (s), 1517 (w), 1439 (w), 1383 (m), 1293 (m), 1125 (s), 1072 (m), 1053 (s), 962 (s), 882 (s), 727 (s), 684 (m). HRMS: m/z calc. for C4H4N3[ 35Cl]2 ([M + H] +) 163.9782, found 163.9783, ∆ = 0.6 ppm. Microanalysis: calc. (found) for C4H4N3Cl2 C 29.30% (29.41%), H 1.84% (1.82%), N 25.62% (24.61%), Cl 43.23 (43.44%). 202 Experimental 3,6-Dichloro-3-hydrazinylpyridazin-4-amine (91)302 N N Cl H2N HN NH2 A mixture of 3,6-dichloropyridazin-4-amine (88) (3.0 g, 18.2 mmol), hydrazine hydrate (27 mL) and water (8 mL) was heated in two portions in sealed vials under microwave irradiation at 100 ◦C for 3 h. The reaction mixture was allowed to cool to room temperature, and the resulting slurry collected by filtration, washing on the filter with water (10 mL). The title compound (2.2 g, 13.6 mmol, 75 %) was collected as a white solid. 1H NMR (400 MHz, DMSO-d6): δ /ppm = 7.27 (br. s, 1 H), 6.36 (s, 1 H), 6.24 (br. s, 2 H), 4.23 (br. s, 2 H). 13C NMR (100 MHz, DMSO-d6): δ /ppm = 143.93, 137.58, 103.59, 91.83 (CH). IR (neat): ν /cm−1 = 3317 (w), 3285 (w), 3101 (m), 3045 (w), 2984 (m), 2931 (w), 2763 (w), 2272 (w), 2254 (w), 2216 (w), 2199 (w), 2168 (w), 2138 (w), 2027 (w), 1988 (w), 1980 (w), 1972 (w), 1958 (w), 1670 (w), 1577 (s), 1558 (m), 1456 (m), 1417 (w), 1394 (w), 1344 (w), 1308 (w), 1276 (m), 1177 (w), 1145 (w), 1100 (m), 991 (m), 960 (m), 879 (m), 824 (m), 761 (w), 695 (m). HRMS: m/z calc. for C4H7N5[ 35Cl] ([M + H]+) 160.0390, found 160.0396, ∆ = 3.7 ppm. 6-Chloro-3-methyl-[1,2,4]triazolo[4,3-b]pyridazin-8-amine (76)235 N N N N Cl H2N A mixture of 3,6-dichloro-3-hydrazinylpyridazin-4-amine (91) (2.4 g, 15 mmol) and acetic acid (30 mL) was heated in two portions in sealed vials under microwave irradiation at 120 ◦C for 2 h. The solvent was removed and the combined residue recrystallized from 10 % water in ethanol. The title compound (1.35 g, 7 mmol, 49 %) was collected as a pale yellow crystalline solid, m.p. 233.5 – 234.0 ◦C (decomp.). 1H NMR (400 MHz, DMSO-d6): δ /ppm = 7.85 (br. s, 2 H), 6.09 (s, 1 H), 2.48 (s, 3 H). 13C NMR (100 MHz, DMSO-d6): δ /ppm = 149.77, 147.16, 144.31, 139.79, 94.01 (CH), Experimental 203 9.90 (CH3). IR (neat): ν /cm−1 = 3451 (m), 3299 (w), 3214 (w), 3103 (m), 3073 (m), 1651 (s), 1596 (w), 1563 (s), 1529 (m), 1466 (s), 1421 (s), 1411 (s), 1379 (m), 1339 (m), 1243 (m), 1188 (w), 1129 (s), 1051 (m), 994 (w), 929 (s), 810 (s), 794 (m), 745 (m), 718 (s), 669 (m). HRMS: m/z calc. for C6H7N5[ 35Cl] ([M + H]+) 184.0390, found 184.0392, ∆ = 1.1 ppm. 2-Methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)aniline (100)303 B OO NH2 (3-Amino-4-methylphenyl)boronic acid (0.50 g, 3.3 mmol) and pinacol (0.43 g, 3.6 mmol) in acetonitrile (5 mL) was stirred at 25 ◦C for 18 h. The solvent was removed and the resulting solid triturated with CH2Cl2 to afford the title compound (0.5 g, 65 %) as a pale orange crystalline solid. An analytical sample was recrystallised from ethanol, m.p. 115.2 – 115.4 ◦C (ethanol) [lit: 107 – 108 ◦C303]. 1H NMR (400 MHz, CDCl3): δ /ppm = 7.16 (d, J = 7.2 Hz, 1 H, H-4), 7.12 (s, 1 H, H-8), 7.07 (d, J = 7.2 Hz, 1 H, H-5), 3.57 (br. s, 2 H, NH2), 2.18 (s, 3 H, H-6 ′), 1.33 (s, 12 H, H-1). 13C NMR (100 MHz, CDCl3): δ /ppm = 144.18, 130.12 (CH), 125.99, 125.35 (CH), 121.17 (CH), 83.70, 24.98 (CH3), 17.72 (CH3), (One carbon resonance was not visible). IR (neat): ν /cm−1 = 3421 (w), 3352 (w), 3003 (w), 2979 (w), 2931 (w), 1632 (w), 1607 (w), 1564 (w), 1516 (w), 1454 (w), 1419 (m), 1378 (m), 1366 (m), 1349 (s), 1320 (m), 1297 (m), 1285 (m), 1260 (s), 1214 (w), 1203 (m), 1169 (m), 1137 (s), 1104 (m), 1064 (w), 1035 (w), 997 (m), 967 (m), 922 (w), 892 (w), 855 (s), 832 (w), 812 (s), 787 (w), 753 (w), 727 (m), 701 (m), 685 (s), 672 (m). HRMS: m/z calc. for C13H21[ 11B]NO2 ([M + H] +) 234.1665, found 234.1662, ∆ = −1.3 ppm. 204 Experimental N-(2-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl) methanesulfonamide (102e) 4 5 8 B OO 1 N H S 9 OO 6' Compound 100 (1.5 g, 6.6 mmol), mesyl chloride (1.0 mL, 13 mmol, 2 equiv.), and pyridine (1.0 mL, 13 mmol) in acetonitrile (4 mL) were stirred at 25 ◦C for 1 h. The solvent was removed and the solid product partitioned between ethyl acetate (10 mL) and aqueous HCl (0.2 M, 10 mL). The aqueous phase was extracted with ethyl acetate (10 mL), and the combined organic phases dried (MgSO4), concentrated and recrystallized from ethanol to afford the title compound (637 mg, 50 %) as an off-white solid, m.p. 146.5 – 146.8 ◦C (ethanol). 1H NMR (400 MHz, CDCl3): δ /ppm = 7.77 (s, 1 H, H-8), 7.60 (d, J = 7.6 Hz, 1 H, H- 4), 7.26 (d, J = 7.6 Hz, 1 H, H-5), 6.10 (br. s, 1 H, NH), 3.06 (s, 3 H, H-9), 2.38 (s, 3 H, H-6′), 1.33 (s, 12 H, H-1). 13C NMR (100 MHz, CDCl3): δ /ppm = 135.76, 133.30, 130.99, 130.25, 115.64, 84.11 (CH), 40.37 (CH3), 25.02 (CH3), 18.60 (CH3), (One carbon resonance was not visible). IR (neat): ν /cm−1 = 3277 (w), 2980 (w), 1611 (w), 1512 (w), 1474 (w), 1449 (w), 1405 (m), 1370 (m), 1352 (m), 1317 (s), 1283 (w), 1271 (w), 1215 (w), 1191 (w), 1161 (m), 1149 (s), 1131 (m), 1089 (m), 1038 (w), 1000 (w), 985 (m), 963 (m), 931 (w), 919 (w), 853 (m), 838 (w), 828 (w), 797 (w), 776 (m), 722 (w), 697 (w), 683 (w), 671 (m). HRMS: m/z calc. for C14H23[ 11B]NO4S ([M + H] +) 312.1441, found 312.1435, ∆ = −1.9 ppm. Microanalysis: calc. (found) for C13H20BNO2 C 54.03% (53.83%), H 7.13% (7.07%), N 4.50% (4.51%). Experimental 205 General Procedure A for sulfonamide formation Compound 100, the aryl sulfonyl chloride (1.1 equiv.), and pyridine (0.5 mL, 2 equiv.) in ethanol (4 mL) were stirred at 25 ◦C for 18 h. The solvent was removed and the solid product partitioned between ethyl acetate (5 mL) and water (5 mL). The organic phase was dried (MgSO4), concentrated and recrystallized from ethanol. 2-Chloro-N-(2-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) phenyl)benzenesulfonamide (102a) 4 5 8 B OO 1 N H S 13 12 11 10 Cl OO 6' Prepared according to General Procedure A on 3.3 mmol scale. Recrystallisation from ethanol (two rounds) afforded the title compound (914 mg, 68 %) as a colourless crystalline solid, m.p. 177.6 – 177.8 ◦C (ethanol). 1H NMR (400 MHz, CDCl3): δ /ppm = 7.99 (dd, J = 8.0 Hz and 1.6 Hz, 1 H, H-10), 7.54 (dd, J = 8.0 Hz and 1.3 Hz, 1 H, H-13), 7.51 – 7.41 (m, 3 H, H-4, H-8 and H-12), 7.32 (td, J = 8.0 Hz and 1.3 Hz, 1 H, H-11), 7.13 (d, J = 7.4 Hz, 1 H, H-5), 6.72 (br. s, 1 H, NH), 2.29 (s, 3 H, H-6′), 1.27 (s, 12 H, H-1), (One carbon resonance was not visible). 13C NMR (100 MHz, CDCl3): δ /ppm = 137.35, 136.01, 133.98 (CH), 133.68, 132.92 (CH), 132.07 (CH), 131.75 (CH), 131.67, 130.69 (CH), 130.34 (CH), 127.19 (CH), 83.91, 24.97 (CH3), 18.27 (CH3). IR (neat): ν /cm−1 = 3371 (w), 2980 (w), 1615 (w), 1453 (w), 1436 (w), 1380 (s), 1372 (s), 1354 (s), 1331 (s), 1317 (s), 1284 (w), 1251 (w), 1235 (w), 1217 (w), 1160 (s), 1144 (s), 1127 (s), 1111 (s), 1087 (s), 1042 (s), 996 (w), 967 (s), 947 (w), 916 (w), 880 (w), 853 (s), 828 (w), 771 (s), 751 (s), 719 (s), 710 (w), 696 (w), 683 (w), 665 (s). HRMS: m/z calc. for C19H23[ 11B][35Cl]NO4S ([M + H] +) 408.1208, found 408.1199, ∆ = −2.2 ppm. Microanalysis: calc. (found) for C19H22BClNO4S C 55.97% (55.92%), H 5.69% (5.61%), N 3.44% (3.49%), Cl 8.69% (8.89%). 206 Experimental 3-Chloro-N-(2-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)phenyl)benzenesulfonamide (102b) 4 5 8 B OO 1 N H S 9 10 11 13OO 6' Cl Prepared according to General Procedure A on 3.3 mmol scale. Recrystallisation from ethanol (two rounds) afforded the title compound (741 mg, 55 %) as a colourless crystalline solid, m.p. 138.4 – 138.6 ◦C (ethanol). 1H NMR (400 MHz, CDCl3): δ /ppm = 7.73 (t, J = 1.9 Hz, 1 H, H-14), 7.64 – 7.47 (m, 4 H, H-4, H-8, H-10 and H-12), 7.37 (t, J = 7.9 Hz, 1 H, H-11), 7.15 (d, J = 7.5 Hz, 1 H, H-5), 6.22 (br. s, 1 H, NH), 2.10 (s, 3 H, H-6′), 1.32 (s, 12 H, H-1). 13C NMR (100 MHz, CDCl3): δ /ppm = 141.36, 136.70, 135.29, 133.59 (CH), 133.46, 133.11 (CH), 132.19 (CH), 130.74 (CH), 130.35 (CH), 127.64 (CH), 125.58 (CH), 84.05, 25.00 (CH3), 18.14 (CH3), (One carbon resonance was not visible). IR (neat): ν /cm−1 = 3371 (w), 2980 (w), 1615 (w), 1578 (w), 1560 (w), 1510 (w), 1453 (w), 1436 (w), 1380 (s), 1372 (s), 1354 (s), 1331 (s), 1317 (s), 1284 (m), 1251 (m), 1235 (w), 1217 (w), 1160 (s), 1144 (s), 1127 (s), 1111 (m), 1087 (m), 1042 (m), 996 (m), 967 (m), 947 (m), 916 (w), 880 (w), 853 (s), 828 (m), 771 (s), 751 (s), 719 (m), 710 (m), 696 (w), 683 (m), 665 (s). HRMS: m/z calc. for [C19H23[ 11B][35Cl]NO4S] + ([M + H]+) 408.1208, found 408.1202, ∆ = −1.5 ppm. Microanalysis: calc. (found) for C19H22BClNO4S C 55.97% (55.93%), H 5.69% (5.64%), N 3.44% (3.54%), Cl 8.69% (8.71%). Experimental 207 4-Chloro-N-(2-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) phenyl)benzenesulfonamide (102c) 4 5 8 B OO 1 N H S 11 10OO 6' Cl Prepared according General Procedure A on 2.7 mmol scale. Recrystallisation from ethanol (two rounds) afforded the title compound (852 mg, 77 %) as a colourless crystalline solid, m.p. 199.1 – 199.3 ◦C (ethanol). 1H NMR (400 MHz, CDCl3): δ /ppm = 7.62 (d, J = 8.6 Hz, 2 H, H-10), 7.55 (d, J = 7.5 Hz, 1 H, H-4), 7.47 (s, 1 H, H-8), 7.40 (d, J = 8.6 Hz, 2 H, H-11), 7.15 (d, J = 7.5 Hz, 1 H), 6.14 (br. s, 1 H, NH), 2.09 (s, 3 H, H-6′), 1.32 (s, 12 H, H-1). 13C NMR (100 MHz, CDCl3): δ /ppm = 139.54 (C-12), 138.10 (C-9), 136.94 (CAr), 133.58 (CH, CAr), 133.52 (CAr), 132.47 (CH, CAr), 130.73 (CH, CAr), 129.33 (CH, C-11), 128.96 (CH, C-10), 84.05 (C-2), 24.99 (CH3, C-6 ′), 18.16 (CH3, C-1), (One carbon resonance was not visible). IR (neat): ν /cm−1 = 3219 (w), 2977 (w), 2162 (w), 2033 (w), 2010 (w), 2000 (w), 1988 (w), 1976 (w), 1614 (w), 1586 (w), 1575 (w), 1515 (w), 1477 (w), 1446 (w), 1409 (m), 1383 (m), 1372 (m), 1350 (s), 1333 (s), 1278 (m), 1218 (w), 1188 (m), 1164 (s), 1145 208 Experimental (s), 1129 (s), 1111 (m), 1089 (s), 1037 (w), 1013 (m), 997 (w), 965 (m), 936 (m), 920 (m), 884 (w), 854 (m), 836 (m), 822 (m), 798 (m), 756 (s), 723 (m), 696 (s), 681 (s). HRMS: m/z calc. for C19H23[ 11B][35Cl]NO4S ([M + H] +) 408.1208, found 408.1198, ∆ = −2.5 ppm. Microanalysis: calc. (found) for C19H22BClNO4S C 55.97% (55.94%), H 5.69% (5.64%), N 3.44% (3.41%), Cl 8.69% (8.56%). The structure was confirmed by X-ray crystallographic analysis, and deposited at Cambridge University with the unique reference SL1242. 3,4-Dichloro-N-(2-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)phenyl)benzenesulfonamide (102d) 4 5 8 B OO 1 N H S 10 11 14OO 6' Cl Cl Prepared according General Procedure A on 3.3 mmol scale. Recrystallisation from ethanol (two rounds) afforded the title compound (922 mg, 63 %) as a colourless crystalline solid, m.p. 176.2 – 176.4 ◦C (ethanol). 1H NMR (400 MHz, CDCl3): δ /ppm = 7.81 (s, 1 H, H-14), 7.57 (d, J = 7.5 Hz, 1 H, H-4), 7.51 (m, 2 H, H-10 and H-11), 7.40 (s, 1 H, H-8), 7.18 (d, J = 7.5 Hz, 1 H, H-5), 6.29 (br. s, 1 H, NH), 2.17 (s, 3 H, H-6′), 1.31 (s, 12 H, H-1). 13C NMR (100 MHz, CDCl3): δ /ppm = 139.28, 137.86, 137.23, 133.83 (CH), 133.69, 133.18, 132.35 (CH), 131.11 (CH), 130.91 (CH), 129.56 (CH), 126.57 (CH), 126.36, 84.09, 24.99 (CH3), 18.29 (CH3). IR (neat): ν /cm−1 = 3202 (w), 2975 (w), 1617 (w), 1520 (w), 1452 (w), 1424 (m), 1394 (w), 1386 (w), 1356 (s), 1339 (s), 1310 (m), 1272 (m), 1217 (w), 1192 (m), 1167 (s), 1141 (s), 1129 (s), 1090 (s), 1033 (m), 998 (w), 963 (m), 942 (m), 914 (w), 889 (w), 855 (m), 822 (s), 813 (s), 798 (m), 767 (w), 700 (s), 677 (s). HRMS: m/z calc. for C19H24[ 11B][35Cl]2NO4S ([M + H]+) 442.0818, found 442.0806, ∆ = −2.7 ppm. Microanalysis: calc. (found) for C19H23BCl2NO4S C 51.61% (51.46%), H 5.02% (4.92%), N 3.17% (3.14%), Cl 16.03% (15.76%). Experimental 209 General Procedure B for Aryl Coupling Triazolopyridazine 76 (0.1 mmol), the boronic ester (1.1 equiv.), potassium phosphate (4 equiv.) and [1,1′-bis(di-tert-butylphosphino)ferrocene]dichloropalladium(II) (10 mol%) were combined with n-butanol / water (7 : 3 v/v, 2 mL) in a sealed vial. The solvent was degassed under vacuum by three freeze-thaw cycles, and the vial was then backfilled with N2. The reaction mixture was heated to 100 ◦C for 4 h under microwave irradiation. The solvent was removed, and the resulting mixture redissolved in methanol / water (1 : 1 v/v) and purified by preparative HPLC using a methanol / water gradient. N-(5-(8-Amino-3-methyl-[1,2,4]triazolo[4,3-b]pyridazin-6-yl)-2-meth- ylphenyl)-2-chlorobenzenesulfonamide (103b) 5 N N N N 1 12 9 8 10' N H S 17 16 15 14 OO H2N Cl Prepared according to General Procedure B above. Purification by HPLC afforded the title compound (15 mg, 35 %) as a white solid. 1H NMR (400 MHz, CDCl3): δ /ppm = 8.07 (dd, J = 7.9 Hz and 1.5 Hz, 1 H, H-19), 7.79 (d, J = 1.8 Hz, 1 H, H-12), 7.64 – 7.44 (m, 3 H, H-8, H-15 and H-9 or H-22), 7.36 (t, J = 7.4 Hz, 1.5 Hz, 1 H, H-21), 7.25 (d, J = 7.4 Hz, 1 H, H-9 or H-22), 6.98 (br. s, 1 H, NH), 6.41 (s, 1 H, H-5), 5.57 (br. s, 2 H, NH2), 2.78 (s, 3 H, H-1), 2.34 (s, 3 H, H-10 ′). 13C NMR (126 MHz, CDCl3): δ /ppm = 154.16, 148.03, 141.15, 140.02, 136.97, 134.84, 134.70, 134.24 (CH), 132.92, 131.82 (CH), 131.82 (CH), 131.54, 131.49 (CH), 127.29 (CH), 124.45 (CH), 121.18 (CH), 93.69 (CH), 17.69 (CH3), 10.00 (CH3). HRMS: m/z calc. for C19H18N6O2S[ 35Cl] ([M + H]+) 429.0900, found 429.0901, ∆ = 0.2 ppm. 210 Experimental N-(5-(8-Amino-3-methyl-[1,2,4]triazolo[4,3-b]pyridazin-6-yl)-2-meth- ylphenyl)-3-chlorobenzenesulfonamide (103c) 5 N N N N 1 12 9 8 10' N H S 18 16 15 14 OO H2N Cl Prepared according to General Procedure B above. Purification by HPLC afforded the title compound (1 mg, 4 %) as a white solid. 1H NMR (400 MHz, CDCl3): δ /ppm = 7.85 (d, J = 1.9 Hz, 1 H, H-12), 7.81 (t, J = 1.9 Hz, 1 H, H-18), 7.73 (dd, J = 8.0 Hz and 1.9 Hz, 1 H, H-14), 7.64 (d, J = 8.0 Hz, 1 H, H-16), 7.55 (d, J = 8.2 Hz, 1 H, H-8), 7.40 (t, J = 8.0 Hz, 1 H, H-15), 7.26 (d, J = 8.2 Hz, 1 H, H-9), 6.48 (s, 1 H, H-5), 6.41 (br. s, 1 H, NH), 5.43 (br. s, 2 H, NH2), 2.81 (s, 3 H, H-1), 2.10 (s, 3 H, H-10′). 13C NMR (126 MHz, CDCl3): δ /ppm = 153.99, 148.13, 141.23, 141.16, 140.02, 135.33, 135.07, 134.42, 133.62, 133.28 (CH), 131.47 (CH), 130.44 (CH), 127.25 (CH), 125.29 (CH), 125.22 (CH), 123.27 (CH), 93.72 (CH), 17.50 (CH3), 10.00 (CH3). HRMS: m/z calc. for C19H18N6O2S[ 35Cl] ([M + H]+) 429.0900, found 429.0909, ∆ = 2.1 ppm. N-(5-(8-Amino-3-methyl-[1,2,4]triazolo[4,3-b]pyridazin-6-yl)-2-meth- ylphenyl)-4-chlorobenzenesulfonamide (103d) 5 N N N N 1 12 9 8 10' N H S 14 15 OO H2N Cl Prepared according to the General Procedure B above. Purification by HPLC afforded the title compound (12 mg, 32 %) as a white solid. Experimental 211 1H NMR (500 MHz, DMSO-d6): δ /ppm = 9.81 (br. s, 1 H, NH), 7.73 – 7.52 (m, 7 H), 7.46 (br. s, 2 H, NH2), 7.27 (d, J = 8.0 Hz, 1 H, H-9), 6.45 (s, 1 H, H-5), 2.61 (s, 3 H, H-1), 2.06 (s, 3 H, H-10′). 13C NMR (126 MHz, DMSO-d6): δ /ppm = 153.35 (C-6), 146.70 (C-2), 142.85 (C-3), 140.12 (C-16), 139.86 (C-4), 137.24 (C-13), 135.62 (C-10), 134.08 (C-7), 131.10 (CH, C-9), 129.29 (CH, C-15), 128.50 (CH, C-14), 123.69 (CH, C-12), 91.45 (CH, C-5), 17.66 (CH3, C-10 ′), 9.53 (CH3, C-1), (C-8 and C-11 were not visible). HRMS: m/z calc. for C19H18[ 35Cl]N6O2S ([M + H] +) 429.0900, found 429.0894, ∆ = −1.4 ppm. N-(5-(8-Amino-3-methyl-[1,2,4]triazolo[4,3-b]pyridazin-6-yl)-2-meth- ylphenyl)-3,4-dichlorobenzenesulfonamide (103e) 5 N N N N 1 12 9 8 10' N H S 18 15 14 OO H2N Cl Cl Prepared according to the General Procedure B above. Purification by HPLC afforded the title compound (9 mg, 13 %) as a white solid. 1H NMR (400 MHz, DMSO-d6): δ /ppm = 10.00 (br. s, 1 H, NH), 7.84 (d, J = 2.1 Hz, 1 H, H-18), 7.75 (d, J = 8.3 Hz, 1 H, H-15), 7.64 (dd, J = 8.4 Hz and 2.1 Hz, 1 H, H-14), 7.54 (d, J = 1.9 Hz, 1 H, H-12), 7.41 (br. s, 2 H, NH2), 7.46 – 7.33 (m, 1 H, H-8), 7.19 (d, J = 7.8 Hz, 1 H, H-9), 6.44 (s, 1 H, H-5), 2.60 (s, 3 H, H-1), 2.10 (s, 3 H, H-10′). 13C NMR (126 MHz, DMSO-d6): δ /ppm = 154.13 (C-6), 146.67 (C-2), 142.74 (C-3), 139.97 (C-4), 134.49 (C-11), 133.97 (C-13), 133.82 (C-7), 131.47 (C-16), 131.19 (CH, C-15), 130.58 (CH, C-9), 128.19 (CH, C-18), 126.67 (CH, C-14), 91.73 (CH, C-5), 18.23 (CH3, C-10′), 9.58 (CH3, C-1), (C-8, C-10, C-12 and C-17 were not visible). HRMS: m/z calc. for C19H17[ 35Cl]2N6O2S ([M + H] +) 463.0511, found 463.0505, ∆ = −1.3 ppm. 212 Experimental Ethyl (6-(3-((2-chlorophenyl)sulfonamido)-4-methylphenyl)-3-meth- yl-[1,2,4]triazolo[4,3-b]pyridazin-8-yl)carbamate (108b) 5 N N N N 1 12 9 8 10' N S 17 16 15 OO H2N O O Cl 20 21 To a solution of compound 103b (1 mg), N,N-dimethylaminopyridine (3 mg) and triethy- lamine (8 µL) in tetrahydrofuran (5 mL) under an N2 atmosphere was added a solution of ethyl chloroformate in tetrahydrofuran (0.1 M, 0.4 mL) in two portions. The mixture was stirred at room temperature for 20 minutes. Aqueous citric acid (0.1 M, 5 mL) was added, and the mixture concentrated under reduced pressure. The residue was dissolved in methanol / water (1 : 1 v/v) and purified by HPLC to afford the title compound (<1 mg). 1H NMR (400 MHz, CDCl3): δ /ppm = 8.43 (d, J = 8.1 Hz and 1.9 Hz, 1 H), 8.10 (d, J = 1.9 Hz, 1 H), 7.88 (dd, J = 8.0 Hz and 1.9 Hz, 1 H), 7.66 – 7.56 (m, 2 H), 7.56 – 7.49 (m, 1 H), 7.45 (d, J = 8.0 Hz, 1 H), 6.52 (s, 1 H), 5.45 (s, 2 H), 4.25 – 4.04 (m, 2 H), 2.83 (s, 3 H), 2.49 (s, 3 H), 1.13 (t, J = 7.1 Hz, 3 H). Experimental 213 Ethyl (6-(3-((3-chlorophenyl)sulfonamido)-4-methylphenyl)-3-meth- yl-[1,2,4]triazolo[4,3-b]pyridazin-8-yl)carbamate (108c) 5 N N N N 1 12 9 8 10' N S 18 16 15 OO H2N O O Cl 20 21 To a solution of compound 103c (1 mg), N,N-dimethylaminopyridine (5.6 mg) and triethy- lamine (8 µL) in tetrahydrofuran (5 mL) under an N2 atmosphere was added a solution of ethyl chloroformate in tetrahydrofuran (0.1 M, 0.2 mL). The mixture was stirred at room temperature for 10 minutes. Aqueous citric acid (0.1 M, 5 mL) was added, and the mixture concentrated under reduced pressure. The residue was dissolved in methanol / water (1 : 1 v/v) and purified by HPLC to afford the title compound (<1 mg). 1H NMR (400 MHz, CDCl3): δ /ppm = 8.10 (t, J = 1.9 Hz, 1 H, H-18), 8.03 (d, J = 7.8, 1 H, H-14), 7.91 (dd, J = 8.0 Hz and 1.9 Hz, 1 H, H-8), 7.73 – 7.64 (m, 2 H, H-12 and H-16), 7.55 (t, J = 8.0 Hz, 1 H, H-15), 7.46 (d, J = 8.0 Hz, 1 H, H-9), 6.50 (s, 1 H, H-5), 5.50 (s, 2 H, NH2), 4.31 – 4.07 (m, NH2, H-20), 2.82 (s, 3 H, H-1), 2.37 (s, 3 H, H-10 ′), 1.19 (t, J = 7.1 Hz, 3 H, H-21). 13C NMR (126 MHz, CDCl3): δ /ppm = 153.73, 151.65, 148.15, 141.26, 140.79, 140.55, 140.00, 135.58, 134.99, 134.92, 134.12 (CH), 131.77 (CH), 130.14 (CH), 129.16 (CH), 128.12 (CH), 127.79 (CH), 127.46 (CH), 93.66 (CH), 64.00 (CH2), 18.10 (CH3), 14.08 (CH3), 10.03 (CH3). 214 Experimental Ethyl (6-(3-((4-chlorophenyl)sulfonamido)-4-methylphenyl)-3-meth- yl-[1,2,4]triazolo[4,3-b]pyridazin-8-yl)carbamate (108d) 5 N N N N 1 12 9 8 10' N S 14 15 OO H2N ClO O 18 19 To a solution of compound 103d (10 mg) and triethylamine (10 µL) in tetrahydrofuran (10 mL) was added a solution of ethyl chloroformate in tetrahydrofuran (0.1 M, 0.5 mL). The mixture was stirred at room temperature for 5 minutes. Aqueous citric acid (0.1 M, 5 mL) was added, and the mixture concentrated under reduced pressure. The residue was extracted into ethyl acetate (2×10 mL), and the combined organic phases were dried (MgSO4), filtered, and concentrated under reduced pressure to afford the title compound (11 mg, 95 %) as a white solid. 1H NMR (400 MHz, CDCl3): δ /ppm = 8.06 (d, J = 8.7 Hz, 2 H, H-14), 7.89 (dd, J = 8.0 Hz and 1.9 Hz, 1 H), 7.65 (d, J = 1.9 Hz, 1 H), 7.57 (d, J = 8.7 Hz, 2 H, H-15), 7.45 (d, J = 8.0 Hz, 1 H, H-9), 6.49 (s, 1 H, H-5), 5.50 (br. s, 2 H, NH2), 4.27 – 4.06 (m, 2 H, H-18), 2.82 (s, 3 H, H-1), 2.34 (s, 3 H, H-10′), 1.18 (t, J = 7.1 Hz, 3 H, H-19). 13C NMR (126 MHz, CDCl3): δ /ppm = 153.78, 151.77, 148.10, 141.24, 140.81, 140.42, 139.99, 137.51, 135.61, 134.88, 131.71 (CH), 130.72 (CH), 129.19 (CH), 128.06 (CH), 127.88 (CH), 93.66 (CH), 63.87 (CH2), 18.11 (CH3), 14.12 (CH3), 10.01 (CH3). HRMS: m/z calc. for C22H22[ 35Cl]N6O4S ([M + H] +) 501.1112, found 501.1133, ∆ = 4.2 ppm. Experimental 215 Ethyl ((3,4-dichlorophenyl)sulfonyl)(5-(8-((ethoxycarbonyl)amino)-3- methyl-[1,2,4]triazolo[4,3-b]pyridazin-6-yl)-2-methylphenyl)carbamate (109e) 5 N N N N 1 12 9 8 10' N S 18 15 OO H N Cl Cl O OO 21 20 O24 23 To a solution of 103e (4 mg) and triethylamine (10 µL) in dichloromethane (5 mL) was added ethyl chloroformate (0.1 mL). The mixture was stirred at room temperature for 5 minutes. Water (2 mL) was added, and the solvent removed under reduced pressure. Purification by HPLC afforded the title compound (2 mg, 38 %). 1H NMR (500 MHz, DMSO-d6): δ /ppm = 8.20 (d, J = 2.2 Hz, 1 H, H-18), 8.09 (s, 1 H, H-5), 8.05 (d, J = 8.5 Hz, 1 H, H-15), 8.01 (dd, J = 8.0 Hz and 1.9 Hz, 1 H, H-8), 7.94 (dd, J = 8.5 Hz and 2.2 Hz, 1 H, H-14), 7.76 (d, J = 1.9 Hz, 1 H, H-12), 7.63 (dd, J = 8.0 Hz, 1 H, H-9), 4.27 (q, J = 7.1 Hz, 2 H, H-23), 4.23 – 4.02 (m, 2 H, H-20), 2.73 (s, 3 H, H-1), 2.32 (s, 3 H, H-10′), 1.30 (t, J = 7.1 Hz, 3 H, H-24), 1.09 (t, J = 7.1 Hz, 3 H, H-21). 13C NMR (126 MHz, DMSO-d6): δ /ppm = 171.59, 156.02, 152.86, 148.75, 147.50, 140.57, 139.09, 138.28, 137.98, 135.36, 134.32, 132.26, 132.08 (CH), 131.95 (CH), 130.62 (CH), 128.58 (CH), 128.21 (CH), 127.58 (CH), 100.75 (CH), 64.56 (CH2), 64.09 (CH2), 17.57 (CH3), 14.39 (CH3), 13.85 (CH3), 9.56 (CH3), (One carbon resonance was not visible). HRMS: m/z calc. for C25H25N6O6S[ 35Cl]2 ([M + H] +) 607.0933, found 607.0912, ∆ = −3.5 ppm. 216 Experimental Chemical Probes for CECR2 6-Chloro-3-methyl-[1,2,4]triazolo[4,3-b]pyridazine (129)233 4 5 N N NN 1 Cl To 3-chloro-6-hydrazino pyridazine (250 mg, 1.75 mmol, Alfa Aesar) was added acetic acid (2.5 mL) and the mixture heated at 120 ◦C for 1 hour in a sealed vial under microwave irradiation. The solvent was removed, and the remaining acetic acid removed by azeotropic drying with toluene. The title compound was obtained (270 mg, 1.60 mmol, 92 %) as a white solid, m.p. 105 – 107 ◦C (ethyl acetate / hexane) [lit: 106 – 107 ◦C233]. 1H NMR (400 MHz, CDCl3): δ /ppm = 8.02 (d, J = 9.6 Hz, 2 H, H-5), 7.07 (d, J = 9.6 Hz, 2 H, H-4), 2.77 (s, 3 H, H-1). 13C NMR (100 MHz, CDCl3): δ /ppm = 149.19 (C-6), 147.36 (C-2), 142.74 (C-3), 126.39 (CH, C-5), 121.76 (CH, C-4), 9.78 (CH3, C-1). IR (neat): ν /cm−1 = 3029 (w), 1813 (w), 1605 (w), 1538 (w), 1513 (m), 1464 (m), 1441 (w), 1406 (w), 1383 (m), 1364 (m), 1330 (m), 1172 (w), 1152 (m), 1139 (w), 1081 (m), 1030 (m), 996 (m), 930 (m), 813 (s), 784 (m), 755 (s), 669 (w), 658 (m). LC-MS: tR = 1.04 min, m/z 169.15 ([M + H]+). HRMS: m/z calc. for [C6H6N4 35Cl]+ ([M + H]+) 169.0276, found 169.0270, ∆ = −3.1 ppm. General Procedure A for the synthesis of sulfonamides The sulfonyl chloride (256 mg, 1 mmol) was dissolved in n-butanol, CPME or DME (8 mL). QuadraPure-dimethylamine (250 mg, 0.75 equiv.) was added and the mixture stirred at 25 ◦C for 15 minutes. The amine (1 mmol) was added, with rapid formation of a crystalline precipitate. The mixture was stirred at 25 ◦C for 1 hour, and then water (1 mL) was added. The amine resin was removed by filtration, and then the solvents were removed to afford the product as a colourless crystalline solid. Experimental 217 4-((4-Bromophenyl)sulfonyl)morpholine (125a) 3 2 Br S N O 6 5 O O Prepared according to General Procedure A above, and collected as the hydrochloride salt (357 mg, quant.). Free base m.p. 150.2 – 150.5 ◦C (dimethoxyethane / water). 1H NMR (400 MHz, CDCl3): δ /ppm = 9.97 (br. s, NH+), 7.69 (d, J = 8.7, 2 H, H-2 or H-3), 7.62 (d, J = 8.7, 2 H, H-2 or H-3), 3.78 – 3.70 (m, 4 H, H-6), 3.04 – 2.96 (m, 4 H, H-5). 13C NMR (100 MHz, CDCl3): δ /ppm = 134.40 (C-1 or C-4), 132.61 (CH, C-2 or C-3), 129.43 (CH, C-3 or C-3), 128.36 (C-1 or C-4), 66.18 (CH2, C-6), 46.07 (CH2, C-5). IR (neat): ν /cm−1 = 2971 (w), 2920 (w), 2904 (w), 2867 (w), 2761 (w), 2720 (w), 2161 (w), 1574 (w), 1466 (w), 1453 (m), 1431 (w), 1390 (w), 1347 (s), 1332 (m), 1299 (w), 1276 (w), 1261 (m), 1228 (w), 1216 (w), 1188 (w), 1179 (w), 1161 (s), 1123 (m), 1108 (s), 1093 (s), 1069 (s), 1049 (w), 1021 (w), 1009 (m), 970 (w), 942 (s), 925 (m), 911 (w), 872 (w), 849 (m), 826 (s), 753 (s), 715 (m). LC-MS: tR = 4.52 min, m/z 307.94 ([M + H]+). HRMS: m/z calc. for [C10H13NO3S 79Br]+ ([M + H]+) 305.9800, found 305.9801, ∆ = 0.3 ppm. 4-((3-Bromophenyl)sulfonyl)morpholine (122a) 4 5 13 S N O 8 7 Br O O Prepared according to General Procedure A above, and collected as the hydrochloride salt (338 mg, quant.). Free base m.p. 151.3 – 151.5 ◦C (CPME). 1H NMR (400 MHz, CDCl3): δ /ppm = 7.89 (t, J = 1.5 Hz, 1 H, H-1), 7.75 (dt, J = 7.9 Hz and 1.5 Hz, 1 H, H-3), 7.68 (dt, J = 7.9 Hz and 1.5 Hz, 1 H, H-5), 7.43 (t, J = 7.9 Hz, 1 H, H-4), 3.78 – 3.71 (m, 4 H, H-8), 3.06 – 2.98 (m, 4 H, H-7). 13C NMR (100 MHz, CDCl3): δ /ppm = 137.18 (C-2 or C-6), 136.10 (CH, C-3), 130.64 (CH, C-1 or C-4), 130.56 (CH, C-1 or C-4), 126.30 (CH, C-5), 123.27 (C-2 or C-6), 66.02 (CH2, C-8), 45.95 (CH2, C-7). 2868 (w), 2860 (w), 2161 (w), 1979 (w), 1574 (w), 1467 (w), 1453 (m), 1390 (w), 1363 218 Experimental (w), 1347 (s), 1332 (m), 1299 (w), 1276(w), 1261 (m), 1217 (w), 1188 (w), 1178 (w), 1161 (s), 1123 (w), 1108 (s), 1093 (s), 1069 (s), 1021 (w), 1009 (m), 970 (w), 942 (s), 925 (m), 849 (m), 826 (s), 753 (s), 716 (m), 708 (m). LC-MS: tR = 4.48 min, m/z 307.88 ([M + H]+). HRMS: m/z calc. for [C10H13NO3S 79Br]+ ([M + H]+) 305.9800, found 305.9814, ∆ = 4.6 ppm. 2-((4-Bromophenyl)sulfonyl)-1,2,3,4-tetrahydroisoquinoline (125b) 3 2 Br S N 5 6 13 11 10 9 8 O O Prepared according to General Procedure A above, and collected as the hydrochloride salt after recrystallisation from ethanol (176 mg, 0.453 mmol, 45 %). Free base, m.p. 153.2 – 154.4 ◦C (ethanol). 1H NMR (400 MHz, CDCl3): δ /ppm = 7.70 (d, J = 8.7 Hz, 1 H, H-2 or H-3), 7.66 (d, J = 8.7 Hz, 1 H, H-2 or H-3), 7.20 – 6.99 (m, 4 H, H-8 to H-11), 4.28 (s, 1 H, H-13), 3.39 (t, J = 6.0 Hz, 1 H, H-5), 2.93 (t, J = 6.0 Hz, 1 H, H-6). 13C NMR (100 MHz, CDCl3): δ /ppm = 135.82 (C-4), 133.04 (C-7), 132.53 (CH, C-2), 131.43 (C-12), 129.26 (CH, C-3), 128.99 (CH, C-8), 128.03 (C-1), 127.03 (CH, C-10), 126.60 (CH, C-11), 126.45 (CH, C-9), 47.60 (CH2, C-13), 43.83 (CH2, C-5), 28.89 (CH2, C-6). IR (neat): ν /cm−1 = 2160 (w), 1576 (w), 1498 (w), 1467 (w), 1428 (w), 1393 (w), 1356 (w), 1334 (m), 1272 (w), 1244 (w), 1162 (m), 1134 (w), 1121 (w), 1090 (m), 1068 (m), 1023 (m), 1009 (m), 954 (m), 921 (m), 838 (w), 822 (m), 771 (m), 743 (s), 706 (m). HRMS: m/z calc. for [C15H14O2N 79Br23Na32S]+ ([M + Na]+) 373.9821, found 373.9803, ∆ = −4.8 ppm. 4-((4-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)sulfonyl) morpholine (126a) 3 2 B S N 5 6 O O O 2' O O Experimental 219 4-Bromobenzenesulfonyl chloride (256 mg, 1 mmol) was dissolved in n-butanol (4 mL) and heated to 50 ◦C. Morpholine (0.2 mL, 2 mmol) was added, and the mixture allowed to cool to room temperature. A precipitate formed upon cooling. Bis(pinacolato)diboron (279 mg, 1.1 mmol, 1.1 equiv.), KOAc (294 mg, 3 mmol, 3 equiv.), Pd2(dba)3 (21 mg, 20 µmol, 0.02 equiv.) and X-Phos (19 mg, 40 µmol, 0.04 equiv.) were added. The resulting mixture was degassed by sparging with N2 (1 bar) for 15 minutes, and then heated at 150 ◦C for 1 hour in a sealed vial under microwave irradiation, and then allowed to cool to room temperature. The solvent was removed and the solid product recrystallised from ethanol to afford the title compound (43 mg, 0.12 mmol, 12 %) as a colourless crystalline solid, m.p. 170.1 – 170.4 ◦C (ethanol). 1H NMR (400 MHz, CDCl3): δ /ppm = 7.97 (d, J = 7.87 Hz, 2 H, H-2), 7.73 (d, J = 7.87 Hz, 2 H, H-3), 3.76 – 3.69 (m, 4 H, H-6), 3.03 – 2.95 (m, 4 H, H-5), 1.36 (s, 12 H, H-2′). 13C NMR (100 MHz, CDCl3): δ /ppm = 137.41 (C-4), 135.46 (CH, C-2), 126.91 (CH, C-3), 84.63 (C-2′), 66.22 (CH2, C-6), 46.12 (CH2, C-5), 25.02 (CH3, C-2′), (C-1 was not observed). IR (neat): ν /cm−1 = 2161 (w), 1449 (w), 1392 (m), 1352 (s), 1295 (w), 1260 (m), 1212 (w), 1168 (s), 1138 (m), 1115 (s), 1101 (m), 1079 (s), 1018 (m), 964 (w), 940 (s), 852 (m), 822 (w), 745 (s), 732 (s), 703 (m), 670 (m). LC-MS: tR = 4.89 min, m/z 354.00 ([M + H]+). HRMS: m/z calc. for C16H25 11BNO5 32S]+ ([M + H]+) 354.1547, found 354.1546, ∆ = −0.3 ppm. Microanalysis: calc. (found) for C16H24BNO5S C 54.54% (54.39%), H 6.85% (6.81%), N 3.97% (4.11%). 2-((4-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)sulfonyl)- 1,2,3,4-tetrahydroisoquinoline (126b) 3 2 S N 5 6 13 11 10 9 8 BO O 2' O O Compound 125c (105 mg, 0.3 mmol) was combined with bis(pinacolato)diboron (114 mg, 0.45 mmol, 1.5 equiv.), KOAc (88 mg, 0.9 mmol, 3 equiv.), Pd2(dba)3 (6 mg, 6 µmol, 0.02 equiv.) and X-Phos (6 mg, 12 µmol, 0.04 equiv.). CPME (12 mL) was added and the 220 Experimental resulting mixture degassed by sparging with N2 (1 bar) for 15 minutes, and then heated at 110 ◦C for 1 hour in a sealed vial under microwave irradiation. The title compound was obtained as a colourless crystalline solid (98 mg, 244 mmol, 49 %) after recrystallisation from hot ethanol, m.p. 168.5 – 169.7 ◦C (ethanol). 1H NMR (400 MHz, CDCl3): δ /ppm = 7.95 (d, J = 8.0 Hz, 2 H, H-2 or H-3), 7.82 (d, J = 8.0 Hz, 2 H, H-2 or H-3), 7.19 – 6.97 (m, 4 H, H-8 to H-11), 4.25 (s, 2 H, H-13), 3.36 (t, J = 5.9 Hz, 2 H, H-5), 2.93 (q, J = 5.9 Hz, 2 H, H-6), 1.35 (s, 12 H, H-2′). 13C NMR (100 MHz, CDCl3): δ /ppm = 138.66 (C-4), 135.46 (CH, C-2), 133.17 (C-7), 131.67 (C-12), 128.95 (CH, C-8), 126.89 (CH, C-10), 0 126.77 (CH, C-3), 126.51 (CH, C-11), 126.47 (CH, C-9), 84.59 (C-1′), 47.63 (CH2, C-13), 43.87 (CH2, C-5), 29.03 (CH2, C-6), 25.02 (CH3, C-2 ′), (C-1 was not observed). IR (neat): ν /cm−1 = 1391 (w), 1353 (s), 1335 (s), 1270 (w), 1165 (s), 1144 (m), 1121 (w), 1096 (m), 1077 (m), 1069 (m), 1024 (w), 1017 (w), 953 (m), 922 (m), 856 (w), 839 (w), 823 (w), 814 (w), 766 (m), 733 (s), 714 (w), 671 (w), 656 (s). LC-MS: tR = 5.42 min, m/z 400.01 ([M + H]+). HRMS: m/z calc. for [C21H26O4N 11B23Na32S]+ ([M + Na]+) 422.1568, found 422.1555, ∆ = −2.97 ppm. Microanalysis: calc. (found) for C21H25O4NBS C 63.17% (63.24%), H 6.56% (6.38%), N 3.51% (3.80%). General Procedure B for one-pot synthesis of target molecules The sulfonyl chloride (256 mg, 1 mmol) was dissolved in n-butanol (8 mL). QuadraPure- dimethylamine (250 mg, 0.75 equiv.) was added and the mixture stirred at 25 ◦C for 15 minutes. The amine (1 mmol) was added, with rapid formation of a crystalline precipitate. The mixture was stirred at 25 ◦C for 1 hour, and then water (1 mL) was added. The amine resin was removed by filtration, and then the solvents were removed to afford a solid product. The solid (generally approx. 0.75 mmol) was combined with bis(pinacolato)diboron (286 mg, 1.13 mmol, 1.5 equiv.), KOAc (221 mg, 2.25 mmol, 3 equiv.), Pd2(dba)3 (16 mg, 15 µmol, 0.02 equiv.) and X-Phos (14 mg, 30 µmol, 0.04 equiv.). CPME (12 mL) was added and the resulting mixture degassed by sparging with N2 (1 bar) for 15 minutes, and then heated at 110 ◦C for 1 hour in a sealed vial under microwave irradiation, and then allowed to cool to room temperature. The solvents were removed under reduced pressure and then the reaction mixture was redis- Experimental 221 solved in n-butanol / water (9 : 1 v/v, 10 mL). Triazolopyridazine 129 (76 mg, 0.46 mmol, 0.9 equiv.) and K3PO4 (386 mg, 1.8 mmol, 3.6 equiv.) were added and the resulting mixture degassed by sparging with N2 (1 bar) for 15 minutes, and then heated at 100 ◦C for 2 hours in a sealed vial under microwave irradiation. The reaction mixture was allowed to cool to room temperature, concentrated, loaded onto KP-NH silica, and then purified by column chromatography using KP-NH silica, using a gradient of ethyl acetate / methanol (9 : 1 v/v) and n-hexane: 0 : 1 for 4 CV, then 0 : 1 to 2 : 3 over 40 CV. 4-((4-(3-Methyl-[1,2,4]triazolo[4,3-b]pyridazin-6-yl)phenyl)sulfonyl) morpholine (128a) 8 9 5 4 N N N N 1 S N O 12 11 O O Obtained using General Procedure B above. Purification by column chromatography afforded the title compound (7 mg, 0.019 mmol, 2 % over 3 steps) as a white solid. 1H NMR (500 MHz, CDCl3): δ /ppm = 8.22 (d, J = 9.7 Hz, 1 H, H-4), 8.18 (d, J = 8.3 Hz, 2 H, H-8), 7.94 (d, J = 8.3 Hz, 2 H, H-9), 7.56 (d, J = 9.7 Hz, 1 H, H-5), 3.80 – 3.74 (m, 4 H, H-12), 3.12 – 3.03 (m, 4 H, H-11), 2.90 (s, 3 H, H-1). 13C NMR (126 MHz, CDCl3): δ /ppm = 151.84 (C-6), 147.84 (C-2), 143.34 (C-3), 138.98 (C-10), 137.55 (C-7), 128.77 (CH, C-8), 128.17 (CH, C-9), 125.82 (CH, C-4), 118.41 (CH, C-5), 66.23 (CH2, C-12), 46.12 (CH2, C-11), 10.01 (CH3, C-1). IR (neat): ν /cm−1 = 2860 (w), 2160 (w), 2024 (w), 1551 (w), 1524 (w), 1454 (w), 1392 (w), 1346 (m), 1329 (m), 1309 (m), 1260 (m), 1166 (s), 1128 (m), 1113 (s), 1071 (m), 1042 (w), 1014 (w), 990 (w), 946 (s), 848 (m), 827 (m), 763 (s), 738 (s), 710 (m). LC-MS: tR = 4.18 min, m/z 360.02 ([M + H]+). HRMS: m/z calc. for [C16H18N5O3 32S]+ ([M + H]+) 360.1130, found 360.1135, ∆ = 1.4 ppm. 222 Experimental 2-((4-(3-Methyl-[1,2,4]triazolo[4,3-b]pyridazin-6-yl)phenyl)sulfonyl)- 1,2,3,4-tetrahydroisoquinoline (128b) 8 9 5 4 N N N N 1 S N 11 12 19 17 16 15 14 O O Obtained using General Procedure B above. Purification by column chromatography and subsequent recrystallisation from chloroform / ethanol afforded the title compound (7 mg, 0.017 mmol, 2 % over 3 steps) as a white solid, m.p. 232.3 – 233.4 ◦C (decomp.). 1H NMR (400 MHz, CDCl3): δ /ppm = 8.20 (d, J = 9.7 Hz, 1 H, H-4), 8.15 (d, J = 8.3 Hz, 2 H, H-8), 8.02 (d, J = 8.3 Hz, 2 H, H-9), 7.55 (d, J = 9.7 Hz, 1 H, H-5), 7.21 – 7.02 (m, 4 H, H-14 to H-17), 4.35 (s, 2 H, H-19), 3.46 (t, J = 5.9 Hz, 2 H, H-11), 2.95 (t, J = 5.9 Hz, 2 H, H-12), 2.89 (s, 3 H, H-1). 13C NMR (100 MHz, CDCl3): δ /ppm = 151.91 (C-6), 147.81 (C-2), 139.05 (C-10), 138.68 (C-7), 133.05 (C-13), 131.39 (C-18), 129.03 (CH, C-14), 128.56 (CH, C-8), 128.11 (CH, C-9), 127.09 (CH, C-16), 126.65 (CH, C-17), 126.44 (CH, C-15), 125.73 (CH, C-4), 118.45 (CH, C-5), 115.63 (C-3), 47.62 (CH2, C-19), 43.89 (CH2, C-11), 28.95 (CH2, C-12), 10.00 (CH3, C-1). IR (neat): ν /cm−1 = 2160 (w), 2035 (w), 1968 (w), 1524 (w), 1388 (w), 1339 (m), 1308 (w), 1274 (w), 1164 (m), 1131 (w), 1117 (w), 1106 (w), 1092 (w), 1073 (w), 1048 (w), 1025 (w), 998 (w), 956 (w), 919 (m), 857 (w), 841 (w), 820 (m), 769 (m), 746 (s), 724 (w), 707 (w), 672 (w). LC-MS: tR = 4.69 min, m/z 405.98 ([M + H]+). HRMS: m/z calc. for [C21H20N5O2 32S]+ ([M + H]+) 406.1338, found 406.1349, ∆ = 2.7 ppm. Experimental 223 6-(4-((2-Ethylpiperidin-1-yl)sulfonyl)phenyl)-3-methyl-[1,2,4]triazolo [4,3-b]pyridazine (128c) 8 9 5 4 N N N N 1 S N 14 13 12 11 17 O O Obtained using General Procedure B above. Purification by column chromatography afforded the title compound (16 mg, 0.042 mmol, 4 % over 3 steps) as a white solid, m.p. 179.4 – 179.8 ◦C (ethyl acetate / hexane / methanol). 1H NMR (400 MHz, CDCl3): δ /ppm = 8.19 (d, J = 9.7 Hz, 1 H, H-4), 8.13 (d, J = 8.5 Hz, 2 H, H-8), 8.02 (d, J = 8.5 Hz, 2 H, H-9), 7.57 (d, J = 9.7 Hz, 1 H, H-5), 4.00 (q, J = 6.8 Hz, 1 H, H-15), 3.83 (dd, J = 14.0 Hz and 4.6 Hz, 1 H, H-11), 3.04 (td, J = 14.0 Hz and 2.7 Hz, 1 H, H-11), 2.91 (s, 3 H, H-1), 1.73 – 1.47 (m, 7 H, H-12, H-13, H-14 and H-16), 1.38 – 1.13 (m, 1 H, H-12), 0.90 (t, J = 7.5 Hz, 3 H, H-17). 13C NMR (100 MHz, CDCl3): δ /ppm = 151.95 (C-6), 147.76 (C-2), 144.39 (C-3), 143.37 (C-10), 137.84 (C-7), 127.97 (CH, C-8 or C-9), 127.81 (CH, C-8 or C-9), 125.57 (CH, C-4), 118.47 (CH, C-5), 54.98 (CH, C-15), 40.93 (CH2, C-11), 27.31 (CH2, C-14), 24.76 (CH2, C-12, C-13 or C-16), 22.65 (CH2, C-12, C-13 or C-16), 18.47 (CH2, C-12, C-13 or C-16), 11.15 (CH3, C-17), 10.00 (CH3, C-1). IR (neat): ν /cm−1 = 3123 (w), 3068 (w), 2941 (w), 2871 (w), 2527 (w), 2418 (w), 2179 (w), 2164 (w), 2144 (w), 2033 (w), 2019 (w), 1962 (w), 1609 (w), 1552 (w), 1527 (w), 1493 (w), 1468 (w), 1397 (m), 1374 (m), 1360 (w), 1330 (s), 1311 (m), 1291 (m), 1270 (w), 1218 (w), 1194 (w), 1162 (s), 1152 (s), 1092 (m), 1071 (w), 1058 (m), 1038 (m), 1012 (m), 993 (m), 950 (s), 917 (m), 904 (m), 865 (m), 851 (w), 837 (m), 818 (s), 775 (m), 760 (m), 734 (s), 686 (m). LC-MS: tR = 4.69 min, m/z 386.02 ([M + H]+). HRMS: m/z calc. for [C19H24N5O2 32S]+ ([M + H]+) 386.1651, found 386.1649, ∆ = −0.5 ppm. Microanalysis: calc. (found) for C19H23N5O2S C 59.20% (59.16%), H 6.01% (5.88%), N 18.17% (17.75%). 224 Experimental 6-(4-((4-Ethylpiperazin-1-yl)sulfonyl)phenyl)-3-methyl-[1,2,4]triazolo [4,3-b]pyridazine (128d) 8 9 5 4 N N N N 1 S N 11 12 N 13 14 O O Obtained using General Procedure B above. Purification by column chromatography and subsequent recrystallisation from ethanol afforded the title compound (11 mg, 0.028 mmol, 3 % over 3 steps) as a white solid. 1H NMR (500 MHz, CDCl3): δ /ppm = 8.21 (d, J = 9.7 Hz, 1 H, H-4), 8.14 (d, J = 8.3 Hz, 2 H, H-8), 7.93 (d, J = 8.3 Hz, 2 H, H-9), 7.54 (d, J = 9.7 Hz, 1 H, H-5), 3.17 – 3.03 (m, 4 H, H-11), 2.90 (s, 3 H, H-1), 2.59 – 2.50 (m, 4 H, H-12), 2.41 (q, J = 7.2 Hz, 2 H, H-13), 1.03 (t, J = 7.2 Hz, 3 H, H-14). 13C NMR (126 MHz, CDCl3): δ /ppm = 151.93 (C-6), 147.84 (C-2), 143.35 (C-3), 138.78 (C-10), 137.57 (C-7), 128.79 (CH, C-8 or C-9), 128.04 (CH, C-8 or C-9), 125.78 (CH, C-4), 118.44 (CH, C-5), 52.03 (CH2, C-13), 51.91 (CH2, C-12), 46.25 (CH2, C-11), 12.06 (CH3, C-14), 10.01 (CH3, C-1). IR (neat): ν /cm−1 = 1547 (w), 1521 (w), 1454 (w), 1388 (w), 1330 (m), 1307 (m), 1262 (w), 1168 (s), 1152 (s), 1101 (m), 1065 (w), 951 (s), 926 (m), 820 (m), 761 (m), 740 (s). LC-MS: tR = 3.52 min, m/z 387.03 ([M + H]+). HRMS: m/z calc. for [C18H23N6O2 32S]+ ([M + H]+) 387.1603, found 387.1594, ∆ = −2.3 ppm. Experimental 225 6-(3-((2-Ethylpiperidin-1-yl)sulfonyl)phenyl)-3-methyl-[1,2,4]triazolo [4,3-b]pyridazine (132a) 8 9 10 12 5 4 N N NN 1 S N 16 15 14 19 O O Obtained using General Procedure B above. Purification by column chromatography using KP-NH silica afforded the title compound (43 mg, 0.122 mmol, 12 % over 3 steps) as a white solid, m.p. 160.9 – 161.3 ◦C (ethyl acetate / hexane / methanol). 1H NMR (400 MHz, CDCl3): δ /ppm = 8.44 (s, 1 H, H-12), 8.29 – 8.10 (m, 2 H, H-4 and H-8), 8.00 (d, J = 7.7 Hz, 1 H, H-10), 7.68 (t, J = 7.7 Hz, 1 H, H-9), 7.57 (d, J = 9.7 Hz, 1 H, H-5), 3.99 (q, J = 7.1 Hz, 1 H, H-17), 3.79 (dd, J = 14.0 Hz and 4.2 Hz, 1 H, H-13), 3.03 (td, J = 14.0 Hz and 2.5 Hz, 1 H, H-13), 2.87 (s, 3 H, H-1), 1.78 – 1.14 (m, 8 H, H-14, H-15, H-16 and H-18), 0.88 (t, J = 7.4 Hz, 3 H, H-19). 13C NMR (100 MHz, CDCl3): δ /ppm = 223.24 (C-6), 152.04 (C-2), 143.52 (C-3), 135.59 (C-11), 130.68 (CH, CAr), 130.11 (CH, CAr), 129.03 (CH, CAr), 125.71 (CH, CAr), 125.62 (CH, C-4), 118.43 (CH, C-5), 115.59 (C-7), 54.95 (CH, C-17), 40.89 (CH2, C-13), 27.33 (CH2, C-16), 24.73 (CH2, C-14, C-15 or C-18), 22.63 (CH2, C-14, C-15 or C-18), 18.46 (CH2, C-14, C-15 or C-18), 11.15 (CH3, C-19), 9.97 (CH3, C-1). IR (neat): ν /cm−1 = 2933 (w), 2166 (w), 1980 (w), 1546 (w), 1522 (w), 1472 (w), 1389 (m), 1332 (s), 1315 (s), 1195 (w), 1154 (s), 1104 (m), 1036 (m), 994 (m), 947 (s), 913 (m), 838 (m), 804 (s), 761 (m), 737 (s), 689 (s). LC-MS: tR = 4.68 min, m/z 386.02 ([M + H]+). HRMS: m/z calc. for [C19H24N5O2 32S]+ ([M + H]+) 386.1651, found 386.1641, ∆ = −2.6 ppm. Microanalysis: calc. (found) for C19H23N5O2S C 59.20% (59.39%), H 6.01% (5.97%), N 18.17% (18.01%). 226 Experimental 2-((3-(3-Methyl-[1,2,4]triazolo[4,3-b]pyridazin-6-yl)phenyl)sulfonyl)- 1,2,3,4-tetrahydroisoquinoline (132b) 9 10 128 5 4 N N NN 1 S N 21 14 16 17 18 19O O Obtained using General Procedure B above. Purification by column chromatography and subsequent recrystallisation from ethyl acetate / hexane afforded the title compound (15 mg, 0.037 mmol, 4 % over 3 steps) as a white solid, m.p. 222.9 – 224.5 ◦C (ethyl acetate / hexane). 1H NMR (500 MHz, CDCl3): δ /ppm = 8.40 (dd, J = 1.9 Hz and 1.6 Hz, 1 H, H-12), 8.22 (ddd, J = 7.8, 1.9 Hz and 1.1 Hz, 1 H, H-8), 8.19 (d, J = 9.7 Hz, 1 H, H-4), 8.01 (ddd, J = 7.8, 1.6 Hz and 1.1 Hz, 1 H, H-10), 7.73 (t, J = 7.8 Hz, 1 H, H-9), 7.51 (d, J = 9.7 Hz, 1 H, H-5), 7.16 – 7.12 (m, 2 H, HAr), 7.10 – 7.01 (m, 2 H, HAr), 4.38 (s, 2 H, H-21), 3.49 (t, J = 5.9 Hz, 2 H, H-13), 2.93 (t, J = 6.0 Hz, 2 H, H-14), 2.89 (s, 3 H, H-1). 13C NMR (126 MHz, CDCl3): δ /ppm = 151.76 (C-6), 147.65 (C-2), 143.22 (C-3), 138.42 (C-7 or C-11), 135.71 (C-7 or C-11), 132.90 (C-15), 131.31 (CH, CAr), 131.24 (C-20), 130.13 (CH, CAr), 129.52 (CH, CAr), 128.91 (CH, C-14), 126.98 (CH, C-18), 126.51 (CH, C-19), 126.28 (CH, C-17), 126.20 (CH, CAr), 125.60 (CH, C-4), 118.27 (CH, C-5), 47.51 (CH2, C-21), 43.70 (CH2, C-13), 28.61 (CH2, C-14), 9.88 (CH3, C-1). IR (neat): ν /cm−1 = 3062 (w), 2161 (w), 1553 (w), 1524 (w), 1465 (w), 1397 (w), 1373 (w), 1356 (m), 1335 (s), 1271 (w), 1164 (s), 1122 (m), 1091 (m), 1073 (m), 1020 (m), 995 (w), 957 (m), 926 (m), 835 (m), 796 (m), 759 (s), 742 (s), 723 (s), 686 (s), 675 (m), 658 (m). LC-MS: tR = 4.66 min, m/z 406.00 ([M + H]+). HRMS: m/z calc. for [C21H20N5O2 32S]+ ([M + H]+) 406.1338, found 406.1339, ∆ = 0.2 ppm. Experimental 227 3-Methyl-6-(3-(piperidin-1-ylsulfonyl)phenyl)-[1,2,4]triazolo[4,3-b] pyridazine (132c) 4 5 N N NN 1 8 9 10 12 S N O O 15 14 Obtained using General Procedure B above. Purification by column chromatography afforded the title compound (3.5 mg, 0.01 mmol, 1 % over 3 steps) as a white solid. 1H NMR (500 MHz, CDCl3): δ /ppm = 8.34 (dd, J = 1.8 Hz and 0.6 Hz, 1 H, H-12), 8.24 (ddd, J = 7.8 Hz, 1.8 Hz and 1.1 Hz, 1 H, H-4), 8.20 (d, J = 9.7 Hz, 1 H, H8 or H-10), 7.93 (ddd, J = 7.8 Hz, 1.8 Hz and 1.1 Hz, 1 H, H-5), 7.74 (td, J = 7.8 Hz and 0.6 Hz, 1 H, H-9), 7.57 (d, J = 9.7 Hz, 1 H, H8 or H-10), 3.13 – 3.03 (m, 4 H, H-13), 2.90 (s, 3 H, H-1), 1.73 – 1.63 (m, 4 H, H-14), 1.52 – 1.38 (m, 2 H, H-15). 13C NMR (126 MHz, CDCl3): δ /ppm = 151.88 (C-6), 147.64 (C-2), 143.23 (C-3), 138.04 (C-7 or C-11), 135.57 (C-7 or C-11), 131.07 (CH, CAr), 130.00 (CH, CAr), 129.59 (CH, CAr), 126.23 (CH, CAr), 125.60 (CH, C-4), 118.29 (CH, C-5), 46.96 (CH2, C-13), 25.15 (CH2, C-14), 23.45 (CH2, C-15), 9.88 (CH3, C-1). LC-MS: tR = 4.49 min, m/z 358.04 ([M + H] +). HRMS: m/z calc. for [C17H20N5O2 32S]+ ([M + H]+) 358.1338, found 358.1352, ∆ = −3.9 ppm. 228 Experimental Frontal Affinity Chromatography Pierce Streptavidin Ultralink resin (P/N PL-53113) was obtained from Fisher Scientific. Biotinylated BRD9 and CECR2 were kindly provided by the Structural Genomics Con- sortium, University of Oxford. Upchurch 1 mm×20 mm 15 µL microbore HPLC guard column (P/N C-128) was obtained from Kinesis Ltd. Preparation of Immobilised Enzymes The proteins were supplied in individual vials each containing 10 µL of 0.5 mg/mL protein suspension in aqueous buffer. The contents of 16 vials were combined, and an additional 16×10 µL was used to rinse the vials. A suspension of streptavidin-functionalised acry- lamide resin (60 µL) was added, and the mixture shaken at 4 ◦C for 1 hour. The mixture was then loaded into a 15 µL column using a syringe, with occasional packing using the HPLC pump. FAC-UV / FAC-MS experiments FAC assays were run on a modified Agilent 1100 HPLC system with a 100 µL injection syringe, a 600 µL storage loop and 100 mM ammonium acetate (when the MS was in use), or 100 mM phosphate-buffered saline (PBS) solution (for UV-only experiments) was used as the eluent. Two wavelengths were monitored simultaneously: 220 nm (DMSO) and 254 nm (analyte) using a UV Diode Array Detector. The output from the HPLC was combined with a stream of 0.1 % formic acid in methanol (200 µL/min) and directed into an Expression CMS mass spectrometer (Advion). The CMS was used in positive ESI mode, with selective ion monitoring (SIM, span 1 AMU, dwell time 500 ms). Stock solutions of compounds were prepared at a concentration of 50 mM in DMSO and stored at −20 ◦C. The HPLC system was set to inject 600 µL of 4 µM analyte at 30 µL/min for 120 minutes. References and Notes 1. Szostak, J. W. Chem. Rev. 1997, 97, 347–348, doi: 10.1021/cr9700080. 2. Nefzi, A.; Ostresh, J. M.; Houghten, R. A. Chem. Rev. 1997, 97, 449–472, doi: 10.1021/cr960010b. 3. Newman, D. J.; Cragg, G. M. J. Nat. Prod. 2007, 70, 461–77, doi: 10 . 1021 / np068054v. 4. Feher, M.; Schmidt, J. J. Chem. Inf. Comput. Sci. 2003, 43, 218–227, doi: 10.1021/ ci0200467. 5. Spring, D. R. Org. Biomol. Chem 2003, 1, 3867–3870, doi: 10.1039/b310752n. 6. Schmink, J. R.; Bellomo, A.; Berritt, S. Aldrichim. Acta 2013, 46, 71–80. 7. Accelerating chemical process development at GSK. Radleys Press Release Feb. 2005, http://www.radleys.co.uk/pages/press/accelerating_chemical.html. 8. Sanderson, K. Bloggers put chemical reactions through the replication mill. Nature News Jan. 2013, http://www.nature.com/news/bloggers-put-chemical-reactions- through-the-replication-mill-1.12262. 9. Bosch, C. Nobel Lecture: The development of the chemical high pressure method during the establishment of the new ammonia industry. 1932. 10. Phillips, P. Manufacture of sulphuric acid. British Patent, 6096, 1831. 11. Stankiewicz, A. I.; Moulijn, J. A. Process intensification: transforming chemical engineering. Chem. Eng. Progress 2000, 96, 22–34. 12. Ramshaw, C. The Incentive for Process Intensification In 1st International Confer- ence on Process Intensification for the Chemical Industry, BHR Group: London, UK, 1995. 13. Nagaki, A.; Imai, K.; Kim, H.; Yoshida, J.-i. RSC Adv. 2011, 1, 758–760, doi: 10.1039/c1ra00377a. 14. Bogdan, A. R.; Poe, S. L.; Kubis, D. C.; Broadwater, S. J.; McQuade, D. T. Angew. Chem. Int. Ed. 2009, 48, 8547–8550, doi: 10.1002/anie.200903055. 15. Ley, S. V.; Ladlow, M.; Vickerstaffe, E., The Use of Polymer-Assisted Solution-Phase Synthesis and Automation for the High-Throughput Preparation of Biologically Active Compounds. In Exploiting Chemical Diversity for Drug Discovery, Bartlett, P. 229 230 References and Notes A., Entzeroth, M., Eds.; The Royal Society of Chemistry: 2006; Chapter 1, pp 3–32, doi: 10.1039/9781847552556-00001. 16. Brocklehurst, C. E.; Lehmann, H.; La Vecchia, L. Org. Process Res. Dev. 2011, 15, 1447–1453, doi: 10.1021/op200055r. 17. Audiger, L.; Watts, K.; Elmore, S. C.; Robinson, R. I.; Wirth, T. ChemSusChem 2012, 5, 257–260, doi: 10.1002/cssc.201100372. 18. Brodmann, T.; Koos, P.; Metzger, A.; Knochel, P.; Ley, S. V. Org. Process Res. Dev. 2012, 16, 1102–1113, doi: 10.1021/op200275d. 19. Baumann, M.; Baxendale, I. R.; Ley, S. V.; Nikbin, N.; Smith, C. D.; Tierney, J. P. Org. Biomol. Chem. 2008, 6, 1577–1586, doi: 10.1039/b801631n. 20. Künzle, N.; Solèr, J.-W.; Baiker, A. Catal. Today 2003, 79U˝-80, 503–509, doi: 10.1016/S0920-5861(03)00075-0. 21. Johnson, M. D.; May, S. A.; Calvin, J. R.; Remacle, J.; Stout, J. R.; Diseroad, W. D.; Zaborenko, N.; Haeberle, B. D.; Sun, W.-M.; Miller, M. T.; Brennan, J. Org. Process Res. Dev. 2012, 16, 1017–1038, doi: 10.1021/op200362h. 22. Elvira, K. S.; Casadevall i Solvas, X.; Wootton, R. C. R.; DeMello, A. J. Nature Chem. 2013, 5, 905–915, doi: 10.1038/nchem.1753. 23. Wegner, J.; Ceylan, S.; Kirschning, A. Adv. Synth. Catal. 2012, 354, 17–57, doi: 10.1002/adsc.201100584. 24. Baumann, M.; Baxendale, I. R.; Ley, S. V. Mol. Divers. 2011, 15, 613–630, doi: 10.1007/s11030-010-9282-1. 25. Ley, S. V.; Baxendale, I. R.; Bream, R. N.; Jackson, P. S.; Leach, A. G.; Longbottom, D. A.; Nesi, M.; Scott, J. S.; Storer, R. I.; Taylor, S. J. J. Chem. Soc. Perkin Trans. 1 2000, 3815–4195, doi: 10.1039/b006588i. 26. Wasik, B. In the Programmable World, All Our Objects Will Act as One. Wired USA May 2013, http://www.wired.com/2013/05/internet-of-things-2/. 27. Coughlan, S. Curriculum changes ‘to catch up with world’s best’. BBC News July 2013, http://www.bbc.co.uk/news/education-23222068. 28. Raspberry Pi Foundation. Website: http://www.raspberrypi.org/. 29. Ingham, R. J.; Riva, E.; Nikbin, N.; Baxendale, I. R.; Ley, S. V. Org. Lett. 2012, 14, 3920–3923, doi: 10.1021/ol301673q. 30. Saczewski, J.; Paluchowska, A.; Klenc, J.; Raux, E.; Barnes, S.; Sullivan, S.; Duszyn- ska, B.; Bojarski, A. J.; Strekowski, L. J. Heterocycl. Chem. 2009, 46, 1259–1265, doi: 10.1002/jhet.236. 31. El-Deeb, I. M.; Lee, S. H. Bioorg. Med. Chem. 2010, 18, 3860–3874, doi: 10.1016/j. bmc.2010.04.037. 32. Stanetty, P.; Hattinger, G.; Schnürch, M.; Mihovilovic, M. D. J. Org. Chem. 2005, 70, 5215–5220, doi: 10.1021/jo0505223. References and Notes 231 33. Zimmermann, J. Pyrimidine derivatives and processes for the preparation thereof. US Patent, 5521184, 1996. 34. Agarwal, A.; Srivastava, K.; Puri, S. K.; Chauhan, P. M. S. Bioorg. Med. Chem. 2005, 13, 4645–4650, doi: 10.1016/j.bmc.2005.04.061. 35. Wang, X.; Chakrabarti, P. P.; Ognyanov, V. I.; Pettus, L. H.; Tamir, R.; Tan, H.; Tang, P.; Treanor, J. J. S.; Gavva, N. R.; Norman, M. H. Bioorg. Med. Chem. Lett. 2007, 17, 6539–6545, doi: 10.1016/j.bmcl.2007.09.080. 36. Hopkin, M. D.; Baxendale, I. R.; Ley, S. V. Org. Biomol. Chem. 2013, 11, 1822– 1839, doi: 10.1039/c2ob27002a. 37. Almario Garcia, A.; Ando, R.; Aritomo, K.; Frost, J. R.; Li, A. T.; Shoda, A.; Uehara, F.; Watanabe, K. Preparation of 2-(arylalkylamino)pyrimidones and 2- (heteroarylalkylamino)pyrimidones for preventive and/or therapeutic treatment of a neurodegenerative disease caused by abnormal activity of GSK3β . World Patent, 2001070727, 2001. 38. Masquelin, T.; Sprenger, D.; Baer, R.; Gerber, F.; Mercadal, Y. Helv. Chim. Acta 1998, 81, 646–660, doi: 10.1002/hlca.19980810315. 39. Gayo, L. M.; Suto, M. J. Tetrahedron Lett. 1997, 38, 211–214, doi: 10.1016/S0040- 4039(96)02256-3. 40. Baekeland, L. H. J. Ind. Eng. Chem. 1909, 1, 149–161, doi: 10.1021/ie50003a004. 41. Adams, B. A.; Holmes, E. L. Improvements in and relating to the removal of components or constituents from liquids by adsorption or absorption. British Patent, 450308, 1936. 42. Adams, B. A.; Holmes, E. L. Improvements in and relating to the treatment of liquids and gases by absorption and in particular to treatment of liquids for the removal or exchange of anions. British Patent, 450309, 1936. 43. Herr, D. S. Ind. Eng. Chem. 1945, 37, 631–634, doi: 10.1021/ie50427a011. 44. Merrifield, R. B. J. Am. Chem. Soc. 1963, 85, 2149–2154, doi: 10.1021/ja00897a025. 45. See references within Akelah, A.; Sherrington, D. C. Chem. Rev. 1981, 81, 557–587, doi: 10.1021/cr00046a003. 46. Baxendale, I. R.; Ley, S. V. Bioorg. Med. Chem. Lett. 2000, 10, 1983–1986, doi: 10/d7jxzq. 47. Ley, S. V.; Massi, A. J. Comb. Chem. 2000, 2, 104–107, doi: 10.1021/cc9900697. 48. Habermann, J; Ley, S. V.; Scott, J. S. J. Chem. Soc. Perkin Trans. 1 1999, 1253–1255, doi: 10.1039/a901802f. 49. Hinzen, B.; Ley, S. V. J. Chem. Soc. Perkin Trans. 1 1997, 1907–1908, doi: 10.1039/ A703461J. 50. Vickerstaffe, E.; Warrington, B.; Ladlow, M.; Ley, S. J. Comb. Chem. 2004, 6, 332– 339, doi: 10.1021/cc049977g. 232 References and Notes 51. Baxendale, I. R.; Ley, S. V.; Mansfield, A. C.; Smith, C. D. Angew. Chem. Int. Ed. 2009, 48, 4017–4021, doi: 10.1002/anie.200900970. 52. Kolb, H. C.; Finn, M. G.; Sharpless, K. B. Angew. Chem. Int. Ed. 2001, 40, 2004– 2021, doi: 10/br9nj7. 53. Sherrington, D. J. Assoc. Lab. Autom. 2000, 5, 75–78, doi: 10 . 1016 / S1535 - 5535(04)00066-8. 54. Svec, F.; Fréchet, J. M. J. Ind. Eng. Chem. Res. 1999, 38, 34–48, doi: 10.1021/ ie970598s. 55. Svec, F.; Fréchet, J. M. J. Anal. Chem. 1992, 64, 820–822, doi: 10.1021/ac00031a022. 56. Petro, M.; Svec, F.; Fréchet, J. M. J. Biotechnol. Bioeng. 1996, 49, 355–363, doi: 10.1002/(SICI)1097-0290(19960220)49:4<355::AID-BIT1>3.0.CO;2-O. 57. Altava, B.; Burguete, M. I.; Fraile, J. M.; García, J. I.; Luis, S. V.; Mayoral, J. A.; Vicent, M. J. Angew. Chem. Int. Ed. 2000, 39, 1503–1506, doi: 10/bwj6f5. 58. Krause, J. O.; Lubbad, S. H.; Nuyken, O.; Buchmeiser, M. R. Macromol. Rapid Commun. 2003, 24, 875–878, doi: 10.1002/marc.200300024. 59. Solodenko, W.; Wen, H.; Leue, S.; Stuhlmann, F.; Sourkouni-Argirusi, G.; Jas, G.; Schönfeld, H.; Kunz, U.; Kirschning, A. Eur. J. Org. Chem. 2004, 2004, 3601–3610, doi: 10.1002/ejoc.200400194. 60. Burguete, M. I.; Cornejo, A.; García-Verdugo, E.; Gil, M. J.; Luis, S. V.; Mayoral, J. A.; Martínez-Merino, V.; Sokolova, M. J. Org. Chem. 2007, 72, 4344–4350, doi: 10.1021/jo070119r. 61. Baumann, M.; Baxendale, I. R.; Ley, S. V.; Nikbin, N.; Smith, C. D. Org. Biomol. Chem. 2008, 6, 1587–1593, doi: 10.1039/b801634h. 62. Roper, K. A. Synthesis of Heterocyclic Molecules using Flow and Microwave Technologies. Master’s Thesis, University of Cambridge, 2008. 63. Peters, E. C.; Petro, M.; Svec, F.; Fréchet, J. M. J. Anal. Chem. 1997, 69, 3646–3649, doi: 10.1021/ac970377w. 64. Sherrington, D. C., Preparation, Functionalisation and Characteristics of Polymer Supports. In Polymer Supported Reactions in Organic Synthesis, Hodge, P., Sher- rington, D. C., Eds., 1st ed.; Wiley: Chichester, 1980; Chapter 1, pp 1–82. 65. Olah, G. A.; Burrichter, A.; Rasul, G.; Prakash, G. K. S. J. Am. Chem. Soc. 1997, 119, 12929–12933, doi: 10.1021/ja972553o. 66. Hennel, J. W.; Klinowski, J., Magic-Angle Spinning: a Historical Perspective. In New Techniques in Solid-State NMR, Klinowski, J., Ed.; Topics in Current Chemistry, Vol. 246; Springer Berlin Heidelberg: 2005, pp 1–14, doi: 10.1007/b98646, http: //dx.doi.org/10.1007/b98646. 67. Carberry, J. J.; Bretton, R. H. Axial dispersion of mass in flow through fixed beds. AIChE J. 1958, 4, 367–375, doi: 10.1002/aic.690040327. 68. Liebeskind, L. S.; Srogl, J. Org. Lett. 2002, 4, 979–981, doi: 10.1021/ol0200091. References and Notes 233 69. Lengar, A.; Kappe, C. O. Org. Lett. 2004, 6, 771–774, doi: 10.1021/ol036496h. 70. Dunn, P. J. Chem. Soc. Rev. 2012, 41, 1452–1461, doi: 10.1039/c1cs15041c. 71. O’Boyle, N. Python - the scripting language of chemistry., 2008, http://baoilleach. blogspot.co.uk/2008/03/python-scripting-language-of-chemistry.html. 72. Personal communication from Vapourtec Ltd. 73. Zimmermann, J. Pyrimidin derivatives and process for their preparation. EU Patent, 0564409, 1993. 74. Zimmermann, J.; Sutter, B.; Bürger, H. M. Crystal modification of a N-phenyl-2- pyrimidineamine derivative, processes for its manufacture and its use. US Patent, 6894051, 2005. 75. Buchdunger, E.; Capdeville, R.; Demetri, G. D.; Dimitrijevic, S.; Druker, B. J.; Fletcher, J. A.; Heinrich, M. C.; Joensuu, H.; Silberman, S. L.; Tuveson, D. Treatment of gastrointestinal stromal tumors. US Patent, 6958335, 2005. 76. Szczepek, W.; Luniewski, W.; Kaczmarek, L.; Samson-Lazinska, D.; Szelejew- ski, W.; Skarzynski, M. A process for preparation of imatinib base. World Patent, 2006071130, 2006. 77. Loiseleur, O.; Kaufmann, D.; Abel, S.; Buerger, H. M.; Meisenbach, M.; Schmitz, B.; Sedelmeier, G. N-phenyl-2-pyrimidine-amine derivatives. World Patent, 2003066613, 2003. 78. Kankan, R. N.; Rao, D. R. A process of preparing imatinib. World Patent, 2004074502, 2004. 79. Kompella, A.; Bhujanga Rao, A. K. S.; Venkaiah Chowdary, N.; Srinivas, R. Process for the preparation of the anti-cancer drug imatinib and its analogues. World Patent, 2004108699, 2004. 80. Szakács, Z.; Béni, S.; Varga, Z.; Orfi, L.; Kéri, G.; Noszál, B. J. Med. Chem. 2005, 48, 249–255, doi: 10.1021/jm049546c. 81. Kompella, A. K.; Rachakonda, S.; Gampa, V. G. K.; Bhujanga Rao, A. K. S.; Nannapaneni, V. C. Processes for the preparation of imatinib base and intermediates thereof. World Patent, 2013035102, 2013. 82. Ma Wang, Z.; Zunyuan, W.; Zhen, M.; Zhengrong, S. Process for preparing N- phenyl-2-pyrimidyl amine derivative. Chinese Patent, 1900073, 2007. 83. Xing, L.; Xungui, H.; Wang, Y.; Bekhazi, M.; Krivonos, S.; Danon, E. Imatinib production process. World Patent, 2008135980, 2008. 84. Macdonald, P.; Rossetto, P. Process for the preparation of imatinib. World Patent, 2008051597, 2008. 85. Anli, H.; Xing, L.; Zelikovitch, L.; Kaspi, J. Process for preparing Imatinib. US Patent, 7507821, 2009. 86. Maiti, D.; Fors, B. P.; Henderson, J. L.; Nakamura, Y.; Buchwald, S. L. Chem. Sci. 2011, 2, 57–68, doi: 10.1039/c0sc00330a. 234 References and Notes 87. Liu, Y.-F.; Wang, C.-L.; Bai, Y.-J.; Han, N.; Jiao, J.-P.; Qi, X.-L. Org. Process Res. Dev. 2008, 12, 490–495, doi: 10.1021/op700270n. 88. Leonetti, F.; Capaldi, C.; Carotti, A. Tetrahedron Lett. 2007, 48, 3455–3458, doi: 10.1016/j.tetlet.2007.03.033. 89. Lange, H.; Carter, C. F.; Hopkin, M. D.; Burke, A.; Goode, J. G.; Baxendale, I. R.; Ley, S. V. Chem. Sci. 2011, 2, 765–769, doi: 10.1039/c0sc00603c. 90. Sahoo, H. R.; Kralj, J. G.; Jensen, K. F. Angew. Chem. Int. Ed. 2007, 46, 5704–5708, doi: 10.1002/anie.200701434. 91. O’Brien, A. G.; Horváth, Z.; Lévesque, F.; Lee, J. W.; Seidel-Morgenstern, A.; Seeberger, P. H. Angew. Chem. Int. Ed. 2012, 51, 7028–7030, doi: 10.1002/anie. 201202795. 92. O’Brien, M.; Koos, P.; Browne, D. L.; Ley, S. V. Org. Biomol. Chem. 2012, 10, 7031–7036, doi: 10.1039/c2ob25912e. 93. Rubacha, M.; Rattan, A. K.; Hosselet, S. C. J. Lab. Autom. 2011, 16, 90–98, doi: 10.1016/j.jala.2009.01.002. 94. IDBS E-Workbook Suite, IDBS Business Solutions Ltd. Website: http://www.idbs. com/en/platform-products/e-workbook/. 95. King, A. Waving goodbye to the paper lab book. Chemistry World Nov. 2011, 46–49. 96. King, A. Out on a LIM. Chemistry World Feb. 2014, http : / / www . rsc . org / chemistryworld/2014/02/lims-lab-information-management. 97. Lab Information Management Systems (LIMS), Thermo Fisher Scientific Ltd. Web- site: http://www.thermoscientific.com/en/products/lab-information-management- systems-lims.html. 98. Delaney, N. F.; Rojas Echenique, J. I.; Marx, C. J. J. Lab. Autom. 2013, 18, 171–177, doi: 10.1177/2211068212460237. 99. Peak Analysis and Automation Ltd. (PAA). Website: http://www.paa-automation. com/. 100. PerkinElmer Integration Solutions. Website: http://www.perkinelmer.com/Catalog/ Category/ID/OnPoint%20Integration%20Solutions. 101. infoteam Software AG. Website: http://infoteam.de/. 102. Standards in Laboratory Automation (SiLA). Website: http://www.sila-standard. org/. 103. Bär, H.; Hochstrasser, R.; Papenfub, B. J. Lab. Autom. 2012, 17, 86–95, doi: 10. 1177/2211068211424550. 104. McMullen, J. P.; Stone, M. T.; Buchwald, S. L.; Jensen, K. F. Angew. Chem. Int. Ed. 2010, 49, 7076–7080, doi: 10.1002/anie.201002590. 105. McMullen, J. P.; Jensen, K. F. Org. Process Res. Dev. 2010, 14, 1169–1176, doi: 10.1021/op100123e. References and Notes 235 106. National Instruments LabVIEW. Website: http://www.ni.com/labview/ni-labview/. 107. Uniqsis Ltd. Website: http://www.uniqsis.com/. 108. Syrris Ltd. Website: http://www.syrris.com/. 109. OpenAPC (Open Advanced Process Control). Website: http://www.openapc.com/. 110. MathWorks MATLAB. Website: http://www.mathworks.co.uk/products/matlab/. 111. Parrott, A. J.; Bourne, R. A.; Akien, G. R.; Irvine, D. J.; Poliakoff, M. Angew. Chem. Int. Ed. 2011, 50, 3788–3792, doi: 10.1002/anie.201100412. 112. Smith, C. J.; Nikbin, N.; Ley, S. V.; Lange, H.; Baxendale, I. R. Org. Biomol. Chem. 2011, 9, 1938–1947, doi: 10.1039/c0ob00815j. 113. Moore, J. S.; Jensen, K. F. Org. Process Res. Dev. 2012, 16, 1409–1415, doi: 10. 1021/op300099x. 114. Rueping, M.; Bootwicha, T.; Sugiono, E. Beilstein J. Org. Chem. 2012, 8, 300–307, doi: 10.3762/bjoc.8.32. 115. Python Programming Language. Website: http://www.python.org/. 116. Wilson, J. A. J. Benefits Of Open Source Code., 2013, http://oss-watch.ac.uk/ resources/whoneedssource. 117. SciPy: open-source software for mathematics, science and engineering. Website: http://www.scipy.org. 118. Experimental Flux reactor developed by Syrris Ltd. 119. Hook, B. D.; Dohle, W.; Hirst, P. R.; Pickworth, M.; Berry, M. B.; Booker-Milburn, K. I. J. Org. Chem. 2005, 70, 7558–7564, doi: 10.1021/jo050705p. 120. Vapourtec UV-150 Photochemical Reactor. http://www.vapourtec.co.uk/products/ photochemicalreactor. 121. For example, the FlowSYNTH reactor from Milestone Inc. Website: http://www. milestonesci.com/. 122. User Manual for VICI Microelectric Actuator. http://www.vici.com/support/tn/ tn415.pdf. 123. A wide range of remote desktop tools are available, including: VNC (such as Re- alVNC, http://www.realvnc.com/), NoMachine NX (http://www.nomachine.com/), Microsoft Remote Desktop (Included with Microsoft Windows) and TeamViewer (http://www.teamviewer.com/). These products can allow portable devices to be used for the remote operation of existing control software packages. 124. Arduino Uno Image by SparkFun Electronics. Reproduced under a Creative Com- mons license from: http://en.wikipedia.org/wiki/File:Arduino_Uno_-_R3.jpg. 125. Microsoft .NET Gadgeteer and button Module image by Multichill. Reproduced under a Creative Commons license from: http : / / en . wikipedia . org / wiki / File : SytechMainboardAndGHIButton.png. 236 References and Notes 126. Raspberry Pi image by Jwrodgers, adapted under the Creative Commons Attribution - Share Alike License, from: http://en.wikipedia.org/wiki/File:RaspberryPi.jpg. 127. Intel Galileo image by Regi51. Reproduced under a Creative Commons license from: http://en.wikipedia.org/wiki/File:Embedded_World_2014_Intel_Galileo_01.jpg. 128. Arduino Uno Image by SparkFun Electronics. Reproduced under a Creative Com- mons license from: http://en.wikipedia.org/wiki/File:Arduino_Uno_-_R3.jpg. 129. Microsoft .NET Gadgeteer and button Module image by Multichill. Reproduced under a Creative Commons license from: http : / / en . wikipedia . org / wiki / File : SytechMainboardAndGHIButton.png. 130. Raspberry Pi image by Jwrodgers, adapted under the Creative Commons Attribution - Share Alike License, from: http://en.wikipedia.org/wiki/File:RaspberryPi.jpg. 131. Intel Galileo image by Regi51. Reproduced under a Creative Commons license from: http://en.wikipedia.org/wiki/File:Embedded_World_2014_Intel_Galileo_01.jpg. 132. Arduino prototyping platform. Website: http://www.arduino.cc/. 133. Microsoft Research .NET Gadgeteer prototyping tool. Website: http://research. microsoft.com/en-us/projects/gadgeteer/. 134. Intel Galileo Development Board. Website: http://www.intel.com/content/www/us/ en/intelligent-systems/galileo/galileo-overview.html. 135. Intel Edison Development Board. Website: http://www.intel.co.uk/content/www/uk/ en/do-it-yourself/edison.html. 136. Phidgets Single Board Computer (SBC), P/N 1073_0. Available from http://www. phidgets.com/. 137. Octopus, version 0.1. Ingham, R. 2014. http://www.github.com/richardingham/ octopus/. 138. Ousterhout, J. Why Threads are a Bad Idea (for most purposes) In USENIX Winter Technical Conference, Jan. 1996, http://www.cs.utah.edu/~regehr/research/ouster. pdf. 139. Erb, B. Concurrent Programming for Scalable Web Architectures. Diploma Thesis, Institute of Distributed Systems, Ulm University, Apr. 2012. 140. Dahl, R. node.js In JSConf, 2009, http://s3.amazonaws.com/four. livejournal/ 20091117/jsconf.pdf. 141. Twisted event-driven networking engine. Website: http://www.twistedmatrix.com/. 142. Open Babel: The Open Source Chemistry Toolbox. Website: http://www.openbabel. org/. 143. Python Data Analysis Library. Website: http://pandas.pydata.org/. 144. SimpleCV: Computer Vision platform using Python. Website: http://www.simplecv. org/. References and Notes 237 145. Syme, D.; Petricek, T.; Lomov, D. The F# Asynchronous Programming Model In Principles and Applications of Declarative Languages, ACM SIGPLAN: 2011, http://research.microsoft.com/apps/pubs/default.aspx?id=147194. 146. NumPy (Numerical Python). Website: http://www.numpy.org/. 147. Åström, K. J.; Murray, R. M., PID Control. In Feedback Systems: An Introduction for Scientists and Engineers; Princeton University Press: Princeton, N.J., 2012; Chapter 10, pp 293–314. 148. Ingham, R. J.; Battilocchio, C.; Hawkins, J. M.; Ley, S. V. Beilstein J. Org. Chem. 2014, 10, 641–652, doi: 10.3762/bjoc.10.56. 149. Carey, J. S.; Laffan, D.; Thomson, C.; Williams, M. T. Org. Biomol. Chem. 2006, 4, 2337–2347, doi: 10.1039/b602413k. 150. Dugger, R. W.; Ragan, J. A.; Ripin, D. H. B. Org. Process Res. Dev. 2005, 9, 253– 258, doi: 10.1021/op050021j. 151. Constable, D. J. C.; Dunn, P. J.; Hayler, J. D.; Humphrey, G. R.; Leazer, Jr., J. L.; Linderman, R. J.; Lorenz, K.; Manley, J.; Pearlman, B. a.; Wells, A.; Zaks, A.; Zhang, T. Y. Green Chem. 2007, 9, 411–420, doi: 10.1039/b703488c. 152. García-Álvarez, R.; Crochet, P.; Cadierno, V. Green Chem. 2013, 15, 46–66, doi: 10.1039/c2gc36534k. 153. Saaby, S.; Knudsen, K. R.; Ladlow, M.; Ley, S. V. Chem. Commun. 2005, 2909– 2911, doi: 10.1039/b504854k. 154. Battilocchio, C.; Hawkins, J. M.; Ley, S. V. Org. Lett. 2014, 16, 1060–1063, doi: 10.1021/ol403591c. 155. Battilocchio, C.; Hawkins, J. M.; Ley, S. V. Org. Lett. 2013, 15, 2278–2281, doi: 10.1021/ol400856g. 156. Chorghade, R.; Battilocchio, C.; Hawkins, J. M.; Ley, S. V. Org. Lett. 2013, 15, 5698–5701, doi: 10.1021/ol4027107. 157. Eichhorn, E.; Roduit, J.-P.; Shaw, N.; Heinzmann, K.; Kiener, A. Tetrahedron: Asymm. 1997, 8, 2533–2536, doi: 10/cp2p2d. 158. Piperazine-2-carboxamide (39) listed by Fluorochem (P/N 068934) at £233/g on 14th Apr 2013. 159. U.S. Food and Drug Administration (FDA), PAT Guidance; tech. rep.; 2004. 160. Fabry, D. C.; Sugiono, E.; Rueping, M. Isr. J. Chem. 2014, 54, 341–350, doi: 10. 1002/ijch.201300080. 161. Routh, M. W.; Swartz, P. A.; Denton, M. B. Anal. Chem. 1977, 49, 1422–1428, doi: 10.1021/ac50017a033. 162. Morgan, E.; Burton, K. W.; Nickless, G. Chemometr. Intel. Lab. 1990, 8, 97–107, doi: 10.1016/0169-7439(90)80127-R. 238 References and Notes 163. Schrijver, A., Theory of Linear and Integer Programming; Wiley-Blackwell: Chich- ester, UK, 1998. 164. Cyr, P.; Deng, S. T.; Hawkins, J. M.; Price, K. E. Org. Lett. 2013, 15, 4342–4345, doi: 10.1021/ol4018134. 165. Hogan, P. J.; Hopes, P. A.; Moss, W. O.; Robinson, G. E.; Patel, I. Org. Process Res. Dev. 2002, 6, 225–229, doi: 10.1021/op0101052. 166. Jamieson, C.; Congreve, M. S.; Emiabata-Smith, D. F.; Ley, S. V. Synlett 2000, 2000, 1603–1607, doi: 10.1055/s-2000-7917. 167. Ley, S. V.; Ingham, R. J.; O’Brien, M.; Browne, D. L. Beilstein J. Org. Chem. 2013, 9, 1051–1072, doi: 10.3762/bjoc.9.118. 168. Ingham, R. J. figshare 2013, doi: 10.6084/m9.figshare.881895. 169. Vaxelaire, C.; Winter, P.; Christmann, M. Angew. Chem. Int. Ed. 2011, 50, 3605– 3607, doi: 10.1002/anie.201100059. 170. Gothard, C. M.; Soh, S.; Gothard, N.; Kowalczyk, B.; Wei, Y.; Baytekin, B.; Grzy- bowski, B. Angew. Chem. Int. Ed. 2012, 51, 7922–7927, doi: 10 . 1002 / anie . 201202155. 171. Baxendale, I.; Schou, S.; Sedelmeier, J.; Ley, S. Chem. Eur. J. 2010, 16, 89–94, doi: 10.1002/chem.200902906. 172. Storer, R. I.; Takemoto, T.; Jackson, P. S.; Brown, D. S.; Baxendale, I. R.; Ley, S. V. Chem. Eur. J. 2004, 10, 2529–2547, doi: 10.1002/chem.200305669. 173. Müller, E.; Berger, R.; Blass, E.; Sluyts, D.; Pfennig, A., Liquid–Liquid Extraction. In Ullmann’s Encyclopedia of Industrial Chemistry, 7th ed.; Wiley-VCH Verlag GmbH & Co. KGaA: 2011, doi: 10.1002/14356007.b03_06.pub2. 174. Ripperger, S.; Gösele, W.; Alt, C.; Loewe, T., Filtration, 2. Equipment. In Ullmann’s Encyclopedia of Industrial Chemistry, 7th ed.; Wiley-VCH Verlag GmbH & Co. KGaA: 2011, doi: 10.1002/14356007.n11_n01.pub2. 175. Stichlmair, J., Distillation, 2. Equipment. In Ullmann’s Encyclopedia of Industrial Chemistry, 7th ed.; Wiley-VCH Verlag GmbH & Co. KGaA: 2011, doi: 10.1002/ 14356007.o08_o01. 176. Billet, R., Evaporation. In Ullmann’s Encyclopedia of Industrial Chemistry, 7th ed.; Wiley-VCH Verlag GmbH & Co. KGaA: 2011, doi: 10.1002/14356007.b03_03. 177. Hartman, R. L. Org. Process Res. Dev. 2012, 16, 870–887, doi: 10.1021/op200348t. 178. Koos, P.; Browne, D. L.; Ley, S. V. Green Process. Synth. 2012, 1, 11–18, doi: 10.1515/greenps-2011-0501. 179. Browne, D. L.; Deadman, B. J.; Ashe, R.; Baxendale, I. R.; Ley, S. V. Org. Process Res. Dev. 2011, 15, 693–697, doi: 10.1021/op2000223. 180. Adamo, A.; Heider, P. L.; Weeranoppanant, N.; Jensen, K. F. Ind. Eng. Chem. Res. 2013, 52, 10802–10808, doi: 10.1021/ie401180t. References and Notes 239 181. Syrris Asia FLLEX Module. Website: http : / / syrris . com/flow- products /asia - modules/asia-fllex. 182. Biotage universal phase separator. Website: http:/ /www.biotage.com/product- page/phase-separators. 183. Kashid, M. N.; Harshe, Y. M.; Agar, D. W. Ind. Eng. Chem. Res. 2007, 46, 8420– 8430, doi: 10.1021/ie070077x. 184. Aota, A.; Nonaka, M.; Hibara, A.; Kitamori, T. Angew. Chem. Int. Ed. 2007, 46, 878–880, doi: 10.1002/anie.200600122. 185. Deadman, B. J.; Battilocchio, C.; Sliwinski, E.; Ley, S. V. Green Chem. 2013, 15, 2050–2055, doi: 10.1039/c3gc40967h. 186. Borukhova, S.; Noël, T.; Metten, B.; de Vos, E.; Hessel, V. ChemSusChem 2013, 6, 2220–2225, doi: 10.1002/cssc.201300684. 187. Snead, D. R.; Jamison, T. F. Chem. Sci. 2013, 4, 2822–2827, doi: 10 . 1039 / c3sc50859e. 188. Battilocchio, C.; Baxendale, I. R.; Biava, M.; Kitching, M. O.; Ley, S. V. Org. Process Res. Dev. 2012, 16, 798–810, doi: 10.1021/op300084z. 189. Labbé-Jullié, C; Deschaintres, S; Gully, D; Le Fur, G; Kitabgi, P J. Pharmacol. Exp. Ther. 1994, 271, 267–276. 190. Battilocchio, C.; Deadman, B. J.; Nikbin, N.; Kitching, M. O.; Baxendale, I. R.; Ley, S. V. Chem. Eur. J. 2013, 19, 7917–7930, doi: 10.1002/chem.201300696. 191. Baxendale, I. R.; Cheung, S; Kitching, M. O.; Ley, S. V.; Shearman, J. W. Bioorg. Med. Chem. 2013, 21, 4378–4387, doi: 10.1016/j.bmc.2013.04.075. 192. Nagasawa, H. T.; Elberling, J. A.; Shirota, F. N. J. Med. Chem. 1973, 16, 823–826, doi: 10.1021/jm00265a016. 193. Pascal, R.; Taillades, J.; Commeyras, A. Tetrahedron 1980, 36, 2999–3008, doi: 10.1016/0040-4020(80)88025-2. 194. Paventi, M.; Chubb, F. L.; Edward, J. T. Can. J. Chem. 1987, 65, 2114–2117, doi: 10.1139/v87-351. 195. Ritter, J. J.; Minieri, P. P. J. Am. Chem. Soc. 1948, 70, 4045–4048, doi: 10.1021/ ja01192a022. 196. Pantano, M.; Formaggio, F.; Crisma, M.; Bonora, G. M.; Mammi, S.; Peggion, E.; Toniolo, C.; Boesten, W. H. J.; Broxterman, Q. B. Macromolecules 1993, 26, 1980– 1984, doi: 10.1021/ma00060a028. 197. Dummann, G.; Quittmann, U.; Gröschel, L.; Agar, D. W.; Wörz, O.; Morgenschweis, K. Catalysis Today 2003, 79-80, 433–439, doi: 10/frgnqr. 198. Calcinated Kieselguhr obtained from Sigma-Aldrich Co. 199. Activated charcoal powder obtained from Breckland Scientific Supplies Ltd. 240 References and Notes 200. Hu, D. X.; O’Brien, M.; Ley, S. V. Org. Lett. 2012, 14, 4246–4249, doi: 10.1021/ ol301930h. 201. Prat, D.; Pardigon, O.; Flemming, H.-W.; Letestu, S.; Ducandas, V.; Isnard, P.; Guntrum, E.; Senac, T.; Ruisseau, S.; Cruciani, P.; Hosek, P. Org. Process Res. Dev. 2013, 17, 1517–1525, doi: 10.1021/op4002565. 202. Control script for the 3-step 2-aminoadamantane-2-carboxylic acid synthesis plat- form: https://gist.github.com/richardingham/4ac997b5d720a4f7384f. 203. DIN 53170. Solvents for paints and varnishes – Determination of the evaporation rate. Deutsches Institut für Normung., http://www.din.de/. 204. Schmidt, E. Y.; Vasil’tsov, A. M.; Mikhaleva, A. I.; Zaitsev, A. B.; Afonin, A. V.; Toryashinova, D.-S. D.; Klyba, L. V.; Arndt, J.-D.; Henkelmann, J. ARKIVOC 2003, 35–44. 205. Mascia, S.; Heider, P. L.; Zhang, H.; Lakerveld, R.; Benyahia, B.; Barton, P. I.; Braatz, R. D.; Cooney, C. L.; Evans, J. M. B.; Jamison, T. F.; Jensen, K. F.; Myerson, A. S.; Trout, B. L. Angew. Chem. Int. Ed. 2013, 52, 12359–12363, doi: 10.1002/ anie.201305429. 206. Deadman, B. J. New Tools for Flow Chemistry and the Machine Assisted Synthesis of Pharmaceuticals. PhD Thesis, University of Cambridge, 2013. 207. Schiaffo, C. E.; Dussault, P. H. J. Org. Chem. 2008, 73, 4688–4690, doi: 10.1021/ jo800323x. 208. Sorge, A. R.; Turco, M.; Pilone, G.; Bagnasco, G. J. Propul. Power 2004, 20, 1069– 1075, doi: 10.2514/1.2490. 209. Djaballah, H. Chemical space, high throughput screening and the world of block- buster drugs. Drug Discov. World 2013, 14, 73–78. 210. Rees, D. C.; Congreve, M.; Murray, C. W.; Carr, R. Nat. Rev. Drug Disc. 2004, 3, 660–672, doi: 10.1038/nrd1467. 211. Zartler, D. E.; Swain, D. C.; Pearce, S. Fragment library design: the evolution of fragment-based lead discovery. Drug Discov. World 2012, 13, 19–24. 212. Jhoti, H.; Williams, G.; Rees, D. C.; Murray, C. W. Nat. Rev. Drug Disc. 2013, 12, 644–645, doi: 10.1038/nrd3926-c1. 213. Desai, B.; Dixon, K.; Farrant, E.; Feng, Q.; Gibson, K. R.; van Hoorn, W. P.; Mills, J.; Morgan, T.; Parry, D. M.; Ramjee, M. K.; Selway, C. N.; Tarver, G. J.; Whitlock, G.; Wright, A. G. J. Med. Chem. 2013, 56, 3033–3047, doi: 10.1021/jm400099d. 214. Schneider, G.; Lee, M.-L.; Stahl, M.; Schneider, P. J. Comput. Aided Mol. Des. 2000, 14, 487–494, doi: 10.1023/A:1008184403558. 215. Dial-a-Molecule EPSRC Grand Challenge Network. Website: http://www.dial-a- molecule.org/. 216. Schönherr, H.; Cernak, T. Angew. Chem. Int. Ed. 2013, 52, 12256–12267, doi: 10.1002/anie.201303207. References and Notes 241 217. Guetzoyan, L.; Nikbin, N.; Baxendale, I. R.; Ley, S. V. Chem. Sci. 2013, 4, 764–769, doi: 10.1039/c2sc21850j. 218. Filippakopoulos, P.; Knapp, S. FEBS Letters 2012, 586, 2692–2704, doi: 10.1016/j. febslet.2012.04.045. 219. Bromosporine, a chemical probe developed by the SGC. Biological data and synthe- sis scheme available at: http://www.thesgc.org/chemical-probes/bromosporine. 220. Kasai, K.-i.; Ishii, S.-i. J. Biochem. 1975, 77, 261–264. 221. Tateno, H.; Nakamura-Tsuruta, S.; Hirabayashi, J. Nat. Protoc. 2007, 2, 2529–2537, doi: 10.1038/nprot.2007.357. 222. Chan, N. W. C.; Lewis, D. F.; Rosner, P. J.; Kelly, M. A.; Schriemer, D. C. Anal. Biochem. 2003, 319, 1–12, doi: 10/cfzrjt. 223. Ng, W.; Dai, J.-R.; Slon-Usakiewicz, J. J.; Redden, P. R.; Pasternak, A.; Reid, N. J. Biomol. Screen. 2007, 12, 167–174, doi: 10.1177/1087057106297567. 224. Ng, E. S. M.; Chan, N. W. C.; Lewis, D. F.; Hindsgaul, O.; Schriemer, D. C. Nat. Protoc. 2007, 2, 1907–1917, doi: 10.1038/nprot.2007.262. 225. Calleri, E. et al. J. Med. Chem. 2010, 53, 3489–3501, doi: 10.1021/jm901691y. 226. Beckett, D.; Kovaleva, E.; Schatz, P. J. Protein Sci. 1999, 8, 921–929, doi: 10.1110/ ps.8.4.921. 227. Green, N. M. Methods Enzymol. 1990, 184, 51–67, doi: 10.1016/0076-6879(90) 84259-J. 228. Rossotti, H., The study of ionic equilibria: An introduction; Longman: London, 1978. 229. Lineweaver, H.; Burk, D. J. Am. Chem. Soc. 1934, 56, 658–666, doi: 10.1021/ ja01318a036. 230. Guetzoyan, L.; Ingham, R. J.; Nikbin, N.; Rossignol, J.; Wolling, M.; Baumert, M.; Burgess-Brown, N. A.; Strain-Damerell, C. M.; Shrestha, L.; Brennan, P. E.; Fedorov, O.; Knapp, S.; Ley, S. V. MedChemComm 2014, 5, 540–546, doi: 10.1039/ c4md00007b. 231. Kocevar, M.; Pozgan, F.; Polanc, S. Heterocycles 2001, 54, 1011–1019, doi: 10. 3987/COM-00-S(I)73. 232. Cox, J. M.; Edmondson, S. D.; Harper, B.; Weber, A. E. Preparation of aminopiperid- ines as dipeptidyl peptidase IV inhibitors for the treatment or prevention of diabetes. World Patent, 2006039325, 2006. 233. Sunder, S.; Peet, N. P. J. Heterocycl. Chem. 1980, 17, 1527–1529, doi: 10.1002/jhet. 5570170734. 234. Monge-Vega, A.; Aldana, I.; Ferández-Alvarez, E. J. Heterocycl. Chem. 1981, 18, 1533–1536, doi: 10.1002/jhet.5570180808. 242 References and Notes 235. Alazawe, S.; Elvidge, J. A. J. Chem. Soc. Perkin Trans. 1 1974, 696–698, doi: 10.1039/p19740000696. 236. Albrecht, B. K.; Harmange, J.-C.; Cote, A.; Taylor, A. M. Bromodomain inhibitors and uses thereof. World Patent, 2012174487, 2012. 237. Cugnon de Sevricourt, M.; Robba, M. J. Heterocycl. Chem. 1978, 15, 977–979, doi: 10.1002/jhet.5570150614. 238. Zaher, A. F. A.; Khalil, O. M.; Refaat, H. M. Med. Chem. Res. 2011, 21, 3146–3153, doi: 10.1007/s00044-011-9847-2. 239. Bourotte, M.; Delhomel, J.-F.; Dubernet, M.; Gouy, M.-H. Preparation of derivs. of 6-substituted triazolopyridazines as Rev-Erb agonists for treating inflammatory, circadian rhythm-related, or cardiometabolic disorders. World Patent, 2013045519, 2013. 240. Mizzoni, R. H.; Spoerri, P. E. J. Am. Chem. Soc. 1951, 73, 1873–1874, doi: 10.1021/ ja01148a537. 241. Li, A.-H.; Steinig, A. G.; Kleinberg, A.; Weng, Q.; Mulvihill, M. J.; Wang, J.; Chen, X.; Wang, T.; Dong, H.; Jin, M. Furo- and Thieno [3,2-c] Pyridines. US Patent, 20090197864, 2009. 242. Calderwood, D. J.; Bonafoux, D. F.; Burchat, A.; Ding, P.; Frank, K. E.; Hoe- mann, M. Z.; Mullen, K. D.; Davis, H. M. Novel triazolopyridazines. World Patent, 2009005675, 2009. 243. Bacque, E.; Nemecek, C.; Ugolini, A.; Wentzler, S. 6-(alkyl- or cycloalkyl-triazolo- pyridazine-sulfanyl) benzothiazole derivatives: preparation, application as medica- ments and use as met inhibitors. World Patent, 2011121223, 2011. 244. Dewdney, N. J.; Lou, Y.; Soth, M. Novel imidazo[1,2-a]pyridine and imidazo[1,2- b]pyridazine derivatives. World Patent, 2009077334, 2009. 245. Lucas, M. C.; Goldstein, D. M.; Hermann, J. C.; Kuglstatter, A.; Liu, W.; Luk, K. C.; Padilla, F.; Slade, M.; Villaseñor, A. G.; Wanner, J.; Xie, W.; Zhang, X.; Liao, C. J. Med. Chem. 2012, 55, 10414–10423, doi: 10.1021/jm301367c. 246. Structural Genomics Consortium, Unpublished results. 247. Billingsley, K.; Buchwald, S. L. J. Am. Chem. Soc. 2007, 129, 3358–3366, doi: 10.1021/ja068577p. 248. Nikbin, N. Unpublished results. 249. Newman, M. S.; Gildenhorn, H. L. J. Am. Chem. Soc. 1948, 70, 317–319, doi: 10.1021/ja01181a097. 250. Shioiri, T.; Ninomiya, K.; Yamada, S. J. Am. Chem. Soc. 1972, 94, 6203–6205, doi: 10.1021/ja00772a052. 251. Spano, M. M.; Bickler, J. R. Heterocyclic Amines Purification on a New FLASH Media - KP-NH. Biotage Application Note #40., 2003, http://www.biotage.com/ literature/download/1583_app_note_40.pdf. References and Notes 243 252. Suginome, M.; Ohmura, T., Alkylboronic acid derivative cross-coupling reactions. In Science of Synthesis: Cross Coupling and Heck-Type Reactions 1: C-C Cross Coupling using Organometallic Partners, Molander, G. A., Ed.; Thieme: Stuttgart, 2013; Chapter 1.1.1.2, pp 147–201. 253. Angew. Chem. Int. Ed. 2010, 49, 8300, doi: 10.1002/anie.201006286. 254. Suzuki, A. Angew. Chem. Int. Ed. 2011, 50, 6722–6737, doi: 10 . 1002 / anie . 201101379. 255. Chemler, S. R.; Trauner, D.; Danishefsky, S. J. Angew. Chem. Int. Ed. 2001, 40, 4544–4568, doi: 10/dcfn97. 256. Guram, A. S.; Milne, J. E.; Tedrow, J. S.; Walker, S. D., Alkylboronic Acid Cross- coupling Reactions. In Science of Synthesis: Cross Coupling and Heck-Type Reac- tions 1: C-C Cross Coupling using Organometallic Partners, Molander, G. A., Ed.; Thieme: Stuttgart, 2013; Chapter 1.1.1.1, pp 9–146. 257. Gillis, E. P.; Burke, M. D. J. Am. Chem. Soc. 2007, 129, 6716–6717, doi: 10.1021/ ja0716204. 258. Molander, G. A.; Canturk, B. Angew. Chem. Int. Ed. 2009, 48, 9240–9261, doi: 10.1002/anie.200904306. 259. Clayden, J.; Greeves, N.; Warren, S.; Wothers, P., Organometallic chemistry. In Organic Chemistry; Oxford University Press: Oxford, 2001; Chapter 48, pp 1311– 1343. 260. Cepanec, I., The Suzuki-Miyaura Reaction. In Synthesis of Biaryls; Elsevier: 2004; Chapter 5, pp 139–207, doi: 10.1016/B978-008044412-3/50006-3. 261. Billingsley, K. L.; Anderson, K. W.; Buchwald, S. L. Angew. Chem. Int. Ed. 2006, 45, 3484–3488, doi: 10.1002/anie.200600493. 262. Martin, R.; Buchwald, S. L. Acc. Chem. Res. 2008, 41, 1461–1473, doi: 10.1021/ ar800036s. 263. Strieter, E. R.; Buchwald, S. L. Angew. Chem. Int. Ed. 2006, 45, 925–938, doi: 10.1002/anie.200502927. 264. Miyaura, N. J. Organomet. Chem. 2002, 653, 54–57, doi: 10/ftq26v. 265. Wuts, P. G. M., Greene’s protective groups in organic synthesis, 4th ed.; Wiley- Interscience: Hoboken, N.J., 2007. 266. Itoh, T.; Sato, K.; Mase, T. Adv. Synth. Catal. 2004, 346, 1859–1867, doi: 10.1002/ adsc.200404159. 267. Guetzoyan, L. Unpublished results. 268. Compounds 112 and 113 are described in Rossignol, J. Design and synthesis of a series of bromodomain inhibitors; tech. rep.; University of Cambridge, 2012. 269. Malet-Sanz, L.; Madrzak, J.; Ley, S. V.; Baxendale, I. R. Org. Biomol. Chem. 2010, 8, 5324–5332, doi: 10.1039/c0ob00450b. 244 References and Notes 270. Ikemoto, N.; Liu, J.; Brands, K. M.; McNamara, J. M.; Reider, P. J. Tetrahedron 2003, 59, 1317–1325, doi: 10/fb8zp5. 271. Birum, G. H.; Jansen, R. F. Preparation of arylsulfonamides. US Patent, 4739116, 1988. 272. Reid, J. R.; Dufresne, R. F.; Chapman, J. J. Org. Synth. 1997, 74, 217–223, doi: 10.15227/orgsyn.074.0217. 273. Wolling, M. Unpublished results. 274. Ishiyama, T.; Murata, M.; Miyaura, N. J. Org. Chem. 1995, 60, 7508–7510, doi: 10.1021/jo00128a024. 275. Ishiyama, T.; Miyaura, N. J. Organomet. Chem. 2000, 611, 392–402, doi: 10.1016/ S0022-328X(00)00470-8. 276. CMPE product literature, Zeon Corporation. http://www.zeon.co.jp/business_e/ enterprise/spechemi/spechemi5-13.html. 277. Adamo, C.; Amatore, C.; Ciofini, I.; Jutand, A.; Lakmini, H. J. Am. Chem. Soc. 2006, 128, 6829–6836, doi: 10.1021/ja0569959. 278. Fürstner, A.; Seidel, G. Org. Lett. 2002, 4, 541–543, doi: 10.1021/ol0171463. 279. O’Brien, C. J.; Kantchev, E. A. B.; Valente, C.; Hadei, N.; Chass, G.; Lough, A.; Hopkinson, A. C.; Organ, M. G. Chem. Eur. J. 2006, 12, 4743–4748, doi: 10.1002/chem.200600251. 280. Kantchev, E. A. B.; O’Brien, C. J.; Organ, M. G. Angew. Chem. Int. Ed. 2007, 46, 2768–2813, doi: 10.1002/anie.200601663. 281. Matos, K.; Burkhardt, E. R. Chim. Oggi 2005, 23, 12–17. 282. Ishiyama, T.; Itoh, Y.; Kitano, T.; Miyaura, N. Tetrahedron Lett. 1997, 38, 3447– 3450, doi: 10.1016/S0040-4039(97)00642-4. 283. Glide, version 5.8. Schrödinger LLC, New York, NY, 2012. 284. Huang, D.; Rossini, E.; Steiner, S.; Caflisch, A. ChemMedChem 2014, 9, 573–579, doi: 10.1002/cmdc.201300156. 285. For example, the Accendo Conjure platform performs automated segmented-flow experiments. Website: http://www.accendocorporation.com/. 286. For example, Chemspeed Technologies AG produce a variety of automated synthesis platforms. Website: http://www.chemspeed.com/. 287. Braslau, R.; Rivera, F.; Tansakul, C. React. Funct. Polym. 2013, 73, 624–633, doi: 10.1016/j.reactfunctpolym.2013.01.009. 288. Lücking, U.; Cleve, A.; Haendler, B.; Faus, H.; Köhr, S.; Irlbacher, H. Sustituted (heteroarylmethyl) thiohydantoins as anticancer drugs. World Patent, 2011029537, 2011. 289. Schmidt, H.-W.; Koitz, G.; Junek, H. J. Het. Chem. 1987, 24, 1305–1307, doi: 10.1002/jhet.5570240514. References and Notes 245 290. Bonnet, M.; Flanagan, J. U.; Chan, D. A.; Lai, E. W.; Nguyen, P.; Giaccia, A. J.; Hay, M. P. Bioorg. Med. Chem. 2011, 19, 3347–3356, doi: 10.1016/j.bmc.2011.04.042. 291. Kumar, D.; Kommi, D. N.; Chopra, P.; Ansari, M. I.; Chakraborti, A. K. Eur. J. Org. Chem. 2012, 2012, 6407–6413, doi: 10.1002/ejoc.201200778. 292. Hernández, S.; Moreno, I.; SanMartin, R.; Gómez, G.; Herrero, M. T.; Domínguez, E. J. Org. Chem. 2010, 75, 434–441, doi: 10.1021/jo902257j. 293. Kamal, A.; Dastagiri, D.; Ramaiah, M. J.; Reddy, J. S.; Bharathi, E. V.; Reddy, M. K.; Sagar, M. V. P.; Reddy, T. L.; Pushpavalli, S. N. C. V. L.; Pal-Bhadra, M. Eur. J. Med. Chem. 2011, 46, 5817–5824, doi: 10.1016/j.ejmech.2011.09.039. 294. Liu, Y.; Bai, Y.; Zhang, J.; Li, Y.; Jiao, J.; Qi, X. Eur. J. Org. Chem. 2007, 2007, 6084–6088, doi: 10.1002/ejoc.200700577. 295. Leproult, E.; Barluenga, S.; Moras, D.; Wurtz, J.-M.; Winssinger, N. J. Med. Chem. 2011, 54, 1347–1355, doi: 10.1021/jm101396q. 296. Kagarlitskii, A.; Krichevskii, L.; Suvorov, B. Pharm. Chem. J. 1993, 27, 219–220, doi: 10.1007/BF00773004. 297. Bach, F. L.; Kushner, S.; Williams, J. H. J. Am. Chem. Soc. 1955, 77, 6049–6051, doi: 10.1021/ja01627a075. 298. Sum, Y. N.; Yu, D.; Zhang, Y. Green Chem. 2013, 15, 2718–2721, doi: 10.1039/ c3gc41269e. 299. Koval’skaya, S. S.; Dikusar, E. A.; Kozlov, N. G.; Popova, L. A. Russ. J. Org. Chem. 2001, 37, 1225–1227, doi: 10.1023/A:1013102815024. 300. Eguchi, S.; Arasaki, M. J. Chem. Soc. Perkin Trans. 1 1988, 1047–1050, doi: 10. 1039/p19880001047. 301. Kuroda, Y.; Ueda, H.; Nozawa, H.; Ogoshi, H. Tetrahedron Lett. 1997, 38, 7901– 7904, doi: 10/dh27rp. 302. Liscio, P.; Carotti, A.; Asciutti, S.; Karlberg, T.; Bellocchi, D.; Llacuna, L.; Mac- chiarulo, A.; Aaronson, S. A.; Schüler, H.; Pellicciari, R.; Camaioni, E. J. Med. Chem. 2014, 57, 2807–2812, doi: 10.1021/jm401356t. 303. Qiu, D.; Wang, S.; Tang, S.; Meng, H.; Jin, L.; Mo, F.; Zhang, Y.; Wang, J. J. Org. Chem. 2014, 79, 1979–1988, doi: 10.1021/jo402618r.