38
BOOK OF ABSTRACTS 12 TH DOCDAYS UNIVERSITY OF GRAZ SEPTEMBER 14 TH 15 TH

BOOK OF ABSTRACTS · 2020. 9. 2. · [email protected], 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

  • Upload
    others

  • View
    7

  • Download
    0

Embed Size (px)

Citation preview

Page 1: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

BOOK OF ABSTRACTS

12TH DOCDAYS

UNIVERSITY OF GRAZ

SEPTEMBER 14TH – 15TH

Page 2: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

2

DOCDAYS SUPPORTED BY:

Page 3: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

3

TABLE OF CONTENTS

ONLINE REGISTRATION ............................................................................. 4

SOCIAL EVENING ....................................................................................... 6

SCHEDULE .................................................................................................. 7

PLENARY LECTURE 1 – PROF. RUDOLF PIETSCHNIG. ............................ 8

INDUSTRY TALK – PHILIPP SELIG FROM PATHEON .............................. 10

STUDENT TALKS 1-24 .............................................................................. 12

LIST OF PARTICIPANTS ............................................................................ 37

Content

Page 4: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

4

ONLINE REGISTRATION

IN THE MAIN SLIDE OF THE EVENT WILL BE DISPLAYED A QR CODE THAT EACH

PARTICIPANTS HAVE TO SCAN IN ORDER TO GET THE CREDITS.

THIS HAS TO BE DONE IN THE MORNING (8:45-9:00) AND BEFORE THE STARTING OF THE

AFTERNOON SESSION (13:30-13:45).

THE DAILY REGISTRATION AT THE EVENT IS SIMPLE:

SCAN THE QR CODE WITH YOUR PHONE

OPEN THE LINK WITH YOUR BROWSER

ENTER YOUR NAME AND SURNAME

CONFIRM

Page 5: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

5

INSTRUCTIONS FOR THE MEETING:

The meeting will be hold via “Zoom Webinar”; for those of you who have never used the platform

before, you do need to download Zoom on your computer and create an account. More information

can be found at the link: https://www.youtube.com/watch?v=fMUxzrgZvZQ.

Please during the meeting keep your microphone and camera off, unless you are required otherwise.

It is required of you to share your screen and turn on your camera and microphone during your

presentation. Therefore, have your power point presentation ready and check the correct functioning

of your camera and microphone before the meeting. You will share your screen during the whole

duration of your presentation and question time.

Every speaker has a slot of 20 minutes, which is composed of maximum 15 minutes of talk and 5

minutes of questions. Please keep your talk within the time limit.

In the 5 minutes of questions, every participant has to give a feedback, by filling an online evaluation

sheet. The link for the evaluation sheet will be posted on the online chat, after every talk. Every

participant can express his/her feedback by a single vote (in the form of 1 to 5, 1 being the highest

score, 5 being the lowest) and a voluntary comment or suggestion. Please make sure to express your

vote in the given time, it will not be possible to vote after the chair has closed the vote and has moved

on with the schedule.

After every talk, questions can be asked by using the function “raise your hand” in the online chat. The

chair of the session will then proceed with giving you the possibility to talk (according to the time

schedule). Please speak clear, loud and be concise.

Page 6: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

6

SOCIAL EVENING:

The Social Evening will take place on Tue. September 15th from 6 pm at s'Biergartl. You will get a

personal coupon for food and drinks (you will get more information during the meeting).

s'Biergartl

Schönaugasse 41, 8010 Graz

From: https://www.google.com/maps, 2020

Page 7: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

Schedule

7

MON. September 14th

TUE. September 15th

0845-0900 Online Registration 0845-0900 Online Registration

0900-0915 Opening, Welcome

Prof. Christof Gattringer

Prof. Frank Uhlig

0900-1000 Chair: Weinberger

IT01 Selig from Patheon

ST12 Mata Gomez

ST13 Pöcheim

0915-1000 Chair: Prof. Uhlig

PL01 Prof. Pietschnig

1000-1015 Coffee Break

1000-1015 Coffee Break

1015-1115 Chair: Pöcheim

ST01 Pompei

ST02 Guttmann

ST03 Köckinger

1015-1115 Chair: Guttmann

ST14 Walenta

ST15 Sagmeister

ST16 Lazzarotto

1115-1130 Coffee Break

1115-1130 Coffee Break

1130-1230 Chair: Sagmeister

ST04 Wolfsgrubger

ST05 Schwarz

ST06 Steinegger

1130-1230 Chair: Schlatzer

ST17 Steiner

ST18 Steller

ST19 Eggbauer

1230-1345 Lunch Break

1230-1345 Lunch Break

1345-1445 Chair: Jurkaš

ST07 Breukelaar

ST08 Schlatzer

ST09 Zelzer

1345-1445 Chair: Breukelaar

ST20 Wiedemaier

ST21 Jud

ST22 Fuchs

1445-1500 Coffee Break

1445-1500 Coffee Break

1500-1540 Chair: Steller

ST10 Kodolitsch

ST11 Maierhofer

1500-1540 Chair: Breukelaar

ST23 Bondi ST24 Hoffellner

1540-1555 Concluding remarks

1800 Social Evening s'Biergartl

Page 8: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

8

PLENARY LECTURE

PL01 Prof. Rudolf Pietschnig

Page 9: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

Plenary Lecture

9

Phosphorus - more than a carbon copy

Roman Franz, Fabian Roesler, Denis Kargin and Rudolf Pietschnig*

Institute of Chemistry, University of Kassel

Heinrich-Plett-Straße 40, 34132 Kassel, Germany

[email protected], www.uni-kassel.de/go/hym

The diagonal relationship between phosphorus and carbon has been inspiration for synthetic and

structural chemistry for decades. [1] More recently, the unique and variable bonding situation in many

organophosphorus compounds has be employed to endow molecular materials with fascinating

electronic properties.[2] In our contribution a survey of recent achievements in this area will be

presented with particular focus on reactivity, stereo control and luminescence of organometallic

phosphorus compounds. In vicinity of electron rich metallocene units phosphorus frameworks are

prone to electronic interaction and exchange processes.[3]

Fe

FeFe

Figure 1. The pronounced s-character of lone-pairs at trivalent phosphorus atoms entails subtle interaction in P-

heterocyclic compounds such as phospholes (1) or phospha ferrocenophanes (2, 3) where low valent group 14 elements

(E = Si, Ge, Sn, Pb) are adjacent to P-stereogenic centers.

The authors acknowledge financial support by the DFG (PI 353/8-1, PI 353/9-1, PI 353/ 11-1 and CRC

1319 (ELCH)).

References: [1] K. B. Dillon, F. Mathey, J. F. Nixon, Phosphorus: The Carbon Copy, Wiley, Chichester, England, 1998.

[2] T. Baumgartner, R. Réau, Chem. Rev. 2006, 106, 4681-4727.

[3] A. Lik, D. Kargin, S. Isenberg, Z. Kelemen, R. Pietschnig, H. Helten, Chem. Commun. 2018, 54, 2471-2474

[4] R. Pietschnig, Chem. Soc. Rev. 2016, 45, 5216 - 5231.

Page 10: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

10

INDUSTRY TALK

IT01 Philipp Selig

Patheon, by Thermo Fisher Scientific

Page 11: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

Industry Talk

11

Process Chemistry at Patheon, by Thermo Fisher Scientific

Philipp Selig

Patheon, by Thermo Fisher Scientific; Patheon Austria GmbH & Co KG

St.-Peter-Straße 25, 4040 Linz, Austria

[email protected]

Process Chemistry is the art of developing a laboratory synthesis into a universally applicable, quality-

controlled and safe large-scale production process.

Following a short presentation of our pharmaceutical production site in Linz, we will introduce you to

some of the most important aspects of process chemistry (safety, operability, quality) and shed some

light on the questions: Why do we need process chemistry, and what makes a good production process?

Page 12: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

12

STUDENT TALKS

MON. September 14th

ST01 Simona Pompei

ST02 Robin Guttmann

ST03 Manuel Köckinger

ST04 Andreas Wolfsgruber

ST05 Romana Schwarz

ST06 Andreas Steinegger

ST07 Willem Breukelaar

ST08 Thomas Schlatzer

ST09 Sieglinde Zelzer

ST10 Katharina Kodolitsch

ST11 Maximilian Maierhofer

TUE. September 15th

ST12 Alejandro Mata Gomez

ST13 Alexander Pöcheim

ST14 Martin Walenta

ST15 Peter Sagmeister

ST16 Mattia Lazzarotto

ST17 Alexander Steiner

ST18 Beate Steller

ST19 Bettina Eggbauer

ST20 Fabian Wiedermaier

ST21 Wolfgang Jud

ST22 Andreas Fuchs

ST23 Riccardo Bondi

ST24 Lisa Hoffellner

Page 13: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

ST 01

13

Biocatalytic demethylation of guaiacol derivatives

Simona Pompei, Christopher Grimm, Wolfgang Kroutil*

Institute of Chemistry, University of Graz

Heinrichstrasse 28, 8010 Graz, Austria

[email protected], https://www.uni-graz.at

The ether functionality belongs to the most common and unifying structural features found in nature.[1]

The ubiquitous distribution of the C-O-C bond is widely shown among natural products, as well as in

man-made flavorings, fragrances and pharmaceuticals.[2] Therefore, the formation or breakage of C-O

ether bonds are valuable synthetic transformations. Since the C-O bond energy is 360 kJ/mol,

microbial cleavage of the ether bond is an extraordinary phenomenon and inevitably requires a

considerable investment of energy.[1] Consequently, to cleave the ether bond chemically, strong acids

such as hydrobromic acid (HBr) are generally necessary. Ethers are, in fact, rather stable to hydrolysis

even in the presence of mild acids or bases.

Figure 1. Enzymatic quasi-irreversible demethylation of guaiacol derivatives coupled with thiol-methyl acceptors.

We herein present a biocatalytic approach requiring a methyltransferase reversibly catalyzing both O-

methylation of phenols and demethylation of methyl pheny ethers via a cobalamin cofactor which is

bound to a carrier protein. The two proteins used here originate from the anaerobic bacteria

Desulfitobacterium halfiense. Interestingly, the methylation of thiols was found to be quasi-

irreversible. As this methyl acceptors seemed to act as a “methyl group sink”, the thiols are ideal as

acceptors for a demethylation method.

The role of downstream methyl acceptor was essential to drive the direction of the reaction towards

demethylation. We tuned the reaction parameters (pH, donor and acceptor concentrations, protein

loading) to obtain a rather general method to achieve the demethylation of guaiacol derivatives. As

proof of applicability, the biotransformation was scaled up, whereby hydroxytyrosol was produced in

high purity manner in a single step.

References: [1] G. F. White, N. J. Russell, E. C. Tidswell, Microbiol. Rev. 1996, 60, 216.

[2] E. J. Barreiro, A. E. Kummerle, C. A. Fraga, Chem. Rev. 2011, 111, 5215-5246.

Page 14: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

ST 02

14

Highly Accurate Geometries and Hydrogen-Bonded Energies of Small Systems

Robin Guttmann, A. Daniel Boese*

Institute of Chemistry, University of Graz

Heinrichstrasse 28, 8010 Graz, Austria

[email protected], https://www.uni-graz.at

As hydrogen-bonded systems are highly important in Biology and Chemistry, as almost all reactions

and systems are investigated and present in the liquid phase. In order to describe this phase properly,

the interactions have to be well understood and characterized. In order to do so, we calculate these

systems by state-of-the-art quantum mechanical post-Hartree-Fock methods. However, such

calculations are often not feasible for large systems due to the high computational costs. Therefore,

small model systems are often used as reference data for benchmarking various computational

methods. [1] A very promising benchmark set for intermolecular interaction energies and geometries is

the HB49 reference set, consisting of 49 complexes of small interacting molecules.[2]

In this talk, we will significantly extend and improve upon the existing HB49 reference data. First, we

use the so-called "gold standard" of quantum chemistry CCSD(T) for interaction energies as well as

for geometries. Second, we extend the benchmark set to also include non-equilibrium geometries via

constrained optimizations of 10 different intermolecular distances, leading to potential energy curves and thus a better description of the whole potential energy surface. Complete basis set extrapolations

and inclusion relaxation energies throughout lead to a further improvement of the obtained reference

data.

We find that the performed CCSD(T) geometry optimizations lead to small modifications of the

molecular structures compared to the original HB49 data. The resulting HB49x10 reference set

contains extremely accurate geometries and interaction energies for equilibrium as well as non-

equilibrium structures, which will enable high-quality benchmarks and development of new

computational methods for hydrogen bonds in particular.

Figure 1. Steps for highly accurate CCSD(T) interaction energies and geometries obtained with complete basis set

extrapolations (CBS).

References: [1] P. Morgante, R. Peverati, J. Comput. Chem. 2019, 40, 839-848.

[2] A. D. Boese, Mol. Phys. 2018, 113, 1618-1629.

Page 15: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

ST 03

15

Continuous-flow Synthesis of Aryl Aldehydes by Pd-catalyzed Formylation of

Phenol Derived Aryl Fluorosulfonates Using Syngas

Manuel Köckinger1,2, Christopher A. Hone1,2, Paul Hanselmann3, C. Oliver Kappe1,2* 1Institute of Chemistry, University of Graz, NAWI Graz

Heinrichstrasse 28, 8010 Graz, Austria

[email protected], https://www.goflow.at 2Research Center Pharmaceutical Engineering GmbH (RCPE)

Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology, Lonza AG, CH-3930 Visp, Switzerland

The fluorosulfonate group (-OSO2F) is a new and underutilized highly reactive functional group,[1]

which can be used as versatile intermediate in modern organic synthesis, particularly in the preparation

of biologically active compounds. The properties of fluorosulfonates are very similar to triflates, which

are one of the most commonly employed leaving groups. Aryl Fluorosulfonates (ArOFs) can be made

by the treatment of ArOH with sulfuryl fluoride (SO2F2), an inexpensive commodity chemical that is

widely used as an insecticide. Phenols are a particularly attractive and inexpensive starting material as

they are readily available from biomass. Building on previous knowledge on formylation reactions,[2]

we developed the first Pd-catalyzed formylation of ArOFs with syngas to yield aryl aldehydes using

stoichiometric amounts of syngas in continuous flow (Scheme 1). The optimized conditions were

applicable to a large set of substrates giving good to excellent yield of the corresponding arylaldehyde.

Scheme 1. Formation of Fluorosulfonates and subsequent Pd catalyzed continuous flow carbonylation using syngas in

good to excellent yields.

References: [1] J. Dong, L. Krasnova, M. G. Finn, K. Barry Sharpless, Angew. Chemie - Int. Ed. 2014, 53, 9430–9448.

[2] C. A. Hone, P. Lopatka, R. Munday, A. O’Kearney-McMullan, C. O. Kappe, ChemSusChem 2019, 12, 326–337.

Page 16: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

ST 04

16

Ligand Directed Protein Profiling of Carbohydrate-Processing Enzymes:

From Challenging Synthesis to First Biological Results

Andreas Wolfsgruber, Tanja M. Wrodnigg*

Institute of Chemistry and Technology of Biobased Systems, Graz University of Technology

Stremayrgasse 9, 8010 Graz, Austria

[email protected], https://www.tugraz.at

Carbohydrate-processing enzymes (CPEs) are a group of proteins, which are responsible for the

metabolism of carbohydrates and their conjugates in living organisms. Malfunction of these enzymes

results in diseases, such as, lysosomal storage diseases, immunological diseases, cancer, diabetes or

bacterial as well as viral infections. [1] However, the role and the contribution of the carbohydrate part

is still unknown or unrealised. Regarding this, activity based protein profiling (ABPP) has become a

solid technique for studying and elucidating protein functions in highly complex biological

environments. For instance, Stubbs, Withers and Overkleeft, [2] have developed paradigmatic examples

of such activity-based probes for carbohydrate processing enzymes.

Another approach for protein profiling is to use ligand-directed chemistry. [3] In contrast to ABPP

methods, this strategy allows for traceless and target-selective chemical labeling of proteins while

restoring the enzyme activity for further applications such as real-time monitoring in living cells. In

this case, the probe (Figure 1) is equipped with a recognition part (warhead) that allows for reversible

interaction of the probe with the active site of the enzyme of interest, and a detectable agent (tag) for

identification. The reactive group in between is responsible for the connection of the warhead and the

tag and at the same time behaves as electrophile that can be attacked by an amino acid residue near the

outside of the ligand-binding pocket. It covalently attaches the tag to the target protein. Concomitantly,

the warhead dissociates from the active site, which results in retention of protein activity. [3]

Figure 1. Example of a potential iminosugar based LDPP probe.

Results of our work towards the design, synthesis as well as first biological evaluation of iminosugar-

based probes for activity-based protein profiling of CPEs by ligand directed chemistry will be

presented.

References:

[1] A. E. Stütz, T. M. Wrodnigg, Adv. Carbohydr. Chem. Biochem. 2016, 73, 225-302.

[2] M. N. Gandy, A. W. Debowski, K. A. Stubbs, Chem Commun. 2011, 47, 5037-5039; O. Hekmat, S. He, R.

A. Warren, S. G. Withers, J. Proteome Res. 2008, 7, 3282-3292; J. Jiang, T. J. M. Beenakker, W. W.

Kallemeijn, G. A. van der Marel, H. van den Elst, J. D. C. Codeé, J. M. F. G. Aerts, H. S. Overkleeft, Chem.

Eur. J. 2015, 21, 10861-10869.

[3] S. Tsukiji, M. Miyagawa, Y. Takaoka, T. Tamura, I. Hamachi, Nat. Chem. Biol. 2009, 5, 341-343; Y. Takaoka, A.

Ojida, I. Hamachi, Angew. Chem. Int. Ed. 2013, 52, 4088-4106.

Page 17: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

ST 05

17

Exploring Thiol based Resins for the 3D-Printing of Medical Devices

Romana Schwarz1,2, Thomas Griesser1, Gregor Trimmel2* 1Institute of Chemistry of Polymeric Materials, Montanuniversitaet Leoben,

Otto-Glöckel-Straße 2, 8700 Leoben, Austria. 2Institute of Chemistry and Technology of Materials, Graz University of Technology

Stremayrgasse 9, 8010 Graz, Austria.

[email protected], https://www.tugraz.at

The last years have seen an increasing interest in the development of photo-polymerizable monomers

providing low cytotoxicity for the 3D printing of medical devices. In this PhD thesis, novel concepts

based on the thiol-X photo click chemistry are investigated and evaluated for the realization of

biocompatible photopolymers according to ISO 10993-5 (“Biological evaluation of medical devices -

Part 5: Tests for in vitro cytotoxicity”).

For the production of medical devices, which are in direct contact with human tissue, especially in the

case of mucosa or blood, non-network bound components like photoinitiators or their cleavage

products, but also residual monomers, stabilizers, degradation products or monomer impurities are of

major concern.[1] Many of these components state health risks when released to the human body by

migration processes. This contribution deals with the investigation of photocurable thiol-yne resins

covering several important aspects for the production of medical devices by UV based manufacturing

processes.

It turned out that these thiol based resins offer curing rates similar to acrylates, while providing much

higher conversion and lower monomer cytotoxicity.[2] This reactions leads to highly uniform polymeric

networks exhibiting a sharp and defined thermal glass transition together with outstanding toughness

which makes these polymers interesting for challenging applications such as medical implants.

The herein described monomer systems pave the way towards the individual fabrication of tissue

compatible photopolymers with tunable thermo-mechanical properties.

References:

[1] J. L. Ferracane, Resin composite-state of the art. Dental materials: official publication of the Academy of Dental

Materials 2011, 27, 29-38.

[2] A. Oesterreicher, S. Ayalur‐ Karunakaran, A. Moser, F. H. Mostegel, M. Edler, P. Kaschnitz, G. Pinter, G. Trimmel,

S. Schlögl, T. Griesser, Exploring thiol‐ yne based monomers as low cytotoxic building blocks for radical

photopolymerization. Journal of Polymer Science Part A: Polymer Chemistry 2016, 54, 3484-3494.

Page 18: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

ST 06

18

TADF emitters for self-referenced optical sensing

Andreas Steinegger, Silvia Zieger, Ingo Klimant, Sergey M. Borisov*

Institute of Analytical Chemistry and Food Chemistry, Graz University of Technology

Stremayrgasse 9, 8010 Graz, Austria

[email protected]

Optical sensors based on luminescent materials have become an integral part of analytical chemistry

as they offer distinct advantages over conventional sensing schemes. Sensors for pH, ions, oxygen,

carbon dioxide, and other analytes already have been well established, but the field is still intensively

investigated in search for more advanced probes, expanding the scope of analytes and applications.

Most luminescent materials in these sensors emit prompt fluorescence or phosphorescence.

Thermally activated delayed fluorescence (TADF) is a type of luminescence with intermediate

properties between prompt fluorescence and phosphorescence; similar to fluorescence, the emission

of photons occurs from the excited singlet state and analogously to phosphorescence, the triplet state

with correspondingly long lifetimes is involved. These characteristics of TADF render it very

interesting for sensing applications. Since TADF relies on reverse intersystem crossing, it displays

temperature-dependent behavior and consequentially can enable optical temperature sensing.

Additionally, the inherently long lifetimes of TADF can render the dyes also susceptible to oxygen

quenching, and therefore may make them promising for oxygen sensing. For measurement, the

luminescence decay time is determined, which is inherently self-referenced. By choice of the

immobilization matrix, temperature (Fig. 1, left) or dual sensors for temperature corrected oxygen

sensing can be prepared (Fig. 1, right).

Figure 1. Left: sensor design and temperature dependency of the TADF decay time for two TADF emitters.[1]

Right: calibration planes depicting the luminescence response of a TADF emitting material to temperature and oxygen.[2]

References: [1] S. Zieger, A. Steinegger, I. Klimant, S. M. Borisov, ACS Sens. – in Revision

[2] A. Steinegger. S. M. Borisov, submitted

Page 19: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

ST 07

19

Biocatalytic Reduction of Oximes Using Flavin-Dependent Ene-Reductases

Willem B. Breukelaar, Stefan Velikogne, Silvia Glück-Harter, Wolfgang Kroutil*

Institute of Chemistry, University of Graz

Heinrichstrasse 28, 8010 Graz, Austria

[email protected]

Flavin-dependent ene-reductases (EREDs), such as those of the Old Yellow Enzyme (OYE) family,

are a well described and studied class of enzymes, mostly applied for the enantioselective reduction of

activated C=C double bonds (Figure 1, top) . While a wide substrate scope has been established,[1] the

enzymes are not known for the reduction of C=heteroatom bonds. Recent work in our group has shown

that several ene-reductases reduce the oxime functionality of -keto--oximo esters very efficiently,

yielding tetrasubstituted pyrazines from non-enzymatic cyclisation and oxidation of the product

formed in biotransformation (Figure 1, bottom). Currently, we are working on a better understanding

of the mechanism, as well as application of this reactivity for the chemoenzymatic synthesis of chiral

amines.

Figure 1. General reaction schemes of the well described “traditional” use of EREDs for the reduction of activated C=C

double bonds (top) and the outline of this work, ERED-mediated reduction of an activated oxime to an amine

intermediate, followed by non-enzymatic cyclisation and oxidation to tetrasubstituted pyrazines (bottom).

The oxime reduction has been tested using a small substrate library (8 oximes) and six EREDs. This

has shown that various oximes are transformed with high efficiency (up to 77% product formation

within 24 hours), and that the experiments can easily be scaled up to preparative amounts. Typical

experiments using 2 mmol oxime yield 150-250 mg pyrazine, corresponding to isolated yields of up

to 62%.[2]

Currently, we are exploiting this unusual reactivity by synthesising and testing a library of oximes

derived from asymmetric malonates. Preliminary experiments have shown substrates of this

description to be accepted by the enzyme and transformed into the corresponding primary amine. This

work will be expanded further by establishing a substrate library and quantitative determination of

product formation and enantiomeric excess.

References: [1] Toogood, H. S.; Gardiner, J. M.; Scrutton, N. S. ChemCatChem 2010, 2, 892–914

[2] S. Velikogne, W. B. Breukelaar, F. Hamm, R. A. Glabonjat, S. Glück, K. Francesconi, W. Kroutil,

manuscript in preparation.

Page 20: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

ST 08

20

Palladium-Catalyzed S-Allylation as Bioorthogonal Strategy

Thomas Schlatzer1,*, Julia Kriegesmann2, Christian F. W. Becker2, Rolf Breinbauer1 1Institute of Organic Chemistry, Graz University of Technology

Stremayrgasse 9, 8010 Graz, Austria 2Institute of Biological Chemistry, University of Vienna,

Währinger Straße 38, 1090 Vienna, Austria

*[email protected]

Organopalladium species have proven as extremely useful tools for the selective modification of

peptides and proteins under biocompatible conditions. Therefore, numerous protocols based on

organopalladium reagents have recently emerged to artificially modify biomolecules.[1] In vivo many

nascent or folded proteins also undergo post-translational modifications (PTMs). An especially

noteworthy PTM is protein S-prenylation since this process does not only provide the protein with

hydrophobic anchors that allow binding to membranes but also is crucial for the activity of oncogenic

forms of Ras proteins.[2]

Figure 1. Reagent design and reaction mechanism of the Pd-catalyzed S-allylation.

In order to address this challenge, we developed an efficient method based on in situ prepared

π-allylpalladium complexes that are ideally suited for S-allylations overcoming prevalent difficulties

such as catalyst poisoning and thiol oxidation. The generated palladium species are highly functional

group tolerant and exhibit excellent chemo- as well as regioselectivity. This was mechanistically

investigated and applied on a broad set of thiol substrates including natural products such as

cephalosporins.[3] We then expanded the scope of our methodology to biomolecules enabling the

cysteine-selective prenylation (farnesylation, geranylgeranylation) of unprotected peptides and

proteins in aqueous solvents via a native thioether linkage. Therefore, this method offers access to

authentic, post-translationally modified proteins in vitro. Moreover, the broad utility of this new

ligation method was demonstrated by the incorporation of bioconjugation handles, an affinity tag and

a fluorophore into cysteine-containing peptides or proteins.[4]

References: [1] M. Jbara, S. K. Maity, A. Brik, Angew. Chem. Int. Ed. 2017, 56, 10644-10655.

[2] M. H. Gelb, L. Brunsveld, C. A. Hrycyna, S. Michaelis, F. Tamanoi, W. C. van Voorhis, H. Waldmann,

Nat. Chem. Biol. 2006, 2, 518-528.

[3] T. Schlatzer, H. Schröder, M. Trobe, C. Lembacher-Fadum, S. Stangl, C. Schlögl, H. Weber, R.

Breinbauer, Adv. Synth. Catal. 2020, 362, 331-336.

[4] T. Schlatzer, J. Kriegesmann, H. Schröder, M. Trobe, C. Lembacher-Fadum, S. Santner, A. V. Kravchuk,

C. F. W. Becker, R. Breinbauer, J. Am. Chem. Soc. 2019, 141, 14931-14937.

Page 21: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

ST 09

21

Application of a new LC-MS/MS method for determination of Vitamin D

metabolites

Sieglinde Zelzer1,2, Andreas Meinitzer2, Markus Herrmann2, Walter Goessler1 1University of Graz, Institute of Chemistry, Universitätsplatz 1, 8010 Graz, Austria;

2Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University

of Graz, Auenbruggerplatz 15, 8036 Graz

[email protected], https://www.uni-graz.at

In recent years, determination of vitamin D has increased in importance due to the clinical relevance

of vitamin D deficiency and intoxication [1]. Assessment of the vitamin D status including metabolites

remains critical especially for persons with a 24-hydroxylase deficiency. This enzyme plays an

important role in calcium homeostasis by catalyzing the conversion of 25-hydroxyvitamin D3

(25(OH)D3) into 24,25-dihydroxyvitamin D3 (24,25(OH)2D3). Reduced enzymatic activity has been

reported to cause hypercalcemia and nephrotoxic diseases with of too high vitamin D levels [2]. In the

case of a 24-hydroxylase deficiency, it is very important to refrain from any kind of vitamin D

supplementation. To identify a disordered vitamin D metabolism the simultaneous determination of

25(OH)D3 and some other hydroxylated metabolites in serum samples is necessary. We established a

new LC-MS/MS method based on 4-Phenyl-1,2,4-triazole-3,5-dione (PTAD) derivatization with high

sensitivity, selectivity and good accuracy for 25(OH)D3, 25-hydroxyvitamin D2 (25(OH)D2) and

24,25(OH)2D3. The method was verified by external controls provided by the Vitamin D External

Quality Assessment Scheme (DEQAS) which is traceable to the National Institute for Standards and

Technology [3]. The clinical utility of this method has been confirmed in a patient with 24-hydroxylase

deficiency with an extreme low 24,25(OH)2D3 concentration. In the sample, we found an additional

analyte peak next to the 24,25(OH)2D3 response which was identified as 25,26-dihydroxyvitamin D3.

A comparison study between our LC-MS/MS method and the IDS-iSYS 25OHDS assay, an

immunoassay used in clinical routine showed a mean bias of -16.6 %. [4]. A possible cross reactivity

of the 24,25(OH)2D3 could explain this discrepancy.

Figure 1. Typical chromatogram for determination of 24,25-dihydroxyvitamin D3 concentration and the new identified

vitamin D metabolite 25,26-dihydroxyvitamin D3

References: [1] M. Herrmann, CL. Farrell, I. Pusceddu, N. Fabregat-Cabello, E. Cavalier, Clin Chem Lab Med. 2017, 55, 3-26.

[2] K. P. Schlingmann, M. Kaufmann, S. Weber, A. Irwin, C. Goos, U. John, et al, New Engl. J. Med. 2011, 365, 410-21.

[3] N. Binkley, CT. Sempos, J. Bone Miner Res. 2014, 29, 1709-14.

[4] S. Zelzer, W. Goessler, M. Herrmann M, J. Lab. Precis. Med. 2018, 3, 99.

24,25(OH)2D3

25,26(OH)2D3

Compound Precursor ion / Product ion (m/z)

24,25(OH)2D3 574.2 / 298.0

25,26(OH)2D3 574.1 / 298.1

Page 22: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

ST 10

22

Construction of coordination polymers with bifunctional linkers

Katharina Kodolitsch1, Ana Torvisco2 and Christian Slugovc1 1 Institute for Chemistry and Technology of Materials, Graz University of Technology,

Stremayrgasse 9, 8010 Graz, Austria 2 Institute of Inorganic Chemistry, Graz University of Technology, Stremayrgasse 9, 8010 Graz,

Austria

In terms of the fabrication of Metal Organic Frameworks (MOFs), the assembly with desired structural

types is totally dependent on the judicious choice of appropriate organic spacers. In this regard, organic

linkers with N and/or O donors often have been employed as effective building blocks.[1]

Herein we report a truly green way to prepare potentially bidentate ligands namely 3-(1H-imidazol-1-

yl)propanoic acid derivates. The two-step synthesis includes an aza-Michael reaction of the imidazole

derivatives with enoates and an ester cleavage using one equivalent of NaOH in water. The obtained

ligands have been used for the preparation of coordination polymers with various metal salts from

water solutions, whereby herein special focus will be placed on zinc.

Starting from aqueous zinc(II) solutions and adding two equivalents of the ligand dissolved in water

led to the instantaneous formation of white precipitates. Of particular interest is the 2-methylimidazole

based linker because it forms a microcrystalline, microporous precipitate consisting of 1D coordination

polymers which are organized in a 3D scaffold held together by hydrogen bonding with crystal water

(see Figure above). The material does not lose its porosity and crystallinity upon drying. Molecules

bearing carboxylic acid groups such as the protein BSA or fluorescein could be incorporated into the

crystals. In contrast, the imidazole and 3-phenylimidazole based linkers initially form amorphous

precipitates which show permanent porosity upon drying. Prolonged heating of the amorphous

precipitate at 80°C leads to the formation of a crystalline material consisting of 2D coordination

polymer sheets linked via hydrogen bonding to form a 3D scaffold. Drying of these crystals however

led to the loss of crystallinity. However, encapsulation of guests (proteins, dyes) into the initial

precipitate is still feasible.

The contribution will deal with a structural description of the materials and a presentation of their

properties such as surface area, thermal stability, solubility in water and several puffer systems and

will discuss the encapsulation of guests.

References:

[1] S. Chen, Q.Liu, Y.Zhao, R.Qiao, L. Sheng, Z. Liu, S. Yang, C. Song, Cryst. Growth Des, 2014, 14, 3727-3741.

Page 23: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

ST 11

23

A Fluorescent Optical Ammonia Sensor–Suitable for Online Bioprocess

Monitoring

Maximilian Maierhofer, Veronika Rieger, Torsten Mayr*

Institute of Analytical Chemistry and Food Chemistry, Graz University of Technology

Stremayrgasse 9/II, 8010 Graz, Austria

[email protected], https://www.tugraz.at/institute/acfc/home/

Optical sensors have found numerous applications in the last decades, e.g. optical sensors for oxygen

and pH are established in bioprocess monitoring. In bio processing ammonia is another key parameter

due to its toxicity at certain concentration levels.[1] Since this compound is often a by-product in

bioprocessing, online monitoring is desired. However, sensors for monitoring ammonia or ammonium

in bioreactors are rare. We present an ammonia sensor (Fig. 1 (b)) suitable for bioprocess monitoring.

Our system is based on an acid-base concept including a fluorescent pH-sensitive dye.[2] The sensing

layer is covered by a hydrophobic porous membrane, which excludes hydrophilic interfering materials.

Our target analyte, ammonia (NH3), diffuses through the barrier to the protonated dye whereby it

deprotonates the dye and switches off the NIR-emission. Read-out is performed with a commercially

available compact phase fluorimeter combined with optical fibers. Dual-lifetime referencing (DLR)

acts as detection method and Egyptian blue as reference material. A sensor performance in the range

of total ammonia concentration (TAC) from 1 to 100 mmol L-1 is demanded. By exchanging the dye,

the dynamic range of the sensor is shifted towards other concentrations opening different application

fields like environmental monitoring and chemical reaction monitoring. Depending on temperature

and ammonia concentration the response time t90 and the recovery time vary from 20 s up to 4 min

(Fig 1 (a)). The sensor performance is not influenced sufficiently by varying temperature (Fig. 1 (c)).

The sensor materials are chosen to withstand β-sterilization. Further experiments on other sterilization

methods will be studied in future. This sensor is planned to monitor reactions in bioreactor systems.

0 5000 10000 15000 20000 25000

5

10

15

20

25

30

35

40

0.03

10

dp

hi / °

Time / s

100

10

1

3

0.3

0.1

1

3

0.3

0.1

0.03

0.1 1 10 100 1000

0.0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

0.8

0.9

1.0

25°C

35°C

45°C

No

rm. co

t(p

hi)

TAC / mmol L-1

(a) (b) (c) Figure 1. (a) Sensor response at 25°C at seven different ammonia concentrations in mg L-1 (b) Optical ammonia sensor

attached to an optical fiber with a stainless screw-cap, diameter 5 mm; (c) Calibration plots of ammonia sensor monitored

at three different temperatures for TAC in mmol L-1. Mean values and standard deviations obtained from four sensors.

References:

[1] B. Timmer, W. Olthuis, A. van den Berg, Sensors and Actuators B: Chemical 2005, 107, 666–677.

[2] M. Strobl, T. Rappitsch, S. M. Borisov, T. Mayr, I. Klimant, Analyst 2015, 140, 7150–7153.

Page 24: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

ST 12

24

Acyl Azide Generation and Amide Bond Formation in Continuous-Flow for the

Synthesis of Peptides

Alejandro Mata, U. Weigl, O. Floegel, Christopher A. Hone and C. Oliver Kappe*

Continuous Flow Synthesis and Processing (CCFLOW), Research Center Pharmaceutical

Engineering GmbH (RCPE) Inffeldgasse 13, Graz 8010, Austria.

Institute of Chemistry, University of Graz Heinrichstrasse 28, 8010 Graz, Austria

[email protected], https://www.goflow.at

Acyl azides are valuable synthetic intermediates for the preparation of amides, amines, isocyanates,

ureas, ketenimines and carbodiimides.1 One particularly important application is their use for the

synthesis of peptides and proteins.2 There are a number of synthetic strategies to form peptides from

their corresponding amino acids.3 However, many of the traditional amide bond formation protocols

do not reach the high requirements of enantioselectivity necessary for active pharmaceutical ingredient

(API) synthesis. The acyl azide method is perhaps the best method for racemization-free peptide

segment condensation, which cannot be so readily achieved by other amide coupling methods.2,4

Figure 1. Acyl azide method for the preparation of dipeptides: 1) hydrazide formation; 2) acyl azide generation; and 3)

peptide coupling

A disadvantage of the acyl azide method is its rather laborious synthetic sequence that involves the

handling of a highly dangerous byproduct as azides anions. Despite the advantages described above,

the acyl azide method for the synthesis of peptides has been abandoned and it is strictly off-limits in

industrial scale. To overcome these safety concerns, continuous flow chemistry is presented as a key

tool to implement “forbidden” chemistry into safe processes.5 We envisioned a continuous-flow

protocol that would address the safe handling and quenching of such highly unstable and potentially

explosive intermediates. Microreactor technology enables the safe generation of acyl azides and their

reaction in situ in a telescoped process. A biphasic environment made possible the separation of azide

anions from the desired dipeptides and their posterior and immediate quench. The protocol has been

used to prepare various dipeptides (5 examples) free of epimerization (<1%). In combination with boc-

cleavage protocol (also investigated in continuous flow), we extended the scope to the formation of a

tripeptide.

References: [1] a) M. Balci, Synthesis 2018, 50, 1373-1401; b) S. Bräse, C. Gil, K. Knepper and V. Zimmermann, Angew.

Chem. Int. Ed. 2005, 44, 5188-5240.

[2] a) J. Meienhofer, in The Peptides: Analysis, Synthesis, Biology, Academic Press, London, UK, 1979, vol.

1, Ch. 4, pp. 197-239; b) Y. S. Klausner and M. Bodanszky, Synthesis 1974, 549-559.

[3] A. El-Faham and F. Albericio, Chem. Rev. 2011, 111, 6557-6602.

[4] a) K. Hofmann, A. Jöhl, A. E. Furlenmeier and H. Kappeler, J. Am. Chem. Soc. 1957, 79, 1636-1641; b) I.

A. L. Ali, Arkivoc 2008, 78-89; c) S. M. El Rayes, I. A. I. Ali and W. Fathalla, Arkivoc, 2008, 86-95;

[5] a) B. Gutmann, D. Cantillo and C. O. Kappe, Angew. Chem. Int. Ed. 2015, 54, 6688-6728; b) M.

Movsisyan, E. I. P. Delbeke, J. K. E. T. Berton, C. Battilocchio, S. V. Ley and C. V. Stevens, Chem. Soc. Rev.

2016, 45, 4892-4928.

Page 25: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

ST 13

25

A Game of Reactivity – Why a Silandiide is Stable but the corresponding

Monosilanide is not

Alexander Pöcheim, Judith Baumgartner*, Christoph Marschner*

Institute of Inorganic Chemistry, Graz University of Technology

Stremayrgasse 9/IV, 8010 Graz, Austria

[email protected], https://www.uni-graz.at

Some 20 years ago, a method for an easy and direct access toward potassium silanides was

developed.[1] Since these days tremendous progress has been made as the initially on non-

functionalized oligosilanes based method was extended to a large scope of functionalities. In fact,

potassium silanides in proximity to halogen-, amino- or methoxy substituents offer an astounding

chemistry, where the main product depends on the applied reaction conditions.[2, 3]

Figure 1. Reaction of a 2,5-oligosilanyl substituted silole with potassium tert-butoxide leads to a labile monosilanide,

which can rearrange to an allyl anion or react further toward a silandiide depending on the applied reaction conditions.

Here, we describe the quite unexpected reactivity of a 2,5-oligosilanylsubstituted silole with potassium

tert-butoxide. The formed monosilanide is labile toward rearrangement yielding an allyl anion. This

rearrangement requires an unusual C-H activation in ortho-position of the attached phenyl moiety.

However, if the desilylation step toward the second silanide is sufficiently fast, for instance in the

presence of 18-crown-6, the silandiide is accessible which is not prone to rearrangement.

References: [1] C. Marschner, Eur. J. Inorg. Chem. 1998, 221–226.

[2] I. Balatoni, J. Hlina, R. Zitz, A. Pöcheim, J. Baumgartner, C. Marschner, Molecules 2019, 24, 3823.

[3] I. Balatoni, J. Hlina, R. Zitz, A. Pöcheim, J. Baumgartner, C. Marschner, Inorg. Chem. 2019, 58, 14185–

14192.

Page 26: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

ST 14

26

Metal(loid)enrichment of Pleurotus ostreatus

Martin Walenta, Simone Braeuer, Walter Goessler*

Institute of Chemistry – Analytical Chemistry, University of Graz

Universitätsplatz 1/I, 8010 Graz, Austria

[email protected], https://chemie.uni-graz.at/de/analytische-chemie/forschung/ache/

Mushrooms do not only decompose and recycle organic matter, they also play a necessary role in the

global nutrient cycle and are therefore important components of every ecosystem in the world. Fungi,

neither plants nor animals, interact very strongly with their environment for example in the form of

symbiosis or as parasites. Furthermore, mushrooms become more and more important as a source of

protein, carbohydrates, vitamins and necessary trace elements such as selenium, iron or cobalt.

Pleurotus ostreatus were found to be of economic importance, since they have rather undemanding

cultivation conditions, a strong growth, can accumulate trace elements and have some interesting

medical applications such as inhibitor of tumor growth and inflammation or as a mean of lowering

blood pressure and blood lipid concentration.[1,2]

The aim of this study is to improve or find appropriate growing conditions for these mushrooms while

enriching them with different trace elements or their compounds. We started with selenium because

the enrichment of Pleurotus mushrooms with this element is well studied.[3-6]

The substrate consisting of 18 kg straw, 1.3 kg millet overgrown with mycelium and 0.8 kg lime were

homogenized in a mixing machine. For enrichment, 125 ml of a sodium selenite solution with various

concentrations were added during the homogenization resulting in a selenium concentration in the

substrate of 0.34 mg/kg and 0.76 mg/kg respectively. Next, 3 kg of the substrate were placed into six

separate plastic bags and the inoculated packs were incubated for three weeks at 25°C in the dark. For

fructification, they were placed in a fruiting room with a controlled temperature of 16°C and an air

humidity of 85 %. The mushrooms were collected over three flushes during a period of three months.

Addition of selenium did not influence the harvest volume.

After freeze-drying and milling to 0.1 mm, samples were digested with nitric acid. Thereafter,

36 elements were determined with inductively coupled plasma mass spectrometry.

First results showed that the enrichment was a success. The control mushrooms had a selenium

concentration between 0.038±0.001 to 0.095±0.002 mg/kg, while the enriched mushrooms had a

selenium concentration of 0.92±0.03 to 2.2±0.1 mg/kg and 1.4±0.1 to 4.1±0.2 mg/kg respectively. The

Se concentration in the first flush was always the lowest, even in the control mushrooms. Besides

selenium other essential trace elements such as Zn and Cu were also significantly accumulated in the

mushrooms, making these elements promising candidates for further enrichment experiments.

References: [1] A. Gregori, M. Švagelj, J. Pohleven, Food Technol. Biotechnol. 2007, 45, 238-249.

[2]L. Ma, Y. Zhao, J. Yu, H. Ji, A. Liu, International journal of biological macromolecules 2018, 111, 421.

[3] G. Kaur, A. Kalia, H. S. Sodhi, J Food Biochem 2018, 42, e12467.

[4] P. Bhatia, F. Aureli, M. D'Amato, R. Prakash, S. S. Cameotra, T. P. Nagaraja, F. Cubadda, Food chemistry 2013, 140,

225.

[5] P. Niedzielski, M. Mleczek, M. Siwulski, P. Rzymski, M. Gąsecka, L. Kozak, Eur Food Res Technol 2015, 241, 419.

[6] A. P. d. Oliveira, J. Naozuka, Microchemical Journal 2019, 145, 1143.

Page 27: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

ST 15

27

A Modular Flow Platform for Real Time Control of Multistep API Synthesis

Using Model-Based Strategies

Peter Sagmeister, Rene Lebl, Jason D. Williams, David Cantillo and C. Oliver Kappe

Institute of Chemistry, University of Graz

Heinrichstrasse 28, 8010 Graz, Austria

[email protected], http://goflow.at/

In recent years continuous flow processing has attracted the interest of pharmaceutical manufacturers

for reasons including shortened synthetic routes, improved quality and enhanced sustainability

profiles. One important benefit of a continuous flow regime is the ease of implementation of process

analytical technology (PAT) tools for real time monitoring of process parameters and critical quality

attributes (CQAs) for single and multistep syntheses.[1] The chemical and pharmaceutical industries

are encouraged by regulatory agencies, such as the US Food and Drug Administration (FDA),[2] to

integrate process analytics and control technology (PACT) to continuous manufacturing, from an early

stage in development.[3, 4]

Figure 1. Conceptual representation of the model reaction and the process platform.

We showcase PACT in a multistep synthesis of the active pharmaceutical ingredient (API) mesalazine.

This three step synthesis of the API includes hazardous chemistry, use of an extreme process window

(elevated temperature and pressure) and multiphase chemistry. In the first stage, the process was

carefully designed in a modular continuous flow microreactor system and a PAT strategy was

implemented. Fundamental process understanding was developed by performing kinetic studies,

design of experiment (DoE) optimization and dynamic experiments for each individual step. Based on

this data, a parametric model and an artificial intelligence (AI) model have been developed to

implement predictive control strategies. In the second stage a central supervisory control and data

acquisition (SCADA) platform was implemented to unify analytical data acquired from the PAT tools

and control the process itself. Automatic tuning of the critical process parameters during the chemical

reaction, enabled by the control system, will enhance the robustness of the process. An automated

control strategy will anticipate system failures or deviations from the quality attributes and react before

they occur.

References: [1] P. Sagmeister, J. D. Williams, C. A. Hone, C. O. Kappe React. Chem. Eng. 2019, 4, 1571-1578

[2] FDA. PAT Guidance for Industry – A Framework for Innovative Pharmaceutical Development Manufacturing and

Quality Assurance, FDA, Rockville, MD, 2004, p. 16.

[3] L. L. Simon, H. Pataki et al. Org. Process Res. Dev. 2015, 19, 3−62

[4] C. Herwig Anal. Bioanal. Chem. 2020, in press

Page 28: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

ST 16

28

Synthesis of Natural Products and Key Pharmaceuticals via 2-Oxoglutarate

Dependent Dioxygenases

Mattia Lazzarotto1, Lucas Hammerer1, Peter Hartmann1, Michael Hetmann 2, Karl Gruber 2,

Wolfgang Kroutil 1 * and Michael Fuchs 1 * 1 Institute of Chemistry, Organic and Bioorganic Chemistry, University of Graz, 8010 Graz, Austria

2 Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria

[email protected]

The synthesis of lignan natural products is essential to the development of new pharmaceuticals and

recently we could show that 2-oxoglutarate dependent dioxygenases (2-ODDs) can facilitate key-steps

in the synthetic pathway towards this compound class [1]. 2-ODDs are non-heme FeII dioxygenases

that activate molecular oxygen via 2-oxogluratate in order to perform a wide range of oxidative

reactions. Despite being an interesting class of enzymes these enzymes are seldom used for biocatalytic

purposes. Nevertheless, the chemoenzymatic synthesis of deoxy-, epi-, and Podophyllotoxin has been

developed in our laboratories. Remarkably, a 2-ODD from Podophyllum hexandrum is implemented

for the stereoselective cyclisation of the natural product’s scaffold starting from its precursor yatein.

This enzyme has shown an impressive biocatalytic performance as the reaction has been upscaled to

two grams to prove the scalability of the biocatalytic key step. This chemoenzymatic synthesis of epi-

podophyllotoxin is a novel, simple route, which is high yielding and low in step count (32% overall

yield over 4 steps) [1].

Figure 1. Natural products obtained via 2-ODDs biotransformations.

Moreover, we are now focused on other 2-ODDs, namely the Hyoscyamine 6β-Hydroxylase (H6H) a

key enzyme in the biosynthesis of scopolamine and the AsqJ from Aspergillus nidulans which is

involved in the synthesis of quinolone antibiotics. By screening the conditions of the

biotransformation, addition of additives, deactivation and mechanistic investigations we want to gain

a deeper understanding of the mechanistic principles inherent to this class of enzymes that may provide

an easier implementation of 2-ODDs in chemoenzymatic routes for relevant synthetic purposes. In fact

we are trying to apply the AsqJ for a chemoenzymatic synthesis of Tipifarnib [2]. In conclusion, with

our work we want to prove that 2-ODD are underestimated with regard to their biocatalytic potential.

References: [1]: Lazzarotto M., Hammerer L., Hetmann M., Borg A., Schmermund L., Steiner L., Hartmann P., Belaj F., Kroutil W.,

Gruber K., Fuchs M. Chemoenzymatic Total Synthesis of Deoxy-, epi -, and Podophyllotoxin and a Biocatalytic Kinetic

Resolution of Dibenzylbutyrolactones. Angew. Chem. Int. Ed. (2019), 58, 8226- 8230.

[2]: Angibaud P.R., Venet M. G., Filliers W., Broeckx R., Ligny Y. A., Muller P., Poncelet V.S., End D. W. Synthesis

Routes Towards the Farnesyl Protein Transferase Inhibitor ZANESTRATM. Eur. J. Org. Chem. (2004), 479-486.

Page 29: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

ST 17

29

Photochemical benzylic brominations in continuous flow:

in situ generation of Br2, intensification and scale up

Alexander Steiner, Jason D. Williams, C. Oliver Kappe*

Institute of Chemistry, University of Graz, Heinrichstrasse 28, 8010 Graz, Austria

Center for Continuous Flow Synthesis and Processing (CC FLOW), Research Center

Pharmaceutical Engineering GmbH (RCPE), Inffeldgasse 13, 8010 Graz, Austria.

[email protected], www.goflow.at

Benzyl bromides are important building blocks in the pharmaceutical, agrochemical and materials

industries. Their synthesis is usually accomplished by radical bromination, initiated either thermally

or photochemically. The use of N-bromosuccinimide (NBS), a stable and easy to handle crystalline

reagent, reduces the productivity due to the limited solubility and lower reactivity of NBS and leads to

increased solvent wastage.[1,2,3] Elemental bromine has the advantage of higher reactivity, however, its

use is complicated by storage issues, high vapor pressure, toxicity and the need for chlorinated

solvents. Flow technology, combined with the concept of in situ generation of the hazardous reagent,

can overcome these drawbacks, enabling fast and efficient bromination reactions under highly process

intensified conditions.

Figure 1. Schematic representation of the experimental setup: Combination of the two aqueous streams forms bromine

inside the reactor, which is mixed with the organic substrate stream. Irradiation by LEDs at 405 nm initiates the radical

bromination to yield the desired benzylic bromide.

Herein, we report the development of a Br2 generator for the photochemical benzylic bromination of

toluene derivatives in a Corning Advanced-Flow Lab Photoreactor.[4] The Br2 is generated from

concentrated aqueous hydrobromic acid and sodium bromate streams, providing exceptional mass

efficiency. Efficient mixing in the reaction plate ensures fast extraction of the bromine into the organic

phase, which consists exclusively of the neat substrate. Reaction times as low as 15 to 18 seconds for

monobromination or 100 seconds for a dibromination could be achieved, resulting in excellent yields

of the corresponding products. A scale-out run of the monobromination of 2,6-dichlorotoluene was

performed, producing 1.17 kg of the benzyl bromide in only 230 minutes processing time, with an

overall yield of 95% and a remarkably low PMI of 3.08. Furthermore, experiences in scaling up this

process by a factor of ~20, in a small production-scale reactor, will be described.

References: [1] F. Sabuzi, G. Pomarico, B. Floris, F. Valentini, P. Galloni, V. Conte, Coord. Chem. Rev. 2019, 385, 100-136.

[2] I. Saikia, A. J. Borah, P. Phukan, Chem. Rev. 2016, 116, 6837-7042.

[3] D. Cantillo, C. O. Kappe, React. Chem. Eng. 2017, 2, 7-19.

[4] A. Steiner, J. D. Williams, O. de Frutos, J. A. Rincón, C. Mateos, C. O. Kappe, Green Chemistry 2020, 22, 448-454.

Page 30: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

ST 18

30

A Molecular Construction Kit for Metalloid Tin Clusters:

From Organotin hydrides to polyhedral compounds

Beate G. Steller, and Roland C. Fischer*

Institute of Inorganic Chemistry, Graz University of Technology

Stremayrgasse 9/V, 8010 Graz, Austria

[email protected], https://www.ac.tugraz.at

The number and structural diversity of polyhedral metalloid tin clusters RxSnn (n>x) remains still fairly

small. This extraordinary class of compounds has aroused great interest not only because of the

intrinsic beauty of the molecules, but also due to their unique electronic properties and bonding

situations. Their cluster core geometries frequently adopt atomic arrangements observed in the solid

states of their constituents and therefore can serve as molecular models for reactions at the surface of

the bulk element. [1] Additionally, metalloid tin clusters have recently gained interest in the context of

small molecule activation.[2]

Among other strategies, these compounds were accessed via the thermolysis of oligo-tin fragments,[3]

reduction of tin halides or amides,[4] dehydrogenation of organotin(IV) trihydrides,[5]

disproportionation of tin(I) halides[6] or the derivatisation of ZINTL ions.[7]

In course of our studies, we developed a straightforward one step synthetic protocol based on tin/tin

bond formation from organotin di- and trihydrides R4-nSnHn and low oxidation state diamides of

group 14 via the elimination of readily removable H-NR2. (Figure 1) Thereby, this concept allows not

only a large variation of the tin hydride as well as the amide species, but also like in a construction kit

the individual combination of these to access metalloid tin clusters. Several examples of these obtained

clusters will be presented.

Figure 1. Schematic illustration of cluster formation starting from tin hydrides and group 14 element amides.

References: [1] a) A. Schnepf, in: Structure and Bonding, Clusters – Contemporary Insight in Structure and Bonding, (Ed. S. Dehnen),

Springer, Cham, Germany, 2017, 174, 135-200. b) M. Driess, H. Nöth (Eds.) Molecular Clusters of the Main Group

Elements, Wiley-VCH, Weinheim, Germany, 2004.

[2] P. Vasko, S. Wang, H. M. Tuononen, P. P. Power, Angew. Chem. - Int. Ed. 2015, 54, 3802-3805.

[3] a) L. R. Sita, I. Kinoshita, J. Am. Chem. Soc. 1991, 113, 1856-1857 and J. Am. Chem. Soc. 1992, 114, 7024-7029.

[4] a) M. Wagner, M. Lutter, B. Zobel, W. Hiller, M. H. Prosenc, K. Jurkschat, Chem. Commun. 2015, 51, 153. b) N.

Wiberg, H.-W. Lerner, H. Nöth, W. Ponikwar, Angew. Chem. Int. Ed. 1999, 38, 1103. c) P. Prabusankar, A. Kempter, C.

Gemel, M.-K. Schröter, R. A. Fischer, Angew. Chem. Int. Ed. 2008, 47, 7234-7237. d) E. Rivard, J. Steiner J. C. Fettinger,

J. R. Giuliani, M. P. Augustine, P. P. Power, Chem. Commun. 2007, 32, 4919-4912.

[5] J.-J. Maudrich, C. P. Sindlinger, F. S. W. Aicher, K. Eichele, H. Schubert, L. Wesemann, Chem. – A Eur. J. 2016, 23,

2192-2200.

[6] C. Schrenk, A. Schnepf, Rev. Inorg. Chem. 2014, 34, 93-118.

[7] S. C. Sevov, J. M. Goicoechea, Organometallics 2006, 25, 5678-5692.

Page 31: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

ST 19

31

Exploring the potential of chanoclavine synthase in Ergot alkaloid pathway

Bettina Eggbauer, Bianca Hengel, Peter Macheroux, Jörg Schrittwieser,Wolfgang Kroutil

Institute of Chemistry, University of Graz

Heinrichstraße 28, 8010 Graz

Ergot alkaloids constitute a group of indole-derived mycotoxins that are produced in various

filamentous fungi. Due to the potential pharmacological activities for pharmaceutical applications, the

chemical nature of this biomolecules and the biosynthetic routs have been studied for a long time.[1][2]

The biosynthesis of these generally quite diverse compounds follows a route via the intermediate

chanoclavine-1, which is produced by a ring closure step through two enzymes, a FAD-dependent

oxidoreductase (EasE) and a heme-binding catalase (EasC) after prenylation via a prenyltransferase

(DmaW) (Figure 1). Studies on these key enzymes, relying on complementation in fungal strains,

revealed EasE to be responsible for 1,3-diene formation and EasC the essential enzyme for oxidative

ring closure.[3,4] Although, a lot of characterization has been done, the difficulties in EasE protein

expression hampered in vitro studies. In order to facilitate the studies of ergot alkaloids and to

implement an in vitro production platform, one aim of this PhD thesis is the reconstitution of the early

steps in the pathway. Investigating heterologous protein expression, optimization of each step and

additional organic synthesis should finally give reasonable conversion to desired intermediates for

isolation.

Figure 2: Conserved initial part of ergot alkaloid pathway, synthesizing chanoclavine-l

Moreover, with the aim of ergot alkaloids as pharmaceutical ingredient, another focus of the research

will be based on production of semi-synthetic ergot-alkaloid derivatives. Implementation of the in vitro

reconstituted chanoclavine synthase and further organic synthesis should lead to active ingredients like

1-propyl-agroclavine for pharmaceutical application.

References:

[1] J. J. Chen, M. Y. Han, T. Gong, J. L. Yang, P. Zhu, RSC Adv. 2017, 7, 27384–27396.

[2] C. A. Young, C. L. Schardl, D. G. Panaccione, S. Florea, J. E. Takach, N. D. Charlton, N. Moore, J. S. Webb, J.

Jaromczyk, Toxins (Basel). 2015, 7, 1273–1302.

[3] C. A. F. Nielsen, C. Folly, A. Hatsch, A. Molt, H. Schröder, S. E. O’Connor, M. Naesby, Microb. Cell Fact. 2014,

13, 1–11.

[4] Y. Yao, C. An, D. Evans, W. Liu, W. Wang, G. Wei, N. Ding, K. N. Houk, S. S. Gao, J. Am. Chem. Soc. 2019,

141, 17517–17521.

Page 32: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

ST 20

32

Synthesis and Characterization of Manganese-

Based Hydrogenase Model Systems

Fabian Wiedemaier, Nadia C. Mösch-Zanetti*

Institute of Chemistry, University of Graz

Schubertstrasse 1, 8010 Graz, Austria

[email protected], https://www.uni-graz.at

The [Fe-Fe]-hydrogenase catalyzes the reversible interconversion between molecular hydrogen and

protons (Scheme 1). Its active site (Figure 1a) consists of two sulphur bridged iron atoms which show

the in biology rare case of carbonyl and cyanido ligation.[1,2]

Scheme 1. Reaction catalyzed by [Fe-Fe]-hydrogenase

Besides Fe-based functional models[3] recently also Mn complexes are investigated towards their

capability to catalyze the reduction of protons to molecular hydrogen whereby usually an electrode

acts as the electron source.[4] The electrocatalytically active, manganese-based model systems

investigated in our group (e.g. 1) are based on thiopyridazin ligands which allow the formation of

sulphur bridged Mn-dimers similar to the enzyme. A facial tris-carbonyl pattern on each Mn atom

mimics the carbonyl and cyanido ligation. The catalytical characteristics of those model systems

investigated by electrochemical methods will be presented.

Figure 1. a) Active site of the [Fe-Fe]-hydrogenase, [1] b) Manganese-based functional model mimicking the active site

of [Fe-Fe]-hydrogenase

References: [1] D. Schilter, J. M. Camara, M. T. Huynh, S. Hammes-Schiffer, T. B. Rauchfuss, Chem. Rev. 2016, 116, 8693.

[2] D. J. Evans, C. J. Pickett, Chem. Soc. Rev. 2003, 32, 268.

[3] Y.-C. Liu, K.-T. Chu, R.-L. Jhang, G.-H. Lee, M.-H. Chiang, Chem. Commun. 2013, 49, 4743.

[4] V. Kaim, S. Kaur-Ghumaan, Eur. J. Inorg. Chem. 2019, 5041.

Page 33: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

ST 21

33

Electrifying Radical Trifluoromethylations and Oxytrifluoromethylations of

Unactivated Scaffolds

Wolfgang Jud1,2, C. Oliver Kappe1,2, and David Cantillo1,2 1 Institute of Chemistry, University of Graz, NAWI Graz, Heinrichstrasse 28, Graz, Austria

2 Center for Continuous Flow Synthesis and Processing (CC FLOW), Research Center

Pharmaceutical Engineering GmbH (RCPE), Inffeldgasse 13, 8010 Graz, Austria

[email protected], https://www.goflow.at

Synthetic electrochemistry has seen a resurgence in recent years as a method to effect redox processes

in a controlled manner by simply applying electricity to the reaction medium.[1] Electrochemical

methods have the capacity to generate radicals and other high energy intermediates under mild,

typically ambient conditions, which can provide with new strategies to solve challenging synthetic

transformations. C-H trifluoromethylations of unactivated substrates such as arenes and olefins are

examples of reactions that can only be tackled via radical chemistry.[2] Trifluoromethylations, in

general, are among the most pursued transformations in organic and medicinal chemistry during the

past decade due to the enhanced biological properties provided by the fluorinated moiety. Generation

of CF3 radicals from suitable reagents can be accomplished using thermal or photochemical methods,

as well as via stoichiometric amounts of redox reagents (e.g. tBuOOH). Electrochemical methods are

a very attractive alternative, as such redox processes can be enabled by electricity, which avoids the

use of often toxic or expensive reagents and reduces the generation of waste.

Herein, we present electrochemical procedures for the generation of CF3 radicals under ambient

conditions, which have been successfully applied to trifluoromethylations and

oxytrifluoromethylations of unactivated scaffolds. While the trifluoromethylation can be considered

as a simple radical addition/single-electron transfer sequence, the oxytrifluoromethylation entails a

paired electrolysis process in which the intermediate generated by the anodic oxidation event reacts

with the product of the concurrent cathodic reduction. Details on the reaction mechanisms including

cyclic-voltammetry data and radical trapping experiments will also be presented.

Figure 1. Electrolysis enables the release of trifluoromethyl radicals from suitable sources devoid of toxic or expensive

reagents, which subsequently can be trapped by unactivated substrates such as arenes or olefins.

References: [1] M. Yan, Y. Kawamata, P. S. Baran, Chem. Rev. 2017, 117, 13230; (b) S. R. Waldvogel, A. Wiebe, T.

Gieshoff, S. Möhle, E. Rodrigo, M. Zirbes, Angew. Chem. Int. Ed. 2018, 57, 5594.

[2] a) D. Staveness, I. Bosque, C. R. J. Stephenson, Acc. Chem. Res. 2016, 49, 2295; b) T. Koike, M. Akita,

Acc. Chem. Res. 2016, 49, 1937.

Page 34: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

ST 22

34

Advanced testing of odorants in plastic materials

Andreas Fuchs1, Stefanie Engleder1, Jürgen Huber1, Theresa Kaltenbrunner1, Erich Leitner2* 1 Borealis Polyolefine GmbH

St.-Peter-Straße 25, 4021 Linz, Austria

[email protected], www.borealisgroup.com 2 Institute of Analytical Chemistry and Food Chemistry, Graz University of Technology

Stremayrgasse 9/2, 8010 Graz, Austria

[email protected], www.analytchem.tugraz.at

What is the most appropriate analysis technique to test for odour-active compounds in plastic materials

and where to start? In recent years, many advances were made regarding the instrumental analysis of

emission and odour testing. Hyphenation of different (automated) sample preparation regimes with

gas chromatography (GC) and mass spectrometry (MS) systems for emission testing were developed.

It was found that a commonly used approach typically involves solid phase microextraction (SPME)

coupled with GCxGC/MS systems. Contrarily to that, there might be more sensitive ways to perform

emission tests including emission chambers or advanced adsorbents. GC-olfactometry (GC-O),

another well-known technique in this respect would even involve the human being in the test setup but

was not discussed here [1-7].

The aim of this study was to compare and to investigate the potential of various emission testing setups

in order to characterise odorants in plastic materials. Different system from “fast and simple”

approaches to more complex one like two dimensional comprehensive GCxGC were used. In the

discussion a critical evaluation of the pros and cons of the different methods will be given. Where are

potential limitations in sensitivity and selectivity? What might influence method precision and

accuracy? Where does discrimination need to be considered?

With this study, a comprehensive overview of currently known approaches to instrumental analysis of

odour-active substances is presented. The advantages but also the limitations of the various analysis

setups are shown and discussed. Case studies involve e.g. headspace, thermodesorption and headspace

sorptive extraction using Twister® or MonoTrapTM systems as well as comprehensive GCxGC/TOF-

MS analyses.

References: [1] J. B. Phillips, J. Xu, Journal of Chromatography A 1995, 703, 327-334.

[2] G. Schomburg, Journal of Chromatography A 1995, 703, 309-325.

[3] M. Adahchour, J. Beens, R. J. J. Vreuls, U. A. T. Brinkman, TrAC Trends in Analytical Chemistry 2006, 25, 438-

454.

[4] M. Adahchour, J. Beens, R. J. J. Vreuls, U. A. T. Brinkman, TrAC Trends in Analytical Chemistry 2006, 25, 540-

553.

[5] E. Reingruber, J. Reussner, C. Sauer, A. Standler, W. Buchberger, Journal of Chromatography A 2011, 1218,

3326-3331.

[6] Ó. Ezquerro, B. Pons, M. a. T. Tena, Journal of Chromatography A 2002, 963, 381-392.

[7] R. V. Emmons, R. Tajali, E. Gionfriddo, Separations 2019, 6, 39.

Page 35: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

ST 23

35

Bioinspired Tungsten-Acetylene Model Complexes for Acetylene Hydratase

Riccardo Bondi, Nadia C. Mösch-Zanetti*

Institute of Chemistry, University of Graz

Schubertstraße 1III, 8010 Graz, Austria

[email protected], https://www.uni-graz.at

Amid all the tungsten-dependent enzymes, acetylene hydratase (AH) is the only one that catalyzes a

non-redox reaction, which is the net hydration of acetylene to acetaldehyde. The active site of this

enzyme is composed of a tungsten(IV) center coordinated by four sulfur atoms deriving from two

molybdopterin cofactors (MPT), a water molecule and a cysteine residue (Figure 1a).[1] In order to

gain deeper insight into the mechanism of AH and to develop a functional model of this enzyme, the

bioinspired ligand 3-chloropyridine-2-thiolate (3-ClSPy) was used (Figure 1b). This ligand was chosen

to investigate the effect of an electron withdrawing substituent on the reactivity of the complexes.

Figure 1. a) Active site of AH. b) Bioinspired ligand used for this project: 3-chloropyridine-2-thiolate (3-ClSPy).

The synthesis of a suitable tungsten(IV) model complex started from the literature known metal

precursor [W(CO)3Br2(NCMe)2] (Scheme 1).[2] The initial reaction with NaL (L = 3-ClSPy) led to the

formation of [W(CO)3(L)2], via a straightforward salt metathesis, and the following treatment with

acetylene resulted in the synthesis of [W(CO)(C2H2)(CHCH-L)(L)]. The latter complex derives from

an intramolecular nucleophilic attack of the nitrogen atom of the ligand on the coordinated C2H2.[3]

Nevertheless, this product of insertion can be converted into [W(CO)(C2H2)(L)2] by heating it to 45 °C.

With a slightly different procedure, it was also possible to gain access to [W(CO)(C2H2)(L)2], avoiding

the formation of the product of insertion. Finally, the desired mononuclear tungsten(IV) complex

[WO(C2H2)(L)2] was obtained by oxidation of [W(CO)(C2H2)(L)2]. Additionally, a comparison to less

electron withdrawing systems will be discussed.

Scheme 1. Synthetic ways to gain access to tungsten(IV) model complex.

References: [1] G. B. Seiffert, G. M. Ullmann, A. Messerschmidt, B. Schink, P. M. H. Kroneck, O. Einsle, Proc. Natl. Acad. Sci. U

S A 2007, 104, 3073–3077.

[2] L. M. Peschel, F. Belaj, N. C. Mösch-Zanetti, Angew. Chem. Int. Ed. 2015, 54, 13018–13021.

[3] C. Vidovič, L. M. Peschel, M. Buchsteiner, F. Belaj, N. C. Mösch-Zanetti, Chem. Eur. J. 2019, 25, 14267–14272.

Page 36: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

ST 24

36

Paper as food packaging material – a natural barrier?

Lisa Hoffellner, Erich Leitner*

Institute of Analytical Chemistry and Food Chemistry, Graz University of Technology

Stremayrgasse 9, 8010 Graz, Austria

[email protected], https://www.tugraz.at

Foodstuff can be packaged in many different materials and forms depending on various requirements.

In the packaging sector of dry food, paper and board are currently the most commonly used materials.

Although the use of paper is highly desired, because it has many favorable properties, including its

economic and ecological friendly characteristics, its application is also challenging. Food packaging

materials should be sufficiently inert to preclude the transfer of substances from the packaging into the

food, and it should preserve the organoleptic properties of the food products [1]. Paper can be made

from different fiber purities and sources and is a very complex three dimensional network, with pores

of different sizes. Therefore, it is often regarded as a permeable layer with limited barrier properties

[2]. Complex transfer processes can occur through migration or permeation. Especially low molecular

weight and volatile substances are of concern, because they might transfer from and through the

packaging into the packaged goods and vice versa. To be suitable as a food packaging material, paper

should protect the packaged goods from unwanted transfer of chemicals.

The aim of our work is to gain a fundamental understanding of the interdependence of transport

mechanisms of the paper matrix and volatile food aroma compounds. This may help to better

understand the paper network and to predict the behavior of a selected paper for a specific use. We

developed a fast and simple method based on gas chromatography and flame ionization detection

(GC/FID) or mass spectrometric detection (GC/MS) to measure the adsorption and desorption

behavior of selected volatile organic compounds on various virgin fiber paper samples. Considering

that paper fibers are composed of polar macromolecules, we distinguished between two compound

classes, polar and non-polar aroma compounds. Polar volatiles exhibit a high affinity, i.e., they strongly

adsorb on the surface of the investigated paper samples. Against the widespread theory, that paper

cannot act as a barrier layer, the polar aroma compounds used in our study were not transported through

the test paper samples. This might indicate that a food packaging made from virgin fiber paper can

protect the food up to a certain degree from unwanted transfer of polar chemicals.

References: [1] Parliament THEE, Council THE, The OF, Union P. L 338/4. 2004;(1935):4-17

[2] Geueke, B. (2016). Paper and board. Retrieved February 6, 2020, from

https://www.foodpackagingforum.org/food-packaging-health/food-packaging-materials/paper-and-board

Page 37: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

LIST OF PARTICIPANTS

37

NAME AFFILIATION

1 Flock Michaela TU

2 Kroutil Wolfgang KFU

3 Pietschnig Rudolf PL, UK

4 Uhlig Frank TU

5 Philipp Selig Patheon, by Thermo Fisher Scientific

PHD STUDENTS

NAME AFFILIATION

6 Astria Efwita TU

7 Bierbaumer Sarah KFU

8 Bondi Riccardo KFU

9 Brandner Lea Alexandra TU

10 Breukelaar Willem KFU

11 Brudl Christoph TU

12 Burger Tobias TU

13 Cigan Emmanuel KFU

14 Civita Donato KFU

15 Ćorović Miljan KFU

16 Dalfen Irene TU

17 Edinger David KFU

18 Eggbauer Bettina KFU

19 Fischer Susanne TU

20 Frieß Michael TU

21 Fuchs Andreas TU

22 Fuchs Stefanie TU

23 Glotz Gabriel KFU

24 Goni Freskida TU

25 Gößler Gerhard KFU

26 Grimm Christopher KFU

27 Grössl Doris TU

29 Guttmann Robin KFU

30 Hallwirth Franz TU

31 Hartmann Peter KFU

32 Hoffellner Lisa TU

33 Hogrefe Katharina TU

34 Jovanovic Milica KFU

35 Jud Wolfgang KFU

36 Jurkaš Valentina KFU

37 Klokic Sumea TU

38 Kodolitsch Katharina TU

39 Köckinger Manuel KFU

40 Ladenstein Lukas TU

41 Lazzarotto Mattia KFU

42 Lembacher-Fadum Christian TU

43 Maierhofer Maximilian TU

44 Mata Gomez Alejandro KFU

Page 38: BOOK OF ABSTRACTS · 2020. 9. 2. · manuel.koeckinger@rcpe.at, 2Research Center Pharmaceutical Engineering GmbH (RCPE) Inffeldgasse 13, 8010 Graz, Austria 3Microreactor Technology,

LIST OF PARTICIPANTS

38

45 Müller Philipp TU

46 Pfleger Georg KFU

47 Pöcheim Alexander TU

48 Pommer Reinhold TU

49 Pompei Simona KFU

50 Pontesegger Niklas TU

51 Prieschl Michael KFU

52 Prohinig Jennifer TU

53 Püschmann Sabrina TU

54 Rappitsch Tanja TU

55 Ratzenböck Karin TU

56 Redolfi Sebastian TU

57 Russegger Andreas TU

58 Sagmeister Peter KFU

59 Schallert Viktor TU

60 Schlatzer Thomas TU

61 Schuh Lukas TU

62 Schreiner Till TU

63 Schweda Bettina TU

64 Simic Stefan KFU

65 Sorgenfrei Frieda KFU

66 Steinegger Andreas TU

67 Steiner Alexander KFU

68 Steller Beate TU

69 Schwarz Romana TU

70 Swoboda Alexander KFU

71 Tjell Anders TU

72 Vakalopoulou Efthymia TU

73 Von Keutz Timo KFU

74 Walenta Martin KFU

75 Weinberger Gernot TU

76 Wernik Michaela KFU

77 Wied Peter TU

78 Wiedemaier Fabian KFU

79 Wieser Philipp Aldo TU

80 Windischbacher Andreas KFU

81 Wolfsgruber Andreas TU

82 Yang Wei TU

83 Zelzer Sieglinde MedUni

84 Zukic Erna KFU