scispace - formally typeset
Open AccessBook ChapterDOI

Multi-Enzymatic Cascades In Vitro

Reads0
Chats0
TLDR
The combination of enzymatic reactions in a simultaneous or sequential fashion by designing artificial synthetic cascades allows for the synthesis of complex compounds from simple precursors as mentioned in this paper, which can also lead to a drastic reduction of the produced waste.
Abstract
The combination of enzymatic reactions in a simultaneous or sequential fashion by designing artificial synthetic cascades allows for the synthesis of complex compounds from simple precursors. Such multi-catalytic cascade reactions not only bear a great potential to minimize downstream processing steps but can also lead to a drastic reduction of the produced waste. With the growing toolbox of biocatalysts, alternative routes employing enzymatic transformations towards manifold and diverse target molecules become accessible. In vitro cascade reactions open up new possibilities for efficient regeneration of the required cofactors such as nicotinamide cofactors or nucleoside triphosphates. They are represented by a vast array of two-enzyme cascades that have been designed by coupling the activity of a cofactor regenerating enzyme to the product generating enzyme. However, the implementation of cascade reactions requires careful consideration, particularly with respect to whether the pathway is constructed concurrently or sequentially. In this regard, this chapter describes how biocatalytic cascades are classified, and how such cascade reactions can be employed in order to solve synthetic problems. Recent developments in the area of dynamic kinetic resolution or cofactor regeneration and showcases are presented. We also highlight the factors that influence the design and implementation of purely enzymatic cascades in one-pot or multi-step pathways in an industrial setting.

read more

Content maybe subject to copyright    Report

University of Groningen
Multi-Enzymatic Cascades In Vitro
Schmidt, Sandy; Schallmey, Anett; Kourist, Robert
Published in:
Enzyme Cascade Design and Modelling
DOI:
10.1007/978-3-030-65718-5_3
IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from
it. Please check the document version below.
Document Version
Publisher's PDF, also known as Version of record
Publication date:
2021
Link to publication in University of Groningen/UMCG research database
Citation for published version (APA):
Schmidt, S., Schallmey, A., & Kourist, R. (2021). Multi-Enzymatic Cascades In Vitro. In S. Kara, & F.
Rudroff (Eds.),
Enzyme Cascade Design and Modelling
(pp. 31-48). Springer International Publishing.
https://doi.org/10.1007/978-3-030-65718-5_3
Copyright
Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the
author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).
The publication may also be distributed here under the terms of Article 25fa of the Dutch Copyright Act, indicated by the “Taverne” license.
More information can be found on the University of Groningen website: https://www.rug.nl/library/open-access/self-archiving-pure/taverne-
amendment.
Take-down policy
If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately
and investigate your claim.
Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the
number of authors shown on this cover page is limited to 10 maximum.
Download date: 09-08-2022

Multi-Enzymatic Cascades In Vitro
3
Sandy Schmidt, Anett Schallmey, and Robert Kourist
Abstract
The combination of enzymatic reactions in a
simultaneous or sequential fashion by design-
ing articial synthetic cascades allows for the
synthesis of complex compounds from simple
precursors. Such multi-catalytic cascade
reactions not only bear a great potential to
minimize downstream processing steps but
can also lead to a drastic reduction of the
produced waste. With the growing toolbox of
biocatalysts, alternative routes employing
enzymatic transformations towards manifold
and diverse target molecules become accessi-
ble. In vitro cascade reactions open up new
possibilities for efcient regeneration of the
required cofactors such as nicotinamide
cofactors or nucleoside triphosphates. They
are represented by a vast array of
two-enzyme cascades that have been designed
by coupling the activity of a cofactor
regenerating enzyme to the product generating
enzyme. However, the implementation of cas-
cade reactions requires careful consideration,
particularly with respect to whether the path-
way is constructed concurrently or sequen-
tially. In this regard, this chapter describes
how biocatalytic cascades are classied, and
how such cascade react ions can be employed
in order to solve synthetic problems. Recent
developments in the area of dynamic kinetic
resolution or cofactor regeneration and
showcases are presented. We also highlight
the factors that inuence the design and imple-
mentation of purely enzymatic cascades in
one-pot or multi-step pathways in an industrial
setting.
Keywords
Biocatalysis · Enzymatic cascades · In vitro
biotransformations · Enzymes · Cofactor
regeneration
3.1 Introduction
Reaction systems combining two or more chemi-
cal steps in one pot without isolation of reaction
intermediates are commonly referred to as
cascades [1]. In such systems, individual chemi-
cal steps can be enzyme catalyzed or involve
chemical (metal or organo-) catalysts. Accord-
ingly, multi-enzymatic cascades include several
S. Schmidt
Groningen Research Institute of Pharmacy, Chemical and
Pharmaceutical Biology, Groningen, The Netherlands
e-mail: s.schmidt@rug.nl
A. Schallmey
Technische Universität Braunschweig, Institute for
Biochemistry, Biotechnology and Bioinformatics,
Braunschweig, Germany
e-mail: a.schallmey@tu-braunschweig.de
R. Kourist (*)
Graz University of Technology, Institute of Molecular
Biotechnology, Graz, Austria
e-mail: kourist@tugraz.at
#
Springer Nature Switzerland AG 2021
S. Kara, F. Rudroff (eds.), Enzyme Cascade Design and Modelling,
https://doi.org/10.1007/978-3-030-65718-5_3
31

biocatalytic steps, which can be either performed
simultaneously or sequentially. In the rst case,
also called concurrent cascade or tandem reac-
tion, all enzymes and reagents are present from
the beginning of the reaction, meaning that all
reaction steps take place at the same time. In
contrast, in a sequential multi-enzymatic cascade,
certain enzymes and/or reagents are added at a
later point in time after a certain sequence is
completed. Hence, all reaction steps of a sequen-
tial cascade are still performed in one pot but must
be separated in time. The latter might be neces-
sary if, e.g., two or more enzymes of the same
cascade require different reaction conditions, one
enzyme is inhibited by a compound appearing
prior or later in the sequence, or to prevent unde-
sired side reactions if cross-reactivities of the
involved enzymes occur. Moreover, in vitro and
in vivo multi-enzymatic cascades are distin-
guished depending on the biocatalyst preparation
(compare also Chap. 4). Whereas in in vivo
cascades all enzymes of the cascade are included
in whole living cells, in vitro cascades make use
of isolated enzymes in puried form, as cell-free
extracts, freeze-dried preparations, immobilized
versions, etc. [2].
In addition, cascade reactions in general can
exhibit different topologies (Fig. 3.1)[1, 3]. In a
linear c ascade (Fig. 3.1a), the product of one
chemical step serves as substrate of the
subsequent chemical step. This is probably the
most straightforward type of cascade reaction as
it avoids the isolation of (unstable) reaction
intermediates with the nal goal to increase the
overall product yield while saving time and
resources.
Additionally, a linear cascade can be used to
shift an unfavorable reaction equilibrium of one
step by combination with a subsequent irrevers-
ible reaction step that pulls the product out of the
reaction. A recent example is the combination of
the hydroxynitrile lyase from Manihot esculenta
for the synth esis of optically pure (S)-4-
methoxymandelonitrile with the Candida
antarctica lipase A-catalyzed acylation of the
formed α-cyanohydrin yielding a stable ester
product (Scheme 3.1)[4]. This way, the equilib-
rium of the hydrocyanation reaction could be
shifted towards product formation and isolation
of the unstable cyanohydrin intermediate was
avoided.
Next to linear cascades, also orthogonal
(Fig. 3.1b), cyclic (Fig. 3.1c), and parallel,
interconnected (Fig. 3.1d) cascades have been
described. In an orthogonal enzyme cascade, the
conversion of a substrate into the desired product
is coupled with a second reaction to remove one
or more by-products. An example is the combina-
tion of a transaminase with lactate dehydroge-
nase, where the by-product pyruvate (when
using alanine as amine donor) of the
transaminase-catalyzed reaction is further
converted to lactic acid in order to shift the equi-
librium of the transaminase reaction (Scheme 3.2)
[5]. In a cyclic cascade, one enantiomer out of a
racemic substrate mixture is converted to an inter-
mediate product, which is then transformed back
to the racemic starting material yielding the
unreacted substrate enantiomer as nal product.
The same applies if the unreacted substrate enan-
tiomer is racemized to yield enantiomerically
pure product (dynamic kinetic resolution).
Hence, cyclic cascades are commonly applied in
deracemization processes, e.g. of amino acids,
hydroxy acids, or amines [6, 7]. Finally, in a
parallel, interconnected cascade, two separate
biocatalytic reactions are connected by comple-
mentary cofactor requirements of the two
enzymes. Therefore, parallel, interconnected
cascades are commonly associated with cofactor
recycling systems. While in one biocatalytic step
a substrate is transform ed into the desired prod-
uct, a cheap co-substrate is converted to a
co-product in the parallel enzyme-catalyzed step
to recycle the required cofactor for the rst
enzyme.
As briey mentioned, cascade reactions offer
several advantages compared to conventional
reaction schemes [1]. This includes the avoidance
of operational work up steps, which saves time,
resources, and reagents, can reduce waste forma-
tion and, at the same time, allows for higher nal
product yields. Additionally, different reaction
steps can be smartly combined in a cascade to
solve synthetic problems of a reaction sequence
such as cofactor regeneration, shift of reaction
32 S. Schmidt et al.

equilibria, in situ generation of toxic or instable
reagents, etc. On the other hand, the setup of
efcient cascade reactions is usually a complex
and challenging task [1]. Compatible reaction
conditions have to be identied and, in case of a
simultaneous cascade, the reaction rates of indi-
vidual steps have to be balanced. Moreover, pos-
sible problems, such as the formati on of
undesired side products due to cross-reactivities
of catalysts or the inhibition of an enzyme by a
compound appearing earlier or later in the reac-
tion sequence, can occur that have to be
addressed. Multi-enzymatic cascades are usually
easier to establish than chemoenzymatic or
chemo-catalytic cascade reactions as enzymes
commonly work in aqueous reaction media and
often display similar temperature and pH
requirements for optimal performance. Neverthe-
less, also several examples for the successful
combination of enzymatic and chemical reaction
steps in a cascade have been described in litera-
ture (compare also Cha p. 5) [8].
In an impressive way, nature successfully
evolved a multitude of highly complex conc urrent
cascade reactions. Living organisms built every
minute thousands of highly complex molecules
from simple precursors in an astonishing variety
and efciency. To achieve such high efciencies,
individual enzymatic transformations are
arranged in cascading sequences (biosynthetic
S
P
1
P
2
Cat. 1
Cat. 2
S
P
1
Cat. 1
P
2
P
2
Cat. 2
A
)
B)
C)
S
1
S
2
P
1
S
2
Cat. 1
Cat. 2
S
P
1
Cat. 1
S
2
P
2
Cat. 2
cofactor
red/ox
cofactor
ox/red
D)
Fig. 3.1 Possible
topologies of cascade
reactions. (a) Linear. (b)
Orthogonal. (c) Cyclic. (d)
Parallel, interconnected.
The scheme was adapted
from [1]
Scheme 3.1 Combination of Manihot esculenta hydroxynitrile lyase (MeHNL) and Candida antarctica lipase A (CAL
A) in a linear cascade to shift the reaction equilibrium of the rst hydrocyanation reaction [4]
3 Multi-Enzymatic Cascades In Vitro 33

pathways) in living cells [9]. This strategy can be
mimicked in vitro by the design of articial meta-
bolic pathways [10] through combination of mul-
tiple isolated enzymes in a homogeneous phase or
by combination of two or more catalytic activities
in a single protein, e.g., by fusion of genes
encoding different enzymes or by crosslinking
several enzymes [1114]. Recently, a novel
approach has been mentioned in literature
named systems biocata lysis, which aims for the
in vitro setup of synthetic metabolic cycles for the
production of valuable compounds [15].
To illustrate the synthetic potential of multi-
enzyme cascades, but also potent ial challenges in
the development of cascade reactions, different
cascade examples have been selected and are
described in more detail in Sect. 3.2. Perhaps the
most frequent motivation for currently used
enzyme cascades is the concurrent regeneration
of (redox-) cofactors or expensive reagents. A
second application is the combination of
isomerizing enzymes or catalysts with highly
enantioselective enzymes for dynamic kinetic
resolutions, which allow to overcome the yield
limitation of 50% of kinetic resolutions. Cofactor
regeneration and dynamic kinetic resolutions
require concurrent cascades, which often makes
it very challenging to provide optimal reaction
conditions for both catalysts used. To overcome
compatibility issues, compartmentalization is a
possibility to enable different operating
conditions for all reaction steps of a concurrent
or step-wise cascade [8].
3.2 Cascades to Solve Synthetic
Problems
3.2.1 Combination of Selective
Enzymatic Steps
with Isomerizing Reactions
Due to their availability and ease to use,
hydrolases mainly constituted the rst wave of
biocatalysts in the rst biocatalytic processes
[16]. Kinetic resolution of racemic mixtures was
often the preferred reaction form (Fig. 3.2)
[17]. While this reaction is simple and robust, it
suffers from an intrinsic limitation of 50% yield,
which in turn requires the physical separation of
substrate and product. The addition of a second
catalyst, which racemizes the unreacted substrate,
but not the product (Fig. 3.2) in a cyclic cascade
allows for the complete conversion of starting
material to the desired product enantiomer. The
depletion of the faster-reacting substrate enantio-
mer leads to an increased conversion of the
slower-reacting substrate enantiomer as its rela-
tive concentration increases, which reduces the
optical purity of the product. To prevent this, the
racemization should be one order of magnitude
faster than the enantioselective reaction. This
makes the racemization steps often the bottleneck
of dynamic kinetic resolution reactions.
In 1997, Bäckvall et al. combined Ruthenium-
catalyzed hydrogen transfer reactions for the race-
mization of aryl aliphatic secondary alcohols with
their lipase-catalyzed kinetic resolution in a
R
1
R
2
O
R
1
R
2
NH
2
transaminase
+
H
3
N
COO
-
O
COO
-
lactate
dehydrogenase
HO
COO
-
NAD
+
NADH
+ H
+
cofactor
regeneration
co-substrate
co-product
Scheme 3.2 Combination
of transaminase and lactate
dehydrogenase in an
orthogonal cascade to shift
the reaction equilibrium of
the transaminase-catalyzed
reaction
34 S. Schmidt et al.

Citations
More filters
Journal ArticleDOI

Design and biocatalytic applications of genetically fused multifunctional enzymes.

TL;DR: Fusion proteins, understood as those created by joining two or more genes that originally encoded independent proteins, have numerous applications in biotechnology, from analytical methods to metabolic engineering as mentioned in this paper .
References
More filters
Journal ArticleDOI

Industrial biocatalysis today and tomorrow

TL;DR: Biocatalytic processes can now be carried out in organic solvents as well as aqueous environments, so that apolar organic compounds aswell as water-soluble compounds can be modified selectively and efficiently with enzymes and bioc atalytically active cells.
Journal ArticleDOI

Engineering the third wave of biocatalysis

TL;DR: Applications of protein-engineered biocatalysts ranging from commodity chemicals to advanced pharmaceutical intermediates that use enzyme catalysis as a key step are discussed.
Journal ArticleDOI

Borrowing hydrogen in the activation of alcohols

TL;DR: In this paper, the metal catalyst returned the hydrogen to the transformed carbonyl compound, leading to an overall process in which alcohols can be converted into amines, compounds containing CC bonds and β-functionalised alcohols.
Journal ArticleDOI

Biocatalysis for pharmaceutical intermediates: the future is now

TL;DR: This review describes how demands are being addressed to make biocatalysis successful, particularly by the use of micro-scale technology for high-speed catalyst screening and process development alongside discipline integration of biology and engineering with chemistry.
Journal ArticleDOI

Industrial applications of enzyme biocatalysis: Current status and future aspects.

TL;DR: Recent progress in enzyme biocatalysis is reviewed, and the trends and strategies that are leading to broader industrial enzyme applications are discussed.
Related Papers (5)
Frequently Asked Questions (14)
Q1. What are the contributions in this paper?

Kara and Schallmey this paper classified two-enzymatic cascade reactions into two types: concurrent cascade and tandem cascade. 

The system using an acetate kinase and a hexokinase or a glycerol kinase with acetyl phosphate as donor is one of the most frequently used examples for ATP regeneration in biocatalytic in vitro reactions [60–62]. 

The direct oxidation of cycloalkanes to cycloalkanones employing a P450 monooxygenase and an ADH represents another successful enzyme cascade that is redox neutral or redox self-sufficient [40]. 

In an orthogonal enzyme cascade, the conversion of a substrate into the desired product is coupled with a second reaction to remove one or more by-products. 

One-pot processes in general offer the advantage of high enantioselectivities while circumventing the need for multiple steps, thus being highly efficient. 

An intrinsic challenge for an efficient regeneration of the cofactors (recycling between 100 and 106 times) is given by the usually low long-term stability of the cofactors, even if a complete cascade is cofactor neutral and regenerates the cofactor during the course of its reaction [33, 53]. 

The enzymatic racemization of the inexpensive racemic hydantoin is crucial to achieve a complete conversion of the starting material to the product. 

possible problems, such as the formation of undesired side products due to cross-reactivities of catalysts or the inhibition of an enzyme by a compound appearing earlier or later in the reaction sequence, can occur that have to be addressed. 

Saving unit operations for the isolation and purification of intermediate products allows for tremendous savings in terms in cost, energy, and waste formation. 

Another advantage of one-pot cascade reactions is represented by the possibility to start from simple, inexpensive, and achiral starting materials. 

The combination of an ADH with a BVMO for the conversion of cyclohexanol to ε-CL has been strongly investigated since ε-CL is a valuable precursor for polymer synthesis [42–48]. 

Kara and coworkers developed a new class of redox-neutral reactions designated as convergent cascade involving a bi-substrate and a single product without intermediate formation was described [50]. 

An alternative strategy for the synthesis of optically pure amino acids lies in the combination of N-acyl amino acid racemases (NAAAR) with stereoselective N-acetyl amino acid acylases (Scheme 3.5). 

Multi-enzymatic cascades are usually easier to establish than chemoenzymatic or chemo-catalytic cascade reactions as enzymes commonly work in aqueous reaction media and often display similar temperature and pH requirements for optimal performance.