Reconstruction and biologic activity of mouse monoclonal

73
UNIVERSITY OF TARTU FACULTY OF SCIENCE AND TECHNOLOGY INSTITUTE OF MOLECULAR AND CELL BIOLOGY DEPARTMENT OF MICROBIOLOGY AND VIROLOGY Aleksei Suslov Reconstruction and biologic activity of mouse monoclonal antibodies inhibiting RNA-dependent RNA-polymerase of the Hepatitis C Virus Master of Science thesis Supervisor: Andrei Nikonov, M. Sc. Department of Biomedical Technology, Institute of Technology, University of Tartu TARTU 2010

Transcript of Reconstruction and biologic activity of mouse monoclonal

UNIVERSITY OF TARTU

FACULTY OF SCIENCE AND TECHNOLOGY

INSTITUTE OF MOLECULAR AND CELL BIOLOGY

DEPARTMENT OF MICROBIOLOGY AND VIROLOGY

Aleksei Suslov

Reconstruction and biologic activity of mouse

monoclonal antibodies inhibiting RNA-dependent

RNA-polymerase of the Hepatitis C Virus

Master of Science thesis

Supervisor: Andrei Nikonov, M. Sc. Department of Biomedical Technology,

Institute of Technology,

University of Tartu

TARTU 2010

2

Table of contents

ABBREVIATIONS ............................................................................................................................................... 4

INTRODUCTION ................................................................................................................................................ 6

1. BACKGROUND ......................................................................................................................................... 9

1.1. ANTIBODY STRUCTURE AND FUNCTION ............................................................................................ 9

1.1.1. Antibodies – primary effector molecules in humoral immunity ........................................ 9

1.1.2. Antibody structure .......................................................................................................... 10

1.1.3. Organization of immunoglobulin genes .......................................................................... 13

1.1.4. Fine structure of variable regions. .................................................................................. 16

1.1.5. Canonical structures ........................................................................................................ 17

1.2. DESIGNING ANTIBODIES FOR THERAPY ........................................................................................... 19

1.2.1. Chimeric and humanized mAbs ....................................................................................... 19

1.2.2. Intracellular antibodies (intrabodies) .............................................................................. 21

2. EXPERIMENTAL PART ............................................................................................................................ 24

2.1. AIMS OF THE THESIS .................................................................................................................. 24

2.2. METHODS ............................................................................................................................... 24

2.2.1. Plasmids, bacterial strains and media. ........................................................................... 24

2.2.2. Total RNA isolation with TRIzol reagent ......................................................................... 25

2.2.3. Cloning procedures: ........................................................................................................ 26

2.2.3.1. Reverse Transcription-Polymerase Chain Reaction (RT-PCR) ................................................... 26

2.2.3.2. Restriction Digestion of DNA (RD) ............................................................................................ 27

2.2.3.3. Ligation ..................................................................................................................................... 28

2.2.4. DNA sequencing and analysis. ........................................................................................ 28

2.2.5. Plasmid construction ....................................................................................................... 28

2.2.5.1. Site-directed mutagenesis ........................................................................................................ 28

2.2.5.2. Restriction-free cloning. ........................................................................................................... 30

2.2.5.3. Design and construction of chimeric 7G8 mAb genes .............................................................. 32

2.2.5.4. Design and construction of SapI vector system. ...................................................................... 32

2.2.5.5. Generation of F-2A bicistronic constructs ................................................................................ 33

2.2.5.6. Grafting of 7G8M FR1 regions to 7G8H antibody..................................................................... 34

2.2.6. Production and purification of recombinant mAbs ......................................................... 34

2.2.7. HCV RdRp purification ..................................................................................................... 36

2.2.8. Protein analysis and quantification methods .................................................................. 36

2.2.8.1. Protein quantification using Bradford protein assay ................................................................ 36

3

2.2.8.2. Western blot ............................................................................................................................ 37

2.2.9. Enzyme-linked Immunosorbent Assay (ELISA)................................................................. 37

2.2.10. Changing mAb buffer by ultrafiltration .......................................................................... 38

2.2.11. Primer-dependent RdRp assays ...................................................................................... 38

3. RESULTS AND DISCUSSION .................................................................................................................... 40

3.1.1. Cloning of antibody variable regions .............................................................................. 40

3.1.2. Construction of chimeric 7G8 antibody genes ................................................................ 42

3.1.3. Generation of a vector system for high-throughput construction of recombinant

antibody genes. ............................................................................................................... 44

3.1.4. Construction of a panel of recombinant 7G8 and 8B2 monoclonal antibodies. .............. 48

3.1.5. Design and construction of bicistronic mAb expression vectors utilizing the F-2A peptide

sequence. ........................................................................................................................ 49

3.1.6. Expression and purification of enzymatically active NS5B polymerase. ......................... 52

3.1.7. Biologic activity of recombinant 7G8 and 8B2 mAbs ...................................................... 53

3.1.7.1. Biologic activity of recombinant mAbs in antigen binding assay (ELISA).................................. 54

3.1.7.2. Biologic activity of recombinant mAbs in primer dependent RdRp assay. ............................... 55

3.1.8. Identification of mutations, possibly enhancing the biologic activity of 7G8M mAb. ..... 59

3.1.9. Exchanging FR1 regions of 7G8H antibody for those of 7G8M. ...................................... 60

SUMMARY ..................................................................................................................................................... 61

RESÜMEE ....................................................................................................................................................... 63

ACKNOWLEDGEMENTS .................................................................................................................................. 65

REFERENCES ................................................................................................................................................... 66

4

Abbreviations

(+)ssRNA single-strand RNA of positive polarity

Ab antibody

ADCC antibody-dependent cellular cytotoxicity

ATP adenosine triphosphate

bp basepair

BPV-1 Bovine Papilloma Virus type-1

BSA bovine serum albumin

CDR complementarity-determining region

CIAP calf intestine alcaline phosphatase

CLL chronic lymphocytic leukemia

CMV Cytomegalovirus

cpm counts per minute

CTCL cutaneous T-cell lymphoma

CTP cytidine triphosphate

DEPC diethylpyrocarbonate

DTT dithiotreitol

EBV Epstein-Barr Virus

ECL enhanced chemiluminescense

EGTA ethylene glycol tetraacetic acid

ELISA enzyme-linked immunosorbent assay

ER endoplasmic reticulum

F(ab) fragment, antigen binding

F(c) fragment, crystallizable

FDA Food and Drug Administration

FMDV Foot-and-Mouse Disease Virus

FR framework

GF/C glass fiber/cellulose

GMP guanosine monophosphate

GTP guanosine triphosphate

HAMA human anti-mouse antibodies

HC heavy chain

HCV Hepatitis C Virus

HPLC high performance liquid chromatography

HRP horseradish peroxydase

IACT Intracellular Antibody Capture Technology

Ig immunoglobulin

IPTG isopropyl-b-D-thiogalactoside

LC light chain

mAb monoclonal antibody

M-MuLV Moloney Murine Leukemia Virus

NFDM non-fat dry milk

Ni-NTA nickel-nitriloacetic acid

NLS nuclear localization signal

NS3 non-structural protein 3

NS5B non-styructural protein 5 B

nt nucleotide

5

NTP nucleotide triphosphate

OD optical density

ORF open reading frame

PBS phosphate buffered saline

PVDF polyvinylidene fluoride

RD restriction digest

RdRp RNA-dependent RNA-polymerase

RSV Respiratory Syncytial Virus

RT reverse transcriptase

RT-PCR reverse transcription polymerase chain reaction

scFv single-chain fragment variable

SDM site-directed mutagenesis

SDS sodium dodecyl sulfate

SDS-PAGE SDS-polyacrilamide gel electrophoresis

TBSV Tomato Bushy Stunt Virus

TCA trichloroacetic acid

UTP uridine triphosphate

6

Introduction

Antibodies (Abs), or immunoglobulins, are Y-shaped protein molecules capable of

recognition and discrimination of hundreds of millions of self and nonself substances (termed

antigens) in a host organism. Antibodies are the central component of adaptive immune

system and can be found on the membrane of B-cells (as B-cell receptors (BCR)), or in blood,

in a secreted soluble form [1]. All BCR molecules on the membrane of a single B-cell are

identical, making them specific to only one antigen. After first encounter with its antigen, B-

cell can differentiate into plasma cell and start secretion of large amounts of the same

antibody counteracting the invading microorganism. This might happen only when specific

costimulatory signals are present and antigen is identified as non-self entity by innate immune

system component (for review see Janeway 1989 [2]).

Antibodies are heterodimeric molecules, typically consisting of four polypeptide

chains – two identical heavy chains (~50 kDa) and two identical light chains (~25 kDa) [3-5].

Each light chain is linked to a heavy chain, and the heavy chains are linked to each other by

disulfide bonds [4]. The N-terminal parts of heavy and light chains (the arms of the Y-shaped

molecule) are termed as F(ab) (fragment, antigen binding) and contain regions of high

variability in amino acid sequence [6]. These regions are known as “variable domains” (VH

for heavy chain, and VL for light chain). Each B-cell clone expresses antibody with variable

domains different from those of any other antibody, expressed by any other B-cell clone. Each

variable domain contains unique complementarity determining regions (CDR). The CDRs of

both heavy and light chains form loops of various conformations and interact directly with the

antigen [7-8]. The remaining parts of the heavy and light chains are termed as constant

regions. Constant domains of the antibody heavy chains (constituting the stem of the Y-

shaped molecule) are called F(c) region (fragment, crystallizable). The F(c) region triggers

different effector functions (such as activation of complement system, phagocytosis, cell lysis

by cytotoxic lymphocytes) – cellular mechanisms aimed at elimination of antigens [1, 9].

The great diversity of the antibody repertoire allows isolation of mAbs against

virtually any possible antigen. Due to their exquisite specificity and high affinity to a

particular antigen, antibodies are of great interest for therapeutical applications. After

introduction of hybridoma technology (generation of immortal hybrid cells from myeloma

and mAb-producing B-cell) [10] a lot of murine mAbs with therapeutic potential was

generated and launched into clinical trials. Unfortunately, the vast majority of these mAbs

7

failed, because they were ineffective in activation of effector functions in humans. Induction

of the anti-immunoglobulin response, which triggered rapid clearance of mAbs from patient’s

blood was one of the primary issues associated with antibodies deployment as therapeutics.

Such anti-mAb response was termed human anti-mouse antibodies response (HAMA).

Further attempts to make the antibodies less immunogenic and more effective were

undertaken mostly in two directions. First, human hybridomas were generated. Second, mAb

sequence was modified to make it more “human”-like, while maintaining its antigen-binding

specificity. The latter technique included generation of chimeric mAbs in which murine

variable regions were joined to human constant regions (chimerization), and humanization of

antibodies by reshaping its surface (grafting only the CDRs from murine antibody to an

antibody of a human source). Chimeric and humanized mAbs proved to be effective in

clinical studies also reducing HAMA response, at least in some cases. Development of fully

human antibody production technologies (such as phage display and transgenic mice) in

1990-s has lessened the interest towards chimerization and humanization of murine mAbs.

However, humanized antibodies constituted approximately half of all mAbs being in phase 3

clinical studies in 2005 [11].

Further advances in genetic engineering techniques allowed the use of antibodies in a

new format, such as antigen-binding fragments (Fab) or single-chain fragment variable (scFv)

molecules, while retaining the antigen-binding properties of the original mAb. scFv molecules

represent the VH and VL domains of an antibody, joined together with a flexible amino acid

linker. If correctly designed, such molecules do not need the intra-domain disulfide bonds and

can fold properly in the reducing cellular environment and function inside the cell [12].

Antibodies or antibody fragments that perform their functions inside the cell – in cytoplasm,

nucleus or endoplasmic reticulum (ER) – are called intracellular antibodies, or “intrabodies”.

Intrabodies functioning in ER are also termed as “retained antibodies”, because they contain a

ER retention signal. Due to their small size and ability to function inside cell, scFv antibodies

have gained popularity during the last two decades, stimulating the development of

technologies aimed at isolation of functional intrabodies from large recombinant scFv

libraries.

The theoretical part of this study is dedicated to antibody role in immunity, its

structure-function relationship, therapeutic potential of monoclonal antibodies and several

technologies used for design of therapeutical antibodies and intrabodies.

8

Experimental part describes cloning of antibody variable domains, construction of

chimeric mouse/human and mouse recombinant antibodies, expressed whether from one or

two promoters, and biologic activity of these recombinant constructs compared to their

hybridoma counterparts.

9

1. Background

1.1. Antibody structure and function

1.1.1. Antibodies – primary effector molecules in humoral immunity

Antibodies are glycoproteins, which belong to immunoglobulin superfamily, and are

frequently called immunoglobulins. Antibodies can recognize and bind an antigen – a foreign

substance that can be introduced to an organism for example during viral or microbial

infection. Antibodies bind their antigens with very high affinity, at the same time being highly

specific. Adaptive immune system has and is capable of generating large repertoire of

antibodies specific for various antigens. These immunity properties are vital for the host

organism survival.

Antibodies are expressed by mature B-lymphocytes initially in a form of a membrane

receptor, or B-cell receptor (BCR) [1]. When a B-cell first time encounters an antigen that

matches its BCR, the antigen binding together with proper costimulatory signals (for review

see Janeway 1989 [2]) induces several signal transduction pathways, which result in

activation of B-cell – expression of genes responsible for augmentation of its immunological

functions. After receiving of additional signals from T-helper cells, the B-cell starts to

proliferate (clonal expansion) and its progeny differentiates into plasma cells or into memory

cells [13-14]. Memory B cells have longer life span than the naïve B-cell (one that has not

previously encountered antigen) and express the same membrane-bound antibody as their

parent B-cell. These cells are needed for fast induction of immune response in case of

recurrent introduction of the same antigen. Plasma cells, on the contrary, live only for few

days, but during that time they produce an enormous amount of a secreted form of the

antibody. Secreted antibodies are the major effector molecules of humoral immunity. They

circulate in body fluids, where they find the antigen and neutralize it by blocking its activity,

or, bound to the antigen, activate various effector functions that mediate the clearance of the

antigen from the organism.

Antigens are usually complex molecules (with the exception of haptens) and contain

multiple structures on their surface where antibodies can bind. The part of the antigen

recognized by an antibody is called “epitope” or “antigenic determinant”. Epitopes can be

10

linear or conformational. In case of a peptide antigen, linear epitope is a continious stretch of

amino acids, whereas conformational epitope is a particular three-dimensional structure on the

surface of the antigen, that can be formed from different parts of the polypeptide chain.

Normally, one antibody can recognize only one epitope. Consequently, different antibodies

generated during immune response may bind to different epitopes of the antigen, without

interferring with each other.

Each plasma cell expresses a unique antibody during its whole life cycle. Such

antibody is termed as monoclonal antibody. Polyclonal antibodies are mixture of monoclonal

antibodies secreted by single type B-cells in response to a specific antigen. Polyclonal

antibodies recognize different epitopes on the same antigen.

1.1.2. Antibody structure

The name “immunoglobulins” is derived from the initial finding that antibodies

migrate with globular proteins during the electrophoresis of the antibody-containing serum

[15]. By structure, antibodies are heterodimeric multichain glycoproteins typically consisting

of four peptide chains: two identical heavy chains (HC) of ~50 kDa molecular weight, and

two identical light chains (LC) of ~25 kDa molecular weight (Figure 1) [3-5]. One light and

one heavy chain form a heterodimer, held together by covalent disulfide (S-S) bond [4], and

several noncovalent interactions. These light-heavy chain complexes in turn dimerize to form

a functional Y-shaped antibody molecule (Figure 1). Heavy chains of these LC-HC

heterodimers are also linked to each other by disulfide bridges and noncovalent interactions

[4]. The „arms“ of the molecule are responsible for the antigen binding and are called F(ab)

fragments (Figure 1) (fragment, antigen binding) [16-17]. The “stem” of this Y-like structure

is called F(c) fragment (Figure 1) (fragment, crystallizable). It is capable of binding to

specific receptors on a cell membrane and is responsible for antibody effector functions, for

example, activation of proteins of the complement system [9]. These structural and functional

properties were established by proteolytic cleavage of immunoglobulins with pepsin or papain

(Figure 2) and examination of the biological activity of obtained products [9, 16-20].

11

Figure 1. Schematic diagram of immunoglobulin structure. N-terminal portions of both chains are variable

regions (VH and VL), which are responsible for antigen binding. The remainders of the molecule are constant

regions, which define the antibody isotype and subclass (for light chain). Hinge region providing flexibility to

antigen-binding regions is drawn in black. The region below hinge is called Fc and it is responsible for activation

of effector functions. Figure and legend source: Kuby Immunology 5-th edition [1].

The N-terminal fragments (~110 aa in length) of both heavy and light chains, the

“tips” (or the “hands”) of the Y-shaped molecule arms, exhibit great sequence diversity

between antibodies of different specificities. These regions are called variable regions (or V

regions) and designated as VH and VL for heavy and light chain, respectively (Figure 1) [1].

The remaining parts of the antibody chains are termed as constant regions (CH and CL).

Constant regions of heavy chains consist of three (or four) structurally conserved constant

domains – CH1, CH2, CH3 (and CH4) [1]. Light chains contain only one constant domain,

adjacent to the variable region. The CH1 domain of the constant chain (juxtaposed to the VH

region) associates with CL domain of the light chain, and together they serve as a stable basis

for paired VH and VL domains, which contain antigen-binding sites.

12

Figure 2. Prototype structure of IgG, showing disulfide bonds. Fragments produced by various treatments

(pepsin, papain) are also indicated. Figure and legend source: Kuby Immunology 5-th edition [1].

Based on the amino acid sequences of their constant regions, antibody heavy chains

are classified into five groups, called isotypes – α, µ, γ, δ, and ε. Within the bounds of a

particular isotype, sequences of constant chains are very conserved, with the exception of α

and γ isotypes, which can have minor differences and therefore are divided into subisotypes

(γ1, γ2, etc.). The type of the heavy chain defines the class of the immunoglobulin molecule –

IgA(α), IgG(γ), IgM(µ), IgD(δ) or IgE(ε). IgG and IgA class antibodies are divided into

subclasses, depending of the subisotype of the heavy chain. In humans, there are 2 different

subisotypes for α chain, and 4 subisotypes for γ chain. The respective immunoglobulin

subclasses are IgA1, IgA2, IgG1, IgG2, IgG3 and IgG4. For light chains there are only two

types of constant regions – kappa (κ) and lambda (λ). On the basis of the few amino acid

substitutions, λ chains are divided into subtypes - λ1, λ2, λ3, (λ4) - 4 subtypes in humans and

3 subtypes in mice. Each immunoglobulin class can have either κ or λ light chain, but never

both in the same molecule. Heavy chains that consist of three CH domains also have a

proline-rich spacer region – hinge - between CH1 and CH2 domains (Figure 1). Hinge is very

13

flexible and allows a wide-range motion of a F(ab) fragment relative to both F(c) and to the

other F(ab) [21-22].

Immunoglobulin variable regions are homologous in overall structure among different

antibodies, but are very diverse in their amino acid sequence in antibodies of different

specificities, with the major part of this diversity falling into the sequences of the antigen-

binding sites. At the same time constant regions are very conserved in their sequence and can

be divided into small number of different types and subtypes. Due to this diversity in variable

regions, very large repertoire of antibodies can be generated in a single organism. The reason

for this is very simple: antigens can be very different in structure, and in order to be effective

against all of them, the immune system should have huge enough number of different

potential antigen binders prior to antigen exposure, so that virtually any antigen could be

bound by pre-existing antibodies as soon as possible. At the same time each antibody has to

be capable of activating series of effector functions. This explaines the limited number of

constant region sequences and their conservation. Summarizing all of the described features

of antibody structure, it becomes clear that these molecules are designed in such a way, that

they could be on one hand easily adjusted to rapidly changing conditions (introduction of new

pathogens, or mutation of old ones) and on the other hand could always perform the basic

activatory functions. Mechanism for generation of this very interesting duality should be

different from mechanism for generation the majority of other molecules, but at the same time

evolutionarily conserved on a gene level. The organization of immunoglobulin genes

completely explains this phenomenon.

1.1.3. Organization of immunoglobulin genes

Each type of the antibody chains (the κ and λ light chains and the heavy chains) is

encoded by different gene families, which locate in separate chromosomes. These gene

families are also known as multigene families, because they consist of multiple gene segments

of different types (Figure 3). The gene segments are rearranged with each other in the process

of B-cell maturation by means of recombination. Only one segment (from the pool of multiple

sequences) of each type is present in a final gene that is transcribed into mRNA and expressed

as the immunoglobulin chain. The phenomenon of this DNA rearrangement was first

discovered by Tonegawa in 1976 [23]. His group isolated genomic DNA from mouse embryo

cells and from differentiated plasmocytoma cells (type of B-cell cancer, that produces κ-light

chains), digested the DNA with restriction enzyme and analyzed the products by Southern

14

blot method, using a radioactively labelled κ-light chain mRNA and its 3’-prime end half as

probes. Hybridization patterns for the genomes of embryo cells and plasmocytoma cells were

completely different. In case of rearranged DNA (plasmocytoma) one large fragment

hybridized to both probes, whereas in germ-line (embryo cells) two smaller fragments were

identified, from which one hybridized to both probes, and another to only full mRNA probe.

From the results of this experiment it was deduced, that sequences coding for variable and

constant regions are located separately in genomic DNA, and joined together to create the

continious gene in differentiating lymphocytes [23]. This hypothesis was confirmed in

consequent studies [24-26]. The sequencing of corresponding chromosomal regions further

proved the “multigene” structure of the immunoglobulin genes and allowed to define more

precisely their organization, quantity and spatial distribution [27-28].

Figure 3. Organization of the immunoglobulin germ-line gene segments in the mouse: (a) λ-light chain, (b) κ-

light chain and (c) heavy chain. The light chains are encoded by V, J and C gene segments. The heavy chain is

encoded by V, J, D and C gene segments. The distances in kilobases (kb) between different segments are shown

below each chain diagram. Figure and legend source: Kuby Immunology 5-th edition [1]

Now it is known, that the heavy chain gene family contains four different types of

segments – V (variable), D (diversity), J (joining) and C (constant) and the light chain family

– only three types: V, J and C (either κ or λ) (Figure 3). During the somatic recombination,

these fragments are joined together into one continious transcriptional unit (Figure 4).

Rearranged VDJ fragment corresponds to the heavy chain variable region, and VJ fragment –

to the variable region of the light chain. The C fragments encode the respective constant

regions. All of the known Ig gene families also contains a short sequence upstream to each V

region gene, coding for 19 aa long leader (or signal) peptide (L), which is required for the

15

transport of the newly synthesized peptide to the endoplasmatic reticulum (ER) where it

undergoes the correct assembly and glycosylation. Before the antibody is secreted or sent to

the membrane of the B-cell, the signal peptides are cleaved from both H and L chains, and

assembled antibody molecule does not contain any leader sequences.

Figure 4. The process of the heavy chain gene rearrangement and RNA processing. A DH to JH and VH to DHJH

joinings are necessary to generate functional heavy chain gene. Rearrangement of light chains is analogous,

except the absence of D segment. Each C gene is drawn as a single coding sequence; in reality, each is organized

as a series of exons and introns. Rearrangement of the light chain gene occurs similarly. Figure and legend

source: Kuby Immunology 5-th edition [1]

During the process of somatic recombination, first, assembly of V-(D)-J fragments

takes place, then, following transcription and mRNA splicing, VDJ or VJ fragment is joined

to the corresponding C segment. The sequences coding for the constant regions are presented

by only one gene segment for each isotype of heavy chain, one segment for Cκ chain and 4

segments for the λ constant chain. This fact explains the constancy of these regions in all

antibodies. The number of V, (D) and J segments is much higher. For example, human heavy

chain variable regions contain 44 different VH segments with an open reading frame [27], 27

D segments [29] and 9 J segments [30]. By recombination of these segments a huge number

16

of rearranged genes can be generated. Further diversification of the antibody repertoire

involves a process known as somatic hypermutation, during which mutations are introduced at

a high rate into the VH and VL coding regions of rearranged immunoglobulin genes [31].

Taken together, both the somatic recombination and hypermutation contribute greatly to the

antibody diversity of a single organism.

1.1.4. Fine structure of variable regions.

Although, the sequences of variable regions can be very different, the overall structure

of these domains is conserved. Analysis of antibody molecule structure by x-ray

crystallography [7, 32-39] revealed that immunoglobulin domains are packed into a particular

type of protein structure known as the immunoglobulin fold. Each domain contains two β-

pleated sheets, which pack face to face ~10 Å apart from each other, with the angle of 30°

between them (Figure 5a). These β-pleated sheets are linked by a conserved disulfide bridge

and consist of antiparallel β-strands (Figure 5a), which are stabilized laterally by hydrogen

bonds and are terminally connected by loops of various length (Figure 5a, 5b). In variable

domains these loops are encoded by the 6 hypervariable regions (CDRs) – 3 in VH and 3 in

VL – and form the antigen-binding site (Figure 5a, 5b). The loops are designated as H1, H2

and H3 for VH domain, and L1, L2 and L3 for VL domain.

Figure 5. A) The schematical structure of immunoglobulin VL domain. Strands of β-sheet are epresented by

ribbons. Hypervariable loops are labelled as L1, L2 and L3. L2 and L3 are hairpin loops that link adjacent β-

sheet strands. L1 links two strands that are part of different β-sheets. The VH domain and its hypervariable loops

H1, H2 and H3 have homologous sequences. B) The arrangement of the 6 hypervariable regions that form the

antibody binding site. The squares indicate the position of residues at the ends of the β-sheet strands in the

framework regions. Figure and legend source: Chothia and Lesk (1987) [8].

17

First attempts to correlate the amino acid sequences of variable domains with their

possible structure and function were made by Kabat and colleagues in 1970 [6]. As more and

more sequence data for variable regions were appearing, they could be compared in order to

reveal the structure-function relationship of these domains, responsible for antigen binding.

To systematize the sequence data available for V regions at that time, Kabat’s group created a

database of the aligned amino acid sequences of light chain variable regions and analyzed the

amino acid variabilities at each position. As a result, they have identified three regions of

maximal variation [6]. When the database was supplemented with sequences of heavy chain

variable regions, the same pattern of variability in these sequences was observed. These

hypervariable segments were originally called hypervariable regions and were suggested to

be the antigen-binding sites of the antibody molecule, what was later proved by x-ray

crystallographic studies [7, 32-39]. They correspond to the loops that connect the β-strands

and make direct contact with the antigen. These loops are designated as H1, H2, H3 and L1,

L2, L3 for VH and VL domains, respectively. Because the antigen-binding site is

complementary to the antigen structure, these hypervariable regions are also called the

complementarity-determining regions (CDR). The remainders of the variable region

sequences are known as framework regions (FR) and make up a backbone to which the

hypervariable loops are attached. The framework regions have much lesser variation in amino

acid sequences. They provide the rigid scaffold necessary for maintaining the structural

integrity of the molecule. Several amino acid residues at specific positions were identified,

which can also play important role in CDR conformations [40-43]. This means that despite

main variability in antigen-binding sites is generated by hypervariable regions, framework

regions can “fine-tune” the CDR orientation and, consequently, antibody affinity to antigen.

1.1.5. Canonical structures

Another important finding made by Kabat when comparing the sequences of the

hypervariable regions then known, was the discovery of several conserved residues in these

sequences – 13 in VL regions and 7 in VH regions [44]. It was suggested that these residues

might be responsible for the structure of the hypervariable regions, not for specificity. It was

also supposed that these amino acids have certain positions in the antibody variable regions

and are needed to give a specific conformation to the main-chains of the hypervariable loops.

Padlan and Davies analyzed immunoglobulins of known structure and showed, that in several

cases the hypervariable regions of the same size, but with different sequences have the same

18

main-chain conformations [45]. Another analysis of immunoglobulin sequences made by

Padlan revealed that some residues in hypervariable regions, and residues buried within the

domains in the known structures, are conserved [46]. In 1987, Chothia and Lesk examined

immunoglobulins with known atomic structures and identified several residues that through

various interaction with each other or ability to assume unusual values of torsion angles, were

primarily responsible for the main chain conformations of the hypervariable regions in those

structures [8]. The conformations are determined by the interaction of a few residues at

certain sites in the hypervariable regions, and, in some cases, in the framework regions.

Subsequent analysis of the sequences of immunoglobulins of unknown structures showed,

that many of them contained at least one set of residues responsible for a specific

hypervariable conformation (from those observed). These observations suggested, that despite

the sequence variability, most of the hypervariable loops in immunoglobulins have a limited

number of main-chain conformations which were called “canonical structures”. Possible

canonical structures were described for all hypervariable regions, except the H3 loop [8, 47].

The sequence coding for this loop is located at the junction of rearranged V, D and J

segments, therefore it has much more structural variety than all other hypervariable regions.

Using this canonical structure model, Chothia and colleagues tried to predict the structures of

the hypervariable regions of the D1.3 antibody prior to the experimental determination. It was

found that this antibody had the hypervariable regions of the same size, as in previously

analized structures, and the same, or similar, amino acids at key positions determining the

type of the canonical structure [48]. The comparison of the predicted structure with the

obtained crystal structure showed that the conformations of 4 of 6 hypervariable regions had

been predicted correctly, demonstrating the efficiency of this model [48].

The fact, that at least five of six hypervariable regions of any antibody have only a

small discrete set of conformations that can be easily predicted is of the utmost importance for

the antibody engineering. Understanding, which key residues affect the main chain

conformations of the hypervariable region, it is possible to transfer the antigen-binding

specificity from one antibody to another, and at the same time reshape the surface of the host

antibody to match it with the donor antibody if it is needed for corect orientation of the

hypervariable loops. This feature can be extremely useful in the engineering of therapeutic

antibodies (will be discussed below).

19

1.2. Designing antibodies for therapy

1.2.1. Chimeric and humanized mAbs

Soon after the discovery of the antibodies, their therapeutic potential became obvious.

Antibodies could help in solving many medical problems, such as organ grafting, cancer

therapy, or treatment of viral and microbial infections. The first report describing the method

allowing to produce monoclonal antibodies in large quantities was received from Köhler and

Milstein in 1975 [10]. The method involved generation of murine hybridomas – cell lines,

produced by fusion of a mAb-producing B-cell with a myeloma cell. Resulting hybrid cells

were immortal and produced high enough amounts of the monoclonal antibodies. Despite the

initial procedure of creating hybridomas was unreliable, the technique made revolution in the

field of immunology and became a general method for production of monoclonal antibodies.

Many mAbs against various antigens were generated since that time aimed to be used as

therapeutics. However, when these murine monoclonal antibodies entered the clinical study,

their limited potential to be used as therapeutics became evident. Murine antibodies were

identified as antigens by patients’ immune system and were rapidly cleared from blood by the

immune response involving the human anti-mouse antibodies (HAMA). They were also

ineffective in activation of effector functions in humans. However, this obstacle did not

diminish interest towards these very promising molecules, but rather stimulated the

development of various methods and technologies for production of therapeutic mAbs. These

methods were aimed whether at a development fully human antibodies using the hybridoma

technology (what was not really successful at the beginning), or at lessening the antibody

immunogenicity through decreasing the percentage of murine sequences in antibody by

changing them to sequences of human origin. Two main techniques in this approach were

chimerization and humanization of murine mAbs.

20

Figure 6. Schematic drawing of murine, chimeric and humanized antibodies. Murine sequences are shown in

blue and human sequences in red. Source: Clark, M (2000) [49].

Chimeric mAbs are antibody molecules that contain variable regions derived from a

murine source, and constant regions derived from a human source (Figure 6). The first reports

concerning the creation of chimeric mAbs and their biologic activity were received from

Morrison et al. [50] and Boulianne et al. [51] in 1984. They have shown that chimeric

antibodies maintain their biologic activity at a level comparable with that of original murine

mAbs. In the animal models it was shown that replacing the murine Fc region could reduce

the host immune response, however chimeric mAbs could still induce the anti-idiotypic

responses (responses against antibody variable regions), although these were delayed and

weaker [52-53]. Despite the chimeric mAbs were superior to murine mAbs in terms of

therapeutic application, the number of chimeras entering the clinical studies starting from

1987, was lower than that for murine antibodies. However, their approval success rate was

much higher than that for murine mAbs. According to the data available on July 2005, there

were 5 chimeric mAbs being used worldwide in medicine for treatment of various diseases

[11].

Chimeric mAbs did not receive a wide spread occurrence probably, because the

strategy of humanization, which was first applied only few years after the first chimeric mAb

construction seemed to be superior to the chimerization. Humanization approach was based

on assumption, that greater percentage of fully human sequences in mAb would provide

greater reduction in HAMA response. In a process of humanization, only those regions that

make direct contact with the antigen (CDRs) are grafted into a framework of a human

antibody (Figure 6). The first report of successfully humanized antibody, which maintained its

functions after the procedure was received in 1986 [54]. The first successfully humanized

21

therapeutic antibody was produced in 1988 by a group of scientists from Cambridge

University [55]. They have humanized the rat antibody CAMPATH-1 (raised against human

lymphocyte proteins) that might have a therapeutic application in treatment of chronic

lymphocytic leukemia (CLL). The humanized mAb had entered the clinical studies and turned

out to be effective in patients with non-Hodgkin’s lymphoma, and at the same time it did not

induce the anti-immunoglobulin response [56]. The humanized CAMPATH mAb was finally

approved as therapeutic by U. S. Food and Drug Administration (FDA) in 2001, and now it is

used under the name Alemtuzumab for the treatment of chronic lymphocytic leukemia (CLL),

cutaneous T-cell lymphoma (CTCL) and T-cell lymphoma [57-58]. This first successful

humanization was followed by many similar works on humanization of various mAbs against

cancerous or viral antigens [59-66]. However, the humanization turned out to be a difficult

process, and many humanized mAbs reported had a reduced affinity to the antigen, compared

to the original mAb [55, 60, 62, 65, 67-68]. As it is been mentioned in “Canonical structures”

section, specific framework residues are responsible for the main chain conformations of

variable loops and it has to be considered during humanization [68-69]. All critical FR

residues should be also transferred from donor mAb to the final constructs. The prediction of

the conformation and identification of all important FR residues requires a thorough analysis

of sequence and structure of donor and host mAbs, as well as a careful choice of the donor

framework. Although, after the humanization the percentage of donor amino acids in acceptor

mAb may be the same as for chimeric mAbs, humanized antibodies are yet less immunogenic,

probably because of the more “human-like” overall structure.

Development of fully human antibody production technologies (such as phage display

and transgenic mice) in early 1990s has lessened the interest towards chimerization and

humanization of murine mAbs. However, humanized antibodies constituted the majority of

mAbs entered clinical studies during the period from 1992 to 2002 years [11]. As of data

available in July 2005, humanized antibodies made up approximately half of all mAbs being

in phase 3 clinical studies [11].

1.2.2. Intracellular antibodies (intrabodies)

Naturally occurring antibodies are extracellular molecules. They are synthesized in

endoplasmic reticulum (ER) and secreted into the blood, where they bind antigens and

activate different effector function via the interaction of the F(c) domain with proteins of the

complement system [9], or with receptors present on the membrane of white blood cells. Even

22

without activation of the effector functions, antibodies are able to neutralize antigens in

different ways by simply binding to them. Abs can prevent various molecular interactions, for

example, block the activity of an enzyme by locking it in an nonfunctional conformation or

blocking its active site(s), or bind to substrate molecules, for instance, viral DNA or RNA,

preventing their access to the enzyme. So, it is reasonable to suppose, that all of these effects,

that could be achieved outside the cell should be achievable inside the cell, as well. This

assumption stimulated the development of intracellular antibodies (or intrabodies). The term

“intrabody” refers to an antibody (or a part of antibody) that functions within the bounds of

the outer membrane of a cell – in the cytoplasm, in the nucleus or in the ER. The first report

confirming that antibody can efficiently function inside a cell was received in 1980, when it

had been shown that after microinjection of purified antibodies into individual cells they

could block the function of the target molecule [70]. Later, it was demonstrated that

functional antibodies could be expressed in mammalian cells, and could be targeted to the

nucleus by utilizing the nuclear localisation signal (NLS) [71]. However, this was rather an

exception. The majority of attempts to express antibodies in cytoplasm had failed, because the

reducing environment of cell cytoplasm prevents the formation of inter- and intradomain

disulfide bridges and, consequently, correct folding of the antibody [72-73]. As activation of

the effector functions is not required from the antibody, functioning inside a cell, “scFv”

(single-chain fragment variable) became the most preferable format for design of intracellular

Abs. scFv is a molecule of ~25 kDa molecular weight, and it is composed of antibody heavy

and light chain variable domains, connected with a flexible amino acid linker (Figure 7). The

adoption of this format removed at least one problem – formation of inter-chain disulfide

bridges, because the domains of the scFv molecule are linked physically, what facilitates their

correct association. However, this technological advance by itself was not enough to generate

stable intrabodies with high expression levels.

Figure 7. Schematic drawing of a typical scFv molecule. VH (blue) and VL (orange) domains are shown,

connected with a peptide linker. N-terminus and C-terminus are designatet as N and C, respectively.

23

Development of a strategy for prediction of stabilizing mutations in immunoglobulin

domains, and implication of a consensus engineering approach (engineering of antibody

domains based on consensus sequences), finally led to the construction of stable scFv

molecules that could be efficiently expressed [74-76]. The approach was based on the analysis

of available immunoglobulin sequence data, determination of consensus sequences and

introduction of stabilizing mutations identified by this analysis. Simultaneously, it was shown,

that scFv possessing a stable framework can fold correctly and function without the disulfide

bond [12]. Further development of in vivo selection strategy for functional intrabodies –

Intracellular Antibody Capture Technology (IACT) – which utilized the modified yeast two-

hybrid screening system [77], allowed isolation of various stable and functional intrabodies

from artificially created recombinant scFv libraries [78-79]. Analysis of their sequences

revealed great similarity of their framework structures and allowed to define the stable

consensus framework that could be used for intrabody engineering and generation of new de

novo intrabody libraries [78-80]. In subsequent experiments it was demonstrated, that grafting

of foreign CDRs into this framework yielded highly stable and functional intrabodies [81].

Thus, intrabody consensus framework seems to be a powerful platform for engineering

intracellular antibodies with various specificities or generation of synthetic intrabody libraries

based on this highly stable sequence.

Starting from 1990, many intrabodies with proved efficiency in blocking

intermolecular interactions was developed (for references see [82-83]). Of particular interest

are several antiviral intrabodies, for example scFv against the NS3 protein of Hepatitis C

Virus (HCV), which is capable of inhibiting the viral replication [84-85], or intrabodies

against RNA-dependent RNA polymerase (RdRp) of Tomato Bushy Stunt Virus (TBSV), that

are capable of protecting plants against the viral infection [86]. The latter intrabodies are also

capable of inhibiting the RdRp-s of heterologous closely related viruses, and even of binding

the HCV RdRp [86]. TBSV and HCV belong to the same supergroup and the replication

strategies of these viruses are similar, what means, that the same approach may work in case

of HCV or other (+)ssRNA virus.

First and last, intrabodies are very promising biomolecules to be used as intracellular

therapeutics. Owing to the development of in vivo intrabody capture technologies the

generation of highly engineered intrabodies is recently becoming a reliable and robust

procedure with great potential and may result in a new wave of therapeutics on the market in

nearest future.

24

2. Experimental part

2.1. Aims of the thesis

The main objective of this study was the construction of various recombinant forms of

mouse monoclonal antibodies 7G8 and 8B2, previously identified in our lab and shown to be

capable of inhibiting the activity of HCV RNA-dependent RNA-polymerase in vitro [87], and

demonstration of their biologic activity. This work is an intermediate step in preparation of

these antibodies to a possible therapeutical use in future. Following specific aims were bound.

First, amplification of variable regions of mAbs 8B2 and 7G8 from hybridoma cDNA.

Second, construction of recombinant mouse/human and mouse/mouse 7G8 and 8B2 genes

using expression vectors (developed by Icosagen Group) with pre-cloned mouse and human

antibody constant regions. Third, development of a high-throughput system for construction

of recombinant antibody genes. Fourth, generation of single-promoter expression constructs

for recombinant 7G8 and 8B2 antibodies, based on Foot-and-Mouse Disease virus 2A peptide

and furin cleavage site. Fifth and sixth, purification of enzymatically active HCV RdRp

(NS5B) for biochemical assays and comparison of the biologic activity of generated

recombinant mAbs to that of original hybridoma 7G8 and 8B2 mAbs.

2.2. Methods

2.2.1. Plasmids, bacterial strains and media.

pJet 1.2/blunt (2974 bp) (Fermentas) - positive selection cloning vector containing a

lethal restriction enzyme gene that is disrupted by ligation of a DNA insert into the cloning

site. Contains the pMB1 rep element responsible for the replication of plasmid, amp

resistance gene, multiple cloning site (MCS). In this work used as an intermediate host for

cloning of antibody variabe regions from hybridoma.

pBluescript II SK+ (Stratagene) - Contains f1 helper phage origin of replication, amp

resistance gene, and MCS within LacZ gene fragment, which allows white/blue screening. In

this work used as an intermediate vector for almost all cloning procedures.

pCMV (Icosagen Group) – Expression vector, that consists essentially of 3 parts –

antibiotic resistance marker (Km), Bovine Papilloma Virus type-1 (BPV-1) E2 or Epstein

25

Barr Virus (EBV) EBNA sequence, which allows this plasmid not to get lost during cell

division, and gene expression cassette(s) (in this case – Ab expression cassettes). Expression

is driven by Cytomegalovirus (CMV) early promoter

pCMV_SV40pA_h-mAb2_42 (here – pCMV42) – pCMV vector with cloned codon-

optimized human IgG1 heavy and kappa light chains

pCMV_SV40pA_m-mAb1_65 (here – pCMV65) - pCMV vector with cloned codon-

optimized mouse IgG1 heavy and kappa light chains

pCMV_SV40pA_m-mAb2_83 (here – pCMV83) - pCMV vector with cloned codon-

optimized mouse IgG2a heavy and kappa light chains

pET-19bNS5b[Con1] (Merck) – Expression vector, containing a HCV NS5B coding

gene, cloned under the control of the bacteriophage T7 promotor. Also contains a

decahistidine tag coding sequence, fused directly to the 5’ end of the NS5B gene.

Bacterial strains:

E.coli DH5α - F- endA1 glnV44 thi-1 recA1 relA1 gyrA96 deoR nupG Φ80dlacZΔM15

Δ(lacZYA-argF)U169, hsdR17(rK- mK

+), λ–. Growth and purification of plasmid DNA.

E.coli BL-21 (DE-3) (Stratagene) - F– ompT gal dcm lon hsdSB(rB

- mB

-) λ(DE3 [lacI

lacUV5-T7 gene 1 ind1 sam7 nin5]). Expression and purification of HCV NS5B polymerase.

All bacterial cultures were grown in Luria-Bertani (LB, Difco) medium (10 g/l bacto-

tryptone, 5 g/l bacto-yeast extract, 10 g/l NaCl) supplied with an appropriate antibiotic at a

final concentration of 100 µg/ml for Amp, and 50 µg/ml for Km.

2.2.2. Total RNA isolation with TRIzol reagent

Total RNA was isolated from hybridoma cell lines using commercially available

ready-to-use TRIzol reagent (Invitrogen). The reagent is a mono-phasic solution of phenol

and guanidine isothiocyanate. TRIzol can disrupt cells and dissolve cell components, at the

same time maintaining the integrity of the RNA. After addition of chlorophorm and

centrifugation, the sample solution is divided into three phases. The upper (aqueous) phase

contains the desired RNA which can be easily recovered by precipitation with isopropyl

alcohol.

Hybridoma cells (~107), producing mAbs 7G8, 8B2 and 10D6 were centrifuged at

270×g for 5 minutes and pellets were homogenized in 1 ml of TRIzol reagent (Invitrogen).

The suspensions were centrifuged for 10 minutes at 12 000×g at +4 °C to remove insoluble

material. After centrifugation supernatants were transferred to new tubes and incubated 5

26

minutes at room temperature to permit the complete dissociation of nucleoporin complexes.

Following incubation, 200 µl of chloroform (per 1 ml of TRIzol) was added to each sample

and tubes were shaken vigorously by hand for 30 seconds. After a 15-min centrifugation at

12000×g at +4°C, the RNA-containing aqueous phase of each sample was transferred to a

new 1.5 ml tube. Further, 500 µl of isopropanol (per 1 ml of TRIzol used for initial

homogenization) was added to each sample and tubes were incubated for 15 min at room

temperature and then centrifuged for 40 min at 16000×g at room temperature (5415R,

Eppendorf) to precipitate and pellet the RNA. Following precipitation, supernatants were

discarded and pellets were washed four times with 1 ml of 70% ethanol (15 min at 16000 ×g

at room temperature). After the last washing step ethanol was discarded and the tubes were

stored opened for 15-20 min at room temperature to remove residual ethanol. Following the

incubation, the RNA was dissolved in 100 µl of RNase-free H2O and additionally incubated

for 30 min at 37 °C in closed tubes to allow complete dissolving of the RNA. RNA yields

were quantified using ND-1000 spectrophotometer (NanoDrop Technologies, Inc.). The

purified RNA yield was up to 180 µg per ~107 hybridoma cells. The 260/230 nm ratio was

2.0, indicationg high enough degree of purity. The integrity of the RNA was analyzed using

denaturing formaldehyde gel-electrophoresis (Figure 11)

2.2.3. Cloning procedures:

2.2.3.1. Reverse Transcription-Polymerase Chain Reaction (RT-PCR)

Reverse transcription of hybridoma total RNA was performed using RevertAid™ First

Strand cDNA Synthesis Kit (Fermentas), that utilises RevertAid™ Reverse Transcriptase

(genetically modified M-MuLV RT). Reactions were performed in a reaction buffer provided

by manufacturer and contained ~1 µg of hybridoma total RNA, 20 units of RiboLock™

RNase Inhibitor, 1 mM dNTP mix, 0.5 µM oligo(dT)18 primer and 20 units of RT.

Diethylpyrocarbonate (DEPC)-treated H2O was added to a total volume of 20 µl.

First, template RNA was mixed with primer in a total volume of 12 µl and denatured

for 15 min at 65 °C. After that, tubes were briefly chilled on ice and all other components

were added. Reaction mixes were incubated for 1 h at 50 °C. Following incubation the

reactions were terminated by heating at 70 °C for 5 min. Reaction products were used directly

for amplification of antibody variable regions by PCR.

27

PCR reactions were performed in a total volume of 50 µl. Reaction mix contained

template DNA (or cDNA), 0.2 µM dNTP mix, 0.5 µM primers, 1x reaction buffer and 1 U of

DNA polymerase – Taq (Fermentas) or Phusion (Finnzymes). During the reaction setup tubes

were constantly held on ice. For Phusion DNA polymerase initial denaturation lasted 30 sec

followed by amplification cycle of 25-35 rounds: 10 sec denaturation at 98 °C, 15-30 sec

primer annealing at 55-72 °C (depending on the annealing temperature of primer) and

extension (time depended on a fragment length, considering sysnthesis rate ~15-30 sec/1000

nt) at 72 °C. If Taq DNA polymerase was used, initial denaturation was performed at 94 for 3

min and during amplification cycles denaturation lasted 1 min at 94 °C. Elongation time was

also increased accordingly to the systhesis rate of Taq polymerase (~1 min/1000 nt).

Reactions were accomplished by final 10 min extension step and stored at -20 °C until

needed.

2.2.3.2. Restriction Digestion of DNA (RD)

DNA digestion reactions were performed using enzymes from Fermentas, according to

manufacturer’s instructions. Enzyme quantity was chosen depending on the amount of DNA.

Generally, 0.5-2 µg of DNA was digested with a 5- to 10-fold excess of enzyme (considering

that 1 unit of enzyme cleaves 1 µg of DNA in 1 h at 37 °C) in a total volume of 20 µl in

appropriate buffer for at least 1 h. When needed, DNA was dephosphorylated after digestion

using Calf Intestine Alcaline Phosphatase (CIAP), (Roche). 3 µl of CIAP (1 u/µl) were added

directly to RD mix and incubated at 37°C for 30 min. The enzyme was inactivated by heating

at 80 °C for 20 min.

Products of PCRs or DNA digestion were resolved on 0.8% agarose gel (or 2%

agarose gel for fragments <200 nt in length), and fragments of correct size were extracted

with „JETquick Gel Extraction Spin Kit“ (Genomed) or „NucleoSpin Extract II Kit“

(Macherey-Nagel) following manufacturer’s recommendations. Alternatively, PCR products

were purified with „JETquick PCR Product Purification Kit“ (Genomed). Purified DNA was

used for downstream applications, such as ligation or PCR.

28

2.2.3.3. Ligation

Ligations occurred in total volume of 20 µl using 5 U of phage T4 DNA ligase

(Fermentas). Approximately 10-20 ng of vector DNA was used per reaction. Molar ratio of

insert to vector was 3 to 1. Reactions were performed at room temperature for 1 h. Up to ¼ of

ligation mix was further used for transformation of chemically competent E.coli DH5α cells.

Bacterial transformation was performed using heat shock method. Transformed cells

were inoculated on LB plates containing an appropriate antibiotic – ampicillin (Amp, final

concentration – 100 µg/µl) (Roche), or kanamycin (Km, final concentration – 50 µg/µl),

depending on a plasmid resistance gene. Plasmid DNA was purified from transformed cells

using NucleoSpin Plasmid QuickPure Kit (Macherey-Nagel), which utilises basic lysis

method. Purified DNA was dissolved whether in Milli-Q water (Millipore) or in Tris-HCl

buffer (pH 8.5).

All cloning procedures were performed exactly as they are described under this

chapter, unless otherwise indicated.

2.2.4. DNA sequencing and analysis.

All constructs generated by PCR were sequenced using sequencing facilities (ABI

3130xl Genetic Analyzer and ABI 3730xl DNA Analyzer) provided by Institute of Molecular

and Cell Biology. Multiple sequence alignments were performed using ClustalW [88]

program provided on European Bioinformatic Institute (EBI) server

(http://www.ebi.ac.uk/Tools/clustalw2/) and analyzed with BioEdit 7.0 software (Tom Hall,

Ibis Therapeutics).

2.2.5. Plasmid construction

2.2.5.1. Site-directed mutagenesis

For site-directed mutagenesis (SDM) of pBSK internal SapI restriction site a modified

approach of Sergeant and Mikaelian [89] was used (described in Wu et al. [90]). (Figure 9)

Primers pBSK_for, pBSK_rev and pBSK_SapI_mut were used in this experiment

(Figure 8).

29

Figure 8. A scheme of SapI site containing region and primers used for site-directed mutagenesis with their

annealing sites indicated. pBSK_for and pBSK_rev are flanking primers, and pBSK_mut is a mutagenic primers.

Restriction sites SacI and XceI used for further subcloning of amplified product are also depicted, along with

SapI restriction site.

Reannealing temperature and temperature at which the pBSK_for and pBSK_rev

primers do not bind to the plasmid DNA were determined practically using gradient PCR.

Further, the mutagenesis was performed in a single PCR reaction using all three primers and

pBSK vector with following PCR program:

1. 94 °C - 1 min

2. 94 °C - 45 sec

3. 72 °C (-1 °C per cycle) - 1 min

4. 72 °C - 1 min

5. GOTO 2 REP 10

6. 94 °C - 45 sec

7. 62 °C - 1 min

8. 72 °C - 1 min

9. GOTO 6 REP 6

10. 94 °C - 45 sec

11. 80 °C (Tra) - 45 sec

12. 42 or 52 °C - 1 min

13. 72 °C - 1 min

14. GOTO 11 REP 3

15. 72 °C - 2 min

16. GOTO 6 REP 6

17. HOLD AT 4 °C

The resulting PCR product of correct size was purified from gel, digested with SacI +

XceI restriction enzymes and ligated to a pBSK digested with the same enzymes, producing

plasmid pBSKmut. Incorporation of the mutation was controlled by SapI digestion.

Reaction mix (100 µl):

10x Phu buffer - 10 µl

2 mM dNTP mix - 10 µl

pBSK_for - 6 µl (60 pmol)

pBSK_rev - 6 µl (60 pmol)

pBSK_SapI_mut - 0.8 µl (8 pmol)

Template DNA - 1.2 µl (120 ng)

Pfu polymerase - 1 µl (2 U)

H2O - 65 µl

30

Figure 9. A schematic drawing of the mutagenesis process. During the first step, a single stranded mutagenic

DNA (mtDNA) is amplified from a plasmid with high Tm mutagenic primer and with high annealing

temperature at which flanking primers cannot anneal. During the second step, the megaprimer is produced with

one of the flanking primers from the smDNA template at low annealing temperature and low denaturing

temperature, at which where the plasmid DNA is not denatured. By repeating the above described procedures

(step 3), the full-length mutated DNA is synthesized first with a megaprimer at high annealing temperature and

then a complementary strand is synthesized by another flanking primer at low denaturing and low annealing

temperatures. Redrawn and modified from Wu et al. (2005) [90]

2.2.5.2.

2.2.5.2.

2.2.5.2.

2.2.5.2.

2.2.5.2.

2.2.5.2.

2.2.5.2.

2.2.5.2. Restriction-free cloning.

A restriction-free cloning method described by Ent and Löwe (2006). was used in this

work for construction of several plasmids such as SapI site containing vectors, F2A peptide

containing vectors and chimeric pCMV42 7G8H vector. This is a two-step PCR-based

approach, which involves amplification of the desired insert with specific primers in such

way, that the ends of the resulting fragment are complementary to the host vector at the

desired site of insertion. The amplification product serves further as a pair of primers in the

linear amplification reaction around a circular plasmid, thus eliminating the need for

restriction endonuclease recognition sequences (Figure 10).

31

Figure 10. Schematic representation of restriction-free cloning process. 1. The fragment to be inserted is first

amplified by a pair of primers, which are partially complementary to the unique insertion sites of the host vector

(red and blue fragments), isolated from dam+ strain. 2. The resulting PCR product serves further as a pair of

primerrs in a linear amplification reaction around a circular plasmid. Once annealed to the vector DNA

polymerase (Phusion in our case) extends and incorporates the gene into a nicked circular molecule (dashed

lines). After the digestion of parental vector with DpnI, the remaining circular double-nicked and double-

stranded DNA is transformed to an appropriate host cell. Redrawn and modified from van den Ent and Löwe

(2005) [91].

At first, a fragment to be inserted was amplified with a pair of ~50 nt, HPLC-purified

primers. In such primers about a half of sequence is complementary to desired insert and

another half is analogous to a host vector sequence upstream or downstream from the site of

insertion. Following the amplification, fragment of correct size was gel-purified and used as a

pair of primers in a linear amplification reaction. When the reaction was completed, buffer

32

was changed to water and the PCR products were digested with 20 U of DpnI for 2 h to

remove methylated parental DNA. Products of correct size were purified from agarose gel and

used directly for transformation. The correctness of insertion and absence of undesired

mutations possibly introduced during PCR was controlled by sequencing.

2.2.5.3. Design and construction of chimeric 7G8 mAb genes

All operations with antibody heavy and light chains were performed in an intermediate

vector – pBluescript II SK+ (pBSK). Human IgG1 heavy chain (together with the downsteam

intron sequence) and kappa light chain were cloned to MCS of pBSK vectors from pCMV42

vector using restriction enzymes XhoI + EcoRI for HC (~2500 bp) and XhoI + Cfr42I for LC

(~750 bp), generating plasmids pBSK42HC and pBSK42LC, respectively. Variable heavy

(VH) and variable light (VL) region coding sequences were changed in these vectors for those

of 7G8 mAb using RF-cloning method. Briefly, 7G8 VH and VL sequences were amplified

from pJET 1.2 vectors using primers HC_7G8_up_site_ins + HC_7G8_down_site_ins for

HC, and LC_7G8_up_site_ins + LC_7G8_down_site_ins for LC (Table 1). The resulting

products were purified from gel and used as a pair of primers (or, a „megaprimer“) in linear

amplification reactions using respective pBSK plasmids as templates (pBSK42HC for heavy

chain and pBSK42LC for light chain). After 2 h DpnI digestion the PCR products were

transformed to E.coli DH5α cells. Purified plasmids were controlled by sequencing. For

eucaryotic mAb expression, modified heavy and light chains were transferred back to

pCMV42 vector using enzymes Eco72I + BshTI for HC and Bsp119I + Cfr42I for LC,

producing a plasmid named pCMV_7G8H.

2.2.5.4. Design and construction of SapI vector system.

Sequences, coding for human HC and LC were transferred to pBSKmut from

pCMV42 just as described previously, generating plasmids pBSKmutHC42 and

pBSKmutLC42, respectively.

Sequences coding for murine IgG1 and IgG2a heavy chains were transferred to

pBSKmut from vectors pCMV65 (IgG1) and pCMV83 (IgG2a). Plasmids were digested with

SacI enzyme and corresponding fragments were gel-purified and ligated to pBSKmut digested

with the same enzyme, generating plasmids pBSKmutHC65 and pBSKmutHC83.

33

Sequence coding for murine light chain was transferred to pBSKmut in the same way

as the human light chain coding sequence, generating plasmid pBSKmut_mLC. Light chain

coding sequence of pCMV83 vector was identical to that of pCMV65 vector. Presence and

correct orientation of insert were confirmed with restriction digest (RD) analysis.

SapI restriction sites were inserted into generated pBSKmut vectors (bearing whether

heavy or light antibody chains) using RF-cloning strategy. First, VH and VL regions were

amplified with SapI restriction site containing primers, that were also partly complementary to

host vectors. Further, these amplification products were used in a linear amplification reaction

to generate plasmids with desired sites. Two SapI restriction sites in opposite orientations

were introduced into each target plasmid. One at the junction between leader sequence and

VH or VL sequence (antisense orientation), and another (sense orientation) whether at a

junction between VH (or VL) and constant region, or inside of a constant region of heavy

chain (CH), few nucleotides downstream the VH/CH junction (only for pBSKmutHC65 and

pBSKmutHC 83 vectors). Generated constructs are listed in a Table 1. Generation of a panel

of recombinant mAb expression constructs using SapI vector system.

Variable heavy and variable light regions of mAbs 7G8 and 8B2 were amplified from

corresponding pJet 1.2 plasmids/hybridoma cDNA using primers containig SapI restriction

site sequences (Table 1). PCR products of correct size were purified and digested with SapI.

pBSKmutSapI vectors containing different heavy and light chains were also digested with

SapI enzyme and ligated together with corresponding SapI-digested PCR fragments. Ligation

products were then transformed into chemically competent DH5α cells. Absence of mutations

was verified by sequencing. Further, HC and LC with changed VH and VL regions were

trasnferred from pBSKmutSapI vectors to pCMV vectors for expression exactly as described

in previous sections. Generated expression constructs were named pCMV_7G8M,

pCMV_8B2M and pCMV_8B2H. For the expression of chimeric 7G8 previously generated

pCMV_7G8H vector was used. The „H“ letter stands for human constant regions and the „M“

letter – for murine constant regions.

2.2.5.5. Generation of F-2A bicistronic constructs

For generation of F-2A constructs pBSK vectors with previously cloned antibody

heavy and light chains (see “Design and construction of SapI vector system”.) were used as

HC and LC donors. Heavy chains were cut out from the pCMV vectors using enzymes

HindIII and SalI. Light chains were cut out with XhoI and Cfr42I. Considering that enzymes

34

SalI and XhoI leave the same overhangs after cleavage, both HC and LC could be then

simultaneously ligated to pBSK vectors cut with HindIII and Cfr42I. The linker containing

DNA coding for furin cleavage site, SGSG spacer and F-2A peptide was inserted between the

heavy and light chains using RF-cloning method. Then, the whole sequence coding for HC

and LC (with a short linker between them) was transferred from each pCMV vector to pBSK

vector, generating pBSKF2A plasmids (Table 1).

A DNA sequence coding for a furin cleavage site, SGSG linker and Foot-and-Mouth

Disease Virus (FMDV) 2-A peptide (F-2A) was kindly provided by Radi Tegova (Icosagen

Group). The sequence was amplified with specific primer pairs (Table 1) and purified from

2% agarose gels. Resulted fragments were fused to pBSKF2A vectors directly between heavy

and light chains using the RF-cloning method. The whole sequence – HC-F-2A-LC – was

transferred from each pBSK vector to pCMV vectors for expression using Eco72I and Cfr42I

enzymes (generated plasmids – pCMVF2A, Table 1). All constructs generated by PCR were

analyzed by sequencing.

2.2.5.6. Grafting of 7G8M FR1 regions to 7G8H antibody

The VH and VL regions were transferred from pCMV vector bearing the 7G8M

sequence to the pCMV vector containing 7G8H sequence. For transfer of the VH region

enzymes Eco72I and Bpu1102I were used. Resulted constructs were sequenced, and correct

plasmid was used for transfer of the 7G8M VL region. Both, donor and host vectors were

digested with BpiI, which cleaves given plasmids at three sites, leaving different sticky ends

at each cleavage site. The fragment, containing the desired sequence of 7G8M was then re-

ligated with two remaining fragments obtained from the pCMV_7G8H digestion. Generated

constructs were analyzed by sequencing and clones bearing the correct plasmid were chosen

for expression.

2.2.6. Production and purification of recombinant mAbs

The novel technology, called QMCF Technology, developed by Icosagen Group

(Estonia) was utilized in this study for the large-scale production of murine- and chimeric

recombinant monoclonal antibodies. In particular, secretable recombinant mAbs were

produced by mammalian cells, based on chinese hamster ovary (CHO) and purified by protein

G sepharose (GE Healthcare) affinity chromatography at Icosagen facility.

35

Table 1. Constructs generated during this study (primer sequences are available upon request).

F- forward R- reverse

Primers (sequences are available

upon request) Method

Generated constructs (intermediate)

Expression constructs

mAb/ Comments

F ----> VH(for)

Ligation mAb VH regions: pJET_8B2_VH pJET_7G8_VH

R <---- VH(back) or VH_rev_IgG1, or VH_rev_IgG2a

F ----> VLκ(for) mAb VL regions: pJET_8B2_VL pJET_7G8_VL

R <---- VLκ(back) or VK_rev_CLk

F ----> HC_7G8_up_site_ins

RF-Cloning

pBSK_HC_7G8H

--> pCMV42_7G8H 7G8H R <---- HC_7G8_down_site_ins

F ----> LC_7G8_up_site_ins pBSK_LC_7G8H

R <---- LC_7G8_down_site_ins

F ----> HC65ForSapI

RF-cloning

pBSKmut_HC65_SapI_FR

HC65 - mouse IgG1 heavy chain

R <---- HC65RevFRSapI

F ----> HC65ForSapI pBSKmut_HC65_SapI_ Const

R <---- Hc65RevIgG1SapI

F ----> HC83ForSapI pBSKmut_HC83_SapI_FR

HC83 - mouse IgG2a heavy chain

R <---- HC83RevFRSapI

F ----> HC83ForSapI pBSKmut_HC83_SapI_ Const

R <---- HC83RevIgG2aSapI

F ----> HC70ForSapI

pBSKmut_HC42_SapI

HC42 - human IgG1 heavy chain

R <---- HC70RevFRSapI

F ----> LCAllForSapI

pBSKmut_mLC_SapI

mLC - mouse kappa light chain

R <---- LC6583RevSapI

F ----> LCAllForSapI pBSKmut_hLC_SapI

hLC - human light chain

R <---- LC70RevSapI

F ----> 8B2VH6570ForSapI Ligation pBSKmut_HC42_8B2VH

--> pCMV42_8B2H 8B2H R <---- VHAllRevFRSapI

F ----> 8B2VLForSapI Ligation pBSKmut_hLC_8B2VL

R <---- VL70RevSapI

F ----> 8B2VH83ForSapI Ligation pBSKmut_HC83_8B2VH_FR

--> pCMV83_8B2M_ FR

8B2M

R <---- VHAllRevFRSapI

F ----> VLAllForSapI Ligation pBSKmut_mLC_8B2VL

R <---- VHAllRevFRSapI

--> pCMV83_8B2M_ Const

F ----> 8B2VH83ForSapI Ligation pBSKmut_HC83_8B2VH_const

R <---- VH83RevIgG2aSapI

F ----> VH6570ForIgG1SapI Ligation pBSKmut_HC65_7G8VH

--> pCMV65_7G8M 7G8M R <---- VH65RevIgG1SapI

F ----> VLAllForSapI Ligation pBSKmut_mLC_7G8VL

R <---- VL6583RevSapI

F ----> F2A_hIgG1_fw RF-cloning

pBSK_8B2H_F2A and pBSK_7G8H_F2A

pCMV_8B2H_F2A and pCMV_7G8H_F2A

8B2H_F2A

R <---- F2A_rev_all 7G8H_F2A

F ----> F2A_mIgG2a_fw RF-cloning

pBSK_8B2M_F2A

pCMV_8B2M_F2A 8B2M_F2A R <---- F2A_rev_all

F ----> F2A_mIgG1_fw RF-cloning

pBSK_7G8M_F2A

pCMV_7G8M_F2A 7G8M_F2A R <---- F2A_rev_all

36

2.2.7. HCV RdRp purification

HCV RdRp was purified using the original protocol of Binder et al, (2007) [92] with

minor modifications made to meet the conditions of our lab. Plasmid pET-19b was

transformed to chemically competent E. coli BL-21 (DE-3) cells. Bacterial cells were grown

in LB medium at 37°C. When the optical density at 600 nm reached 0.7-0.8, NS5B expression

was induced by adding Isopropyl-β-D-Thiogalactoside (IPTG) to a final concentration of 0.6

mM and bacterial culture was shifted to a room temperature. After 8 h of induction, cells were

pelleted by centrifugation at 6000×g for 10 min, washed once with 1xPBS and resuspended in

40 ml of Lysis Buffer I (LBI: 100 mM Tris-HCl [pH 8.0], 100 mM NaCl, 1mM MgCl2, 2%

Triton X-100, 2 mg/ml lysozyme (Amresco), 3 kU of DNase (Calbiochem), and protein

inhibition cocktail (Roche)). After a 30 min incubation at +4 °C lysate was centrifuged at

20000×g for 10 min at +4 °C, supernatant was removed and the pellet was resuspended in 5-7

ml of Lysis Buffer II (LBII: 20 mM Tris-HCl [pH 7.5], 500 mM NaCl, 2% Triton X-100, 10

mM imidazole, 30% glycerol, 10 mM 2-mercaptoethanol, protein inhibition cocktail

(Roche)). The suspension was further sonicated on ice in 1-ml aliquots 5 times for 20 seconds

at an output control setting of 6 using Bandelin Sonopuls HD2070 sonifier. After 30 min

centrifugation at 21,100×g at +4°C, the supernatant (cleared lysate) was mixed with 400 µl of

1:1 Ni-nitriloacetic acid (Ni-NTA) agarose slurry (QIAGEN), incubated for 1 h at a rotary

shaker at +4 °C and centrifuged for 5 min at 500×g. Supernatant was discarded and Ni-NTA

matrix with bound NS5B was packed onto the column (Bio-Rad), washed 4 times with 4 ml

of LB II containing 50 mM imidazole and then eluted with 1 ml (5×200 µl fractions) of LB II

containing 250 mM imidazole. Eluate was stored in small aliquots at -80°C. Fractions were

analysed on 12% SDS-PAGE and protein concentration was determined in Bradford protein

assay (Bio-Rad) using BSA standart.

2.2.8. Protein analysis and quantification methods

2.2.8.1. Protein quantification using Bradford protein assay

Protein quantification was performed by a modified Bradford method using Bio-Rad

protein assay dye. 5 µl of BSA dilutions (for generation of a standart curve) and NS5B

samples were mixed with 800 µl of H2O in 1.5 ml tubes, then 200 µl of the Bio-Rad protein

assay dye (pre-warmed to room temperature) was added to each tube. Reactions were

37

incubated for 20 min at room temperature and OD was read at 595 nm using a Heλios

(Thermo) spectrophotometer. Protein concentration were calculated from the standard curve

equation.

2.2.8.2. Western blot

NS5B samples, containing known amount of protein were mixed with the same

volume of 2x Laemmli buffer [93] and denatured for 10 min at 95°C prior to SDS-PAGE.

After resolving the samples on 12% polyacrilamide gel, proteins were transferred to a PVDF-

membrane (Millipore) using a semi-dry transfer method in a Trans-Blot SD apparatus (Bio-

Rad). Further, membrane was incubated for 1-2 hours in a blocking solution I (BSI: 5% w/v

Non-fat dry milk (NFDM), 0.1% Tween-20, 1xPBS) and then incubated with a primary mAb

specific to the NS5B in a blocking solution II (BSII: 2% NFDM, 0.1% Tween-20, 1xPBS).

After that, the membrane was washed 3 times for 15 min with a washing solution (50 mM

Tris-HCl pH 7.5; 150 mM NaCl; 0.1% Tween-20) and incubated for 1 h with a secondary

mAb (anti-mouse or anti-human HRP-conjugated IgG) in the blocking solution II. After 3x15

min washings, NS5B specific signals were developed using ECL-Plus kit (Amersham

Pharmacia Biotech) and detected by exposing the membrane to a roentgen film (Fuji).

2.2.9. Enzyme-linked Immunosorbent Assay (ELISA)

ELISA was performed according to Pfaff et al. [94]:

96-well microtiter plates (Nunc) were coated with peptides (5 µg/µl) or full-length

NS5B (0.25 µg/µl) and incubated overnight at 37 °C. After incubation, wells were washed

twice with 200 µl of 1xPBS and saturated with 1% BSA in PBS (30 min at 37 °C). 50 µl of

mAb dilutions (or hybridoma supernatant dilutions – 1:10 to 1:100000) from 0.1 pg/µl to 10

ng/µl were placed in the wells and incubated for 1 h at 37 °C. After washing 5 times with 200

µl of PBS/Tween-20 (0.05%), samples were incubated for another 1 h with 100 µl of

horseradish peroxidase (HRP) conjugated rabbit anti-mouse (at a ratio of 1:10000), or goat

anti-human IgG (at a ratio of 1:8000) in 1% BSA/PBS. After several washings with

PBS/Tween-20 (0.05%) wells received 100 µl of a substrate solution (Bio-Rad) containing

3,3’,5,5’-tetramethylbenzidine in dimethylformamide and hydrogen peroxide at a ratio of

10:1. After development of deep blue color, reactions were stopped with 1N H2SO4 and OD

was read at 450 nm with a “Sunrise” (Tecan) multiscan photometer.

38

2.2.10. Changing mAb buffer by ultrafiltration

For RdRp assays mAb buffer was changed to TN (10 mM Tris; 100 mM NaCl) using

Amicon Ultra-4 Centrifugal Filter Units with Ultracel-10 membrane (Millipore). First, units

were equilibrated by centrifugation with 4 ml of TN for 30 min at 4000×g at room

temperature in a swinging-bucket type rotor (Sigma). Then, flowthrough was discarded and

mAb solution (~200 µg mAb) diluted with TN to a total volume of 4 ml was added. Units

were centrifuged for 12-15 min at 4000×g at room temperature to get residual volume of

approximately 100 µl. Then, another 4 ml of TN were added and centrifuged. The procedure

was repeated 4 times and residual solution, containing concentrated antibody, was transferred

to a new 1.5 ml tube. After buffer exchange mAbs were stored at +4°C. Concentrations of

mAbs were determined with a ND-1000 spectrophotometer at 280 nm using an IgG antibody

extinction coefficient of 1.37. Alternatively, concentration was measured using Bradford

protein assay (Bio-Rad) with IgG standard (Pierce).

2.2.11. Primer-dependent RdRp assays

RdRp assays were performed essentially as described by Nikonov et al. [87]

For all RdRp assays mAb buffer was changed to TN (as described above). Poly (rC)

template was mixed with oligo (rG)12 primer, denatured for 2 min at 95°C, and incubated for

5 min on ice before use.

Primer-dependent RdRp assays were performed in 1xRP buffer (10 mM Tris-HCl, pH

7.5; 12.5 mM KCl; 2.5 mM MgCl2; 0.5 mM DTT; 0.5 mM EGTA, pH 7.5) and contained

~0.12 µmol of HCV NS5B polymerase, various mAb dilutions (0.6 – 1.2 µM), 0.4 µg of

poly(rC) pre-annealed to 4 pmol of oligo(rG)12 primer, 10 units of RiboLock Rnase Inhibitor

(Fermentas), NTP – CTP, ATP and UTP at a final concentration of 0.5 mM and 1 µCi of [α-

32P]-GTP per reaction. First, NS5B and mAbs were preincubated at +4°C for 10 min, then

pre-annealed primer-template mix was added and incubation was prolonged for another 10

min. After that, reactions were initiated by addition of NTP (CTP, ATP, UTP and [α-32

P]-

GTP). The reactions were incubated for 1 h at 25°C and then stopped by addition of 1 ml of

ice-cold 10% trichloroacetic acid (TCA), 0.5% tetrasodium pyrophosphate (Na4PPi) and 100

µg of herring sperm DNA. After 30 min incubation on ice, samples were filtered through

glass-microfiber GF/C filters (Whatmann and Schelicher & Schuell) and washed 2 times with

5 ml of 1% TCA/0.1% Na4PPi solution. After that, filters were placed to scintillation vials and

39

dried for 15 min at 65 °C. The activity of NS5B polymerase was then measured by liquid

scintillation counting of [32

P]-GMP incorporated into the synthesized RNA (bound to the

GF/C filters) using ScintiSafe 3 liquid scintillation cocktail (Fisher Scientific) and

WinSpectral counter (Wallac). The primer-dependent assay for wild type and mutant (GND)

NS5B polymerases was performed in the same manner, but with different concentrations of

the enzyme and without mAbs (thus, the first 10-min preincubation step was omitted).

40

3. Results and discussion

3.1.1. Cloning of antibody variable regions

Two mouse hybridoma cell lines 7G8.1 and 8B2.1 producing monoclonal antibodies

(mAbs) 7G8 (IgG1) and 8B2 (IgG2a) were generated previously in our lab. 8B2 and 7G8

specifically inhibited RdRp activity of HCV NS5B polymerase [87]. We decided to clone

variable regions of these mAbs. For this purpose, genomic DNA extracted from hybridoma

cell line producing specific mAb could not be used, because it contained non-transcribed

DNA encoding nonspecific antibody fragments. That is why hybridomas total RNA was used

to isolate antibody sequences corresponding to variable regions of 7G8 and 8B2.

Subsequently, total RNA was used for reverse transcription-polymerase chain reaction (RT-

PCR) as described in “methods” section.

Total RNA was extracted from hybridomas with TRIzol reagent and its integrity was

analyzed on agarose denaturing gel. The preparations had two sharp bands, corresponding to

28S and 18S rRNA, and the 28S band is approximately as twice intence as 18S band. Taken

together with the absence of a low molecular weight smear (below 5S), these data indicated

that the total RNA was intact.

Figure 11. Denaturing formaldehyde RNA gel-electrophoresis. ~1 µg samples of total hybridoma RNA were run

on 1% denaturing agarose gel. The 28S and 18S bands (indicated by arrows) are sharp, with 28S band being

approximately twice as intense as 18S. The absence of smear below 5S band demonstrates that there is no

degraded low molecular weight RNA.

For amplification of antibody heavy and light chain variable domains degenerate 5’

primers designed by Wang et al. were used (Table 1) [95]. The design of these primers is

41

based on a relatively conserved sequence in the framework one (FR1) regions. The primers

have degeneracies at 7 positions in order to cover the majority of sequence combinations at

these regions. The rest of these primers is complementary to the most conserved nucleotides

in FR1. Two different types of 3’ primers were used: first – primers, complementary to the

framework 4 (FR4) regions, that have a high degree of conservation (design was based on

Orlandi et al., [96]). Second type – primers, complementary to isotype-specific constant

region sequences [95], (see Figure 12).

Figure 12. Schematic representation of antibody variable region with primer annealing sites. Framework regions

are labelled as FR and complementarity-determining regions as CDR with numbers indicating the order of these

regions. L – leader peptide sequence. Primer binding positions are shown as arrows.

As seen from Figure 13A, majority of mAb variable regions could be succesfully

amplified with primers, complementary to framework regions. Amplification of 8B2 VH

region, however, was less efficient, than in case of the other mAbs. This problem was solved

by utilizing the 3’ primer, complementary to mouse IgG2a constant chain (Figure 13B). As

further sequence analysis showed, two amino acids in FR4 of 8B2 VH region were different

from the most frequent ones (on which the design of the initially used 3’ primer was based).

This explained why this region could not be amplified with the proofreading PhusionTM

DNA

polymerase.

42

Figure 13. Amplified antibody variable regions. A) VH and VL regions of mAbs 8B2, 7G8 and 10D6 (control),

amplified with Taq DNA polymerase using degenerate forward primers and reverse primers complementary to

framework regions. Specific bands corresponding to the variable regions are shown with arrow. B) VH region of

8B2 mAb, amplified with PhusionTM DNA polymerase, with the help of reverse primer complementary to

IgG2a constant chain. The amplified fragment is ~90 nt longer then those on Figure 13A, because it contains a

part of the constant region sequence.

Control reaction without reverse transcriptase, without RNA template and without

both enzyme and RNA (Figure 13) did not yield any amplification products, indicating that

there was no DNA contamination during RT-PCR. Subsequently, amplified fragments were

inserted into pJet 1.2/blunt vector for sequencing generating following constructs:

pJet7G8VH, pJet7G8VL, pJet8B2VH, pJet8B2VL. Multiple sequence alignment and analysis

allowed to identify clones with the consensus sequence – those, not containing any undesired

mutations that could be possibly introduced by PCR. Clones containing the consensus

sequences for given antibodies were chosen for further manipulations.

3.1.2. Construction of chimeric 7G8 antibody genes

Antibodies, capable of inhibiting key enzymes in HCV replication, are clearly of great

interest, because of their possible use in anti-HCV therapy. However, murine mAbs, if

administered to human, are effectively recognized by the host immune system as foreign

43

antigens, which leads to rapid clearance of these mAbs from the individual’s organism. There

are some approaches, first described and applied more than 20 years ago, that can help to

eliminate the problems associated with this defense mechanism. For example, murine

antibodies can be chimerized or humanized (reshaped). Humanization is described in details

in the theoretical part of this thesis. The term „chimeric“, when applied to an antibody, means

that the variable regions of this antibody are from one organism (for example, mouse), and all

other regions are from host organism (for example from human) (Figure 6). Such antibody

would contain only ~25% of original murine antibody sequence, what could significantly

lessen the chance of being recognized by a human immune system. There are many examples

of antibodies that were successfully chimerized or humanized, and currently used in antiviral

or anti-cancer therapy (see „background“ part). Though, humanization can make antibody

even more human-like, than chimerization, it is much more difficult process. That is why we

pursued the construction of chimeric antibodies first, to prove that our mAbs can function in

this format, and that all the mAb gene cloning, engineering and expression technology works

in our hands.

Initially the approach was applied only to one mAb (7G8) in order to get a „proof of

principle“ that the whole system for creation of recombinant antibodies in such way works.

Due to the use of the degenerated primers we were unable to determine the native sequence

for the 1st, 3

rd, 5

th and 6

th amino acids of VH, and 4

th, 7

th and 8

th amino acids of VL regions of

our mAbs. Thus, during this pilot experiment of 7G8 chimerization we decided to change

these amino acids for those of a published humanized anti-RSV mAb [63], because the fact

that this mAb maintained its biologic activity after humanization seemed to increase our

chances of success.

Expression vector pCMV_7G8H, containing 7G8 mAb variable region sequences

inserted between the leader peptide coding fragment (needed to direct the antibody to ER for

packing and secretion) and the constant region sequence of corresponding chains, was

constructed as described in „Methods“ section (Figure 18). This chimeric 7G8 antibody was

analyzed in ELISA and Western blot assays for histidine-tagged bacterially expressed NS5B-

binding ability. As could be seen from Figure 14, chimeric 7G8 reacted strongly in Western

blot assay, whereas its activity in ELISA was relatively weak (what later turned out to be the

issue of the secondary antibody used in this experiment (data not shown)). These experiments

demonstrate that the recombinant antibody generated using the described approach

maintained its affinity to the antigen. So, it was decided to generate the high-throughput

44

vector system for generation of similar recombinant constructs not just for 7G8 and 8B2

mAbs, but for virtually any murine antibody of either IgG1 or IgG2a isotype.

Figure 14. Chimeric 7G8 activity. A) A photo of ELISA test results for chimeric 7G8 (7G8H) ability to

recognize NS5B RNA polymerase in native conformation. The supernatant dilutions of a cell culture transfected

with 7G8H expressing plasmid were used in this experiment. Supernatant from a non-transfected culture and an

anti-BPV-1 E2 region antibody were used as negative controls (shown with arrows). B) Western blot with 7G8H

and NS5B polymerase. Amount of input NS5B is shown at the top.

3.1.3. Generation of a vector system for high-throughput construction of

recombinant antibody genes.

Although, the RF-cloning method is very good for generating difficult constructs, it

cannot be used in a high-throughput manner, because of the need of expensive high-quality

primers and also because all constructs generated by PCR need additional sequence control.

Considering these facts, there clearly was a need for a system, which would allow easy and

rapid generation of recombinant antibody genes. One of the simpliest and most wide-spread

techniques in molecular cloning is the use of restriction enzymes, so the RD-based approach

was developed in collaboration with Icosagen Group (Estonia) and used for the construction

of such system. For that purpose, a restriction enzyme named SapI (or LguI) was chosen. SapI

recognition site is 7 bp long, which makes it more rare, compared to typically occuring 6 and

4 bp long restriction sites. The site is also non-palindromic and the cleavage occurs

downstream of the recognition sequence, leaving sticky ends, which usually differ from each

other, preventing the re-circularization of the digested sequence and allowing the ligation of

45

the gene of interest in correct orientation (see Figure 15). Due to the fact, that the cleavage

site differs from the recognition sequence, the restriction sites can be removed after digestion.

Thus, products of the subsequent ligation do not contain any foreign sequences.

Figure 15. SapI recognition site and digestion specifics. SapI recognition sequence is drawn in red in both direct

and reverse orientations. Adjacent nucleotides designated with N, meaning any of four dNTPs. The scissors

icons and a broken line depict the way DNA is cleaved by SapI. In the lower row, SapI cleavage products with

protruding 5’ ends are shown.

Utilizing the RF-cloning method, SapI restriction sites were introduced to pBSK

vectors with following pre-inserted antibody chains: human light and heavy (IgG1 isotype)

chains, murine IgG1 and IgG2a heavy chains, and murine kappa light chain (Table 1). The

naturally occurring SapI cleavage site of the plasmid was eliminated by site-directed

mutagenesis (see “methods“). The 5’ SapI site was inserted in reverse orientarion in such way

that cleavage site located right at the junction between signal peptide and VH (or VL) region.

The 3’-end SapI restriction site was inserted in direct orientation so that it located at the

junction between the VH (or VL) region and the constant region of corresponding chain. For

mouse IgG1 and IgG2a additional constructs were generated. These constructs differed from

the previously described in a way that 3’-end SapI sites were introduced into their constant

regions, in order to allow the ligation of VH fragments amplified with the help of the

VH_rev_IgG1 and VH_rev_IgG2a primers (Table 1, Figure 12, Figure 16). All generated

constructs are schematically drawn in Figure 16.

46

Figure 16. Schematic representation of generated SapI restriction sites containing vectors. The positions of the

SapI sites are shown as blue pentagons, where the tip of each pentagon determines the orientation of the

restriction site (also indicated by arrows at the top of the scheme). L – leader sequence, VH – variable heavy

region, VL – variable light region (kappa), CH – constant heavy region, CL – constant light region. pBSK HC or

LC – pBluescript II SK(+) plasmids containing respective HC or LC genes.

Next, primers containing the SapI restriction sites were designed on a basis of the

primers used for initial cloning of antibody variable domains. The VH and VL regions

amplified with these primers would contain the SapI site in the correct orientation. Also, the

sticky ends, left after digestion with SapI restrictase would be exactly complementary to those

of respective vectors (digested in the same way). At large, utilization of such vector-primer

system allows to amplify variable region from virtually every mouse hybridoma and join them

directly to constant regions of human origin. This system might be further customized to

allow the creation of mAb chimeras of any two or more organisms. Because given project is

47

therapy oriented, only human Ab chains were used as foreign constant region donors. All

generated plasmids were controlled by sequencing for the presence of the desired insertion.

The principle of the SapI system is shown schematically on Figure 17.

Figure 17. Scheme, representing the process of generation of recombinant antibody genes using the SapI vector-

primer system. Variable region amplified from hybridoma cDNA shown in green, host variable region – in gold.

Scheme of amplification of variable regions from hybridoma cDNA (with primer pairing sites) is drawn in a

dashed box. SapI site-containing primers are shown as arrows – green part is complementary to the FR regions

of variable domain and blue part – is a SapI site-containing sequence. SapI restriction sites in PCR products and

in host vector are shown as blue pentagons, where tip of each pentagon indicates the orientation of the

corresponding site. Scissors icons indicate where the cleavage occurs. L – leader peptide.

48

3.1.4. Construction of a panel of recombinant 7G8 and 8B2 monoclonal

antibodies.

Utilizing the above described system, a panel of recombinant 7G8 and 8B2 mAbs was

generated, which included chimeric mouse/human (containing mouse variable regions and

human constant regions) antibodies and their mouse/mouse (containing mouse variable

regions fused to codon-optimized mouse constant chains) analogues (Figure 18A). All

variable regions could be efficiently amplified with Phusion DNA polymerase, using SapI

containing primers. All fragments and vectors containing SapI restriction site were

successfully digested and ligated in correct orientation without the requirement of any

optimization. Recombinant mouse mAbs were constructed in order to compare their biologic

activity with chimeric and hybridoma mAbs biologic activities. Two variants of mouse 8B2

antibody were constructed. First, VH region was amplified with the help of 3’-end primer

complementary to FR4 region. Second, the same region was created by using 3’-end primer

complementary to IgG2a constant region (Table 1). These constructs differed by two amino

acids in FR4 regions and were designated respectively 8B2MF and 8B2Mconst. As the

subsequent experiments did not reveal any difference in their function (data not shown), only

one of them, 8B2MF (further referred to as 8B2M), was used for further experiments.

Chimeric 8B2 was designated as 8B2H and 7G8 chimeric and murine mAbs as 7G8H and

7G8M, respectively. The recombinant mAb genes were cloned to pCMV vectors for

expression purposes. The expression vectors, containing the above described antibody genes

are listed in Table 1.

49

Figure 18. Schematic representation of recombinant antibody genes, generated in this study. A) Recombinant

genes for two-promoter expression and B) “bicistronic” F2A recombinant constructs for single-promoter

expression. L – leader sequence, VH and VL – variable heavy and light regions, CH and CL constant heavy and

light regions, 2A – Foot-and-Mouse Disease Virus 2A peptide encoding sequence. Orange and green boxes are

sequences coding for furin cleavage site and SGSG spacer, respectively. Schematic drawings of expression

vectors with the relative positions of gene expression cassettes are shown under the corresponding gene schemes.

3.1.5. Design and construction of bicistronic mAb expression vectors

utilizing the F-2A peptide sequence.

As pCMV expression vectors utilize two promoters, separately for mAb heavy and

light chains, the molar ratio of the expressed chains is hard to control and with high

probability is not equal, due to a competition between promoters. In this case, efficiency of

production of functional antibody dimers is limited by the efficiency of transcription from the

50

less successful promoter. Besides affecting the total yield of functional antibody, excessory

immunoglobulin chains may also form aggregates and cause precipitation of other proteins,

what can adversely impact the therapeutic potential of the antibody. Thus, for optimal

production and use ratio of expressed antibody chains should be 1:1.

In order to overcome this two-promoter issue, we decided to construct so called

bicistronic mAb expression vectors, according to Fang et al. [97], in which both heavy and

light chain are expressed as a single open reading frame. In such vector, the 5’ end of the gene

encoding the light chain is connected with the 3’ end of the heavy chain through a sequence

directing the synthesis of a short peptipe linker (Figure 19). This linker consists of three parts.

First part is represented by the cleavage site of furin, – a serine protease normally presented in

animal cells [98-99]. Second part is a flexible four amino acid SGSG spacer, shown to

improve the furin cleavage rate [100]. And the last part is the 24 amino acid 2A peptide of the

Foot-and-Mouth Disease Virus (FMDV) (Figure 19). This type of a peptide was first

identified among picornaviruses, in the aphtoviruses sub-group, a typical example of which is

FMDV [101-102]. In these viruses multiple proteins are derived from a large polyprotein

encoded by a single open reading frame. 2A peptides contain the consensus motif at the C-

terminus (Asp-Val/Ile-Glu-X-Asn-Pro-Gly-Pro). The unique feature of this peptide is that

during translation the synthesis of peptide bond between glycine and the last proline residues

does not occur, resulting in the ribosome skipping to the next codon and cleavage of the

nascent protein between Gly and Pro (Donnelly et al., 2001).

Figure 19. Scheme of a typical “bicistronic” antibody gene for expression from a single promoter. Sequence

encoding furin cleavage site, SGSG spacer and FMDV 2A peptide is shown between the dashed lines. Scissors

indicate the sites where the polypeptide chain break occurs. L – leader peptide.

In the case of bicistronic mAb expression constructs, 2A peptide mediates separation

of light chain from heavy chain, leaving the proline residue fused to the N-terminus of the

51

light chain. The remaining part of the peptide linker is then cleaved from the C-terminus of

the heavy chain by cellular furin protease (Figure 19, 20). However, two additional amino

acids derived from the furin cleavage site (Arg and Ala) remain fused to the C-terminal end of

the heavy chain (Figure 20). Fortunately, these additional amino acids seem to have no effect

on the mAb functionality, at least as reported by Fang et al, [97]. Using this approach both

chains constituting mAb could be produced at equimolar ratio.

Figure 20. Scheme of the polypeptide processing during translation of a transcribed bicistronic antibody gene,

and biosynthesis of a corresponding antibody. Furin cleavage site and amino acids that remain fused to the

antibody after assembly are drawn in orange. The course of the expression and processing is indicated at the

right by arrows. Modified from Fang et al. (2005) [97]

The constructs containing the above described sequences (Figure 18B) were generated

on the basis of the pCMV expression vectors, as described in “methods”. The stop codon at

the 3’ end of the heavy chain coding sequence was eliminated during the cloning. The

sequences of the whole resulting ORFs were controlled by sequencing and transferred to the

pCMV plasmids, generating constructs pCMV7G8H-F2A, pCMV8B2H-F2A, pCMV7G8M-

F2A and pCMV8B2M-F2A. A scheme of typical F2A expression construct is presented in

Figure 19.

52

3.1.6. Expression and purification of enzymatically active NS5B

polymerase.

pET19b vector containing the NS5B gene under the control of bacteriophage T7

promoter (kindly provided by Andrei Nikonov) was used for wild type NS5B (wtNS5B) and

its inactive mutant (GND) overexpression in E.coli BL21 (DE3) strain. The resulting protein

had a molecular weight of approximately 66 kDa and contained a decahistidine tag fused to its

N-terminal end. The growth and purification of wtNS5B (or GND) was performed according

to Binder et al. [92], with minor modifications. The expression of the protein was confirmed

by SDS-PAGE, using non-induced cultures as a control (Figure 21A). Samples of expression

culture were taken at different time points – 4, 9, 12 and 18 hours after induction and analyzed

by SDS-PAGE. Results of this analysis (shown on figure 21A) indicate that the optimal time

for expression of NS5B at room temperature is between 4 and 9 hours, and after this time

amount of the product starts to decrease. Therefore, all expression cultures were further

induced for ~8 hours. Following the purification procedure, the eluate, washing and

flowthrough fractions were analyzed by SDS-PAGE (Figure21B). As it is seen on the Figure

21B, the protein was purified to near homogeneity with the majority of the protein coming out

in 2-nd and 3-rd fractions (lanes F1-F5). The flowthrough (lane Ft) contains the major part of

unbound cellular proteins, whereis only trace amount of proteins is seen in the washing

fraction, demonstrating the specificity of the Ni-NTA matrix to His-tagged NS5B.

Figure 21. NS5B A) expression and B) purification. Shown are 12% SDS-polyacrilamide gels stained with

coomassie brilliant blue (R-250). Marker proteins sizes are indicated at the left (in kDa). A) samples of NS5B

expression culture taken at various time points after induction are analyzed by SDS-PAGE, M – marker, NI –

non-induced control. 4h, 9h, 12h and 18h – hours post induction. NS5B specific band is indicated with an arrow

at the right. B) Different fractions collected during the purification of NS5B are analyzed along with series of

BSA dilutions for empirical quantification of NS5B yield. The amounts of input BSA are indicated above the

correspondent lanes. Ft – flowthrough fraction of NS5B, W – washing fraction, F1 – F5 are elution fractions.

NS5B specific band is indicated with an arrow at the right.

53

Relatively high content of NS5B in the flowthrough fraction can be explained by

inefficient disruption of cell membranes during homogenisation. Due to a highly hydrophobic

region at the C-terminus, majority of NS5B was found to be membrane-associated. Thus,

incomplete disruption of cell membranes could result in large amount of the protein remained

membrane-associated, what, in its turn, could interfere with the binding to Ni-NTA matrix.

Generally, purification of NS5B from 1 litre culture yielded approximately 0.5 mg. Enzymatic

activity of purified NS5B was controlled in a primer-dependent RdRp assay, using liquid

scintillation counting of radioactively labelled GMP, incorporated into synthesized RNA

(Figure 22). As it is seen on a figure 22, NS5B activity grows linearly when the enzyme

amount is being increased up to 800 ng and reaches a plateau at higher concentrations,

achieving the upper limit of approximately 170000 cpm. On the other hand, when the same

amounts of the GND mutant were used, only residual radioactivity of ~5000-7000 cpm could

be detected. These results indicate, that the purified wtNS5B is enzymatically active and pure

enough to be used in biochemical assays at concentrations up to 800 ng per reaction (since

there are no signs of activity inhibition at these concentrations) for testing the biologic activity

of different mAbs.

Figure 22. NS5B enzymatic activity. The amount of specific radioactivity (in counts per minute) incorporated

into the synthesized RNA (Y-axis) is plotted against the amount of NS5B in reaction (X-axis). Activity of NS5B

mutant inactive form (GND) is shown as black triangles, wtNS5B – white rectangles.

3.1.7. Biologic activity of recombinant 7G8 and 8B2 mAbs

The recombinant antibodies could be efficiently produced in CHO cells, using the

Icosagen QMCF technology (described in “methods” section), yielding up to 2.5 mg of mAb

for constructs utilizing two promoters. Interestingly, the yield was better for chimeric

54

antibodies (1.5 – 2.5 mg) than for murine ones (less than 1 mg). This could be possibly

explained by a predominance of expression from promoter of the light chain coding gene.

Resulting lower concentration of expressed heavy chains (F(c) regions) could reduce the

efficiency of antibody purification by a protein G affinity chromatography. Nevertheless,

yields of the antibodies produced using the QMCF technology (Icosagen Group) are

comparable with those can be obtained from hybridomas.

Expression of antibodies from bicistronic constructs resulted in ~2-fold increase in

antibody yields, indicating the possible benefits of this single-cassette expression system.

However, it was shown, that in case of high expression level, the amount of cellular furin is

not sufficient to cleave all synthesized protein molecules. This indicates that additional furin

might be provided in order to improve cleavage efficiency. Thus, this approach still requires

additional optimization, before it can be routinely used.

3.1.7.1. Biologic activity of recombinant mAbs in antigen binding assay

(ELISA).

The ELISA tests were performed in order to show whether the ability of recombinant

7G8 and 8B2 mAbs to bind the antigen (NS5B) is comparable with that of the hybridoma-

produced mAbs. ELISA was preferred to western blot method, because in this assay the

antigen remains in its native conformation. Also, comparing to WB, ELISA is a quantitative

method, where the efficiency of mAb binding is estimated by directly measuring the intensity

of the final sample solution color. Results of these ELISA experiments with different

recombinant and hybridoma mAbs are shown in Figure 23.

Figure 23. ELISA assay with A) 8B2 and B) 7G8 hybridoma and recombinant mAbs. ELISA specific signal

intensity (OD450, Y-axis) is plotted against mAb concentration (X-axis). 8B2.1, 7G8.1 – hybridoma mAbs.

8B2H, 7G8H – chimeric mAbs. 8B2M, 7G8M – recombinant native murine mAbs.

55

As it can be judged from these results, for the 8B2 mAbs the signal could be detected

starting from mAb concentration of 0.1 ng/µl, becoming saturated at a 10 ng/ml

concentration. The recombinant 8B2H and 8B2M mAbs show the same pattern of binding

efficiency, whereas the hybridoma 8B2 (8B2.1) gives a weaker signal at a concentration

between 0.1 and 10 ng/µl (probably due to the use of the old mAb stock). In case of the 7G8

mAbs, the specific signal could be detected already at 0.01 ng/µl mAb concentration,

achieving saturation at 1 ng/µl. For all three 7G8 antibodies used in the experiment the

binding profiles were somewhat different. Strength of the signal (at a mAb concentration of 1

ng/µl) decreases in the following order: 7G8M > 7G8H > 7G8.1. A weaker signal for

hybridoma 7G8 can be explained (at least in part) by the usage of the old mAb stock, whereas

difference between the antigen-binding activities of 7G8H and 7G8M antibodies is of another

origin (will be discussed later). All-in-all, these data indicate that recombinant mAbs

constructed in this study are capable of binding the native NS5B with at least the same

efficiency as hybridoma mAbs. Moreover, the recombinant 7G8M mAb shows even stronger

binding pattern, than its chimeric and hybridoma analogues.

3.1.7.2. Biologic activity of recombinant mAbs in primer dependent RdRp

assay.

The standart RdRp assay is a continuous polymerisation reaction, which consists of

repetitive initiation and elongation cycles. NS5B is a processive enzyme, capable of initiating

the elongation polymerization reaction from pre-annealed template/primer RNA substrate.

Polycytidylic acid (poly(C)) with randomly pre-annealed guanosine 12-meric

oligonucleotides ((rG)12 primer) is usually used in the RdRp assay because this

template/primer hybrid is shown to be utilized by NS5B with the highest efficiency [103]. In

contrast to, for example, highly processive DNA polymerases, HCV NS5B RNA polymerase

has noticeably lower processivity. NS5B constantly dissociates from the template/primer

substrate and then re-binds in order to initiate a new elongation cycle. Different 7G8 and 8B2

recombinant antibodies along with hybridoma mAbs were examined in this assay for their

ability to inhibit the primer-dependent synthesis of RNA by NS5B in vitro. The results are

presented in Figure 24.

56

Figure 24. Primer-dependent RdRp assay A) with 8B2 and 7G8 recombinant and hybridoma mAbs and B) with

8B2 F2A and 7G8 F2A mAbs + hybridoma mAbs. The order of the reagent addition is indicated at the top of the

graphs. X-axis shows the molar ratio of mAbs to NB5B in reaction. Y-axis displays the amount of specific

radioactivity incorporated into synthesized RNA (in counts per minute). The means of at least two indipendent

experiments with standart deviations are indicated. 8B2.1, 7G8.1, 9A2.1, 6G5.1 – hybridoma mAbs. Antibodies

designated with “H” letter – chimeric mAbs, and with “M” letter – recombinant native murine mAbs.

Obtained data clearly indicate that 7G8 and 8B2 mAbs, both recombinant and

hybridoma-produced, inhibit the RNA synthesis by the NS5B polymerase in a dose-dependent

manner in this in vitro assay. All 8B2 antibodies demonstrate highly similar inhibition

patterns, similar to those described by Nikonov et al. [87]. These mAbs inhibit the NS5B

polymerase approximately 4-5 fold. Starting from more than 2-fold mAb molar excess over

NS5B, the residual activity of the polymerase is only about twice higher than the background

(determined as the average activity of the non-functional NS5B GND mutant (not shown on

the graph)). At the same time, a control 9A2.1 hybridoma mAb seems to have no

57

concentration-dependent inhibitory effect upon the NS5B. A slight decrease in NS5B activity

at higher concentrations of the control mAb could be explained by purely physical

interference with polymerase-substrate interaction.

A different picture can be observed in case of 7G8 mAbs. 7G8H and 7G8.1 do show

the same functional properties, very similar to those described by Nikonov et al. [87],

however, mAb 7G8M does not. This antibody demonstrates an enhanced NS5B inhibiting

activity, compared to all other mAbs tested. At a mAb to NS5B molar ratio of 4 the

polymerase function is inhibited almost completely. Starting from this point the cpm value is

approximating to the background radioactivity value of ~1500 cpm (not shown on the graph).

Control antibody 6G5.1 slightly varies the NS5B activity at molar ratios up to 1:1, however,

at the next points the activity remains more or less constant, indicating that there is no dose-

dependent inhibitory effect upon NS5B.

The F-2A 7G8 and 8B2 monoclonal antibodies, expressed from a single promoter,

were also analyzed in the RdRp inhibition assay. According to the experimental data,

recombinant 8B2_F2A mAbs behave similarly to the hybridomal 8B2 and, consequently,

similarly to above described recombinant 8B2H and 8B2M mAbs (Figure 23). On the

contrary, 7G8 F2A mAbs exhibit a very unexpected behaviour – both, chimeric and murine

antibodies show much greater potency to inhibit the NS5B polymerase in this assay, however,

still lesser than that of recombinant murine 7G8M antibody. Such an enhancement of

recombinant antibodies biologic activity might be possible due to several reasons. First, since

F2A antibodies are expressed from a single promoter resulting in 1:1 molar ratio of both

heavy and light chains a greater concentration of fully dimerized intact antibodies should be

produced. Whereas, in case mAbs are produced using two-gene cassettes, excess undimerized

heavy and/or light chains might interfere with the correct assembly of functional antibody.

Second, because F2A mAbs tended to precipitate during the buffer exchange these molecules

could also trigger the co-precipitation of NS5B and/or RNase inhibitor during the reaction,

and, consequently, diminish the synthesized radioactive RNA yields. The mAb precipitation

could also explain why the 7G8M_F2A antibody (whose sequence is identical to 7G8M mAb)

exhibits lower activity compared to that of 7G8M mAb. The tendency to precipitate, in its

turn, could be caused by incomplete furin cleavage of translated peptides and deficient folding

of the resulting proteins. In this case, the F-2A peptide sequence at the C-terminal part of the

heavy chain can probably bind to other antibody molecules and induce subsequent

precipitation, or physically interfere with antibody packing. On the other hand, in both cases

58

(precipitaion or low relative amount of functional antibody molecules) similar results should

have been probably observed also for 8B2 F2A mAbs, which, however, do not show any

significant improvement (as well as impairment) of their inhibitory activity. The presence of

two additional amino acids at the C-terminus of the antibody heavy chains does not seem to

be the reason also, because it is the terminal part of the constant domain, which is spatially

very far away from the antigen-binding site of the antibodies. Thus, for now the enhanced

inhibitory activity remains a puzzle for us, and additional experiments should be carried out,

in order to make sure, that such behaviour is not just an artefact, caused by the antibody

precipitation.

Thus, several conclusions can be made from these experiments:

1. The recombinant 7G8 and 8B2 mAbs work in the same manner as the

hybridoma mAbs, justifying the approach of antibody engineering applied in our study.

This approach might be exploited also for the construction of other chimeric antibodies of

interest.

2. 7G8_F2A antibodies exhibit an enhanced inhibitory activity, comparing to

hybridoma 7G8 mAb. The reason for this is unknown at present and additional

experiments are needed to reveal the nature of such unexpected behavior. At the same

time, 8B2_F2A antibodies show an inhibition pattern almost identical to that of

hybridoma 8B2 mAb, with only a slightly higher activity at lower mAb concentrations,

what might be a benefit of the one-promoter expression system.

3. The biologic activity of recombinant 7G8M antibody is higher than of its

chimeric and hybridoma analogues. This fact can be possibly explained by several

mutations in FR1 regions of heavy and light chain variable regions, introduced due to the

use of the degenerate primers. It is clear that the constant chains could not be the reason of

this enhancement, because mAbs 7G8H and 7G8.1, which have constant chains from

different sources, function identically to each other. Consequently, the variable regions

should be the case.

4. Another interesting point is the correlation of data obtained for 7G8 and 8B2

recombinant mAbs in ELISA and RdRp inhibition assay. RdRp assay is aimed in the first

place at testing the inhibitory activity of antibodies, whereas ELISA demonstrates the

affinity of the antibody to a given antigen. However, both affinity and biologic activity

have to be closely connected to each other. This is what we see, when comparing the

ELISA and RdRp assay results. 8B2M and 8B2H mAbs exhibit almost identical affinity to

59

NS5B and the same picture is seen in RdRp inhibition experiments – both mAbs are

equally efficient inhibitors in RdRp assay (Figure 23, 24). In case of 7G8H and 7G8M

mAbs it is evident that the inhibitory activity of 7G8M antibody is stronger than that of

7G8H mAb (Figure 24). At the same time, signal intensity obtained in ELISA assay for

7G8M antibody at the concentration of 0.1 ng/µl is higher than for 7G8H at the same

concentration (Figure 23), indicating the higher affinity of 7G8M to NS5B. These results

suppose that the higher affinity of 7G8M to NS5B makes it more efficient inhibitor,

compared to 7G8H (and to hybridoma 7G8, as can be judged from Figure 24).

Unfortunately, the hybridoma mAbs cannot be compared based on results of these

experiments, because different stocks of these antibodies were used in ELISA and RdRp

assays. At this point, it becomes clear that at least for 7G8M mAb higher affinity results in

higher inhibitory activity, and proves that results obtained for 7G8M in RdRp assay are

not an artefact.

3.1.8. Identification of mutations, possibly enhancing the biologic activity

of 7G8M mAb.

7G8H and 7G8M mAbs were constructed by us utilizing different strategies. Former,

was created by RF-cloning method (simultaneously modifying several amino acids in its FR1

regions, (see “methods”)). The latter, 7G8M, was prepared by utilizing degenerate primers,

which could, with high probability, modify several amino acids in the FR1 sequences of the

mAb variable regions (due to the PCR is biased method), specifically – 1, 3, 5 and 6-th amino

acids in VH and 4-th, 7-th and 8-th amino acids in VL (Figure 25).

Figure 25. Sequences of degenerate primers used in this study for amplification of antibody variable regions.

Posible amino acids that can be generate using these primers are shown in italics, above the respective nucleotide

triplets. Upper – VH primer, lower – VL primer. Restriction sites, introduced to the variable region sequences

during amplification are underlined.

60

The sequences of the variable regions of mAbs 7G8H and 7G8M were aligned and

carefully analyzed. The analysis revealed differences between 1st, 3

rd and 5

th amino acids of

VH regions, and between 3rd

and 4th

amino acids of VL regions of the antibodies (Figure 26).

Figure 26. Amino acid sequence alignment of chimeric and murine recombinant 7G8 antibody VH and VL

regions. Mismatching amino acids are shown in bold. Symbols in “consensus” line show the biochemical

similarity of mismatching amino acids. * means the same amino acids, : - high degree of similarity, and space –

completely dissimilar amino acids.

To the very best of our knowledge, there are no published works, showing that these

amino acids could be critical for the overall mAb conformation, or a conformation of its

CDRs. However, some of these residues are involved in the formation of β-sheet structures

through hydrogen bonding to other residues [8]. That is why we decided to graft these amino

acids from 7G8M antibody into the chimeric 7G8 mAb. If the assumption were correct, the

resulting antibody should get an increase in efficiency, similar to that of 7G8M antibody. This

would have allowed us, first, to construct a highly efficient chimeric antibody which could be

possibly used as a therapeutic agent in future, and second – to discover the amino acids,

which may play an important role in conformation of the antibody variable regions.

3.1.9. Exchanging FR1 regions of 7G8H antibody for those of 7G8M.

Grafting of the 7G8M FR1 regions to 7G8H chimeric construct was carried out using

the conventional restrictase-mediated cloning of the whole VH and VL regions (see

“Methods”). Although, this mAb was expressed from two cassettes, the expression gave an

impressive yield of 9 mg of antibody, what is ~2 times more, than it can be achievable using

hybridoma technology. This fact gives us a reason to suppose that mutations introduced to the

FR1 region of chimeric 7G8H antibody could somehow stabilize the variable domains of this

mAb, thereby increasing the stability of the whole immunoglobulin molecule and increasing

the amount of soluble antibody by lessening the risk of precipitation.

61

Summary

Two murine monoclonal antibodies inhibiting HCV RdRp in vitro were previously

identified and extensively described in our lab. The ability to inhibit the key enzyme in HCV

replication cycle demonstrates the therapeutic potential of these mAbs. As murine mAbs tend

to induce anti-immunoglobulin response in humans, they have to be appropriately modified

first and have to maintain their biologic activity after all modifications. Thus, two major tasks

were pursued in this study. First, development of chimeric HCV RdRp-specific mAbs with

preserved biologic activity. Second, development of a customizable chimerization strategy,

that could be used for rapid and reliable chimerization of virtually any other hybridoma-

produced mAbs.

Variable regions of mAbs 8B2 and 7G8 were successfully amplified from hybridoma

cDNA by utilizing degenerate primers. The sequences of the mAb variable regions were

determined by sequencing. Using the same degenerate primers strategy and unique restriction

endonuclease recognition site, a simple and reliable high-throughput system for amplification

of variable regions from hybridomas and joining them directly to human or mouse constant

regions was developed (together with Icosagen Group).

Using the above described system chimeric 7G8 and 8B2 genes, as well as

recombinant mouse 7G8 and 8B2 genes, were constructed and inserted into expression vector

(provided by Icosagen Group) utilizing two separate expression cassettes for both heavy and

light chains. Recombinant mAbs were produced utilizing novel QMCF technology developed

by Icosagen Group, yielding high amounts of protein – comparable and exceeding those

obtained using hybridoma technology.

Additional recombinant constructs of the same mAbs were generated, in which heavy

and light chain genes were connected with a short linker, encoding the furin cleavage site and

FMDV 2A peptide. These recombinant genes could be expressed from a single promoter,

producing mAb heavy and light chains at equimolar ratio, what resulted in approximately 2-

fold increase of antibody yields.

Sufficient amounts of wild type NS5B along with its inactive mutant form (GND)

were purified for use in biochemical experiments. Enzymatic activity of wtNS5B was

demonstrated in RdRp assay. Subsequently, all generated recombinant antibodies were tested

in RdRp assay for their ability to inhibit HCV NS5B polymerase. Antibodies produced using

62

two-promoter system were additionally tested in ELISA assay for their NS5B-binding

affinity. The results of these experiments clearly indicate that there is no loss of biologic

activity for any of the recombinant antibodies produced during this study compared to

hybridoma mAbs. Moreover, one of the recombinant murine mAbs, specifically 7G8M,

showed an enhanced inhibitory activity. This phenomenon was probably caused by several

mutations in its FR1 region, introduced by degenerate primers in PCR amplification stage.

These mutated amino acids were further grafted to the chimeric 7G8 antibody, in order to test

this hypothesis. For the moment, there is no data available on biologic activity of this

antibody and additional experiments still have to be done.

63

C hepatiidi viiruse RNA-sõltuvat RNA-polümeraasi inhibeerivate hiire

monokloonsete antikehade rekonstrueerimine ja bioloogiline aktiivsus

Aleksei Suslov

Resümee

Antikehad ehk immunoglobuliinid on heterodimeersed Y-tähe kujulised glükoproteiinid, mis

seovad spetsiifilisi märklaudmolekule – antigeene. Tüüpiline immunoglobuliini molekul

koosneb neljast polüpeptiidsest ahelast – kahest raskest ahelast (~50 kDa) ja kahest kergest

ahelast (~25 kDa). Raske ja kerge ahel, samuti nendest moodustunud dimeerid, on omavahel

ühendatud disulfiidsidemetega. Erineva spetsiifilisusega antikehade mõlema ahela N-

terminaalsed osad (esimesed ~110 aminohapet) varieeruvad oma järjestuses tugevasti, seega

on need regioonid tuntud kui V (variable) regioonid. Kergete ja raskete ahelate ülejäänud

piirkonnad on väga konserveerunud. Y-kujulise molekuli „käsi“ (ülemist avatud osa)

nimetatakse Fab fragmendiks, mis vastutab antigeeni sidumise eest. Molekuli „jalg“

omakorda vastutab rakus erinevate antigeenidega võitlemise mehhanismide aktiveerimise eest

ja seda nimetatakse Fc regiooniks (Fc retseptori sidumisala). V regioonid koosnevad kahest β-

lehest, mis asetsevad kohakuti. Need β-lehed koosnevad antiparalleelsetest β-ahelatest, mida

stabiliseerivad vesiniksidemed. β-ahelad on omavahel seotud erineva pikkusega lingudega,

mis moodustavad mõlema ahela V regioonide kolm kõige varieeruvamat piirkonda. Neid

linge nimetatakse hüpervarieeruvateks (hypervariable loop) ja tähistatakse raske ahela puhul

H1, H2, H3 ja kerge ahela puhul L1, L2, L3. Kristallstruktuuride analüüs näitas, et mõlema

ahela varieeruvad lingud moodustavadki antigeeni siduva saidi. Kuna need regioonid on

komplementaarsed vastava antigeeni struktuuriga, on need tuntud kui komplementaarsust

määravad regioonid (complementarity determining regions; CDR-id). Ülejäänud varieeruvate

regioonide osad moodustavad stabiilse karkassi, mis hoiab hüpervarieeruvaid regioone õiges

konformatsioonis. Teades aminohappejääke, mis vastutavad nende konformatsioonide

moodustamise eest, on võimalik kanda antigeenset spetsiifilisust ühelt antikehalt teisele ilma

aktseptor-antikeha funktsiooni rikkumata.

Antikeha V regioonide järjestusi kodeerivad geenid koosnevad erinevatest omavahel

juhuslikult rekombineeruvatest geenisegmentidest. Seepärast on võimalike kombinatsioonide

arv väga suur, tekitades suure hulga erineva spetsiifilisusega antikehasid. Varieeruvus ning

väga spetsiifiline ja tugev antigeeni sidumise võime muudavad antikehad terapeutilise

potentsiaaliga molekulideks. Selline idee oli juba ammu, kuid tehnoloogia hiire

monokloonsete (sama spetsiifilisusega) antikehade tootmiseks ilmus alles 35 aastat tagasi.

Tehnoloogia kasutusele võtmise järgselt selgus peagi, et hiire antikehad, manustatuna

inimesele, põhjustavad anti-immunoglobuliin immuunvastuse. Seega hakati arendama

erinevaid strateegiaid, mille eesmärgiks oli vähendada hiire antikehade immunogeensust,

muutes nende järjestusi rohkem inimeselaadseteks. Taoline lähenemine hõlmas kimäärsete

(hiire antikeha varieeruvad regioonid liidetakse inimese antikeha konstantsete regioonidega)

ja humaniseeritud (hiire antikeha CDR-id viiakse inimese antikeha struktuuri) antikehade

konstrueerimist. Viimane strateegia osutus edukaks ning tänapäeval on suurim osa turul

olevatest terapeutilistest antikehadest just humaniseeritud antikehad.

Ühed suure terapeutilise potentsiaaliga antikehad on rakusisesed antikehad. Kuna konstantsed

regioonid ei ole raku sees funktsioneerimiseks vajalikud, konstrueeritakse selliseid antikehi

põhiliselt scFv molekuli kujul. scFv koosneb kerge ja raske ahela varieeruvatest domäänidest,

mis on omavahel seotud peptiidse linkeriga. Viimasel ajal on rakusiseste antikehade

64

tuvastamiseks ja isoleerimiseks arendatud mitmeid tehnoloogiaid. Sellised antikehad (scFv

molekulid) sisaldavad konsensusjärjestust, mis tagab nende stabiilsuse, võimaldades

korrektset struktuuri isegi tavaliselt disulfiidsidemete moodustamist takistavas redutseerivas

rakusiseses keskkonnas. Viimase 20 aasta jooksul on konstrueeritud suhteliselt palju

rakusiseselt töötavaid antikehasid, kuid ükski neist ei ole veel jõudnud turule. Tehnoloogiad,

mis võimaldavad stabiilsete rakusiseste scFv-de isoleerimist, suudavad suure tõenäosusega

seda olukorda lähemas tulevikus muuta.

Antud magistritöö eesmärgiks oli kahe meie laboris eelnevalt kirjeldatud C hepatiidi

viiruse RNA-sõltuvat RNA-polümeraasi inhibeeriva hiire 7G8 ja 8B2 antikeha varieeruvate

regioonide kloneerimine hübridoomi cDNA-st, nende antikehade rekombinantsete kimäärsete

variantide konstrueerimine ja bioloogilise aktiivsuse määramine.

Tuginedes eksperimentaalsel teel saadud andmetele, võib töö tulemused kokku võtta

järgnevalt:

7G8 ja 8B2 varieeruvad regioonid kloneeriti hübridoomi cDNA-st ja liideti kokku kas

inimese või hiire konstantsete regioonidega, genereerides nii kimäärseid kui natiivseid

rekombinantsete antikehade geene. Selleks loodi lihtne ja efektiivne vektor-praimer süsteem,

mis võimaldab amplifitseerida varieeruvaid regioone ükskõik millisest hübridoomist ja liita

need inimese või hiire konstantsete regioonidega.

Konstrueeritud rekombinantsed geenid kloneeriti ekspressioonivektoritesse kahel

viisil: esimesel juhul ekspresseeriti antikeha ahelad kahe eraldi promootori alt, teisel juhul

ühendati mõlema ahela geenid omavahel lühikese järjestusega, mis kodeeris furiini lõikesaiti

ja Foot-and-mouse disease viiruse 2A peptiidi. Ahelate ühendamine võimaldas ekspresseerida

mõlemat geeni ühe transkriptsiooniühikuna, seejuures vahelejääv linkerala tegi võimalikuks

nende post-translatsioonilise eraldamise. Sellise ühe promootori alt ekspresseeritud kerge ja

raske ahela molaarne suhe on 1:1.

Antikehad toodeti, kasutades QMCF tehnoloogiat (välja töötatud Icosagen Group’i

poolt). Ühe promootori alt ekspresseerides oli antikeha saagis umbes 2 korda suurem kui kahe

promootori alt ekspresseerides ja ligikaudu 1,5 korda suurem kui hübridoomi tehnoloogia

puhul.

Metsiktüüpi NS5B RNA polümeraas ja selle inaktiivne mutantne vorm (GND)

puhastati piisavas hulgas ja metsiktüüpi NS5B ensümaatiline aktiivsus näidati in vitro RNA

sünteesi reaktsioonides.

Konstrueeritud antikehade võime blokeerida HCV polümeraasi aktiivsust kontrolliti in

vitro RNA sünteesi katsetes. Kahe promootori kasutamisel ekspresseeritud antikehade

afiinsus NS5B polümeraasile kontrolliti lisaks ka ELISA testi abil. Saadud tulemused

näitavad selgelt, et rekombinantsete antikehade aktiivsus ja afiinsus ei ole langenud ja veelgi

enam, rekombinantse hiire 7G8 antikeha puhul täheldati isegi inhibiitoorse aktiivsuse tõusu,

mis võiks olla seletatav selle antikeha FR1 regioonidesse degenereeritud praimeritega PCR-i

käigus sisestatud mutatsioonidega. Tõestamaks (või ümber lükkamaks) seda hüpoteesi (et just

need mutatsioonid on muutnud antikeha aktiivsust), olid muteerunud aminohapped ümber

tõstetud kimäärse 7G8 antikeha sisse. Praeguseks hetkeks eksperimentaalsed andmed selle

antikeha funktsionaalsuse kohta veel puuduvad.

65

Acknowledgements

This work was carried out at Professor Mart Ustav’s lab at the chair of Microbiology

and Virology, Institute of Technology, Estonia, in collaboration with Icosagen Group. There

is a lot of people who contributed to a greater or lesser extent to this work, which definitely

would not be done without all of them. I would like to thank:

First of all, my supervisor Andrei Nikonov, for introducing me to the world of

Science, teaching me a lot of things, especially to work independently being at the same time

a part of a group and to be accountable for my actions and decisions. Also, for stimulating

talks and keeping me motivated on my way to a master’s degree. You have changed my view

of many things.

My professor Mart Ustav for giving me possibility to work in Your team and for

interesting projects and ideas.

Icosagen team – Andres Tover, Radi Tegova, Anne Kalling, Andres Männik, Polina

Verhovtsova – for technical help, interesting solutions and of course for production of all of

these antibodies in large quantities.

Erkki Juronen – immunogeneetika ja -keemia vanemteadur at General and Molecular

Pathology Institute, – for your co-operation, helpful advices and perfectly working mAbs.

My colleagues – Mihkel Allik – for teaching me various tips and tricks, and for

valuable discussions and advices; also Nele Jaanson, Axel Soosaar, Vallo Varik.

Colleagues from other labs – Joachim Gerholdt – for providing me with always fresh

isotopes; Sergei Kopantšuk – for making my radioactive life easier; also Mardo Kõiv, Ervin

Valk, Evgeni Mihheev for various assistance.

Special thanks to Ly Käsper, Age Utt, Sirle Saul, Teele Karafin for enriching our floor

with these positive vibrations.

Karin Lauga – for reviewing the estonian part of this work.

My friends – for your support and inspiration.

And, of course, my parents for beeing help in all times.

66

References

1. Goldsby, R., Kuby Immunology 5-th

edition. 2003.

2. Janeway, C.A., Jr., Approaching the asymptote? Evolution and revolution in

immunology. Cold Spring Harb Symp Quant Biol, 1989. 54 Pt 1: p. 1-13.

3. Edelman, G.M. and B. Benacerraf, On structural and functional relations between

antibodies and proteins of the gamma-system. Proc Natl Acad Sci U S A, 1962. 48: p.

1035-42.

4. Edelman, G.M. and M.D. Poulik, Studies on structural units of the gamma-globulins. J

Exp Med, 1961. 113: p. 861-84.

5. Fleischman, J.B., R.H. Pain, and R.R. Porter, Reduction of gamma-globulins. Arch

Biochem Biophys, 1962. Suppl 1: p. 174-80.

6. Wu, T.T. and E.A. Kabat, An analysis of the sequences of the variable regions of

Bence Jones proteins and myeloma light chains and their implications for antibody

complementarity. J Exp Med, 1970. 132(2): p. 211-50.

7. Poljak, R.J., et al., The three-dimensional structure of the fab' fragment of a human

myeloma immunoglobulin at 2.0-angstrom resolution. Proc Natl Acad Sci U S A,

1974. 71(9): p. 3440-4.

8. Chothia, C. and A.M. Lesk, Canonical structures for the hypervariable regions of

immunoglobulins. J Mol Biol, 1987. 196(4): p. 901-17.

9. Taranta, A. and E.C. Franklin, Complement fixation by antibody fragments. Science,

1961. 134: p. 1981-2.

10. Kohler, G. and C. Milstein, Continuous cultures of fused cells secreting antibody of

predefined specificity. Nature, 1975. 256(5517): p. 495-7.

11. Reichert, J.M., et al., Monoclonal antibody successes in the clinic. Nat Biotechnol,

2005. 23(9): p. 1073-8.

12. Worn, A. and A. Pluckthun, An intrinsically stable antibody scFv fragment can

tolerate the loss of both disulfide bonds and fold correctly. FEBS Lett, 1998. 427(3):

p. 357-61.

13. Behar, S.M., et al., The molecular origin of anti-DNA antibodies. Int Rev Immunol,

1989. 5(1): p. 23-42.

14. Radic, M.Z., et al., Structural patterns in anti-DNA antibodies from MRL/lpr mice.

Cold Spring Harb Symp Quant Biol, 1989. 54 Pt 2: p. 933-46.

67

15. Tiselius, A. and E.A. Kabat, An Electrophoretic Study of Immune Sera and Purified

Antibody Preparations. J Exp Med, 1939. 69(1): p. 119-131.

16. Ovary, Z. and F. Karush, Studies on the immunologic mechanism of anaphylaxis. II.

Sensitizing and combining capacity in vivo of fractions separated from papain digests

of antihapten antibody. J Immunol, 1961. 86: p. 146-50.

17. Porter, R.R., The formation of a specific inhibitor by hydrolysis of rabbit

antiovalbumin. Biochem J, 1950. 46(4): p. 479-84.

18. Charlwood, P.A., Ultracentrifugal examination of digestion products from rabbit

gamma-globulin. Biochem J, 1959. 73: p. 126-7.

19. Nisonoff, A., G. Markus, and F.C. Wissler, Separation of univalent fragments of

rabbit antibody by reduction of a single, labile disulphide bond. Nature, 1961. 189: p.

293-5.

20. Porter, R.R., The hydrolysis of rabbit y-globulin and antibodies with crystalline

papain. Biochem J, 1959. 73: p. 119-26.

21. Dangl, J.L., et al., Segmental flexibility and complement fixation of genetically

engineered chimeric human, rabbit and mouse antibodies. EMBO J, 1988. 7(7): p.

1989-94.

22. Roux, K.H., et al., Comparisons of the ability of human IgG3 hinge mutants, IgM, IgE,

and IgA2, to form small immune complexes: a role for flexibility and geometry. J

Immunol, 1998. 161(8): p. 4083-90.

23. Hozumi, N. and S. Tonegawa, Evidence for somatic rearrangement of

immunoglobulin genes coding for variable and constant regions. Proc Natl Acad Sci

U S A, 1976. 73(10): p. 3628-32.

24. Rabbitts, T.H. and A. Forster, Evidence for noncontiguous variable and constant

region genes in both germ line and myeloma DNA. Cell, 1978. 13(2): p. 319-27.

25. Seidman, J.G. and P. Leder, The arrangement and rearrangement of antibody genes.

Nature, 1978. 276(5690): p. 790-5.

26. Tonegawa, S., et al., Sequence of a mouse germ-line gene for a variable region of an

immunoglobulin light chain. Proc Natl Acad Sci U S A, 1978. 75(3): p. 1485-9.

27. Matsuda, F., et al., The complete nucleotide sequence of the human immunoglobulin

heavy chain variable region locus. J Exp Med, 1998. 188(11): p. 2151-62.

28. Li, H., et al., Genetic diversity of the human immunoglobulin heavy chain VH region.

Immunol Rev, 2002. 190: p. 53-68.

68

29. Corbett, S.J., et al., Sequence of the human immunoglobulin diversity (D) segment

locus: a systematic analysis provides no evidence for the use of DIR segments,

inverted D segments, "minor" D segments or D-D recombination. J Mol Biol, 1997.

270(4): p. 587-97.

30. Ravetch, J.V., et al., Structure of the human immunoglobulin mu locus:

characterization of embryonic and rearranged J and D genes. Cell, 1981. 27(3 Pt 2):

p. 583-91.

31. Tonegawa, S., Somatic generation of antibody diversity. Nature, 1983. 302(5909): p.

575-81.

32. Epp, O., et al., The molecular structure of a dimer composed of the variable portions

of the Bence-Jones protein REI refined at 2.0-A resolution. Biochemistry, 1975.

14(22): p. 4943-52.

33. Davies, D.R., E.A. Padlan, and D.M. Segal, Three-dimensional structure of

immunoglobulins. Annu Rev Biochem, 1975. 44: p. 639-67.

34. Schiffer, M., et al., Structure of a lambda-type Bence-Jones protein at 3.5-A

resolution. Biochemistry, 1973. 12(23): p. 4620-31.

35. Furey, W., Jr., et al., Structure of a novel Bence-Jones protein (Rhe) fragment at 1.6 A

resolution. J Mol Biol, 1983. 167(3): p. 661-92.

36. Marquart, M., et al., Crystallographic refinement and atomic models of the intact

immunoglobulin molecule Kol and its antigen-binding fragment at 3.0 A and 1.0 A

resolution. J Mol Biol, 1980. 141(4): p. 369-91.

37. Saul, F.A., L.M. Amzel, and R.J. Poljak, Preliminary refinement and structural

analysis of the Fab fragment from human immunoglobulin new at 2.0 A resolution. J

Biol Chem, 1978. 253(2): p. 585-97.

38. Segal, D.M., et al., The three-dimensional structure of a phosphorylcholine-binding

mouse immunoglobulin Fab and the nature of the antigen binding site. Proc Natl Acad

Sci U S A, 1974. 71(11): p. 4298-302.

39. Suh, S.W., et al., The galactan-binding immunoglobulin Fab J539: an X-ray

diffraction study at 2.6-A resolution. Proteins, 1986. 1(1): p. 74-80.

40. Jung, S., et al., The importance of framework residues H6, H7 and H10 in antibody

heavy chains: experimental evidence for a new structural subclassification of antibody

V(H) domains. J Mol Biol, 2001. 309(3): p. 701-16.

69

41. Honegger, A. and A. Pluckthun, The influence of the buried glutamine or glutamate

residue in position 6 on the structure of immunoglobulin variable domains. J Mol

Biol, 2001. 309(3): p. 687-99.

42. Foote, J. and G. Winter, Antibody framework residues affecting the conformation of

the hypervariable loops. J Mol Biol, 1992. 224(2): p. 487-99.

43. Tramontano, A., C. Chothia, and A.M. Lesk, Framework residue 71 is a major

determinant of the position and conformation of the second hypervariable region in

the VH domains of immunoglobulins. J Mol Biol, 1990. 215(1): p. 175-82.

44. Kabat, E.A., T.T. Wu, and H. Bilofsky, Unusual distributions of amino acids in

complementarity-determining (hypervariable) segments of heavy and light chains of

immunoglobulins and their possible roles in specificity of antibody-combining sites. J

Biol Chem, 1977. 252(19): p. 6609-16.

45. Padlan, E.A. and D.R. Davies, Variability of three-dimensional structure in

immunoglobulins. Proc Natl Acad Sci U S A, 1975. 72(3): p. 819-23.

46. Padlan, E.A., Evaluation of the structural variation among light chain variable

domains. Mol Immunol, 1979. 16(5): p. 287-96.

47. Chothia, C., et al., Conformations of immunoglobulin hypervariable regions. Nature,

1989. 342(6252): p. 877-83.

48. Chothia, C., et al., The predicted structure of immunoglobulin D1.3 and its

comparison with the crystal structure. Science, 1986. 233(4765): p. 755-8.

49. Clark, M., Antibody humanisation: a case of 'emperor's new clothes'? Immunology

Today, 2000.

50. Morrison, S.L., et al., Chimeric human antibody molecules: mouse antigen-binding

domains with human constant region domains. Proc Natl Acad Sci U S A, 1984.

81(21): p. 6851-5.

51. Boulianne, G.L., N. Hozumi, and M.J. Shulman, Production of functional chimaeric

mouse/human antibody. Nature, 1984. 312(5995): p. 643-6.

52. Benjamin, R.J., et al., Tolerance to rat monoclonal antibodies. Implications for

serotherapy. J Exp Med, 1986. 163(6): p. 1539-52.

53. Bruggemann, M., et al., The immunogenicity of chimeric antibodies. J Exp Med, 1989.

170(6): p. 2153-7.

54. Jones, P.T., et al., Replacing the complementarity-determining regions in a human

antibody with those from a mouse. Nature, 1986. 321(6069): p. 522-5.

70

55. Riechmann, L., et al., Reshaping human antibodies for therapy. Nature, 1988.

332(6162): p. 323-7.

56. Hale, G., et al., Remission induction in non-Hodgkin lymphoma with reshaped human

monoclonal antibody CAMPATH-1H. Lancet, 1988. 2(8625): p. 1394-9.

57. Alinari, L., et al., Alemtuzumab (Campath-1H) in the treatment of chronic lymphocytic

leukemia. Oncogene, 2007. 26(25): p. 3644-53.

58. Flynn, J.M. and J.C. Byrd, Campath-1H monoclonal antibody therapy. Curr Opin

Oncol, 2000. 12(6): p. 574-81.

59. Co, M.S., et al., Humanized antibodies for antiviral therapy. Proc Natl Acad Sci U S

A, 1991. 88(7): p. 2869-73.

60. Gorman, S.D., et al., Reshaping a therapeutic CD4 antibody. Proc Natl Acad Sci U S

A, 1991. 88(10): p. 4181-5.

61. Routledge, E.G., et al., A humanized monovalent CD3 antibody which can activate

homologous complement. Eur J Immunol, 1991. 21(11): p. 2717-25.

62. Shearman, C.W., et al., Construction, expression and characterization of humanized

antibodies directed against the human alpha/beta T cell receptor. J Immunol, 1991.

147(12): p. 4366-73.

63. Tempest, P.R., et al., Reshaping a human monoclonal antibody to inhibit human

respiratory syncytial virus infection in vivo. Biotechnology (N Y), 1991. 9(3): p. 266-

71.

64. Carter, P., et al., Humanization of an anti-p185HER2 antibody for human cancer

therapy. Proc Natl Acad Sci U S A, 1992. 89(10): p. 4285-9.

65. Woodle, E.S., et al., Humanized OKT3 antibodies: successful transfer of immune

modulating properties and idiotype expression. J Immunol, 1992. 148(9): p. 2756-63.

66. Sato, K., et al., Reshaping a human antibody to inhibit the interleukin 6-dependent

tumor cell growth. Cancer Res, 1993. 53(4): p. 851-6.

67. Queen, C., et al., A humanized antibody that binds to the interleukin 2 receptor. Proc

Natl Acad Sci U S A, 1989. 86(24): p. 10029-33.

68. Kettleborough, C.A., et al., Humanization of a mouse monoclonal antibody by CDR-

grafting: the importance of framework residues on loop conformation. Protein Eng,

1991. 4(7): p. 773-83.

71

69. Singer, II, et al., Optimal humanization of 1B4, an anti-CD18 murine monoclonal

antibody, is achieved by correct choice of human V-region framework sequences. J

Immunol, 1993. 150(7): p. 2844-57.

70. Antman, K.H. and D.M. Livingston, Intracellular neutralization of SV40 tumor

antigens following microinjection of specific antibody. Cell, 1980. 19(3): p. 627-35.

71. Biocca, S., M.S. Neuberger, and A. Cattaneo, Expression and targeting of

intracellular antibodies in mammalian cells. EMBO J, 1990. 9(1): p. 101-8.

72. Biocca, S., et al., Redox state of single chain Fv fragments targeted to the endoplasmic

reticulum, cytosol and mitochondria. Biotechnology (N Y), 1995. 13(10): p. 1110-5.

73. Glockshuber, R., T. Schmidt, and A. Pluckthun, The disulfide bonds in antibody

variable domains: effects on stability, folding in vitro, and functional expression in

Escherichia coli. Biochemistry, 1992. 31(5): p. 1270-9.

74. Steipe, B., et al., Sequence statistics reliably predict stabilizing mutations in a protein

domain. J Mol Biol, 1994. 240(3): p. 188-92.

75. Ohage, E. and B. Steipe, Intrabody construction and expression. I. The critical role of

VL domain stability. J Mol Biol, 1999. 291(5): p. 1119-28.

76. Wirtz, P. and B. Steipe, Intrabody construction and expression III: engineering

hyperstable V(H) domains. Protein Sci, 1999. 8(11): p. 2245-50.

77. Visintin, M., et al., Selection of antibodies for intracellular function using a two-

hybrid in vivo system. Proc Natl Acad Sci U S A, 1999. 96(21): p. 11723-8.

78. Tse, E., et al., Intracellular antibody capture technology: application to selection of

intracellular antibodies recognising the BCR-ABL oncogenic protein. J Mol Biol,

2002. 317(1): p. 85-94.

79. Visintin, M., et al., The intracellular antibody capture technology (IACT): towards a

consensus sequence for intracellular antibodies. J Mol Biol, 2002. 317(1): p. 73-83.

80. Tanaka, T., et al., De novo production of diverse intracellular antibody libraries.

Nucleic Acids Res, 2003. 31(5): p. e23.

81. Tanaka, T. and T.H. Rabbitts, Intrabodies based on intracellular capture frameworks

that bind the RAS protein with high affinity and impair oncogenic transformation.

EMBO J, 2003. 22(5): p. 1025-35.

82. Stocks, M.R., Intrabodies: Production and promise. Drug Discovery Today, 2004.

9(22): p. 960-966.

72

83. Kontermann, R.E., Intrabodies as therapeutic agents. Methods, 2004. 34(2): p. 163-

170.

84. Zemel, R., et al., Inhibition of hepatitis C virus NS3-mediated cell transformation by

recombinant intracellular antibodies. J Hepatol, 2004. 40(6): p. 1000-7.

85. Sullivan, D.E., et al., Construction and characterization of an intracellular single-

chain human antibody to hepatitis C virus non-structural 3 protein. J Hepatol, 2002.

37(5): p. 660-8.

86. Boonrod, K., et al., Single-chain antibodies against a plant viral RNA-dependent RNA

polymerase confer virus resistance. Nat Biotechnol, 2004. 22(7): p. 856-62.

87. Nikonov, A., E. Juronen, and M. Ustav, Functional characterization of fingers

subdomain-specific monoclonal antibodies inhibiting the hepatitis C virus RNA-

dependent RNA polymerase. J Biol Chem, 2008. 283(35): p. 24089-102.

88. Larkin, M.A., et al., Clustal W and Clustal X version 2.0. Bioinformatics, 2007.

23(21): p. 2947-8.

89. Mikaelian, I. and A. Sergeant, A general and fast method to generate multiple site

directed mutations. Nucleic Acids Res, 1992. 20(2): p. 376.

90. Wu, W., et al., A novel PCR strategy for high-efficiency, automated site-directed

mutagenesis. Nucleic Acids Res, 2005. 33(13): p. e110.

91. van den Ent, F. and J. Lowe, RF cloning: a restriction-free method for inserting target

genes into plasmids. J Biochem Biophys Methods, 2006. 67(1): p. 67-74.

92. Binder, M., et al., Identification of determinants involved in initiation of hepatitis C

virus RNA synthesis by using intergenotypic replicase chimeras. J Virol, 2007. 81(10):

p. 5270-83.

93. Laemmli, U.K., Cleavage of structural proteins during the assembly of the head of

bacteriophage T4. Nature, 1970. 227(5259): p. 680-5.

94. Pfaff, E., et al., Antibodies against a preselected peptide recognize and neutralize foot

and mouth disease virus. EMBO J, 1982. 1(7): p. 869-74.

95. Wang, Z., et al., Universal PCR amplification of mouse immunoglobulin gene variable

regions: the design of degenerate primers and an assessment of the effect of DNA

polymerase 3' to 5' exonuclease activity. J Immunol Methods, 2000. 233(1-2): p. 167-

77.

96. Orlandi, R., et al., Cloning immunoglobulin variable domains for expression by the

polymerase chain reaction. Proc Natl Acad Sci U S A, 1989. 86(10): p. 3833-7.

73

97. Fang, J., et al., Stable antibody expression at therapeutic levels using the 2A peptide.

Nat Biotechnol, 2005. 23(5): p. 584-90.

98. van den Ouweland, A.M., et al., Structural homology between the human fur gene

product and the subtilisin-like protease encoded by yeast KEX2. Nucleic Acids Res,

1990. 18(3): p. 664.

99. van de Ven, W.J., et al., Furin is a subtilisin-like proprotein processing enzyme in

higher eukaryotes. Mol Biol Rep, 1990. 14(4): p. 265-75.

100. Yang, S., et al., Development of optimal bicistronic lentiviral vectors facilitates high-

level TCR gene expression and robust tumor cell recognition. Gene Ther, 2008.

15(21): p. 1411-23.

101. Robertson, B.H., et al., Nucleotide and amino acid sequence coding for polypeptides

of foot-and-mouth disease virus type A12. J Virol, 1985. 54(3): p. 651-60.

102. Ryan, M.D., A.M. King, and G.P. Thomas, Cleavage of foot-and-mouth disease virus

polyprotein is mediated by residues located within a 19 amino acid sequence. J Gen

Virol, 1991. 72 ( Pt 11): p. 2727-32.

103. Lohmann, V., et al., Biochemical properties of hepatitis C virus NS5B RNA-dependent

RNA polymerase and identification of amino acid sequence motifs essential for

enzymatic activity. J Virol, 1997. 71(11): p. 8416-28.