complement-mediated killing of cancer...

74
COMPLEMENT-MEDIATED KILLING OF CANCER CELLS Cover: fluorescence microscopical image of cryostat sections of ovarian microtumors treated with antibodies and complement. Cells with yellow fluorescence are killed by complement.

Upload: dangmien

Post on 17-Jul-2018

218 views

Category:

Documents


0 download

TRANSCRIPT

COMPLEMENT-MEDIATED KILLING OF CANCER CELLS

Cover: fluorescence microscopical image of cryostat sections of ovarian

microtumors treated with antibodies and complement. Cells with yellow

fluorescence are killed by complement.

© 2003 by Juha Hakulinen

Printed at Yliopistopaino, Helsinki, Finland

ISBN 952-91-5503-4 (sid.)

ISBN 952-10-0884-9 (pdf)

http://ethesis.helsinki.fi

COMPLEMENT-MEDIATED KILLING OF CANCER CELLS

JUHA HAKULINEN

Haartman Institute

Department of Bacteriology and Immunology

University of Helsinki

Finland

Academic Dissertation

To be publicly discussed, with the permission of the Medical Faculty of the

University of Helsinki, in the small Auditorium of the Haartman Institute,

Haartmaninkatu 3, Helsinki, on Saturday, January 11th, 2003, at 12 o´clock

noon.

SUPERVISOR

Seppo Meri

MD, PhD, Professor

Haartman Institute

Department of Bacteriology and Immunology

University of Helsinki

REVIEWERS

Mikko Hurme

MD, PhD, Professor

University of Tampere Medical School

and

Timo Paavonen

MD, PhD, Docent

Department of Pathology

University of Helsinki

OPPONENT

Olli Lassila

MD, PhD, Professor

Department of Medical Microbiology

University of Turku

5

CONTENTS

ORIGINAL PUBLICATIONS...............................................................................................7

ABBREVIATIONS..............................................................................................................8

ABSTRACT .................................................................................................................... 10

1. INTRODUCTION........................................................................................................ 12

2. REVIEW OF THE LITERATURE................................................................................. 14

2.1 The complement system................................................................................. 14

2.1.1 Overview ..................................................................................................... 14

2.1.2 The alternative pathway.......................................................................... 16

2.1.3 The classical pathway.............................................................................. 17

2.1.4 The lectin pathway.................................................................................... 18

2.1.5 The lytic pathway...................................................................................... 18

2.1.6 Soluble regulators of complement......................................................... 20

2.1.7 Membrane-bound complement regulatory proteins (mCRP)............. 21

2.1.8 Rat complement system.......................................................................... 26

2.2 The immune system and transformed cells................................................. 27

2.2.1 Antigen-presenting cells (APC)................................................................ 27

2.2.2 T lymphocytes ........................................................................................... 28

2.2.3 Natural killer cells (NK cells).................................................................... 29

2.2.4 Tumor antigens ......................................................................................... 30

2.2.5 Antibody responses against tumor-associated antigens (TAA) in

cancer patients in vivo .......................................................................... 30

2.2.6 Antibody-induced effector mechanisms................................................ 31

2.2.7 Tumor antigens and immunotherapy of cancer.................................. 33

3. AIMS OF THE STUDY .............................................................................................. 37

4. MATERIALS AND METHODS .................................................................................. 38

5. RESULTS AND DISCUSSION................................................................................... 41

5.1 mCRP on tumor cells and milk fat globules (MFG; I, II, III, IV)................... 42

5.1.2 Characterization of CD59 on T47D cells and MFG (I, II) ................... 44

6

5.2 Complement-mediated killing of MCF7 and T47D cells (I)......................... 46

5.2.1 Expression of TAA on ovarian cancer cells (III) .................................. 48

5.2.2 Complement-dependent cytotoxicity (CDC) and ovarian cancer cells

(III)............................................................................................................ 49

5.3 CDC and microtumor spheroids (MTS; IV)................................................. 50

5.3.1 The penetration of mAb and complement into MTS .......................... 54

5.4 A rat model to study complement activation in vivo (V)......................... 56

5.4.1 CDC and rat colorectal cancer cells...................................................... 57

5.4.2 C3 deposition on CC531 cells and CDC................................................. 57

5.4.3 Complement activation in situ................................................................. 58

5.4.4 Homing of mAb into tumors and complement activation in vivo..... 59

6. CONCLUSIONS.......................................................................................................... 61

7. ACKNOWLEDGMENTS.............................................................................................. 63

8. REFERENCES............................................................................................................. 65

7

ORIGINAL PUBLICATIONS

This thesis is based on the following original articles, which are referred to in

the text by their Roman numerals. Some unpublished data are also presented.

I Hakulinen J and Meri S. (1994). Expression and function of the

complement membrane attack complex inhibitor protectin (CD59) on

human breast cancer cells. Lab Invest 71, 820-827.

II Hakulinen J and Meri S. (1995). Shedding and enrichment of the

glycolipid anchored complement lysis inhibitor protectin (CD59) into milk

fat globules. Immunology 85, 495-501.

III Bjørge L, Hakulinen J, Wahlström T, Matre R and Meri S. (1997).

Complement regulatory proteins in ovarian malignancies. Int J Cancer

70, 14-25.

IV Hakulinen J and Meri S. (1998). Complement-mediated lysis o f

microtumors in vitro. Am J Pathol 153, 845-855.

V Gelderman K, Hakulinen J, Hagenaars M, Kuppen P, Meri S and Gorter A..

(2002). Membrane-bound complement regulatory proteins inhibit

complement activation by immunotherapeutic mAb in a syngeneic r a t

colorectal cancer model. Submitted.

The articles in this thesis have been reproduced with the permission of the

copyright holders: the United States and Canadian Academy of Pathology (I),

Blackwell Publishing (II), John Wiley & Sons Inc. (III) and the American Society

of Investigative Pathology (IV).

8

ABBREVIATIONS

51Cr Chromium-51

125I Iodine-125

ADCC antibody-dependent cellular cytotoxicity

AF ascitic fluid

Ag antigen

BSA bovine serum albumin

C4bp C4b binding protein

C9DS human serum deficient in C9

CD cluster of differentiation

CDC complement-dependent cytotoxicity

CDCC complement-dependent cellular cytotoxicity

CD35 complement receptor type 1 (CR1)

CD46 membrane cofactor protein (MCP)

CD55 decay-accelerating factor (DAF)

CD59E CD59 isolated from human erythrocytes

CD59M CD59 isolated from breast milk

CD59U CD59 isolated from urine

CD95 Fas receptor

CR1 complement receptor type 1 (CD35)

DAF decay-accelerating factor (CD55)

ECL electrochemiluminescence

FITC fluorescein isothiocyanate

GPE guinea pig erythrocyte

GPI glycosyl-phosphatidylinositol

IF immunofluorescence

kDa kilodalton

mAb monoclonal antibody

MAC membrane attack complex of complement

MASP mannose binding lectin associated serine protease

MBL mannose binding lectin

MCP membrane cofactor protein (CD46)

9

mCRP membrane-bound complement regulatory protein

MFG milk fat globule

Mr relative molecular weight

MTS microtumor spheroid

NHS normal human serum

NK-cell natural killer cell

NP40 Nonidet P40

OD optical density

pAb polyclonal antibody

PBS phosphate buffered saline

PI propidium iodide

PIPLC phosphatidylinositol-specific phospholipase C

PIPLD phosphatidylinositol-specific phospholipase D

SC5b-8 soluble C5b-8 complex

SDS-PAGE sodium dodecyl sulphate polyacrylamide gel

electrophoresis

TAA tumor associated antigen

TCC terminal complement complex

VBS veronal buffered saline

10

ABSTRACT

The human blood complement system is a part of the innate immune system.

The principal function of complement is to defend the host against microbial

infections and other foreign material. Furthermore, complement participates in

the clearence of apoptotic cells and immune complexes from the blood and

other tissues. The activation of complement by antibodies or by a foreign

surface results in opsonization, chemotaxis and direct killing of microbes and

infected cells. In the host, complement activation causes inflammation and

tissue damage. Host cells express complement regulators (CD46, CD55 and

CD59) on their surfaces to restrict harmful complement activation on the cells

to a minimum. However, the same molecules also make complement-mediated

destruction of malignant cells difficult. The purpose of the current study was

to respond to this challenge by using antibodies and complement as an

effector mechanism to destroy malignant cells and by analyzing the

significance of the membrane bound complement regulator CD59 for the

cancer cell survival from complement attack. In the first study, the

complement resistance of breast cancer cells (MCF-7 and T47D) was

examined. It was possible to increase complement-mediated lysis of these cells

by inactivating CD59 on the cell membranes with a specific monoclonal

antibody (I). During the first study it was noticed that CD59 was strongly

stained in the lumina of milk ductules in sections of breast cancer tissue. This,

and the fact that CD59 had been previously detected in the human breast

milk, led to the observation (II) that CD59 in milk is associated with structures

called milk fat globules (MFG). In the third study (III) complement regulators

CD46 and CD59 were found to be strongly expressed on ovarian cancer cells.

Furthermore, it was possible to kill ovarian cancer cells using NHS as a source

of complement after sensitizing the cells with a mAb reacting with an ovarian

tumor associated antigen and neutralizing CD59 on cell surface with specific

mAb.

Although it was possible to use complement and antibodies to destroy

individual breast cancer cells in suspension, many cancers exist as multicellular

11

tumors that are more resistant to complement-mediated killing than single

cells1. To analyze factors that are associated with killing of cells growing in

three-dimensional aggregates multicellular microtumor spheroids (MTS)

established from breast carcinoma (T47D) and ovarian teratocarcinoma (PA-

1) cell lines were used as models to study complement-mediated destruction

of small solid tumors (IV). This study showed differences in penetration o f

complement components and monoclonal antibodies into the tumor tissue.

Furthermore, complement attack against MTS and the neutralization of CD59

on the cell membranes resulted in a reduction of the microtumor volume. The

microtumor spheroid study suggested that complement-mediated killing o f

cancer cells would be effective against individual cells or small clusters o f

malignant cells that may remain after surgical removal of the main tumor. In

the last study (V), rat colorectal cancer cells (CC531) were injected

subcapsularly into the liver of Wag/Rij rats as a model for metastases o f

colorectal carcinoma. A panel of specific mAbs to CC531 cells were tested for

their complement-activating and tumor-homing capacities in vivo. In this study

it was possible to kill the CC531 cells in vitro using mAb and rat serum and t o

activate complement on tumor sections in situ. However, no C3 deposition

was detected on the tumors in vivo indicating a role for complement

regulators.

In conclusion, these studies show that it is possible to destroy single malignant

cells or partially even microtumors with serum complement if the complement

regulator CD59 is inactivated with a specific mAb on the cancer cell

membranes. In addition to cell membranes the glycophospho inositol-anchored

form of CD59 was detected on MFG in vivo. Complement-mediated killing o f

MTS showed that antibodies and C1q are able to penetrate through the

microtumor spheroids but the penetration of C3 is restricted by its strong

activation and covalent binding to cell surfaces. The syngeneic rat model

suggested that complement-activating mAb penetrate into the tumor tissue

but the lack of penetration of C3 into the tumor is a problem also in vivo.

12

1. INTRODUCTION

A quarter of the western world population faces the fact that they get a

malignant tumor during their life time. Although cancer therapies have

improved a lot during the last decades cancer is still fatal for most people.

Immunotherapy has only been little used for the treatment of cancer.

However, it offers one potential approach. Immunotherapy for cancer was

first used as early as 1895 by Hericourt and Richet who attempted to t reat

cancer patients with antitumor antisera prepared in dogs and donkeys.

However this and many other attempts failed to cure the patients. The

invention of the hybridoma technique by Köhler and Milstein in 19752 and the

discovery of new cancer antigens have given new hope for the specific

destruction of tumor cells by monoclonal antibodies (mAb). Much of the

research to improve the cytotoxicity of mAb has focused on conjugating them

with toxins or radionuclides. However, an ideal immunotherapeutic tumor-killing

system would harness the patient´s own effector mechanisms such as

antibody-dependent cellular cytotoxicity (ADCC) or the complement system t o

destroy the malignant cells. Complement belongs to the innate immune system

that, as opposed to the acquired immune system constitutes the nonadaptive

part of the human immune system. The immune system recognizes and

removes foreign material (viruses, bacteria and other micro-organisms) and is

responsible for the clearance of tissue debris resulting from ageing cells o r

trauma. Discrimination of foreign structures from the normal host tissue

components is the key element in both systems. This leads to activation o f

specific mechanisms to eliminate microbes or non-viable cells. The key

components of adaptive immune system are lymphoid cells (B and T cells).

They recognize their targets by multiple specific B cell and T cell receptors.

The acquired immune response is slow (starting from 3 - 5 days) because o f

the need for clones of responding B and T cells to develop. However, the

acquired immunity develops a memory from previous infections. The innate

immunity includes serum complement, natural killer cells and phagocytic cells.

The innate immunity recognizes molecular structures in microbes by receptors

with broad specificity. The structures can be polysaccharides and

13

polynucleotides that are common in microbes but often not found in the host.

An example is bacterial lipopolysacharide (LPS) that directly activates

complement and, when in complex with LPS-binding protein, is recognized by

the macrophage receptor CD14. Because of the broad specificity of the

receptors and no need for clonal expansion the response of the innate

immunity is immediate. Innate and acquired immunities are not completely

separate systems. Innate immunity can trigger adaptive immunity and vice

versa. Phagocytes (macrophages and dendritic cells) present microbial

antigens to T cells to initiate both cell-mediated and antibody-mediated

adaptive immune responses. Similarly, the antibodies produced by adaptive

immunity can activate the classical pathway of complement against a specific

antigenic structure.

While microbes are often promptly destroyed by the immune system,

developing tumor cells are usually not. Cancers result from the outgrowth of a

single malignant host cell. Immune system has to discriminate the malignant

cells from the normal host cells in order to destroy them. However, except fo r

virus-induced tumors there are often no or only few antigenic differences

between normal and malignant cells. The rejection of cancer cells by immune

system is based on tumor-associated antigens (TAA) that are not expressed

on normal cells or their expression is lower. The success of using complement

against malignant cells requires a specific activation of complement against

these cells with mAbs or other molecules that recognize TAA and

understanding of the mechanisms of complement regulation on cancer cells.

14

2. REVIEW OF THE LITERATURE

2.1 The complement system

2.1.1 Overview

Complement is an essential part of the innate immune system. It co-operates

with the adaptive immune system by mediating inflammatory consequences o f

antigen-antibody interactions and contributing to the enhancement of the

humoral response mounted against specific antigens3, 4. The principal function

of complement is to defend the host against microbial invasion of the body.

Furthermore, complement has an important role in the disposal of dead and

apoptotic cells and immune complexes5-7. Complement can discriminate

between self and non-self structures but not as specifically as the acquired

immunity.

The complement system consists of a complex group of proteins that are

present in blood plasma and on cell membranes. The proteins act as

precursor enzymes, effector molecules (Table 1), control proteins o r

receptors (see chapters 2.1.6 and 2.1.7). Most of the complement proteins

circulate in the bloodstream and body fluids as inert precursors. The contact

of the first component with an activator i.e. an immunoglobulin, activating

surface or certain carbohydrate structures, leads to a subsequent activation

of the second one in a precise order depending on the pathway – classical,

alternative or lectin - that is activated. During the activation some of the

proteins acquire enzymatic properties while the others function as effector

molecules and control proteins. C3 is a key-component of the complement

system, since it can be activated through all three pathways3.

The activation of complement results in three main types of biological

responses. The C3b and iC3b fragments8 generated on the target surface

can interact with cell surface receptors of phagocytes to induce complement-

15

dependent cellular cytotoxicity (CDCC)9, 10. The small cleavage fragments

C3a, C4a and C5a are released into the fluid phase. These small bioactive

peptides act as chemotaxins, leukocyte activators and as anaphylatoxins11,

12. Finally, activation of the terminal complement pathway results in the

formation of the membrane attack complex of complement (MAC) on the

target cell membrane13. Because complement is a cytolytic system and since

the pathways include enzymatic events that allow considerable amplification

during activation, the complement cascade must be carefully regulated t o

prevent damage to the host cells. The stringent control mechanisms include

multiple regulatory proteins in blood plasma and on cell membranes. Because

of this, normal host cells can usually resist the cytolytic activity of homologous

or autologous complement14.

16

Table 1. Proteins involved in complement activation

Protein Molecular Serum Functionweight conc.( k D a ) ( µ g / m l )

Classical pathwayC1q 410 70 Binds IgG and IgM; initiates the

classical pathway.C1r 85 34 Cleaves / activates C1sC1s 85 31 Cleaves / activates C4 and C2C2 95 25 Cleaves / activates C3 and C5C4 206 600 Binds C2 during activation

Lectin pathwayMBL 96 x 2-6 5 Binds to carbohydrates:

initiates lectin pathwayMASP-1 83 - Activates MASP-2MASP-2 - - Cleaves C2 and C4

Alternative pathwayFactor B 100 225 Cleaves / activates C3 and C5Factor D 25 1 Cleaves / activates factor BProperdin 153 25 Stabilizes C3bBb convertase

Common for the pathways aboveC3 195 1200 Subunit in alternative pathway

C3/C5 convertase. Binds C5 in convertases. Opsonisation

Lytic pathwayC5 180 85 C5b: initiates membrane attack;

C5a is the major chemotactic/ anaphylatoxic peptide

C6 128 60 C6, C7 and C8 associate withC5b to form a membrane site to which C9 can bind

C7 120 60C8 150 55C9 79 60 Multiple C9 proteins polymerize

to generate transmembrane pore

2.1.2 The alternative pathway

The alternative pathway is a phylogenetically older system than the classical

pathway3. The alternative pathway of complement is triggered by activation

17

of the C3 protein. C3 has an internal thiolester bond that undergoes cleavage

at a slow rate during a hydrolysis reaction with water. The product of this

reaction is C3(H2O), which can form an initial C3 convertase by binding factor

B which becomes cleaved to Bb by factor D15. The C3(H2O)Bb convertase will

cleave C3 to C3a and C3b. The cleavage product C3b can bind to the

microbial or host cell surface by forming a covalent linkage with -OH or -NH2

groups through the exposed thiolgroup. Recently, it has been shown that

phosphatidyl serine on apoptotic cells can activate the alternative pathway7.

C3b binds the complement component factor B to form C3bB, Mg2+ is

required as a catalyst for the reaction. The newly formed complex is a

substrate for the plasma enzyme factor D that cleaves C3bB to generate

C3bBb, the principal C3 convertase, which can cleave C3 to produce more C3b

and C3a. The unhampered operation of the C3 convertase leads to the

deposition of large numbers of C3b molecules on the microbial surface16.

Finally, a C5 convertase is generated when an additional C3b molecule is

recruited to the C3bBb complex. Cleavage of C5 by the C5 convertase

releases C5a and C5b and initiates the lytic pathway.

2.1.3 The classical pathway

The classical pathway of complement is initiated by the interaction of the C1q

subcomponent of C1 with at least two Fc regions of antigen-bound

immunoglobulins (IgG or IgM). Additional activators the classical pathway are

the C-reactive protein17, the serum amyloid P component18 and membrane

blebs on apoptotic cells19. Conformational change in C1q upon binding leads

to the activation of the C1r protease. C1r will proteolytically cleave C1s into

an enzymatically active form. C1s splits C4 exposing a nascent thiolester bond

in the cleavage product C4b. This enables C4b to attach covalently to the cell

surface. C4b has a binding site for C2 and C1s acts on the formed C4b2

complex to create C4b2a, the classical pathway C3 convertase. From this on

the cascade continues similarly to the alternative pathway with one molecule

18

of C3b bound to C4b2a to generate the C5-cleaving enzyme for the initiation

of the lytic pathway.

2.1.4 The lectin pathway

It has been shown that the classical pathway can also be triggered without

antibodies by mannose binding lectin (MBL), a C1q-like molecule that binds t o

mannose and N-acetylglucosamine structures that are present on the

surfaces of microbes20. MBL forms a C1-like complex with serine proteases

MASP-1 and MASP-2. Conformational change in MBL upon binding leads to the

activation of MASPs which cleave C2 and C421, 22. The cascade continues like

the classical pathway.

2.1.5 The lytic pathway

The formation of the C5-convertase through the either alternative or the

classical pathway initiates the membrane attack that results in the generation

of a large aggregate of proteins, the so-called membrane attack complex

(MAC) on the activating surface13. The lytic pathway consists of five proteins

(C5b, C6, C7, C8 and C9) that are present in plasma. Sequential addition o f

C6, C7 and C8 to C5b generates C5b-8, which catalyzes the polymerization o f

C9 to a pore-like structure on the target membrane. The diameter of the

membrane channel varies between 10 Å and 150 Å23. Deposition of C5b-9

complexes on the membranes of nucleated cells results e.g. in the leakage o f

adenine nucleotides ATP, ADP and AMP. Furthermore, the exposure results in

an increase of intracellular Ca2+ and the loss of mitochondrial membrane

potential24. Normally, killing of nucleated cells requires several C5b-9 lesions

per cell25, although C5b-8-mediated cell lysis has also been reported26.

19

Figure 1. A schematic model illustrating the activation pathways o f

complement. The classical pathway is activated by immune complexes

containing IgG or IgM and the alternative pathway is triggered directly by the

microbial surface. The lectin pathway is activated by a surface containing

mannose or N-acetyl glucosamine residues. The pathways lead to the

generation of the membrane attack complex (C5b-9 or MAC) on the cell

surface which damages the cell membrane and kills the microbe. Dashed lines

indicate enzymatic activity. For abbreviations see Table 1.

C1rC1sC4C2

ALTERNATIVE PATHWAY

C3(H2O)BbC1q

Act ivat ing surfaces

CLASSICAL PATHWAYCarbohydrat es

LECTIN PATHWAYIgG, IgM

C3a

MBL

C3bBb

C5b678 (9) n MAC

MASP1MASP2

C4a

C5a

C4b2a C3 C4b2a

C4C2

C5

C4a

C3bBDP

20

2.1.6 Soluble regulators of complement

To prevent excessive or inappropriate consumption of complement

components the activation needs to be carefully controlled. At least six

regulatory proteins are present soluble in blood plasma (Table 2). The plasma

protein C1 inhibitor (C1 INH) binds to the activated C1r2C1s2 enzyme

complex and prevents the cleavage of C4 and C2 by blocking the serine

esterase activities of C1r and especially of C1s27. The C3 convertase C4b2a

is regulated by the serum C4b-binding protein (C4bp), which competes with

C2a for binding to C4b and displaces C2a from the complex28. C4b in complex

with C4bp is susceptible to cleavage by the plasma protein factor I, another

serine esterase enzyme of complement. Recently, it has been suggested that

ovarian cancer cells bind C4bp, which protects the cells from complement

lysis29. Mechanisms analogous to those described above control the

alternative pathway C3 convertase. The plasma protein factor H competes

with factor B and readily displaces Bb from the C3bBb convertase. Factor H

also forms a complex with C3b catalyzing its proteolytic destruction by

factor I30. Two plasma proteins, clusterin (SP40,40)31 and vitronectin (S-

protein)32 bind to the forming C5b-7, C5b-8 and C5b-9 complexes to prevent

their insertion into cell membranes.

21

Table 2. Soluble regulators of complement

Protein Molecular weight Function( k D a )

C1 inhibitor 105 Inhibitor of C1r and C1s

Factor H 150 Inhibition of C3bBb formation. Decay dissociation of C3bBb. Cofactor in C3b cleavage

C4b binding protein 540 Cofactor in C4b cleavage

Factor I 88 Cleavage of C3b, iC3b andC4b

Vitronectin 80 Keeps TCC* in solution(S-protein)

Clusterin 70 Keeps TCC* in solution(SP40,40Apo-J)

*TCC, terminal complement complex

2.1.7 Membrane-bound complement regulatory proteins (mCRP)

Activated complement may also be toxic to the host cells. However, normal

human cells can resist the cytolytic activity of complement by expressing

several regulatory proteins. Four proteins are expressed on cell membranes

(Table 3). The membrane-bound regulators are species-selective protecting

the cells from homologous or autologous complement33, 34. The proteins are

clustered into groups by their reactivity with monoclonal antibodies (CD:

cluster of differentiation).

22

Table 3. Membrane-bound regulators of complement

Protein Molecular Function

Weight (kDa)

CD35, CR1 190, 220 Control of C3bBb/C4b2a

Cofactor in C3b and C4b inactivation

CD46, MCP 58-68 Cofactor in C3b and C4b

inactivation

CD55, DAF 70 Decay of C3/C5 convertases

CD59, protectin 18-25 Inhibition of MAC

The complement receptor type 1 (CR1, C3b receptor, CD35)35 is a

transmembrane protein. It dissociates the key enzymes of the complement

cascade, the C3 and C5 convertases, and promotes proteolytic inactivation o f

C3b and C4b by the plasma serine protease factor I. Factor I cleaves the

alpha chains of C4b and C3b to generate iC4b and iC3b which are incapable o f

forming classical (C4b2a) or alternative (C3bBb) pathway C3 convertases.

Unlike the other cofactors, CR1 acts as a cofactor also in the cleavage o f

iC3b to C3c and C3dg. CR1 is expressed on erythrocytes, neutrophils,

monocytes, B lymphocytes, eosinophils and some T cells36. On the surface o f

phagocytes CR1 functions also as a receptor for C3b, iC3b, C4b and C1q thus

having a role in the clearance of complement-activating immune complexes37.

Membrane cofactor protein (MCP, CD46). MCP binds to accidentally o r

spontaneously deposited C3b and C4b molecules on cell surfaces and

promotes their inactivation by serving as a cofactor for factor I38. MCP is

present on almost all cells except erythrocytes.

Decay-accelerating factor (DAF, CD55) is a glycoprotein that is

attached to cell membranes with a glycosyl-phosphatidyl inositol-(GPI-)

anchor. DAF dissociates the C3 convertases releasing C2a and Bb from the

complexes39. DAF is expressed on most cells including erythrocytes37.

23

Protectin (CD59) was first isolated from human erythrocyte membranes40,

41. CD59 (Fig. 2) has been also referred to as MACIF42, HRF-2043 and MIRL

44. To simplify the nomenclature the functional name protectin for CD59 has

been proposed45. The first direct experimental demonstration of a MAC

inhibitor on human erythrocytes was published in 1988 by Sugita et al.41. The

primary function of CD59, inhibition of the membrane attack complex o f

complement, was shortly established in many laboratories40, 43, 44. CD59

blocks formation of MAC by preventing the C5b-8 catalyzed insertion of C9

into lipid bilayers (Fig. 3)45, 46. In addition to erythrocytes, CD59 is widely

distributed on all other human blood cells40 as well as on endothelial and

epithelial cells of several organs47. CD59 has also been detected on cultured

endothelial cells48, 49 glomerular epithelial cells50 and spermatozoa51. CD59 is

anchored to cell membranes via its GPI-moiety. Soluble, hydrophilic forms o f

CD59, that lack the anchor phospholipid52 have been detected in various body

fluids such as urine, tears and saliva40, 53 and have also been produced in

recombinant form54. In addition to various cell membranes phospholipid-tailed

CD59 has been found in amniotic fluid55 and in seminal plasma, where it has

been shown to be associated with extracellular organelles called

prostasomes56.

24

Figure 2. A schematic model of the polypeptide backbone, oligosaccharide

side chain and GPI-anchor side chains of CD59. The GPI-anchor contains two o r

three acyl chains that anchor the protein to the cell surface.

CC C

C

N

C

C

CC

C

N

C

21

10

32

43

5566

1

77

RR(R)

PIPLC

PIPLD

N-acetyl glucosamine

Fucose

Mannose

Galactose

N-acetyl neuraminic acid

EthanolaminePhosphorusN-acetyl galactosamine

N-glucose

Inositol

R Acyl chainPIPLC Phosphatidylinositol specific phospholipase CPIPLD Phosphatidylinositol specific phospholipase D

Amino acid

25

CD59

C6 C7

C5b C8Cell surface

C9

MAC

A

B

Figure 3. Formation of the membrane attack complex (MAC; C5b-9) o f

complement on the cell membrane (A) and its inhibition by CD59 (B). On the

membranes of foreign targets C5b-8 catalyzes the polymerization of multiple

C9 molecules to form a pore-like lesion (MAC) into the cell membrane. On the

host cells CD59 inhibits the insertion of C9 by binding to C8 (B).

26

2.1.8 Rat complement system

The complement activation pathways among vertebrates are relatively well

conserved. The complement regulatory system in rat resembles that o f

humans: the rat analogues of human CD46, CD55 and CD59 have been

characterized57-59. In addition, a distinct C3 regulator, Crry/p65, has been

identified in rodents. Crry/p65 has both decay-accelerating and cofactor

activity for the C3/C5 convertases. Thus, Crry/p65 restricts activation o f

both the alternative and the classical complement pathway60-62. The mCRPs

are species-selective and protect cells primarily only against homologous

complement. The species selectivity of mCRP requires a syngeneic animal

model to study the role of mCRP in vivo. Xenografts usually induce a massive

activation of complement in the recipient blood against the vulnerable graft63.

The hyperacute rejection is caused by complement-activating xenoreactive

antibodies to endothelial cells64. Human xenoreactive antibodies against pig

xenografts, for example, consist mainly of anti-Galα1-3Gal antibodies, which

occur in IgM, IgG and IgA classes64.

27

2.2 The immune system and transformed cells

The human body continuously confronts foreign material like potentially

pathogenic bacteria and viruses from the enviroment. However, cell-mediated

and humoral immune responses can in most cases discriminate the pathogens

from the host cells and destroy them. Cancers result from the outgrowth o f

genetically transformed cells. It has been difficult to show that the immune

system responds strongly to tumor cells. Mice that lack lymphocytes o r

humans that are deficient in T cells do not differ much in tumor incidence

compared to normal hosts65. Virus-associated tumors are an exception and

can be usually destroyed by the normal immune system65. However, in animal

studies some tumors have elicited immune responses that prevent their

growth. In these studies the tumors have been induced by carcinogenic

chemicals or radiation in inbred strains of animals. The experimental tumors

can be isolated, grown in vitro and injected into recipients to induce cancer.

Some of the induced cancers start to grow and lead to the death of the host

while others regress. It has been possible to immunize animals with irradiated

tumor cells and suppress the growth of the cells injected thereafter indicating

that some immune surveillance against cancer exists65. Three major types o f

cells are important in the immune response to cancer. These cells are the

antigen-presenting cells, T-lymphocytes and natural killer cells.

2.2.1 Antigen-presenting cells (APC)

Antigen-presenting cells (APC) include dendritic cells, monocytes (Mo) and

macrophages (Mø). The function of AP cells is to engulf pathogens or dying

cells and to present their antigens for T cells. The antigens from the cellular

debris or pathogens must first be processed into peptide fragments. These

epitopes are then presented on either class I or class II major

histocompatibility complex (MHC) proteins for T lymphocytes to recognize.

Dendritic cells are the strongest stimulators of the immune system66. They

have receptors for complement C3d, which improve the uptake of C3d-antigen

complexes to the cells for processing4. Dendritic cells have been used in

28

cancer therapy trials by treating isolated dendritic cells from the patient with

material extracted from cancer cells67, 68. The hope is that after releasing

the modified dendritic cells back into the blood stream of the patient they

start an immune response by presenting the cancer antigens to the T cells.

2.2.2 T lymphocytes

T helper lymphocytes (Th; CD4+) and cytotoxic T-lymphocytes (Tc; CD8+) are

responsible for the generation of specific immune responses. Th cells release

cytokines that signal other immune cells to become activated. The cytokines

are released only if the Th lymphocyte recognizes an APC presenting with a

foreign peptide in its MHC as well as co-stimulatory molecules. These cytokines

can stimulate antibody production by B cells and a Tc response. Tc cells are

particularly important for eliminating those cells in our body that have been

infected with various viruses. Tc cells are critical mediators in tumor

immunology68. They also require the target antigen presented on MHC protein

plus co-stimulatory signals from Th cells to become activated. The target

antigens on the tumor cells have been identified to be peptides from tumor cell

proteins. Although Tc cells can recognize tumor-associated antigen fragments

presented on MHC proteins, they usually do not become activated because

these TAAs are recognized as self proteins. Furthemore, cancer cells rarely

present the required co-stimulatory signals to activate Tc cells. The genetic

instability of tumor cells causes further problems. The malignant cells may lose

their tumor antigens by mutation, which lead to the appearance of escape

mutants that avoid the rejection. The tumor cells may also lose their MHC

molecules and thus become invisible for the cytotoxic T cells69. To further

suppress the activation of the immune system some tumor cells may even

start to express immunosupressive cytokines70.

29

2.2.3 Natural killer cells (NK cells)

Another class of potential immune surveillance cells are NK cells that were first

discovered by their ability to identify and kill cancer cells. Unlike T and B

lymphocytes NK cells do not have specific receptors for a particular antigenic

target. NK cells recognize malignant or infected cells that have been coated

with antibodies by binding to the Fc region of the antibody with an Fc

receptor (FcγRIII or CD16) on their surface. CD16 recognizes IgG1 and IgG3

subclass antibodies. NK cells require certain cytokines to become activated

and for proliferation71. The destruction of the target cell is mediated by the

release of cytotoxic granules containing perforin and granzymes. The target

cell dies by apoptosis and/or membrane damage. Furthemore, NK-cells can

discriminate normal cells from cells that do not express adequate amounts o f

MHC-I molecules72. NK cells express a variety of inhibitory receptors that

recognize self-MHC class I molecules. These deliver an inhibitory signal to the

NK cell and prevent an attack against normal cells. They have also stimulatory

receptors that bind to a ligand on a target cell72. The delicate balance

between opposite signals delivered by inhibitory receptors specific for MHC-I

molecules and the stimulatory natural cytotoxicity receptors (NCR; NKp46,

NKp30 and NKp44) regulate the effector functions of NK cells72. Only recently,

ligands for another stimulatory receptor NKG2D have been identified.

Interestingly, NKG2D-ligands (e.g. MIC and Rae1 proteins) are not expressed

by most normal cells but are strongly upregulated on transformed or infected

cells73. Diefenbach et al. have demonstrated that expression of the NKG2D

ligand Rae1 in several murine tumor cell lines resulted in a dramatic rejection

of tumor cells mediated by NK cells and/or CD8+ T cells74.

30

2.2.4 Tumor antigens

The discrimination of cancer cells from normal ones is based on transformed

antigens or structures on the cell surface. Although cancer cells express only

few tumor-specific antigens that are unique to the tumor cells many cancers

express tumor-associated antigens (TAA) that can serve as potential targets

for immune response. TAA can be grouped into oncofetal Ag, tissue-specific

differentiation Ag and tumor-associated carbohydrate and glycolipid Ag. I t

has been shown that melanoma patients have T cells reactive with so called

MAGE antigens75, 76. The antigens of the MAGE family have a limited

distribution in normal adult tissues. An exception is the testis that is an

immunologically privileged site. The most common melanoma antigens gp75,

gp100 or MART1 are overexpressed melanocyte differentiation antigens75, 77.

CA125 and epithelial cell adhesion molecule (Ep-Cam) are expressed by

ovarian and colorectal cancers, respectively. The 791Tgp72 antigen has been

characterized to be overexpressed in tumors including colorectal, gastric and

ovarian carcinomas and osteosarcomas. An overexpression of the 791Tgp72

antigen indicates a poor prognosis in colorectal cancer patients78, 79. In

subsequent studies, the 791Tgp72 antigen showed 100% sequence identity

with the complement regulator CD5580. Another tumor marker that was first

isolated from the urines of bladder cancer patients and is currently used in

diagnosticks (BTA-TRAK test) was later found to be complement factor H81.

2.2.5 Antibody responses against tumor-associated antigens in

cancer patients in vivo

Abnormally high density, mutagenic expression or changes in glycosylation can

make TAA immunogenic and lead to antibody production. Serological immune

responses to TAA has been detected in cancer patients. Often the antibodies

are directed against intracellular antigens (c-myb, c-myc, p21ras and p53)

that are released by necrotic tumor cells82. Breast cancer patients (43%)

31

and 32% of colon cancer patients have been found to be positive for anti-c-

myb and p21ras antibodies, respectively83, 84. However, the detection of anti-

p53 antibodies is usually associated with poor prognosis probably because

these intracellular antigens are not recognized by the immune system in live

cancer cells83. Instead, antibodies against plasma membrane antigens may

have an impact on tumor growth since they can react with live tumor cells.

The presence of some of these antibody/antigen complexes (PEM/MUC1) in

patients has been associated with a better prognosis85. Antibody responses

detected in cancer patients against cell surface antigens are listed in table 4.

Table 4. Antibody response against cell surface antigens in cancer patients82

Antigen Origin of cancer

HER2/neu Breast

PEM/MUC1 Ovary, breast, colorectal, pancreas

T, Tn, sialyl Tn Breast, lung, pancreas

Gangliosides (GM1,

GM2, GD2) Melanoma

2.2.6 Antibody-induced effector mechanisms

Antibodies are multifunctional proteins that have several effector functions

after they bind to their specific antigens. The IgG1 is the most widely used

antibody isotype in the therapy for cancer, because it activates human

complement, recruits NK cells for ADCC, and has an extended half life in

plasma86. However, some studies suggest that IgA also recruits cytotoxic

cells in humans86.

Apoptosis is an important part of the cellular turnover. Cells that have to be

eliminated without inflammation enter apoptosis. Antibodies can kill nucleated

cells by inducing their apoptosis. Cross-linking of the Fas receptor (CD95) on

the target cell membrane by its ligand, FasL, or with anti-Fas antibodies,

induces apoptosis of the cells by activating of caspases87. Induction of Fas-

32

mediated apoptosis by anti-Fas antibodies has been demonstrated in solid

tumors implanted in mice. Unfortunately, treatment with anti-Fas antibodies o r

FasL causes severe damage to the liver87. Complement activation is likely t o

be required for efficient uptake of apoptotic cells within the systemic

circulation. Exposure of phosphatidylserine on the apoptotic cell surface t o

serum has been shown responsible for complement activation and result in

coating the apoptotic cell surface with iC3b7. The binding of macrophage

receptors CR3 (CD11b/CD18) and CR4 (CD11c/CD18) with iC3b results in the

uptake of apoptotic cells7. Furthermore, C1q have been detected to bind

directly to surface blebs of apoptotic human keratinocytes, vascular

endothelial cells and peripheral blood mononuclear cells19.

Antibody-dependent cellular cytotoxicity (ADCC) occurs when an

effector cell binds to a target cell coated with antibodies and kills it. Cells

capable of ADCC include NK-cells, macrophages, neutrophils, eosinophils and

mast cells. ADCC has been studied most extensively in NK-cells. NK cells

express on their cell membranes CD16 molecules (FcγRIII) that bind IgG1 and

IgG3 on the target surface and thereafter release toxic substances into the

intervening space88.

Complement-dependent cytotoxicity (CDC) can lyse cells independently

of ADCC. In CDC the classical pathway is activated by IgM or IgG bound to the

tumor cell surface: subsequently, the formation of MAC causes the lysis o f

tumor cell targets. CDC is thought to be an important action mechanism o f

the anti-CD20 monoclonal antibody that is used therapeutically in the

treatment of lymphoma patients. Malignant cells from B cell lymphoma

patients express CD46, CD55 and CD59 molecules at various levels resulting in

differences in the sensitivity of the cells to complement-mediated lysis. Lysis o f

cells from patients responding poorly to mAb therapy was increased 5- to 6-

fold after blocking both CD55 and CD5989.

33

Complement-dependent cellular cytotoxicity (CDCC) requires prior

activation of complement and deposition of C1q, C3b, iC3b or C4b on the

target cell. These ligands interact with the C1qR, CR1 or CR3 (CD11b/CD18)

receptors on NK-cells, polymorphonuclear leukocytes or macrophages t o

induce their cytotoxic activity90.

Table 5. Antibody effector mechanisms in cell killing90

ADCC CDC CDCC

Init iator IgG IgG, IgM C3b, C4b, iC3b, C1q

Transducer FcγRI, II, III C1q CR1, CR3, C1qR

Effector Mo, Mø, PMN, NK MAC Mo, Mø, PMN, NK

Abbreviations: ADCC, antibody-dependent cellular cytotoxicity; CDC, complement-dependent cytotoxicity; CDCC, Complement-dependent cellular cytotoxicity; C1qR, C1qreceptor; CR1/CR3, complement receptor type 1 or 3; FcγR, Fc receptor for IgG;iC3b, inactivated C3b; MAC, membrane attack complex of complement; Mo, monocyte;Mø, macrophage; NK, natural killer cell; PMN, polymorphonuclear leukocyte.

2.2.7 Tumor antigens and immunotherapy of cancer

Ideally, tumor associated antigens (TAA) could be used as targets fo r

monoclonal antibodies against malignant cells. B cell lymphomas have been

treated successfully with anti-idiotypic monoclonal antibodies that have

become bound to the specific IgG-idiotype on the malignant cell surface. A

humanized mAb against CD20 (Rituximab®) is already in wide use for the

treatment of non-Hodgkin B cell lymphoma. Rituximab was the first

monoclonal antibody officially registered for the treatment of cancer. The

mAb mediates complement-dependent cell lysis, antibody-dependent cellular

cytotoxicity and induces apoptosis91, 92. Trastuzumab, another humanized

monoclonal antibody, is directed against the HER-2/neu receptor.

Trastuzumab has been shown to be most efficient in the treatment of HER-2-

overexpressing metastatic breast cancer. Combination of trastuzumab with

chemotherapy has produced higher response rates and longer survival than

treatment with chemotherapy alone93. This mAb, however, does not act by

activating complement but by blocking growth promoting activities of the

34

HER-2/neu receptor. Some tumor-associated antigens and their monoclonal

antibodies that are in clinical trials are listed in table 6.

Table 6. Non-conjugated monoclonal antibodies against tumor associated

antigens reviewed in94

Cancer Antigen IgG Type Product

Ovarian carcinoma CA125 mouse IgG OvaRex®

Breast carcinoma HER-2/neu humanized IgG1 Herceptin®

Colorectal carcinoma 17-1A mouse IgG2a Panorex®

Chronic lymphatic CD52 humanized IgG Campath-1H®

Leukemia

Non-Hodgkin´s CD20 humanized IgG Mabthera®

lymphoma (rituximab,IDEC-

C2B8)

The major problem in active immunotherapy of cancer is the poor

immunogenicity of cancer cells. Whole tumor cells have been used as vaccines

either mixed with adjuvants or after transfecting them with nonself proteins

or with immunomodulatory factors. However, isolated antigens that are

selectively or abundantly expressed in cancer cells appear to function better

as vaccines than whole cells95. In experimental animals promising results have

been obtained by increasing the immunogenicity of cancer cells with nonself

peptides. Murine tumor lines with major histocompatibility complex (MHC)

class I-positive melanoma and colon carcinoma cells were injected into mice

together with MHC class I-matched peptide ligands of influenza virus. Mice

bearing live melanoma cells and colon carcinoma were efficiently cured by this

treatment96. Th cell responses have been elicited in patients with metastatic

melanoma by subcutaneous injection of antigen-loaded (MAGE-3 tumor

peptides) monocyte-derived dendritic cells97. In melanoma patients the

immunization with gangliosides produced an IgM response but no correlation

with antibody titers or clinical outcome was observed82. However, in

melanoma patients immunized with GM2 and an adjuvant after surgery the

seroconversion correlated with increased survival98. Active immunotherapy

35

using cancer antigens (T and sialyl Tn) to induce antibody responses in

ovarian and breast cancer patients have produced IgM and IgG antibodies

with potential for CDC82. Most of the breast cancer patients have responded

with IgM production and CDC to the the carbohydrate antigen globo H-keyhole

limpet hemocyanin conjugate vaccine admistered together with QS-21

adjuvant99. A human anti-idiotypic antibody that mimics CD55 has been used

successfully in immunization of over 200 colorectal cancer and osteosarcoma

patients. 70% of patients showed CD55-specific immune responses with no

associated toxicity100. Although, natural antibody responses against cell

surface antigens have been detected in cancer patients (Table 4) only few

studies have indicated that complement is inherently activated in tumors in

vivo101. Sporadical deposits of C3 and C5b-9 have been detected in cervical

and breast cancer samples102, 103. This probably indicates a suppressing role

for the complement regulators.

Indeed, problems with the mAbs have been inefficient killing of the tumors as

well as inefficient penetration of the antibodies into solid tumors. To stimulate

tumor killing ability by mAbs toxic molecules like ricin104 and Pseudomonas

toxin105 have been coupled to antibodies. In other approaches mAbs have

been conjugated to chemotherapeutic substances such as adriamycin106 o r

radioisotopes107 in order to concentrate them to the tumor site108. Some

strategies have been designed to target and activate complement against

tumor cells. These include heteroconjugates composed of monoclonal

antibodies and C3b or cobra venom factor (CVF) that activates the

alternative pathway by replacing C3b in the C3 convertase109, 110. The

formed CVFBb complex is insensitive to complement inhibitors. In one study

interferon-treated tumor cells fixed C3b to their surfaces through the

alternative pathway111.

Successful complement activation on tumor cells may have multiple

consequences on the immune responses against the tumors. The cleavage

fragments of complement proteins C3a, C4a and C5a can act as

36

chemotaxins, leukocyte activators and anaphylatoxins to induce inflammation

in the tumor tissue. The larger fragments C3b, iC3b and C3d generated on the

target surface can interact with cell surface receptors of lymphocytes and

phagocytes to induce CDCC. There is evidence that tumor-cell bound C3

enhances the sensitivity of tumor cells to killing by activated macrophages112.

C3 deposition may also increase the antigenicity of the potential tumor

antigens on the tumor cells. Dempsey et al. have shown that fixing C3d into

hen egg lysozyme lowered the threshold level of B cells' response to lysozyme,

thereby increasing its immunogenicity4. Thus, C3d can act as a molecular

adjuvant and may be able to increase the immunogenicity of tumor antigens.

37

3. AIMS OF THE STUDY

The working hypothesis of this thesis was that complement could be used as

an effector mechanism to destroy malignant cells after membrane-bound

complement regulators have been inactivated on their cell surfaces.

Specifically, this study aimed at the following:

1. to analyze the significance of the membrane-bound complement regulator

CD59 for tumor cell survival from complement attack,

2. to determine the sequestration of CD59 into the various physicochemical

compartments of human breast milk,

3. to examine the ability of antibodies and complement components to

penetrate into microtumor spheroids and

4. to set up an animal model to test mAb-induced complement activation in

vivo.

38

4. MATERIALS AND METHODS

Methods and material used in the following studies I-V are described in detail

in the original publications and are listed in Tables 7-10.

Table 7. Antibodies used in studies I-V

Antibody Description Used in512 mo mAb against rat Crry/p65 V6D1 mo mAb against rat CD59 VAnti-C1q rb pAb against hu C1q IVAnti-C3 rb pAb against hu C3 IVAnti-C3c rb pAb against hu C3c IIIAnti-C3-FITC go pAb against rat C3 VAnti-C3bi mo mAb against hu iC3b IVAnti-C5b-9 mo mAb against hu C5b-9 IVBRIC216 mo mAb against hu CD55 I, III, IVBRIC229 mo mAb against hu CD59 I, II, III, IVBRIC230 mo mAb against hu CD55 III, IVC1242 mo mAb against hu TAA: sialylated Tn IIIC241 mo mAb against hu TAA: CA19-9 IIIclone 528 mo mAb against hu HER2/c-erbB-2 Iclone 3G8 mo mAb against hu FcγRIII IVclone 10.1 mo mAb against hu FcγRI IVclone C1KM5 mo mAb against hu FcγRII IVGB24 mo mAb against hu CD46 IJ4.48 mo mAb against hu CD46 III, IVMa552 mo mAb against hu TAA: MUC-1 IIIMG1, MG2, MG3,MG4 mo mAb against CC531 cells VOv185 mo mAb against hu TAA: CA125 IIIOv197 mo mAb against hu TAA: CA125 IIIRDIII7 mo mAb against rat CD55 VR2 rb antiserum against hu CD59 IS2 rb antiserum against hu MCF-7 I, III, IVYTH53.1 rt mAb against hu CD59 I, II, III, IVAbbreviations: Ag, antigen; go, goat; hu, human; mo, mouse; mAb, monoclonalantibody; pAb, polyclonal antibody; rb, rabbit; rt, rat; TAA, tumor associatedAg

39

Table 8. Cell lines used in studies I-V

Cell lines Description Used in6D1 mo hybridoma cell line producing

mAb to rt CD59 VCaov-3 hu ovarian adenocarcinoma cell line IIICC531 transplantable colon adenocarcinoma

of the Wistar derived Wag/Rij strain VMCF-7 hu breast carcinoma cell line IPA-1 hu ovarian teratocarcinoma cell line III, IVSK-OV-3 hu ovarian adenocarcinoma cell line IIISW626 hu ovarian adenocarcinoma cell line IIIT47D hu breast carcinoma cell line I, IVYTH531.1 rt hybridoma cell line (anti-CD59) I, II, III, IVAbbreviations: hu, human; mo, mouse; rt, rat

Table 9. In vivo tumor samples used in studies I, III and V

Cancer Origin Used inColon adenocarcinoma rt liver VDuctal carcinoma hu breast IFibroadenoma hu breast ILobular carcinoma hu breast IMucinous cystadenoma hu ovary IIIMucinous cystadenocarcinoma hu ovary IIIPapillary cystadenocarcinoma hu ovary IIISerous cystadenoma hu ovary IIIAbbreviations: hu, human; rt, rat

40

Table 10. Laboratory methods used in studies I-V

Methods Used inBichinonic acid (BCA) protein concentrationdetermination assay I, IIBiotinylation of YTH53.1 IIIBinding of CD59 to the TCC I, IICC531 tumor induction into rats VCell damage visualization using propidium iodide IVCell culture I, III, IV, VChromium (51Cr) lysis assay I, III, IV, VComplement hemolysis assay I, IIELISA VF(ab´)2 fragment preparation of YTH53.1 mAb IFlow-cytometry analysis (FACS) III, VGel filtration IImmunoaffinity chromatography of CD59 I, IIImmunofluorescense microscopy I, II, III, IV, VImmunoblotting I, II, III, VIncorporation of CD59 into cell membranes I, IIInduction of tumors in rats VIodine (125I) labeling I, IIMicrotumor spheroid generation IVMilk fat globule (MFG) and MFG membrane isolation IINorthern blotting IIIPI-PLC treatment II, IIIPlasma membrane isolation VRNA extraction IIIScanning electron microscopy IVSDS-PAGE I, II, III, VSucrose density gradient ultracentrifugation I, IIAbbreviations: ELISA, enzyme-linked immuno-adsorbent assay; PI-PLC,phosphatidylinositol-specific phospholipase C; SDS-PAGE, sodium dodecylsulfate polyacrylamide gel electrophoresis; TCC, terminal complment complex.

41

5. RESULTS AND DISCUSSION

The discovery of new tumor-associated antigens and developing mAb against

them is beginning to enable the use of complement as an effector mechanism

in cancer therapy. Complement lysis of cancer cells is an important

therapeutic mechanism already in use in the treatment of certain types o f

lymphomas89. However, mCRP on the surface of tumor cells reduce the

efficiency of CDC. Like on normal cells the GPI-anchored CD59 was found to be

expressed on malignant cells (I, III) and MFG (II). Differences in the expression

levels of CD59 and of the other mCRP were observed in tumor samples (I,

III)113.

In vivo, the malignant cells usually grow as multicellular tumors with

intercellular connections between tumor cells that may constitute a barrier

against complement attack. As shown in study IV they may prevent the

penetration of activated complement components into the tumor tissue.

The species selectivity of the activity of complement regulators requires an

homologous animal model for the studies of complement-mediated destruction

of cancer cells in vivo. To improve the efficiency of complement activity in

cancer therapy it is important to understand the complement regulatory

mechanisms of cancer cells. This thesis focused on studying the expression o f

mCRPs, and especially the significance of CD59 in preventing complement-

mediated lysis of breast and ovarian cancer cells. Characteristically fo r

ovarian cancers, the malignant cells often remain confined to the abdominal

cavity and are in direct contact with ascitic fluid (AF) both as peritoneal

implants and as free-floating tumor cells. The individual malignant cells or small

cell clusters complicate the complete surgical removal of the tumor cells. The

hope is that the remaining ovarian cancer cells could be removed by

intraperitoneal immunotherapy with mAbs.

42

5.1 mCRP on tumor cells and MFG (I, II, III, IV)

Immunofluorescence (IF) studies with mAbs against CD46, CD55 and CD59

showed that the complement inhibitors CD46 and CD59 were expressed on

plasma membranes of the breast cancer (MCF7, T47D) and the ovarian

cancer cell lines (Caov-3, PA-1, SK-OV-3 and SW626) studied (I, III). The

expression of CD59 was also confirmed by immunoblotting (I, III) and by

isolating the CD59 molecule from T47D and MCF7 cells (I). CD55 was detected

in an all examined cell lines except on PA-1 cells by IF. However, its expression

level was clearly lower than that of the other complement regulators.

Complement regulator expression was also studied on tumor samples

obtained at mastectomies and ovarectomies (I, III). CD46, CD55 and CD59

were detected in vivo in cryostat sections of solid breast tumors. A total o f

12 specimens (ductal carcinoma: 5 cases, metastases of ductal carcinoma: 2

cases, lobular carcinoma: 3 cases and fibroadenoma of the breast: 2 cases)

were examined by IF microscopy. CD59 was found to be strongly expressed

by all the tumors (I). Staining appeared significantly stronger in the tumor

cells than in the neighboring connective tissue. In cells that showed an epithelial

polarized pattern the strongest staining for CD59 was seen at the apical

membranes. A very strong staining for CD59 was seen within the ducts and

on cell surfaces adjacent to the duct lumen. In all tissues the endothelia o f

blood vessels appeared strongly positive for CD59.

Twentyeight ovarian tumor samples were obtained during ovarectomies ( 3

serous cystadenomas, 13 mucinous cystadenomas, 8 serous papillary

cystadenocarcinomas and 4 mucinous cystadenocarcinomas). CD46 was

expressed on the epithelial cells of all the ovarian tumors. CD55 was

expressed in 21 out of 28 cases. In general CD55 expression was relatively

weak. CD59 was strongly expressed on the epithelial cells of all the ovarian

adenomas and carcinomas. No apparent difference in staining intensity was

detected between adenomas and carcinomas (III). The distribution of CD59

expression on MTS grown from T47D cells was analogous to tumor samples

43

from breast cancers (I, IV). In some studies the expression of the

complement-regulatory proteins CD55, CD46 and CD59 has been found to be

deregulated in cancer, with tumors showing loss of one or more inhibitors and

strong overexpression of the others100.

Since a strong expression of GPI-anchored CD59 was seen in breast duct

epithelia (I) its presence in human milk was also analyzed. Milk fat is

composed primarily of triglycerides that are secreted into milk enveloped by a

membrane derived from the epithelial cells of the mammary gland. The milk f a t

glubule membrane consists of three zones. The two outer layers are

separated from the core by a proteinaceous coat. The narrow middle layer is

an apparent plasma membrane while the outermost layer or glycocalyx is rich

in carbohydrates and corresponds to a typical glycocalyx of a cell114. The

MFG-membranes contain a variety of glycoproteins that have been used t o

raise antibodies able to detect surface antigens on malignant human

mammary cells115. Milk fat globules were isolated from human colostrum and

milk by successive centrifugation (II). In phase contrast microscopy the

apparent MFGs in the washed cream layer appeared heterogeneous in size

and showed no internal structures. Immunostaining with the BRIC229 mAb

showed that the MFG particles were covered with the CD59-antigen. The

CD59-specific fluorescence was not homogeneous but appeared in clusters on

the surface of the MFG particles. Occasionally CD59-specific staining was seen

on aggregates outside the MFG particles. The membranes of MFG are

unstable, and soon after secretion into alveolar lumen the MFG lose some o f

their membranous material through vesiculation116. The inhomogeneous CD59-

containing aggregates outside the MFG-particles (II; Fig. 1C) may represent

material that has become shed off from the membranes. Recently, we

observed that shedding of CD59 and CD46 in vesicles is a common

phenomenon of ovarian and breast cancer cells117.

44

5.1.2 Characterization of CD59 on T47D cells and MFG (I, II)

CD59 from breast cancer cells (I) and MFG particles (II) was purified t o

further characterize it. The MCF7 and T47D cell membrane-extracts were

solubilized with 60 mM n-octyl-ß-D-glucopyranoside. The extracts were

subjected to YTH53.1 anti-CD59 affinity column and the eluted material was

analyzed by SDS-PAGE and immunoblotting. The isolated proteins migrated as

broad bands with apparent molecular weights ranging from 19 to 25 kDa (I).

The pattern was similar to that of CD59 purified from erythrocyte cell

membranes (CD59E) although, depending on the amount of sample loaded in

the gel, the smears of CD59E sometimes extended to 30 kDa (not shown).

The smear for soluble CD59 isolated from human urine (CD59U) started from

a slightly higher Mw (molecular weight; 21 kDa) than that of the lipid-anchored

CD59. Additional 28 and 32 kDa bands in the immunoblots were apparently

due to nonspecific reactivity of the samples with the secondary antibodies,

because these bands were seen also in the controls where the primary

antibodies were omitted. Purified radiolabeled T47D-CD59 became

incorporated into the rabbit erythrocytes indicating that the isolated T47D-

CD59 had not lost its glycophospholipid anchor. Under similar conditions

urinary CD59 did not become associated with the erythrocytes.

CD59 was affinity-purified in the same manner from milk and MFG-membranes

(CD59M) and analyzed by SDS-PAGE and immunoblotting using the BRIC229

mAb (II). In the CD59M preparation the BRIC229 anti-CD59 mAb bound

reproducibly to discrete bands with molecular weights ranging from 19 to 23

kDa. The pattern of distinct bands, usually three or four in number, was seen

in all MFG samples examined. Control CD59E was visible as a diffuse smear

from 18 to about 28 kDa. CD59M resembled CD59 isolated from MCF7 breast

carcinoma cells that also had distinct bands in the immunoblot. In SDS-PAGE

the affinity-purified CD59M migrated as bands with molecular weights of 19,

20, 22 and 23 kDa. When CD59M was treated with endoglycosidase F its Mw

decreased to 14 to 16 kDa when analyzed in a 15 % SDS-PAGE slab gel under

45

reducing conditions. Heterogeneity in the Mw of CD59 is due to variable

branching and sialylation of the N-linked oligosaccharide side chain, which is

linked to the Asn18 residue in the polypeptide chain (Fig. 2) and constitutes

about 25 % of the molecular mass of CD59118.

The affinity-purified T47D-CD59 and CD59M retained their functional activity as

judged by inhibition of complement lysis of guinea pig erythrocytes (GPE) when

incorporated into their membranes. GPE activate complement via the

alternative pathway leading to deposition of MAC on the cell membranes and

eventually cell lysis. CD59 inhibits lysis by binding to the nascent C5b-8-complex

and preventing the insertion and polymerization of C945, 46. When CD59 is

mixed with GPE it spontaneously incorporates into the cell membrane via its

hydrophobic phospholipid anchor. The full activity of CD59 requires the

presence of its glycolipid tail as the soluble molecule is approximately a 200-

fold less efficient inhibitor of cell lysis than the lipid-tailed CD59119.

Incorporation of T47D-CD59 into GPE was found to lead to inhibition of lysis o f

the cells by NHS. The functional activity of T47D-CD59 was equivalent to that

of CD59E (I). The C-lysis inhibitory effect of incorporated T47D-CD59 could be

blocked by F(ab')2 fragments of the YTH53.1 mAb. When 125I-labeled CD59M

was incubated with GPE 29% of the radioactivity became incorporated into

the cells and remained there after repeated cycles of washing (II). Under

similar conditions only 4% of CD59U became associated with the cells.

Furthermore, the lysis of GPE by NHS could be inhibited in a dose-dependent

fashion by associating of different amounts of CD59M on their membranes. Full

inhibition of GPE lysis by 4% NHS started to occur at a CD59M concentration

of approximately 3 µg/ml (II: Fig. 4). The inhibitory activity of CD59E was

similar to that of CD59M giving a 73% inhibition at a concentration of 5 µg/ml.

Functional activities of T47D-CD59 and CD59M were tested further by

examining whether they bind to the terminal complement complexes (TCC). In

a TCC-binding assay complement is activated with inulin in C9-deficient serum.

Functionally active 125I-labeled CD59 binds to the formed SC5b-8 complex.

46

The bound CD59 can be separated from inactive CD59 by high speed

centrifugation in a sucrose gradient. The 125I-labeled CD59M and T47D-CD59

bound to the soluble SC5b-8 complexes similarly as has been shown earlier fo r

CD59E45 and soluble CD59U

53 (I).

It has been shown that melanoma cells constitutively release a functionally

active form of CD59 that contains an anchor and is able to insert into cell

membranes of homologous cells transiently increasing their expression o f

CD59120. A soluble form of CD59, that retains its anchoring ability and

functional properties, has been identified in body fluids and in culture

supernatants of different malignant cells121. The physiological significance o f

CD59 in body fluids or as well in MFG remains unknown. The latter could simply

represent a GPI-anchored protein that has become sloughed off from the cell

membranes during lipid secretion. An intrinsic property of milk in killing and

preventing the invasion of pathogens in the gastrointestinal tract of the child

is accomplished mainly by noninflammatory mechanisms, i.e. by lactoferrin,

lysozyme, fibronectin and IgA122. Although, the components of the

complement system and complement-activating immunoglobulins IgG and IgM

are present in milk at low levels123, deposition of C3 fragments has been

detected on bacteria incubated with milk124. IgA, the most abundant

immunoglobulin in milk, does not activate complement and thereby also limits

inflammatory responses in the gut125. In addition, a number of anti-

inflammatory substances including antioxidants, catalase and histaminase are

present in milk125. This may be particularly relevant since during the post

partum period both the child and the mother are exceptionally prone t o

infections with associated inflammation.

5.2 Complement-mediated killing of MCF7 and T47D cells (I)

The potential effector mechanims of monoclonal antibodies in cancer therapy

include targeting of various toxic molecules, ADCC, CDC or CDCC. mAb have

47

been used in the therapy of lymphomas and melanomas with a limited

success119, 126. In the latter study intravenously administered monoclonal

anti-GD3-ganglioside mAb was observed to become deposited on solid

melanoma tumors and lead to lymphocyte and mast cell infiltration as well as

to local complement deposition. These results give hope that it is possible t o

launch an in vivo complement attack against tumor cells recognized by

antibodies.

Effective killing of the tumor cells requires their resistance to complement must

be overcome. In study (I) the cytotoxic effect of human complement against

MCF7 and T47D was examined in the presence and absence CD59-neutralizing

antibodies. When MCF7 cells were treated with F(ab')2 fragments of the

YTH53.1 mAb prior to sensitization with complement activating antibody (S2)

an increase in antibody-induced complement-mediated killing of the cells was

observed (I). Approximately 12 µg/ml of the YTH53.1 mAb was required t o

obtain full neutralization of the tumor-cell CD59 activity. In the absence of the

sensitizing rabbit antibodies no lysis occurred with the YTH53.1 F(ab')2 alone.

Also, in separate experiments the F(ab')2 fragments did not induce lysis o f

human erythrocytes by human complement, whereas the whole parent

antibody (rat IgG2b), apparently by activating the classical complement

pathway, lysed human erythrocytes in the presence of complement. Using a

specific anti-CD59 mAb and F(ab')2 fragments thereof it was possible t o

inactivate the functional activity of CD59 on cultured breast cancer cells. In

complement-mediated cell lysis experiments the maximum lysis of MCF7 cells

was 30 - 60% depending on the conditions used.

A polyclonal rabbit antibody S2 was found most effective in sensitizing the

T47D and MCF7 cells to complement lysis. A mAb against HER2/c-erbB-2

epidermal growth factor receptor did not sensitize MCF7 or T47D cells

sufficiently to initiate a lytic complement attack. On the other hand, the rabbit

polyclonal antibody was an efficient inducer of complement lysis, which was

enhanced by the anti-CD59 mAb. For sensitization of breast cancer cells t o

complement lysis the choice of an appropriate antibody is thus a critical

48

factor. This became evident also in the later studies (III and V). A majority o f

the mAb produced are of murine origin and many of them are poor activators

of human complement, especially the IgG1 isotype. Humanization of mouse

monoclonal antibodies is expected to widen their use for human cancer

therapy. It is possible to produce humanized mAbs that are functional in

mediating the immune effector functions, consisting of ADCC and CDC127.

Lately it has been shown that the recombinant form of the anti-HER2 mAb

(trastuzumab, Herceptin) activates complement leading to C3 deposition and

complement-mediated lysis of breast carcinoma cells if the mCRP are

neutralized128. The fact that total lysis was not achieved using S2 and anti-

CD59 suggests that the cells have other protection mechanisms than CD59

expression and/or that a more effective complement treatment protocol

(higher doses, longer incubation times) should be used. When studied by

indirect IF microscopy using a mouse mAb against DAF (BRIC216) or MCP

(GB24) both regulators were found to be expressed by MCF7 and T47D cells,

although the expression of DAF was relatively weak on T47D cells. The

expression of functionally active CD59 on breast cancer cells shows that i t

has a role in protecting the malignant cells against lysis by the homologous

complement system and strongly suggests a similar function for it in vivo. The

neutralization of CD59 selectively on tumor cells, while saving the "bystander"

cells, poses a problem which we have attempted to solve by targeting the

anti-CD59 mAb to the tumor cells with the help of the biotin-avidin system129.

Biotinylation of the anti-CD59 mAb converts it into a nonactivator of C130 and

may facilitate its use as an adjuvant antibody in tumor-specific

immunotherapy. As a nonactivator of complement the biotinylated anti-CD59

mAb probably does not cause significant damage to bystander cells, although

it retains its ability to neutralize CD59 on the surface of tumor cells when

attracted to their surfaces by avidin-conjugated tumor-specific mAb.

5.2.1 Expression of TAA on ovarian cancer cells (III)

A critical factor for considering CDC in anti-tumor therapy is the selection o f

an appropriate TAA and mAb against them. Therefore, the expression o f

49

several TAA in ovarian cancer cell lines was studied using flow-cytometric

analysis (FACS). The Caov-3, PA-1, SK-OV-3 and SW626 cell lines had each an

individual expression pattern for the CA19-9, CA125 and MUC-1 antigens. The

Tn antigen was not expressed by any of the cell lines studied. For the

expressed Ags the staining intensities were generally low, and the expression

patterns were heterogeneous. An exception was the SW626 cell line, which

showed very strong expression of CA19-9 on its surface. In a subsequent

complement-activation assay the SW626 cells exposed to the anti-CA19-9

(C241) mAb and NHS showed a strong C3 deposition on the their surface in

FACS analysis. The amount of C3 deposition was much lower on PA-1 than on

SW626 cells, apparently reflecting the difference in CA19-9 antigen density

between the two cell lines. Similarly, the anti-MUC-1 mAb (Ma552) was able t o

induce C3 deposition on SK-OV-3 cells but not on Caov-3 cells. Neither of the

mAbs (Ov185 and Ov197) against CA125 Ag were able to induce C3

deposition on the Caov-3 cells, although the mAbs bound to the cell surface.

Compared with the mAbs the S2 anti serum efficiently induced C3 deposition

on all cell lines studied.

5.2.2 CDC and ovarian cancer cells (III)

In the next set of experiments, CDC induced by C241 mAb against CA19-9 in

SW626 and PA-1 cells was studied. In a chromium lysis assay 10% of the

SW626 cells were killed when sensitized with the C241 mAb and exposed t o

NHS. The neutralization of CD59 using biotinylated YTH53.1 mAb increased

the complement-mediated killing to 30%. Unlike SW626 cells, PA-1 cells were

not lysed by C241 and NHS, even if the complement regulatory function o f

CD59 was blocked. However, the PA-1 cells exposed to S2 were highly

sensitive to CDC (64%), and almost all PA-1 cells were killed (89%) after

simultaneous blocking of CD59. High sensitivity of the PA-1 teratocarcinoma

cells to complement lysis was associated with a relatively low expression of all

the complement regulators examined. These observations illustrate the need

for finding complement-activating mAbs against antigens that are highly

expressed on the ovarian malignant cells. Neutralization of CD59 with

50

biotinylated YTH53.1 reduced the resistance of almost all the cell lines to CDC,

except of the SK-OV-3 cells that remained somewhat resistant.

5.3 CDC and microtumor spheroids (MTS; IV)

MTS are spherical aggregates of cells growing in culture. They resemble tumor

tissue with their intercellular connections representing an intermediate between

monolayer cell cultures and tumors in vivo131, 132. The size of MTS may range

from a few cells to visible structures of 1 - 2 mm in diameter until the growth

is limited by restricted diffusion of oxygen and nutrients. Spheroids have been

used for studying the penetration of antibodies and the effects of lymphokine

activated killer cells against tumor tissue133, 134. In previous studies we

observed that in traditional killing assays cells growing in aggregates were

notably more resistant to C-mediated cytotoxicity than cells in suspension1. In

a killing assay individual PA-1 cells were incubated with a complement-

activating antibody (S2) and a CD59-neutralizing mAb (YTH53.1B). As a

result, 85 % of the cells were lysed during a 30 min exposure to complement,

whereas 64% of the PA-1 cells exposed to S2 activating antibody alone were

lysed. Under similar conditions the lysis of the PA-1 cells in the MTS exposed t o

S2, YTH53.1 and NHS was approximately 8%. In the control spheroids

exposed to S2 and NHS the lysis remained at 4%.

Experiments in study IV allowed further analysis and visualization o f

complement-mediated attack against three-dimensional tumor tissue. A pulsed

complement attack against MTS grown from T47D and PA-1 cells with

targeting antibodies (S2) and complement in the presence of a CD59

neutralizing antibody resulted in progressive cell membrane damage and cell

detachment on the outer layers of the MTS. Full lysis of cores of the spheroids

appeared to be restricted by an arrest of complement activation at the C3

level. To quantify CDC against the cells in the spheroids a chromium (51Cr)

release assay was used. Individual T47D spheroids were incubated with 3 µCi

of 51Cr for 12 hours in 100 µl of cell culture medium. Earlier studies by

51

autoradiography have demonstrated that an overnight incubation leads t o

penetration of 51Cr throughout the spheroids134. The mean cumulative

spontaneous release of chromium during a 24-hour incubation of spheroids in

the RPMI 1640 medium containing 10% heat-inactivated FCS was 13% ± 0.6%

of the total radioactivity (n=4). This was in correlation with the total

remaining activity (90 ± 1.2%) that was counted from each spheroid after

the 24-hour incubation. The spontaneous release of chromium was considered

as background and was subsequently subtracted from the further results. To

study complement-mediated killing of T47D spheroids the S2 antibody was

used for activation the classical pathway of complement and the biotinylated

YTH53.1 mAb (YTH53.1B) for neutralization of the MAC inhibitor CD59 on the

cells. A 24-hour incubation with antibodies and a single dose of NHS resulted in

the release of 16 ± 4% (IV) of the spheroid-bound radioactivity.

By prolonging the time of exposure and replacing the complement components

consumed it was possible to partially overcome the complement resistance o f

the ductal breast carcinoma (T47D) and ovarian teratocarcinoma (PA-1)

spheroids. During the incubation, at 2, 4 and 8 hours the spheroids were

isolated gently by pipetting and placed into new tubes containing fresh NHS

and antibodies to replace the complement components and IgG consumed, the

cumulative 51Cr-release was measured during the assay at the same time

points. In the killing assay, there was a clear lag phase in the 51Cr release

during the first two hours of incubation (IV). T47D spheroids exposed to S2,

YTH53.1B and NHS for two hours showed only 1% lysis. At 4 hours 5% lysis

was achieved. A more significant lysis was not detected until 8 hours o f

incubation (15 ± 0.5%; n = 4). The mean (± SD) cumulative release o f

chromium from the spheroids exposed to S2, YTH53.1B and NHS during 24

hours of incubation was 33% ± 2.3%, while 62 ± 0.45% of 51Cr was counted

to remain in the spheroids. In the control spheroids incubated with S0, normal

rat IgG and NHS for 24 hours (with replacements) the mean cumulative

activity released was only 3 ± 0.96% (n = 4) more than the spontaneous

release during the same time. In the spheroids exposed to either S2 o r

52

YTH53.1B and NHS the mean cumulative 51Cr-releases were 4 ± 1.5% and 5

± 0.6%, respectively (n = 8). The exposure of single T47D cells in suspension

to S2 alone and NHS resulted in 15% lysis of cells in 30 min (I). The

inactivation of CD59 increased the lysis only 13%. Thus, it is possible that the

detachment of the cells makes them more vulnerable to complement. In some

studies it has been shown that detachment of cells from the matrix induces

apoptosis135 that may lead to additional cell killing. Increasing the incubation

time of the T47D spheroids to 48 hours only marginally increased the level o f

spheroid lysis (IV). The increase in the number of complement replacements t o

a total of 11 at 4-hour intervals during 50 hours of incubation did not

significantly enhance the tumor cell lysis either and a plateau phase was

reached after 42 hours (IV). However, since the spontaneous release o f

51chromium was 30% at 48 hours the assay may not have revealed the full

extent of cell damage during prolonged incubations. With the PA-1 spheroids

3% of lysis was observed at 1 hour while at 2 hours already 37% lysis was

achieved (data not shown). Because of a tendency of PA-1 spheroids t o

spontaneously leak chromium during a prolonged treatment the lysis could not

be followed for longer than four hours at which time 45% lysis was observed.

Following complement exposure the surfaces of the spheroids became clearly

less coherent showing swollen and detaching cells as judged by light

microscopy. Photographic images indicated that the volume of the spheroids

was decreased by 28% from 0.194 ± 0.063 to 0.140 ± 0.05 mm3 (mean ±

SD; n=30) during the 24-hour pulsed treatment with antibodies and

complement. The volume of the control spheroids exposed to S0, normal r a t

IgG and NHS decreased by only 5% during the 24-hour incubation (residual

volume: 0.185 ± 0.013 mm3). Scanning electron microscopic analysis after

the complement exposure showed a release of relatively large vesicles from

the cell surfaces and porous remnants of cell membranes after severe cell

damage (IV). No similar changes were detected in the control spheroids

incubated with NHS, S0 and normal rat IgG (IV). In these controls, the surface

of the microtumors remained smooth with a fine microvillar coating.

53

To visualize death of individual cells in the spheroids, some of the spheroids

treated for various times with S2, YTH53.1B and NHS were incubated for one

hour with propidium iodide (PI) before freezing them into liquid nitrogen.

Fluorescence microscopical analysis of cryostat sections of the PA-1

spheroids showed a 2 to 3-cell layer thick frontier of dying cells on the

microtumor surfaces after 2 hours of complement exposure (IV: Fig. 3).

Apparently, some of the cells had become detached from the spheroid

surface. The nuclei of the detached cells were visible in the propidium iodide

stained specimens. New cell layers became exposed to complement after the

dead cell layer had been peeled off during the process. The cell death thus

proceeded as a frontier but was restricted mainly to the first four layers o f

cells on the periphery of the spheroids after a 24-hour exposure. In the

control microtumors, incubated with S0, nonspecific rat IgG and NHS, no dead

cells were observed on the microtumor surfaces. For comparison, in the T47D

microtumors the rim of dead cells did not extend deeper than one or two cell

layers from the surface after two hours of incubation (IV: Fig. 5).

Furthermore, the cells still remained firmly attached to the microtumor tissue.

Following eight hours of exposure to C, the apparently dead T47D cells on the

surface remained adherent to the spheroidal body. After 24 hours o f

incubation, the PI-staining revealed a large pool of damaged cells detaching

from the surfaces of the spheroids. The necrotic cells in the centers of the

largest spheroids became also stained by PI. In the control spheroids that

were exposed to S0, normal rat IgG and NHS, only the necrotic cells in the

center became stained, but no killing of cells on the surface was detected. This

indicated that propidium iodide was able to penetrate into the center of the

spheroids through the intercellular spaces.

The detachment of cells from the spheroid surface may be due to down

regulation of adhesion receptors and/or proteases released from the dying o r

sublytically attacked tumor cells. The proteolytic enzymes may start t o

degrade the extracellular matrix and proteins required for the cellular

adhesion. Activation of metalloproteinases and degradation of extracellular

matrix occur during various physiological and pathological processes. These

54

include conditions that require tissue remodeling such as tissue

morphogenesis, inflammation and wound healing. On the other hand, e.g. the

complement serine esterase C1s has been shown to degrade type I and type

II collagens136 and to activate the zymogen form of the matrix

metalloproteinase 9 enzyme137. The released proteases might also activate

or inactivate complement components, and thereby have an enhancing o r

inhibitory effect on complement killing of the microtumor cells. Leukocyte

matrix metalloproteinases have been shown to inactivate C1 inhibitor that

inactivates the C1r and C1s proteinases138, 139. Metalloproteases may also

cleave the membrane-bound complement regulators. Recently, we observed

that functionally active CD46 is was released from the surfaces of cancer cells

into solution by a metalloproteinase117. Enzyme release thus occurs and may

affect both tumor cell adhesiveness and complement sensitivity.

5.3.1 The penetration of mAb and complement into MTS

Cryostat sections of the complement-treated spheroids were examined using

IF to analyze the penetration of mAb and complement components into MTS.

During a 2-hour incubation the YTH53.1 mAb penetrated into the spheroid

tissue 4 to 5 cell layers deep (IV; Fig. 4). Following an overnight exposure t o

YTH53.1 and NHS the T47D spheroids became totally infiltrated by the

YTH53.1 IgG. In an earlier study, mAbs were also found to be capable o f

penetrating into MTS140. It is likely that the delivery of complement

components and mAb into the spheroids occurs by diffusion through the

intercellular spaces. However, BRIC 229, another mAb against CD59, did not

completely pass through the spheroids during the same time (data not

shown). Because of stronger binding to target antigens, antibodies of higher

affinity are predicted to diffuse more slowly into the tumor. In some tumor cell

killing assays higher-affinity antibodies have demonstrated an improved

performance in CDC. In contrast, the lower-affinity antibody have penetrated

better into MTS133.

55

Somewhat surprisingly, although C1q has a relatively large size (m.w. 460

kDa) it was able to penetrate 2 to 3 cell layers deep during a 2-hour

exposure to YTH53.1 and NHS (IV: Fig. 4) and almost through the spheroids

during 24 hours. The binding of C1q to the Fc part of an antibody is

noncovalent141 analogous to binding of antibodies to an antigen. The

penetration of S2 was slower compared with the infiltration of the YTH53.1

mAb. In two hours the S2 IgG was able to penetrate only through the first

two cell layers. After 24 hours, the S2 IgG penetrated deeper forming a

gradient towards but not until the core of the spheroids. In the controls,

incubated with rat IgG, rabbit preimmune IgG (S0) and NHS, no deposition o f

nonspecific rat IgG, C1q or rabbit IgG could be detected. To exclude the

possibility that antibodies bound to Fc receptors we used 10.1, C1KM5 and

3G8 mAbs for staining of FcγRI, FcγRII and FcγRIII on T47D cells, respectively.

No binding of any of these mAbs to the T47D cells was detected.

Immunofluorescence analysis showed that C3 (stained with the anti-iC3b

antibody) and C5b-9 deposits remained peripheral in the spheroids and were

restricted to the outermost 1 - 2 cell layers after 2 hours and 2 - 4 cell layers

after 24 hours (IV: Fig. 5) of complement exposure. PI staining was in

accordance with the staining for C3 and C5b-9 revealing that the complement-

mediated killing of the cells was limited to the microtumor surface. In the

controls, where S0, nonspecific rat IgG and NHS were used no deposition o f

C3 or C5b-9 were detected after 24 hours of incubation. Accordingly, no

complement-mediated death of cells was seen on the surfaces of similarly

treated microtumors. The main reason for the lack of cellular death in the

deeper parts of MTS may be the consumption of active complement on the

outer layers of the spheroids.

Complement treatment did not completely disrupt the macroscopic structure

of MTS. The cores of the spheroids retained their integrity during incubation

with antibodies and complement for 24 hours. It was thus possible to kill

efficiently cells growing on the surface of the spheroids but cells deeper inside

were protected from complement lysis. The plating of the spheroids after 50

56

hours resulted in the adherence and spreading of the cells over the cell culture

plate surface indicating that the spheroids still contained viable cells. The

results indicate that the resistance of cancer cells growing in the cores of the

spheroids to complement-mediated injury results at least partly from the

restricted spreading of C3 activation into the spheroids. Unlike, C1q the

forming C3b binds covalently with a thioester bond to a nearby cell surface

during complement activation. While C1q can diffuse relatively freely through

the the spheroid the penetration of C3 is limited, first, by rapid activation and

covalent binding and, thereafter, by rapid inactivation of C3b. Restricted

penetration of C3 is thus not due to a limited diffusion but because of its

specific biochemical properties.

A likely reason for the restriction of complement activation on to the spheroid

surface in our model is an effective complement regulation at the C3 level. In

these studies, we did not attempt to neutralize CD46 or CD55 which both

downregulate C3 activation on cell surfaces. Because of its stronger

expression on the spheroids1 CD46 is likely to promote inactivation of both

C4b and C3b that have become bound to the tumor cell surfaces. Thus, CD46

could effectively block C3 convertase function in both the classical and the

alternative pathways. In our unpublished experiments the inactivation of CD46

together with CD59 (using GB24 and YTH53.1 mAbs, respectively) resulted in

60% lysis of the T47D spheroids.

5.4 A rat model to study complement activation in vivo (V)

The species-selective activity of complement inhibitors has been a hindrance t o

investigating the role of membrane-bound complement inhibitors in rodent

models of human cancer. It has been shown that human cancer cell lines are

significantly more sensitive to lysis by rat complement than by human

complement142. Regression of tumors has been observed in nude mice or ra ts

xenografted with human tumor tissue143-145. In the study V we set up a r a t

model to analyze targeted complement activation in vivo. We characterized

57

the expression levels of complement regulatory proteins in a rat colon tumor

cell line (CC531) and the ability of tumor cell binding monoclonal antibodies

(MG1, MG2, MG3,and MG42a) to activate complement on these cells. MG42a is a

recombinant, complement-activating form of MG4. Rats were inoculated

subcapsularly into the liver with the in vitro growing CC531 cancer cell line that

has been chemically induced in the same inbred strain of Wag Rij rats. The

inoculation resulted in tumor growth in the liver providing a reproducible

homologous animal tumor model.

5.4.1 CDC and rat colorectal cancer cells

Like in studies with human cancer cells (I) the flow cytometric analysis o f

expression of rat mCRP on the CC531 cells suggested that the rat colorectal

cancer cells are efficiently protected from complement activation. The CC531

cells expressed CD55 and CD59 analogously to human mCRP, plus Crry/p65

an additional rat complement inhibitor. As expected, CD46 was not detected

on CC531 cells. In rat, CD46 is mostly expressed in testicular cells. The

expression of Crry/p65, CD55 and CD59 on CC531 cells was confirmed by

Western blotting. Immunoblotting of the CC531 cell membranes with the 5I2

mAb, showed two bands of 55 and 65 kDa for Crry/p65. Also two bands, o f

65 and 185 kDa, were detected with the RdIII7 mAb (anti-CD55), as has been

previously described for CD5558, 146. The anti-CD59 (6D1) mAb revealed one

band of 20 kDa.

5.4.2 C3 deposition on CC531 cells and CDC

To choose an antibody for in vivo studies the ability of the MG1, MG2, MG3

and MG42a mAb to activate the classical pathway of the complement system

was examined by a C3-ELISA. All mAb were able to activate the complement

system in this assay, although MG3 was a weaker complement activator than

the other mAbs. However, MG1 and MG2 were not able to activate

complement on the CC531 cells. Activation of the classical pathway o f

58

complement requires an adequate antigen density on the target cells. C1q has

to be bound by at least two IgG molecules for the activation to occur.

Inactivation of Crry/p65 with anti-Crry/p65 F(ab)2 fragments increased C3

deposition three-fold compared to situation where only MG42a was used.

Inactivation of Crry/p65 and CD55 resulted in a synergistic increase in C3

deposition. As expected, the anti-CD59 mAb (6D1) did not have any effect on

C3 deposition. Because of their low complement activation potential on CC531

cells MG1, MG2 and MG3 were excluded from further studies. For the

complement lysis experiments MG42a was used to specifically activate r a t

complement on the CC531 cells and anti-mCRP mAbs to inactivate the

complement regulators on the cells. Activation of complement by MG42a mAb

and blocking of Crry/p65 and CD59 resulted in 55 % lysis of cells. Blocking o f

Crry/p65, CD55 and CD59 did not further increase the lysis. The control

antibody MG4 did not induce complement lysis. In the presence of a

heterologous complement source, (baby rabbit complement; BRC) 83% C-

mediated lysis was observed.

5.4.3 Complement activation in situ

To investigate the ability of MG42a to activate the complement system in situ

tumor tissue sections of untreated rats were incubated with either MG42a o r

MG4 mAb. Normal rat serum (NRS) was used as a source of complement. The

binding of MG42a mAb and deposition of C3 were analyzed by IF microscopy.

Both MG42a and MG4 bound to the tumor cells but only the recombinant

MG42a mAb was able to activate complement on the tumor sections. A clear

tumor cell membrane staining pattern was observed for both MG42a and MG4

mAb. In cells treated with MG42a a membranous-like staining pattern for C3

was observed. The MG42a mAb staining colocalized with that of C3.

59

5.4.4 Homing of mAb into tumors and complement activation in

vivo

Hagenaars et al. recently described that MG1, MG2 and MG4 were able t o

home to the tumor cells when injected intraperitoneally into tumor bearing

rats147. MG3 could neither be demonstrated on tumor cells when injected into

rats nor when incubated on tumor sections. Although the MG3 antigen was

detected by FACS on cultured CC531 cells it was not detected in the

immunoblots of isolated CC531 cell membranes (unpublished results). This

suggested that the antigen for MG3 mAb is not a protein or that its epitope

is lost during membrane processing. To determine if MG42a had a homing

ability comparable to that of MG4, the two mAb were injected into tumor

bearing rats. Tumors were grown for five days and rats were injected with

MG42a, MG4 or PBS. Two days after the mAb injection the tumors were

removed and snap-frozen. MG42a and MG4 were detected in cryostat sections

of CC531 tumors by using immunofluorescence microscopy. However, C3

deposition on tumor cells could not be detected in MG42a or MG4-treated

rats.

The above experiments suggested that complement C3 may not become

deposited or becomes lost from CC531 cell tumors in vivo. The situation may

be similar as in the spheroids where the penetration of C3 into the MTS

remained peripheral (IV). In the experiments with spheroids the tumor cells are

in direct contact with complement compared to situation in vivo where the

complement components first have to cross endothelial cells of blood vessels.

This may restrict complement activation in the in vivo tumor.

In human cervical carcinoma sporadical deposits of C3d and C5b-9 have been

detected on the tissue sections. Furthermore, the C3d deposition was in

association with low expression of CD46 on the tumor cells103. Interestingly, in

the same study it was observed that both C3d and C5b-9 were stained with

60

high intensity on blood vessels of tumor samples. In CC531 cells both CD55

and Crry/p65 are expressed (V). Complement activation in the rat tumor

interior can be restricted by CD55 or Crry/p65 or the surface layers of the

C3b/iC3b-bearing tumor cells may become cleared away by the r a t

phagocytes. These findings support the idea that complement activation on

tumors is efficiently inhibited at the complement convertase level. As

mentioned earlier, CDC is thought to be an important effector mechanism in

the therapeutic treatment of some lymphomas89, 92. However, lymphoma

cells are in direct contact with serum complement and this together with

efficiently sensitizing mAb may overrun the C3-convertase inhibitors.

Taken together this study shows that mAb can reach tumors in vivo but

multiple mechanisms can restrict the access of complement components t o

the tumor cells.

61

6. CONCLUSIONS

The studies included in this thesis demonstrate that human breast (I) and

ovarian tumors (III) strongly express the complement membrane attack

inhibitor CD59, and to a lesser extent CD46 and CD55. With a mAb against

TAA (CA19-9) that is expressed by an ovarian cancer cell line (SW626) it was

possible to kill these cells using NHS as a source of complement (III).

Expression of CD59 apparently makes the tumor cells resistant to CDC, but

this resistance can be partially overcome by specific mAb that neutralize the

activity of CD59 (I, III). Study III indicated that the target antigen density on

the cancer cells is a critical factor for efficient CDC.

CD59 is expressed on the surfaces of normal and malignant cells. It was also

detected in the breast duct epithelial cells, from where it apparently became

shed to breast milk. Within the milk CD59 was present on the milk fat globule

membranes (II). Like on cell membranes CD59 in MFG had a glycolipid anchor

and maintained its functional activity.

Micrometastases and residual malignant cells after surgery constitute an

important clinical problem in cancer therapy. The approach of using

complement against tumors in vivo is currently limited by the shortage o f

tumor cell specific and complement activating antibodies. Many of the mAb

that are produced in murine hybridoma cell lines belong to the IgG1 subclass

that in general is a poor activator of human complement. Another problem is

the activity of complement regulators on the malignant cells. However, as

implied by the current and our earlier studies it is possible to specifically

target complement attack against tumors if suitable mAb are available. As

shown here, mAb (IV, V) and even large complement molecules, like C1q (IV),

can penetrate into the tumor tissue. Partial destruction of microtumors could

thus be achieved by neutralizing the mCRP and optimizing the antibody-

initiated complement attack. For further studies, the spheroid system offers a

useful model.

62

Like human cancer cells rat colorectal cancer cells also express complement

regulators on their membranes (V). It was possible to kill CC531 cells by

activating rat homologous complement with a specific mAb against these

cells. However, the homologous tumors transplanted into the rat were

resistant to complement activation in vivo although the activating mAb homed

into the tumors. This indicates that the homologous complement inhibitors still

pose a difficult obstacle for mAb-therapy of tumors in vivo. However, by

engineering suitable mCRP-neutralizing antibodies and selecting appropriate

tumor-specific C-activating mAbs it is, in principle, possible to target

complement attack against malignant cells.

63

7. ACKNOWLEDGMENTS

The thesis was carried out at the Department of Bacteriology and

Immunology, Haartman Institute, University of Helsinki. I am grateful to the

present and former heads of the department Professor Olli Mäkelä and Acting

Professors Seppo Meri and Risto Renkonen for providing excellent working

facilities.

I am grateful to all those who have been working with me during these years

and made my work possible. I want to express my deepest gratitude to my

supervisor Docent Seppo Meri for his enthusiasm and guidance and an

opportunity to learn much during this time. He has given me much freedom in

the choice of scientific topics and encouraged me to develop my

responsibilities in science.

I am grateful to my reviewers Professor Mikko Hurme and Docent Timo

Paavonen who gave me valuable criticism and comments for improving this

thesis.

I warmly thank all collaborators and coauthors: Line Bjørge and Roald Matre

from the University of Bergen, Norway, T Wahlström, Helsinki University Central

Hospital, as well as Kyra Gelderman, Martin Hagenaars, P.J.K Kuppen and Arko

Gorter, from the Leiden University, the Netherlands.

I am grateful to Docent Aaro Miettinen, MD, Riitta Väisänen, Maiju Solin, Anni

Haltia, Jukka Reivinen, Laura Kerosuo, they have helped and advised me

whenever needeed. Their lab is full of exciting gadgets and antibodies that

have been helpful in performing the experiments. They have also made the

department a fun place to work at. I warmly thank Docent Pentti Kuusela. He

has always been helpful in sharing his knowledge. I am indebted to Docent

Matti Kaartinen who taught me methods in molecular genetics. I warmly thank

Mine Eray for sharing her knowledge in science. I also thank Docent Ilkka

64

Seppälä for his helpful advice. I thank Monica Schoultz, for technical assistance

with the flow cytometric analyses.

I warmly thank my previous and present colleagues. I am grateful and

priviliged for having had the opportunity to work with Timo Lehto, Jorma

Tissari, Antti Väkevä, and Sakari Jokiranta. I thank Taru Meri and Hanna

Jarva for brain-storming and for making work in the lab fun. I am indebted t o

Riina Rautemaa for her positive and friendly attitude and for our fruitful

discussions. I thank Sami Junnikkala for useful discussions and fun moments. I

want to thank our technician Marjatta Ahonen for her technical assistance

and precise work to making work in the lab more organized and easier. I

thank Mervi Närkiö-Mäkelä, Antti Lavikainen, Antti Harjunpää, Antti Alitalo,

Markus Lehtinen, Mikko Holmberg, Jinghui Yang, Zhu-Zhu Cheng, in our group

for the nice moments together. My special thanks go to Pauliina Luimula, and

Jyrki Eloranta, for fun moments outside the lab and Jyrki also for

proofreading my thesis.

Finally, I am most grateful to my friends for making life outside the lab

pleasant and exciting. Special thanks belong to my parents for their guidance

and support.

I deeply acknowledge the financial and material support from the Sigrid

Jusélius Foundation, the Academy of Finland, Finnish Cultural Foundation, Maud

Kuistila Foundation, Finnish Cancer Society, Oskar Öflund Foundation, the

Haartman Institute, the Helsinki University Central Hospital, and the University

of Helsinki.

Helsinki, December 2002

65

8. REFERENCES

1. Bjørge L, Junnikkala S, Kristofferson EK, Hakulinen J, Matre R, Meri S.1997. Resistance of ovarian teratocarcinoma cell spheroids t ocomplement-mediated lysis. Brit J Cancer 75; 1247

2. Köhler G, Milstein C. 1975. Continuous cultures of fused cells secretingantibody of predefined specificity. Nature 256; 495

3. Sunyer JO, Lambris JD. 1998. Evolution and diversity of the complementsystem of poikilothermic vertebrates. Immunol Rev 166; 39

4. Dempsey PW, Allison MED, Akkaraju S, Goodnow CC, Fearon DT. 1996.C3d of complement as a molecular adjuvant: Bridging innate andacquired immunity. Science 271; 348

5. Walport MJ. 2001. Complement. Second of two parts. New Engl J Med344; 1140

6. Walport MJ. 2001. Complement. First of two parts. New Engl J Med344; 1058

7. Mevorach D, Mascarenhas JO, Gershov D, Elkon KB. 1998. Complement-dependent clearance of apoptotic cells by human macrophages. J ExpMed 188; 2313

8. Harrison RA, Lachman PJ. 1980. The physiological breakdown of thirdcomponent of human complement. Mol Immunol 17; 9

9. Ross GD, Medof ME. 1985. Membrane complement receptors specificfor bound fragments of C3. Adv Immunol 37; 217

10. Ramos OF, Patarroyo M, Yefenof E, Klein E. 1989. Requirement o fleukocytic cell adhesion molecules (CD11a-c/CD18) in the enhanced NKlysis of iC3b-opsonized targets. J Immunol 142; 4100

11. Hugli TE. 1984. Structure and functions of anaphylotoxins. SpringerSemin Immun 7; 193

12. Kohl J. 2001. Anaphylatoxins and infectious and non-infectiousinflammatory diseases. Mol Immunol 38; 175

13. Müller-Eberhard HJ. 1986. The membrane attack complex o fcomplement. Annu Rev Immunol 4; 503

14. Morgan BP. 1989. Complement membrane attack on nucleated cells:resistance, recovery and non-lethal effects. Biochem J 264; 1

15. Pangburn MK, Schreiber RD, Müller-Eberhard HJ. 1981. Formation of theinitial C3 convertase of the alternative pathawy. Aquisition of C3b-likeactivities by spontaneous hydrolysis of the putative thioester in nativeC3. J Exp Med 154; 856

16. Fearon DT, Austen KF. 1977. Activation of the alternative complementpathway with rabbit erythrocytes by circumvention of the regulatoryaction of endogenous control proteins. J Exp Med 146; 22

17. Agrawal A, Shrive AK, Greenhough TJ, Volanakis JE. 2001. Topology andstructure of the C1q-binding site on C-reactive protein. J Immunol 166;3998

18. Hicks PS, Saunero-Nava L, Du Clos TW, Mold C. 1992. Serum amyloid Pcomponent binds to histones and activates the classical complementpathway. J Immunol 149; 3689

66

19. Navratil JS, Watkins SC, Wisnieski JJ, Ahearn JM. 2001. The globularheads of C1q specifically recognize surface blebs of apoptotic vascularendothelial cells. J Immunol 166; 3231

20. Lu J, Thiel S, Wiedemann H, Timpl R, Reid KBM. 1990. Binding of thepentamer/hexamer forms of mannan binding protein to zymosanactivates the proenzyme C1r2C1s2 complex, of the classical pathwayof complement, without involvement of C1q. J Immunol 144; 2287

21. Sato T, Endo Y, Matsushita M, Fujita T. 1994. Molecularcharacterization of a novel serine protease involved in activation of thecomplement system by mannose binding protein. Int Immunol 6; 665

22. Thiel S, Vorup-Jensen T, Stover CM, Schwaeble W, Laursen SB, PoulsenK, Willis AC, Eggleton P, Hansen S, Holmskov U, Reid KB, Jensenius JC.1997. A second serine protease associated with mannan-binding lectinthat activates complement. Nature 386; 506

23. Ramm LE, Mayer MM. 1980. Life-span and size of the trans-membranechannel formed by large doses of complement. J Immunol 124; 2281

24. Papadimitriou JC, Ramm LE, Drachenberg CB, Trump BF, Shin ML. 1991.Quantitive analysis of adenine nucleotides during the prelytic phase o fcell death mediated by C5b-9. J Immunol 147; 212

25. Koski CL, Ramm LE, Hammer CH, Mayer MM, Shin ML. 1983. Cytolysis o fnucleated cells by complement: cell death displays multi-hitcharacteristics. P Natl Acad Sci USA 80; 3816

26. Morgan BP, Imagawa DK, Dankert JR, Ramm LE. 1986. Complementlysis of U937, a nucleated mammalian cell line in the absence of C9:effect of C9 on C5b-8 mediated cell lysis. J Immunol 136; 3402

27. Ziccardi RJ. 1982. A new role of C1-inhibitor in homeostasis: Control o factivation of the first component of human complement. J Immunol128; 2505

28. Scharfstein J, Ferreira A, Gigli I, Nussenzweig V. 1978. Human C4b-binding protein. Isolation and characterization. J Exp Med 148; 207

29. Holmberg MT, Blom AM, Meri S. 2001. Regulation of complementclassical pathway by association of C4b-binding protein to the surfacesof SK-OV-3 and Caov-3 ovarian adenocarcinoma cells. J Immunol 167;935

30. Whaley K, Ruddy S. 1976. Modulation of C3b hemolytic activity by aplasma protein distinct from C3b inactivator. Science 193; 1011

31. Jenne JD, Tschopp J. 1989. Molecular structure and functionalcharacterization of a human complement cytolysis inhibitor found inblood and seminal plasma: identity to sulphated glycoprotein 2, aconstituent of rat testis fluid. P Natl Acad Sci USA 86; 7123

32. Podack ER, Müller-Eberhard HJ. 1979. Isolation of human S-protein, aninhibitor of the membrane attack complex of complement. J Biol Chem254; 9808

33. Yamamoto H, Blaas P, Nicholson-Weller A, Hansch GM. 1990.Homologous species restriction of the complement-mediated killing o fnucleated cells. Immunology 70; 422

34. Zhao XJ, Zhao J, Zhou Q, Sims PJ. 1998. Identity of the residuesresponsible for the species-restricted complement inhibitory function o fhuman CD59. J Biol Chem 273; 10665

67

35. Fearon DT. 1979. Regulation of the amplification C3 convertase o fhuman complement by an inhibitory protein isolated from humanerythrocyte membranes. P Natl Acad Sci USA 76; 5867

36. Fearon DT. 1980. Identification of membrane glycoprotein that is theC3b receptor of the human erythrocyte, polymorphonuclear leukocyte,B lymphocyte, and monocyte. J Exp Med 152; 20

37. Liszewski MK, Atkinson JP. 1998. Regulatory proteins of complement. InThe human complement system in health and disease. Volanakis JE,Frank MM, editors. Marcel Dekker Inc., New York. 149.

38. Seya T, Turner JR, Atkinson JP. 1986. Purification and characterizationof a membrane protein (gp45-70) that is a cofactor for cleavage o fC3b and C4b. J Exp Med 163; 837

39. Nicholson-Weller A, Burge J, Fearon DT, Weller PF, Austen KF. 1982.Isolation of a human erythrocyte membrane glycoprotein with decay-accelerating activity for C3 convertases of the complement system. JImmunol 129; 184

40. Davies A, Simmons DL, Hale G, Harrison RA, Tighe H, Lachmann PJ,Waldmann H. 1989. CD59, an LY-6-like protein expressed in humanlymphoid cells, regulates the action of the complement membraneattack complex on homologous cells. J Exp Med 170; 637

41. Sugita Y, Nakano Y, Tomita M. 1988. Isolation from humanerythrocytes of a new membrane protein which inhibits the formation o fcomplement transmembrane channels. J Biochem-Tokyo 104; 633

42. Sugita Y, Tobe T, Oda E, Tomita M, Yasukawa K, Yamaji N, TakemotoT, Furuichi K, Takayama M, Yano S. 1989. Molecular cloning andcharacterization of MACIF, an inhibitor of membrane channel formationof complement. J Biochem-Tokyo 106; 555

43. Okada N, Harada R, Fujita T, Okada H. 1989. A novel membraneglycoprotein capable of inhibiting membrane attack by homologouscomplement. Int Immunol 1; 205

44. Holguin MH, Fredrick LR, Bernshaw NJ, Wilcox LA, Parker CJ. 1989.Isolation and characterization of a membrane protein from normalhuman erythrocytes that inhibits reactive lysis of the erythrocytes o fparoxysmal nocturnal hemoglobinuria. J Clin Invest 84; 7

45. Meri S, Morgan BP, Davies A, Daniels RH, Olavesen MG, Waldmann H,Lachmann PJ. 1990. Human protectin (CD59), an 18,000-20,000 MWcomplement lysis restricting factor, inhibits C5b-8 catalysed insertion o fC9 into lipid bilayers. Immunology 71; 1

46. Rollins SA, Sims PJ. 1990. The complement-inhibitory activity of CD59resides in its capacity to block incorporation of C9 into membrane C5b-9. J Immunol 144; 3478

47. Meri S, Waldmann H, Lachmann PJ. 1991. Distribution of protectin(CD59), a complement membrane attack inhibitor, in normal humantissues. Lab Invest 65; 532

48. Brooimans RA, Van der Ark AAJ, Tomita M, Van Es LA, Daha MR. 1992.CD59 expressed by human endothelial cells functions as a protectivemolecule against complement-mediated lysis. Eur J Immunol 22; 791

49. Meri S, Mattila P, Renkonen R. 1993. Regulation of CD59 expression onthe human endothelial cell line EA.hy 926. Eur J Immunol 23; 2511

68

50. Rooney IA, Davies A, Griffiths D, Williams JD, Davies M, Meri S, LachmannPJ, Morgan BP. 1991. The complement-inhibiting protein, protectin(CD59 antigen), is present and functionally active on glomerularepithelial cells. Clin Exp Immunol 83; 251

51. Rooney IA, Davies A, Morgan BP. 1992. Membrane attack complex(MAC)-mediated damage to spermatozoa: protection of the cells bythe presence on their membranes of MAC inhibitory proteins.Immunology 75; 449

52. Fletcher CM, Harrison RA, Lachmann PJ, Neuhaus D. 1993. Sequence-specific 1H-NMR assignments and folding topology of human CD59.Protein Sci 2; 2015

53. Meri S, Väkevä A, Laari T, Lachmann PJ. 1991. Soluble forms of CD59-antigen: distribution in body fluids and functional activity (abstr). ComplInflamm 8; 193

54. Sugita Y, Ito K, Shiozuka K, Suzuki H, Gushima H, Tomita M, Masuho Y.1994. Recombinant soluble CD59 inhibits reactive haemolysis withcomplement. Immunology 82; 34

55. Rooney IA, Morgan BP. 1992. Characterization of the membrane attackcomplex inhibitory protein CD59 antigen on human amniotic cells and inamniotic fluid. Immunology 76; 541

56. Rooney IA, Atkinson JP, Krul ES, Schonfeld G, Polakoski K, Saffitz JE,Morgan BP. 1993. Physiologic relevance of the membrane attackcomplex inhibitor protein CD59 in human seminal plasma: CD59 ispresent on extracellular organelles (prostasomes), binds cellmembranes, and inhibits complement-mediated lysis. J Exp Med 177;1409

57. Mead R, Hinchliffe SJ, Morgan BP. 1999. Molecular cloning, expressionand characterization of the rat analogue of human membranecofactor protein (MCP/CD46). Immunology 98; 137

58. Hinchliffe SJ, Spiller OB, Rushmere NK, Morgan BP. 1998. Molecularcloning and functional characterization of the rat analogue of humandecay-accelerating factor (CD55). J Immunol 161; 5695

59. Hughes TR, Piddlesden SJ, Williams JD, Harrison RA, Morgan PB. 1992.Isolation and characterization of a membrane protein from r a terythrocytes which inhibits lysis by the membrane attack complex o frat complement. Biochem J 284; 169

60. Takizawa H, Okada N, Okada H. 1994. Complement inhibitor of rat cellmembrane resembling mouse Crry/p65. J Immunol 152; 3032

61. Quigg RJ, Holers VM. 1995. Characterization of rat complementreceptors and regulatory proteins. CR2 and Crry are conserved, andthe C3b receptor of neutrophils and platelets is distinct from CR1. JImmunol 155; 1481

62. Quigg RJ, Holers VM, Morgan BP, Sneed AE. 1995. Crry and CD59regulate complement in rat glomerular epithelial cells and are inhibitedby the nephritogenic antibody of passive Heymann nephritis. J Immunol154; 3437

63. Diamond LE, Quinn CM, Martin MJ, Lawson J, Platt JL, Logan JS. 2001. Ahuman CD46 transgenic pig model for the study of discordantxenotransplantation. Transplantation 71; 132

69

64. Roos A, Essers M, van Gijlswijk-Janssen D, Bovin NV, Daha MR. 2001.Both IgG and IgM anti-pig antibodies induce complement activation andcytotoxicity. Xenotransplantation 8; 3

65. Janeway CJ, Travers P. 1997. Immonobiology: the immunesystem inhealth and disease. Current Biology Ltd., London.

66. Gunzer M, Janich S, Varga G, Grabbe S. 2001. Dendritic cells and tumorimmunity. Semin Immun 13; 291

67. Ockert D, Schmitz M, Hampl M, Rieber EP. 1999. Advances in cancerimmunotherapy. Immunol Today 20; 63

68. Zitvogel L, Mayordomo JI, Tjandrawan T, DeLeo AB, Clarke MR, LotzeMT, Storkus J. 1996. Therapy of murine tumors with tumor peptide-pulsed dendritic cells: dependence on T cells, B7 costimulation andhelper T helper cell-1 associated cytokines. J Exp Med 183; 87

69. Chen HL, Gabrilovich D, Tampe R, Girgis KR, Nadaf S, Carbone DP. 1996.A functionally defective allele of TAP1 results in loss of MHC class Iantigen presentation in a human lung cancer. Nat Gen 13; 210

70. Almand B, Clark JI, Nikitina E, van Beynen J, English NR, Knight SC,Carbone DP, Gabrilovich DI. 2001. Increased production of immaturemyeloid cells in cancer patients: a mechanism of immunosuppression incancer. J Immunol 166; 678

71. Lou Z, Jevremovic D, Billadeau DD, Leibson PJ. 2000. A balance betweenpositive and negative signals in cytotoxic lymphocytes regulates thepolarization of lipid rafts during the development of cell-mediatedkilling. J Exp Med 191; 347

72. Moretta A, Bottino C, Vitale M, Pende D, Cantoni C, Mingari MC, BiassoniR, Moretta L. 2001. Activating receptors and coreceptors involved inhuman natural killer cell-mediated cytolysis. Ann Rev Immunol 19; 197

73. Diefenbach A, Jamieson AM, Liu SD, Shastri N, Raulet DH. 2000. Ligandsfor the murine NKG2D receptor: expression by tumor cells andactivation of NK cells and macrophages. Nat Immun 1; 119

74. Diefenbach A, Jensen ER, Jamieson AM, Raulet DH. 2001. Rae1 and H60ligands of the NKG2D receptor stimulate tumor immunity. Nature 413;165

75. Palermo B, Campanelli R, Mantovani S, Lantelme E, Manganoni AM,Carella G, Da Prada G, della Cuna GR, Romagne F, Gauthier L, Necker A,Giachino C. 2001. Diverse expansion potential and heterogeneous avidityin tumor-associated antigen-specific T lymphocytes from primarymelanoma patients. Eur J Immunol 31; 412

76. Valmori D, Dutoit V, Rubio-Godoy V, Chambaz C, Lienard D, Guillaume P,Romero P, Cerottini JC, Rimoldi D. 2001. Frequent cytolytic T-cellresponses to peptide MAGE-A10(254-262) in melanoma. Cancer Res61; 509

77. Sakai C, Kawakami Y, Law LW, Furumura M, Hearing VJ, Jr. 1997.Melanosomal proteins as melanoma-specific immune targets. MelanomaRes 7; 83

78. Durrant LG, Robins RA, Armitage NC, Brown A, Baldwin RW, HardcastleJD. 1986. Association of antigen expression and DNA ploidy in humancolorectal tumors. Cancer Res 46; 3543

70

79. Durrant LG, Robins RA, Baldwin RW. 1989. Flow cytometric screening o fmonoclonal antibodies for drug or toxin targeting to human cancer. JNatl Cancer I 81; 688

80. Spendlove I, Li L, Carmichael J, Durrant LG. 1999. Decay acceleratingfactor (CD55): a target for cancer vaccines? Cancer Res 59; 2282

81. Kinders R, Jones T, Root R, Bruce C, Murchison H, Corey M, Williams L,Enfield D, Hass GM. 1998. Complement factor H or a related protein isa marker for transitional cell cancer of the bladder. Clin Cancer Res 4;2511

82. Canevari S, Pupa SM, Menard S. 1996. 1975-1995 revised anti-cancerserological response: biological significance and clinical implications. AnnOncol 7; 227

83. Peyrat J-P, Bonneterre J, Lubin R. 1995. Prognostic significance o fcirculating p53 antibodies in patients undergoing surgery fo rlocoregional breast cancer. Lancet 345; 621

84. Cheever MA, Disis ML, Bernhard H. 1995. Immunity to oncogenicproteins. Immunol Rev 145; 33

85. Gourevitch MM, von Mensdorff-Pouilly S, Litvinov SV. 1995. Polymorphicepithelial mucin (MUC-1) containing circulating immune complexes incarcinoma patients. Brit J Cancer 72; 934

86. Dechant M, Valerius T. 2001. IgA antibodies for cancer therapy. CritRev Oncol Hemat 39; 69

87. Timmer T, de Vries EG, de Jong S. 2002. Fas receptor-mediatedapoptosis: a clinical application? J Pathol 196; 125

88. Eccles SA. 2001. Monoclonal antibodies targeting cancer: 'magic bullets'or just the trigger? Breast Cancer Res 3; 86

89. Golay J, Lazzari M, Facchinetti V, Bernasconi S, Borleri G, Barbui T,Rambaldi A, Introna M. 2001. CD20 levels determine the in vitrosusceptibility to rituximab and complement of B-cell chronic lymphocyticleukemia: further regulation by CD55 and CD59. Blood 98; 3383

90. Gorter A, Meri S. 1999. Immune evasion of tumor cells using membrane-bound complement regulatory proteins. Immunol Today 20; 576

91. Grillo-Lopez AJ, White CA, Varns C, Shen D, Wei A, McClure A, DallaireBK. 1999. Overview of the clinical development of rituximab: firstmonoclonal antibody approved for the treatment of lymphoma. SeminOncol 26; 66

92. Harjunpää A, Junnikkala S, S. M. 2000. Rituximab (anti-CD20) therapyof B-cell lymphomas: direct complement killing is superior to cellulareffector mechanisms. Scand J Immunol 51; 634

93. Ligibel JA, Winer EP. 2002. Trastuzumab/chemotherapy combinations inmetastatic breast cancer. Semin Oncol 29; 38

94. Glennie MJ, Johnson WM. 2000. Clinical trials of antibody therapy.Immunol today 21; 403

95. Jager E, Jager D, Knuth A. 2002. Clinical cancer vaccine trials. Curr OpinImmunol 14; 178

96. Schmidt W, Steinlein P, Buschle M, Schweighoffer T, Herbst E, Mechtler K,Kirlappos H, Birnstiel ML. 1996. Transloading of tumor cells with foreignmajor histocompatibility complex class I peptide ligand: a novel generalstrategy for the generation of potent cancer vaccines. P Natl Acad SciUSA 93; 9759

71

97. Schuler-Thurner B, Schultz ES, Berger TG, Weinlich G, Ebner S, Woerl P,Bender A, Feuerstein B, Fritsch PO, Romani N, Schuler G. 2002. Rapidinduction of tumor-specific type 1 T helper cells in metastatic melanomapatients by vaccination with mature, cryopreserved, peptide-loadedmonocyte-derived dendritic cells. J Exp Med 195; 1279

98. Livingston PO, Wong GYC, Adluri S. 1994. Improved survival in stage IIImelanoma patients with GM2 antibodies: A randomized trial o fadjuvant vaccination with GM2 ganglioside. J Clin Oncol 12; 1036

99. Gilewski T, Ragupathi G, Bhuta S, Williams LJ, Musselli C, Zhang XF,Bencsath KP, Panageas KS, Chin J, Hudis CA, Norton L, Houghton AN,Livingston PO, Danishefsky SJ. 2001. Immunization of metastatic breastcancer patients with a fully synthetic globo H conjugate: a phase I trial.P Natl Acad Sci USA 98; 3270

100. Durrant LG, Spendlove I. 2001. Immunization against tumor cell surfacecomplement-regulatory proteins. Curr Opin Investig Drugs 2; 959

101. Irie K, Irie RF, Morton DL. 1974. Evidence for in vivo reaction of antibodyand complement to surface antigens of human cancer cells. Science186;

102. Niculescu F, Rus HG, Retegan M, Vlaicu R. 1992. Persistent complementactivation on tumor cells in breast cancer. Am J Pathol 140; 1039

103. Gelderman KA, Blok VT, Fleuren GJ, Gorter A. 2002. The inhibitory effectof CD46, CD55, and CD59 on complement activation afterimmunotherapeutic treatment of cervical carcinoma cells withmonoclonal antibodies or bispecific monoclonal antibodies. Lab Invest82; 483

104. Hertler AA, Frankel AE. 1989. Immunotoxins: a clinical review of theiruse in the treatment of malignancies. J Clin Oncol 7; 1932

105. FitzGerald DJ, Willingham MC, Cardelli CO, Hamada H, Tsuruo T,Gottesman MM, Pastan I. 1987. A monoclonal antibody-Pseudomonastoxin conjugate that specifically kills multidrug resistant cells. P NatlAcad Sci USA 84; 4288

106. Suzawa T, Nagamura S, Saito H, Ohta S, Hanai N, Kanazawa J, OkabeM, Yamasaki M. 2002. Enhanced tumor cell selectivity of adriamycin-monoclonal antibody conjugate via a poly(ethylene glycol)-basedcleavable linker. J Control Release 79; 229

107. Chinn PC, Leonard JE, Rosenberg J, Hanna N, Anderson DR. 1999.Preclinical evaluation of 90Y-labeled anti-CD20 monoclonal antibody fortreatment of non-Hodgkin's lymphoma. Int J Oncol 15; 1017

108. Tartour E, Ciree A, Haicheur N, Benchetrit F, Fridman WH. 2000.Development of non-live vectors and procedures (liposomes, pseudo-viral particles, toxin, beads, adjuvants...) as tools for cancer vaccines.Immunol Lett 74; 45

109. Reiter Y, Fishelson Z. 1989. Targeting of complement to tumor cells byheteroconjugates composed of antibodies and of the complement C3b.J Immunol 142; 2771

110. Vogel C-W, Müller-Eberhard HJ. 1981. Induction of immune cytolysis:Tumor-cell killing by complement is initiated by covalent complex o fmonoclonal antibody and stable C3/C5 convertase. P Natl Acad SciUSA 78; 7707

72

111. Yefenof E, McConnell I. 1985. Interferon amplifies complement actvationby Burkitt's lymphoma cells. Nature 313; 684

112. Bara S, Lint TF. 1987. The third component of complement (C3) boundto tumor target cells enhances their sensitivity to killing by activatedmacrophages. J Immunol 138; 1303

113. Niehans GA, Cherwitz DL, Staley NA, Knapp DJ, Dalmasso AP. 1996.Human carcinomas variably express the complement inhibitory proteinsCD46 (membrane cofactor protein), CD55 (decay-accelerating factor),and CD59 (protectin). Am J Pathol 149; 129

114. Buchheim W, Welsch U, Patton S. 1988. Electron microscopy andcarbohydrate histochemistry of the human milk fat globule membrane.In Biology of Human Milk, vol. 15. Hanson LÅ, editor. Raven Press, NewYork. 27.

115. Ceriani RL, Blank EW. 1988. Experimental therapy of human breasttumors with 131I-labeled monoclonal antibodies prepared against thehuman milk fat globule. Cancer Res 48; 4664

116. Patton S, Huston GE. 1986. A method for isolation of milk fat globules.Lipids 21; 170

117. Hakulinen J, Junnikkala S, Sorsa T, Meri S. 2002. The complementinhibitor membrane cofactor protein (CD46) is shed from cancer cells invesicles and converted by a metalloprotease into a functionally activesoluble form. ; manuscript

118. Meri S, Lehto T, Sutton CW, Tyynela J, Baumann M. 1996. Structuralcomposition and functional characterization of soluble CD59:heterogeneity of the oligosaccharide and glycophosphoinositol (GPI)anchor revealed by laser-desorption mass spectrometric analysis.Biochem J 316; 923

119. Hale G, Clark MR, Marcus R, Winter G, Dyer MJS, Phillips JM, Riechmann L,Waldmann H. 1988. Remission and induction in non-Hodgkin lymphomawith reshaped human monoclonal antibody Campath-1H. Lancet i; 1394

120. Brasoveanu LI, Fonsatti E, Visintin A, Pavlovic M, Cattarossi I, Colizzi F,Gasparollo A, Coral S, Horejsi V, Altomonte M, Maio M. 1997. Melanomacells constitutively release an anchor-positive soluble form of protectin(sCD59) that retains functional activities in homologous complement-mediated cytotoxicity. J Clin Invest 100; 1248

121. Maio M, Brasoveanu LI, Coral S, Sigalotti L, Lamaj E, Gasparollo A,Visintin A, Altomonte M, Fonsatti E. 1998. Structure, distribution, andfunctional role of protectin (CD59) in complement-susceptibility and inimmunotherapy of human malignancies (Review). Int J Oncol 13; 305

122. Garza C, Butte NF, Goldman AS. 1993. Human milk and infant formula.2nd ed. In Textbook of Pediatric Nutrition. Suskind RM, Lewinter-SuskindL, editors. Raven Press, New York. 33.

123. Goldman AS, Thorpe LW, Goldblum RM, Hanson LÅ. 1986. Anti-inflammatory properties of human milk. Acta Paediatr Scand 75; 689

124. Ogundele MO. 2000. Activation and deposition of human breast-milkcomplement C3 opsonins on serum sensitive Escherichia coli 0111. JReprod Immunol 48; 99

125. Hanson LÅ, Carlsson B, Jalil F, Hahn-Zoric M, Hermodson S, Karlberg J,Mellander L, Raza Khan S, Lindblad B, Thiringer K, Zaman S. 1988.

73

Antiviral and antibacterial factors in human milk. In Biology of HumanMilk, vol. 15. Hanson LÅ, editor. Raven Press, New York. 141.

126. Houghton AN, Mintzer D, Cordon-Carbo C, Welt S, Fliegel B, Vadhan S,Carswell E, Melamed MR, Oettgen HF, Old LJ. 1985. Mouse monoclonalIgG3 antibody detecting GD3 ganglioside: a phase I trial in patients withmalignant melanoma. P Natl Acad Sci USA 82; 1242

127. Nakamura K, Tanaka Y, Shitara K, Hanai N. 2001. Construction o fhumanized anti-ganglioside monoclonal antibodies with potent immuneeffector functions. Cancer Immunol Immun 50; 275

128. Jurianz K, Maslak S, Garcia-Schuler H, Fishelson Z, Kirschfink M. 1999.Neutralization of complement regulatory proteins augments lysis o fbreast carcinoma cells targeted with rhumAb anti-HER2.Immunopharmacology 42; 209

129. Junnikkala S, Hakulinen J, Meri S. 1994. Targeted neutralization of thecomplement membrane attack complex inhibitor CD59 on the surfaceof human melanoma cells. Eur J Immunol 24; 611

130. Jokiranta TS, Meri S. 1993. Biotinylation of monoclonal antibodiesprevents their ability to activate the classical pathway of complement.J Immunol 151; 2124

131. Carlsson J, Yuhas JM. 1984. Liquid-overlay culture of cellular spheroids.In Recent results in cancer research, vol. 95. Acker H, Carlson J, DurandR, Sutherland RM, editors. Springer-Verlag, Berlin. 1.

132. Sutherland RM. 1988. Cell and enviroment interactions in tumormicroregions: The multicell spheroid model. Science 240; 177

133. Huls G, Gestel D, van der Linden J, Moret E, Logtenberg T. 2001. Tumorcell killing by in vitro affinity-matured recombinant human monoclonalantibodies. Cancer Immunol Immun 50; 163

134. Jääskeläinen J, Kalliomäki P, Paetau A, Timonen T. 1989. Effect of LAKcells against three-dimensional tumor tissue. J Immunol 142; 1036

135. Bachelder RE, Wendt MA, Fujita N, Tsuruo T, Mercurio AM. 2001. Thecleavage of Akt/protein kinase B by death receptor signaling is animportant event in detachment-induced apoptosis. J Biol Chem 276;34702

136. Yamaguchi K, Sakiyama H, Matsumoto M, Moria H, Sakiyama S. 1990.Degradation of type I and II collagen by human C1s. FEBS Lett 268;206

137. Sakiyama H, Inaba N, Toyoguchi T, Okada Y, Matsumoto M, Moriya H,Ohtsu H. 1994. Immunolocalization of complement C1s and matrixmetalloproteinase 9 (92 kDa gelatinase/type IV collagenase) in theprimary ossification center of human femur. Cell Tissue Res 277; 239

138. Pemberton PA, Harrison RA, Lachmann PJ, Carrell RW. 1989. Thestructural basis for neutrophil inactivation of C1 inhibitor. Biochem J258; 193

139. Knäuper V, Triebel S, Reinke H, Tschescke H. 1991. Inactivation o fhuman plasma C1-inhibitor by human PMN leucocyte matrixmetalloproteinases. FEBS Lett 290; 99

140. Cheng F-M, Hansen EB, Taylor CR, Epstein AL. 1991. Diffusion andbinding of monoclonal antibody TNT-1 in multicellular tumor spheroids. JNatl Cancer I 83; 200

74

141. Duncan AR, Winter G. 1988. The binding site for C1q on IgG. Nature 332;738

142. Chen S, Caragine T, Cheung NK, Tomlinson S. 2000. CD59 expressed ona tumor cell surface modulates decay-accelerating factor expressionand enhances tumor growth in a rat model of human neuroblastoma.Cancer Res 60; 3013

143. Velders MP, Van Rhinj Cm, Briairie IH, Fleuren GJ, Warnaar SO, LitvinovSV. 1995. Immunotherapy with low and high affinity monoclonalantibodies 17-1A and 323/A3 in a nude mouse xenograft model.Cancer Res 55; 4398

144. Van Dijk J, Uemura H, Beniers AJMC, Peelen WP, Zegveld ST, Fleuren GJ.1994. Therapeutic effects of monoclonal antibody G250, interferonsand tumor necrosis factor, in nude mice with renal-cell carcinomaxenografts. Int J Cancer 56; 262

145. Takahashi H, Nakada T, Nakaki M, Wands RJ. 1995. Inhibition of hepaticmetastases of human colon cancer in nude mice by chimeris SF-25monoclonal antibody. Gastroenterology 108; 172

146. Spiller OB, Hanna SM, Morgan BP. 1999. Tissue distribution of the r a tanalogue of decay-accelerating factor. Immunology 97; 374

147. Hagenaars M, Koelemij R, Ensink NG, van Eedenburg JDH, vanVlierberghe RLP, Eggermont AMM, van de Velde CJH, Fleuren GJ, KuppenPJK. 2000. The development of novel monoclonal antibodies against therat CC531 rat colon adenocarcinoma. Clin Exp Metastas 18; 281