<<

THE IMMUNOMODULATING ACTIVITY OF AND GAMMA-INTERFERON

ON EXPERIMENTAL MURINE INFECTIONS WITH MYCOBACTERIUM MICROTI AND

MYCOBACTERIUM TUBERCULOSIS AND THE INFLUENCE OF GAMMA-INTERFERON

ON THE BACTERICIDAL ACTIVITY OF ISONIAZID AND RIFAMPICIN

A Thesis submitted for the degree of

Doctor of Philosophy

by

SIEW YAN KHOR

The Royal Postgraduate Medical School

University of London

November, 1985 2

ABSTRACT

Though remarkably successful, chemotherapy of tuberculosis still has

to be given for at least six months and relapses due to endogenous

reactivation remain a problem. Immunomodulating agents which may

complement antituberculous drugs could thus play an important role

in shortening or improving current chemotherapy. This thesis

reports on the effects of two immunomodulating agents, levamisole

and murine gamma-interferon (IFN-X), on models of murine

mycobacterial infections.

Levamisole was investigated in an infection of Mycobacterium microti

and did not have any significant effect on the in vivo growth of

M. microti. The effects of IFN-X were examined in an infection of

M. tuberculosis strain H37Rv. Dosage of 1000 units of IFN-X per mouse every three days with the first dose before infection, caused

a statistically significant but small reduction in bacillary growth

in the organs which occurred mainly in the first day after infec­

tion. An increase in dose size, multiple daily dosage before

infection and encapsulation of IFN-X in liposomes did not increase

the effect. IFN-X treatment of mice infected five days previously

did not influence bacillary growth. The administration of IFN-X both before and after infection, did not increase the bactericidal activities of isoniazid or rifampicin. There was no effect of IFN-X on organ counts of mice previously treated for five days with isoniazid. 3

The effects of IFN-Y on the growth of Listeria monocytogenes and

M. microti in peritoneal macrophage monolayers were also examined.

Peritoneal macrophages previously exposed to IFN-Y were activated for listericidal activity. Macrophages incubated with IFN-ft for 48 or 72 hours before infection with M. microti were activated to be bactericidal in the first 15-30 minutes after infection. The addition of IFN-Y after infection with M. microti resulted in an inhibition of growth that occurred after a delay of 24 hours. The addition of IFN-Y did not increase the bactericidal activities of isoniazid or rifampicin. It is unlikely that levamisole or IFN-Y will have a major impact on the treatment of active tuberculosis. 4

ACKNOWLEDGEMENTS

I would like to thank my supervisor, Professor D.A. Mitchison for all his advice, encouragement and helpful discussions throughout

this work. My thanks are also due to Dr D.B. Lowrie and Dr Ann Rees for their interest and advice and Dr A.R. Coates for supplying the recombinant interferon-gamma. I would also like to acknowledge

Mr V.R. Aber for statistical assistance, members of the former MRC

Unit for Laboratory Studies of Tuberculosis for technical advice and the Commonwealth Fellowship Commission in the United Kingdom for the award of the Commonwealth Fellowship.

Finally, I would like to express my appreciation to my family and

Jeremy for all their support and encouragement. 5

CONTENTS PAGE

List of Tables 12

List of Illustrations 14

1. LITERATURE REVIEW AND INTRODUCTION

I. Literature review

1.1 Tuberculosis 18

^.1.1 The mycobacteria 18

1.1.2 Pathogenesis of tuberculosis 21

1.1.3 The immune response to tuberculosis 23

1.1.4 Mycobacterium-macrophage interactions 29

1.1.4.1 Microbicidal mechanisms in macrophages 30

1.1.4.2 Mycobacterial evasion of macrophage killing mechanisms 33

1.1.4.3 Genetic susceptibility 35

1.2 Tuberculosis control 36

1.2.1 BCG Vaccination 37

1.2.2 Case-finding and chemotherapy 39

1.2.2.1 Case-finding 40

1.2.2.2 Chemotherapy 41

1.2.3 Chemoprophylaxis 45

1.3 Immunomodulation and immunomodulating agents 47

1.3.1 Agents derived from microorganisms 48

1.3.2 Agents derived from mammalian cells 50

1.3.3 Synthetic immumomodulating agents 52

1.4 Interferon 54

1.4.1 Description of the interferons 54

1.4.2 Molecules and genes of the interferons 55 6

1.4.3 Mechanisms of action of interferons 56

1.4.4 Immunomodulating effects of the interferons 58

1.4.4.1 Effects on humoral immunity 59

1.4.4.2 Effects on cell surface membrane 59

1.4.4.3 Effects on cell mediated immunity 61

1.5 Levamisole 63

1.5.1 Description of levamisole 63

1.5.2 Immunomodulating effects of levamisole 64

1.5.3 Mechanisms of action of levamisole 66

1.6 Targeting of drugs 67

1.6.1 Description of drug carriers 67

1.6.2 Liposomes 68

1.6.2.1 Types of liposomes 68

1.6.2.2 In vivo interactions of liposomes 69

1.6.2.3 Therapeutic applications of liposomes 70

II. Introductionand objectives of the experimental work 73

1.7 Introduction and objectives 73

2 MATERIALS AND METHODS 78

2.1 Media 78

2.1.1 Bacteriological culture media and supplements 78

2.1.2 Tissue culture media and supplements 79

2.2 Chemicals and reagents 81

2.3 Bacteria 83

2.3.1 Bacterial strains 83

2.3.2 Animal passage and maintenance 83

2.4 Animals 85

2.4.1 Mouse strains 85 7

2.4.2 Random allocation and husbandry of mice 85

2.5 Immunomodulating and chemotherapeutic agents 86

2.5.1 Agents used 86

2.5.2 In vivo administration 86

2.5.2.1 Preparation of liposomes 87

2.5.3 In vitro administration of agents 88

2.6 Preparation of infective inocula 88

2.6.1 Enumeration of total number of bacilli 88

2.6.2 Infective inocula for in vivo experiments 89

2.6.3 Infective inocula for in vitro experiments 89

2.7 Investigation of the T lymphocyte response 91

2.7.1 Proliferative response of splenic T lymphocytes 91

2.7.1.1 Preparation of splenic lymphocytes 91

2.7.1.2 Antigens and mitogens 91

2.7.1.3 Lymphocyte proliferation test 92

2.7.2 Immunoenzymatic staining of splenic T lymphocyte subsets 93

2.7.2.1 Antibodies and sera 93

2.7.2.2 Cytospin preparations 94

2.7.2.3 Preparation of buffers, fixative and substrates 94

2.7.2.4 Immunoenzymatic staining technique 95

2.8 Assessment of growth of mycobacteria in vivo and in vitro 97

2.8.1 Homogenization of organs 97

2.8.2 Enumeration of viable M. tuberculosis and M. microti 98

2.8.3 Enumeration of L. monocytogenes and M. microti in 98

macrophage monolayers

2.9 In vitro techniques used in macrophage microbicidal assays 99

2.9.1 Preparation of monolayers 99

2.9.2 Enumeration of cell suspensions 100 8

2.9.3 Characterization of macrophage monolayers 101

2.9.4 Harvesting of bacteria from the monolayers 102

2.9.5 Analysis of DNA 103

2.10 In vivo models in the assessment of immunomodulating agents 104

2.10.1 Effect of levamisole on M. microti infection in CFLP mice 104

2.10.2 Effect of IFN-X on M. tuberculosis infection in mice 104

2.10.3 Effect of IFN-X in combination with isoniazid and with 105

rifampicin on M. tuberculosis infection in mice

2.11 In vitro assessment of the effects of IFN-Y 106

2.11.1 Effect of saponin on viability of L. monocytogenes 106

and M. microti

2.11.2 Listericidal assay 107

2.11.3 Effect of IFN-X on growth of M. microti in vitro 108

2.11.3.1 Effect of previous exposure of monolayers to IFN-X 108

2.11.3.2 Effect of IFN-Y on the phagocytosis of M. microti 109

2.11.3.3 Effect of IFN-Y added after infection of monolayers 110

with M. microti

2.11.3.4 Effect of IFN-Y in combination with isoniazid and 111

with rifampicin on the growth of M. microti in

macrophages

2.12 Statistical analysis of the data 112

3 THE IMMUNOMODULATING EFFECT OF LEVAMISOLE ON M. MICROTI 113

INFECTION IN MICE

3.1 Preliminary experiments 113

3.1.1 Choice of media for growth of M. microti 113

3.1.2 Course of infection of M. microti in CFLP mice 115

3.2 The effect of levamisole on M. microti infection in CFLP mice 117 9

3.2.1 The effect of levamisole on the growth of M. microti 118

3.2.2 The effect of levamisole on proliferative responses 121

of splenic T lymphocytes

3.2.3 The effect of levamisole on splenic T cell subsets 125

3.3 Discussion 130

4 THE IMMUNOMODULATING EFFECT OF INTERFERON-GAMMA ON 135

M. TUBERCULOSIS INFECTION IN MICE

4.1 Preliminary experiments 136

4.1.1 Sonication of M. tuberculosis 136

4.2 The effect of infective dose size of M. tuberculosis on the 139

growth of bacilli in the lungs and spleens of CFLP mice

4.3 The effect of dose size of IFN-V on the growth of 142

M. tuberculosis in BALB/c mice

4.4 The effect of administration of IFN-^f in liposomes 146

on the growth of M. tuberculosis in mice

4.4.1 The effect of IFN-X administered in liposomes on 146

M. tuberculosis infection in CFLP mice

4.4.2 The effect of the administration of IFN-JT in liposomes 151

on M. tuberculosis infection in BALB/c mice

4.5 The effect of pretreatment of mice with IFN-^ on 156

M. tuberculosis infection in BALB/c mice

4.6, , TUDiscussion . 161

5 THE IMMUNOMODULATING EFFECT OF INTERFERON-GAMMA IN COMBINATION 169

WITH ISONIAZID AND RIFAMPICIN ON MURINE M. TUBERCULOSIS INFECTION

5.1 Assessment of IFN-tf and isoniazid alone and in combination 169

on M. tuberculosis infection in BALB/c mice 10

5.1.1 The effect of IFN-Y and Isoniazid when IFN-Y was 169

administered before and after infection of BALB/c mice

with M. tuberculosis

5.1.2 The effect of IFN-Y and isoniazid when IFN-Y was 175

administered both before and after infection, and only

after infection of BALB/c mice with M. tuberculosis

5.2 The effect of IFN-Y and rifampicin alone and in combination 184

on M. tuberculosis infection in BALB/c mice

5.3 Discussion 188

6 IN VITRO ASSESSMENT OF THE EFFECTS OF INTERFERON-GAMMA 194

6.1 Preliminary experiments 194

6.1.1 Characterization of monolayers 194

6.1.2 The effect of saponin on viability of L. monocytogenes 194

and M. microti

6.2 Listericidal assay 197

6.2.1 The effect of dose size of IFN-V on the listericidal 197

activity of peritoneal macrophages after previous

exposure for 24 hours

6.2.2 The effect of varying the dose size and pretreatment 200

period of IFN-Y on listericidal activity of

peritoneal macrophages

6.3 In vitro assessment of effect of IFN-& on the growth 202

of M. microti

6.3.1 Treatment of macrophage monolayers with IFN-Y before 202

and after infection 11

6.3.1.1 The effect of dose size of IFN-^ added both before and 202

after infection on the growth of M. microti in

macrophages

6.3.1.2 The effect of previous exposure to IFN-Y on the 207

phagocytosis of M. microti

6.3.1.3 The effect of previous exposure of macrophages with 210

IFN-tf on viable intracellular M. microti at intervals

during the phagocytosis period

6.3.2 The effect of IFN-)f added after M. microti infection 215

of peritoneal macrophages

6.3.3 The effect of IFN-Jf and isoniazid alone and in 217

combination on the growth of M. microti in vitro

6.3.4 The effect of IFN-& and rifampicin alone and in 220

combination on the in vitro growth of M. microti

6.4 Discussion 223

7 GENERAL DISCUSSION 229

REFERENCES 240

APPENDIX 1 289 12

LIST OF TABLES PAGE

Viable counts of M. microti on various media under 114 different conditions.

Analysis of variance of levamisole and time on growth 121 of M. microti in CFLP mice

Proliferative responses of splenic T cells of uninfected 122 and infected mice before levamisole treatment

Proliferative responses of splenic T cells of uninfected 123 and infected mice after 2 weeks of levamisole treatment

Proliferative responses of splenic T cells of uninfected 124 and infected mice after 5 weeks of levamisole treatment

Splenic Thy 1.2+ T cells in infected and uninfected mice 127 before and after levamisole treatment

Splenic Lyt 1+ T cells in infected and uninfected mice 128 before and after levamisole treatment

Splenic Lyt 2+ T cells in infected and uninfected mice 129 before and after levamisole treatment

Total counts of M. tuberculosis after sonication 138

Analysis of variance of IFN-tf dose size and time on 143 growth of M. tuberculosis in BALB/c mice

3-way analysis of variance of the effect of IFN-^ dose 150 size, liposomes and time on M. tuberculosis infection in CFLP mice

3-way analysis of variance of the effect of IFN-V dose 155 size, liposomes and time on M. tuberculosis infection in BALB/c mice 13

TABLE 4.5 The effect of IFN-X pretreatment on the uptake of 157

M. tuberculosis in organs of BALB/c mice

TABLE 4.6 Analysis of variance of the effects of IFN-X pretreatment 161

on M. tuberculosis infection in BALB/c mice

TABLE 5.1 3-way analysis of variance of the effects of IFN-X*, 174

time and isoniazid on the growth of M. tuberculosis

in BALB/c mice

TABLE 5.2 3-way analysis of variance of the effects of isoniazid, 179

time and IFN-X given both before and after infection

of BALB/c mice with M. tuberculosis

TABLE 5.3 3-way analysis of variance of the effects of isoniazid, 183

time and IFN-ft given after infection of BALB/c mice

with M. tuberculosis

TABLE 5.4 3-way analysis of variance of the effects of IFN-X, 188

rifampicin and time on M. tuberculosis infection

in BALB/c mice

TABLE 6.1 The effect of saponin on viable counts of 196

L. monocytogenes

TABLE 6.2 The effect of saponin on viable counts of M. microti 197

TABLE 6.3 Analysis of variance of the effects of IFN-X dose size 199

and time on listericidal activity

TABLE 6.4 The effect of previous exposure to IFN-X on viable counts 208

of M. microti in monolayers in 8-chambered slides

TABLE 6.5 The effect of previous exposure to IFN-t# on counts of 210

acid-fast bacilli in macrophage monolayers

TABLE 6.6 3-way analysis of variance of in vitro effects of time, 219

isoniazid and IFN-X on M. microti infection of macrophages 14

TABLE 6.7 3-way analysis of variance of in vitro effects of time, 222

rifampicin and IFN-^ on the growth of M. microti in

macrophages

LIST OF ILLUSTRATIONS

Fig 3.1 Growth of M. microti in lungs and spleens of CFLP mice 116

Fig 3.2 Effect of levamisole on growth of M. microti in lungs of 119

CFLP mice

Fig 3.3 Effect of levamisole on growth of M. microti in spleens 120

of CFLP mice

Fig 3.4 Immunoalkaline phosphatase staining of cytocentrifuged 126

murine splenic T cells with monoclonal Thy 1.2 antibody

Fig 4.1 Effects of sonication on viable counts of mycobacteria 137

Fig 4.2 The effect of infective dose size of IFN-& on growth of 140

M. tuberculosis in lungs of CFLP mice

Fig 4.3 The effect of infective dose size of IFN-V on growth of 141

M. tuberculosis in spleens of CFLP mice

Fig 4.4 Effect of dose size of IFN-X on growth of M. tuberculosis 144

in lungs of BALB/c mice

Fig 4.5 Effect of dose size of IFN-V on growth of M. tuberculosis 145

in spleens of BALB/c mice

Fig 4.6 Viable counts of M. tuberculosis in lungs of CFLP mice 148

after administration of IFN-Jf in liposomes

Fig 4.7 Viable counts of M. tuberculosis in spleens of CFLP mice 149

after administration of IFN-^ in liposomes 15

Fig 4.8 Viable counts of M. tuberculosis in lungs of BALB/c 153

mice after administration of IFN-X in liposomes

Fig 4.9 Viable counts of M. tuberculosis in spleens of BALB/c 154

mice after administration of IFN-fc in liposomes

Fig 4.10 Viable counts of M. tuberculosis in lungs of BALB/c 159

mice after pretreatment with IFN-tf

Fig 4.11 Viable counts of M. tuberculosis in spleens of BALB/c 160

mice after pretreatment with IFN-Y

Fig 5.1 Effect of IFN-V and isoniazid alone and in combination 171

on the growth of M. tuberculosis in lungs of BALB/c mice

when IFN-Y was administered before and after infection

Fig 5.2 Effect of IFN-Y and isoniazid alone and in combination 173

on the growth of M. tuberculosis in spleens of BALB/c mice

when IFN-V was administered before and after infection

Fig 5.3 Effect of IFN-Y and isoniazid alone and in combination 176

on the growth of M. tuberculosis in lungs of BALB/c mice

in the first two days, when IFN-Y was administered before

and after infection

Fig 5.4 Effect of IFN-Y and isoniazid alone and in combination 177

on the growth of M. tuberculosis in spleens of BALB/c mice

in the first two days, when IFN-Y was administered before

and after infection

Fig 5.5 Effect of IFN-X and isoniazid alone and in combination 181

on the growth of M. tuberculosis in lungs of BALB/c mice

when IFN-Y was administered on Days +5 and +7 and

isoniazid from Day 0. 16

Fig 5.6 Effect of IFN-$ and isoniazid alone and in combination 182

on the growth of M. tuberculosis in spleens of BALB/c mice

when IFN-Y was administered on Days +5 and +7 and

isoniazid from Day 0.

Fig 5»7 Effect of IFN-J and rifampicin alone and in combination 185

on the growth of M. tuberculosis in lungs of BALB/c mice

Fig 5.8 Effect of IFN-Y and rifampicin alone and in combination 186

on the growth of M. tuberculosis in spleens of BALB/c mice

Fig 6.1A Wright’s (Diff-Quik) staining of macrophage monolayers 195

established in tissue culture 8-chambered slide for 2 days

Fig 6.IB Non-specific esterase staining of macrophage monolayers 195

established in tissue culture 8-chambered slide for 2 days

Fig 6*2 Effect of dose size of IFN-Y on listericidal activity of 198

peritoneal macrophages after prior exposure for 24 hours

Fig 6.3 Effect of varying the dose size and pretreatment period 201

of IFN-Y on listericidal activity of macrophages

Fig 6.4A Macrophage monolayers after 3 days incubation in BMM 204

Fig 6.4B Macrophage monolayers after 3 days incubation in BMM 204

plus 100 u IFN-Y per ml

Fig 6.5 Effect of dose size of IFN-Y on growth of M. microti 205

in macrophages after prior exposure for 72 hours

Fig 6.6 Effect of dose size of IFN-Y on growth of M. microti 206

in macrophages after prior exposure for 48 hours

Fig 6.7 Distribution of uptake of acid-fast bacilli by 209

macrophages after pretreatment with IFN-Y

Fig 6.8 Effect of previous exposure to IFN-Y on viable 212

intracellular M. microti at intervals during

the phagocytosis period 17

Fig 6.9 Effect of previous exposure to IFN-X on viable 213

intracellular M. microti at intervals during the phago­

cytosis period and their subsequent fate 24 hours later

Fig 6.10 Effect of IFN-)£ added after infection of macrophages 216

with M. microti

Fig 6.11 Effect of IFN-% and isoniazid alone and in combination 218

on growth of M. microti in macrophages

Fig 6.12 Effect of IFN-V and rifampicin alone and in combination 221

on growth of M. microti in macrophages 18

CHAPTER 1

LITERATURE REVIEW AND INTRODUCTION

I. LITERATURE REVIEW

1.1 Tuberculosis

Tuberculosis, once known as the great white plague, is an infectious disease of world-wide distribution. Records indicate that it is an ancient disease recognized by early physicians such as Hippocrates and archaeological evidence show that man has been afflicted with tubercu­

losis for many thousands of years (Keers, 1978). Over a hundred years ago, the causative organism was identified as Mycobacterium tuberculosis

(Koch, 1882) but despite the advances made in chemotherapy and vaccina­ tion, there are still 10 million new cases and 3 million deaths every year (World Health Organization, 1982).

1.1.1 The Mycobacteria

The mycobacteria are a group of non-motile, non-sporing, gram-positive rods that are relatively impermeable to basic dyes but once stained are able to resist decolourization with acids or acid-alcohol. Due to this characteristic property, the mycobacteria are often referred to as acid- fast bacilli. The mycobacteria (reviewed by Barksdale and Kim, 1977) comprise pathogenic as well as saprophytic species. The most important human pathogens, M. tuberculosis, M. bovis, M. africanum and M. leprae are strict pathogens usually transmitted from one person to another, but may cause disease in animals. The species which are occasional patho­ gens have also been called atypical or opportunistic mycobacteria. 19

These species have a saprophytic existence in water or soil, though

M. avium is also a pathogen for birds, pigs and some herbivores. These mycobacteria are less virulent than the strict pathogens for man, and occurrence of disease is thought to be due to host immune deficiency.

Other mycobacteria like M. phlei and M. smegmatis are strict saprophytes and do not cause disease at all.

M. tuberculosis, and probably all mycobacteria are obligate aerobes.

With the exception of M. leprae, M. lepraemurium and M. paratuberculo- sis, all mycobacteria can be grown on a variety of media including simple synthetic media. M. tuberculosis can be grown on very simple media containing only the NH^+ ion as nitrogen source, glucose or glyce­ rol as carbon source, magnesium, phosphates, iron and trace metals.

Growth is inhibited by the presence of fatty acids on the glass and cotton wool plugs which can be neutralized by the addition of adsorbing agents such as whole blood, serum, albumin, egg yolk or activated charcoal. All mycobacteria grow more slowly than most other bacteria.

The saprophytic species have generation times of 2-4 hours while the pathogenic species like M. tuberculosis have doubling times of 18-20 hours under optimal conditions. M. leprae which is highly adapted for parasitism, has a generation time of 12 days.

M. microti causes tuberculosis in voles, and its serological and patho­ genic properties suggest that it is very closely related to M. tubercu­ losis (Wells, 1946). Pathogenic studies have shown that if a large enough dose is used, M. microti will produce progressive and fatal disease in guinea-pigs, rabbits and mice (Corper and Cohn, 1943).

However, it lacks virulence in man and has been used in the form of a live vaccine as an alternative to BCG for vaccination (Medical Research 20

Council, 1956; Sula, 1958). M. microti has a distinctive morphology.

Pleomorphism is seen in vivo, and in culture media, the bacilli are often hooked or curved. It has a slightly slower growth rate than

M. tuberculosis and has been reported to be inhibited by glycerol, particularly in primary isolation (Wells, 1957).

Mycobacteria have cell walls with a unique chemical composition

(Barksdale and Kim, 1977). The mycobacterial cell wall is probably responsible for many of the special characteristics of the genus

including acid-fastness, resistance to host defence mechanisms and adjuvant activity. The cell wall is composed of a basal peptidoglycan structure (murein) covalently linked to an arabinogalactan-mycolate layer to form a complex polymer with a rigid framework which comprises about 60% of the cell wall.

A further 25% of the cell is composed of free lipids including Wax D, cord factor and sulpholipids. These lipids can be extracted with organic solvents and have been extensively investigated because of their diverse biological activities. Wax D is a monomer of the rigid frame­ work containing elements of both the murein and arabinogalactan-mycolate layers, which are thought to be autolysis products of the cell walls.

Cord factor (trehalose mycolate) is highly toxic to mice and destroys mitochondrial membranes. It is also found in the cell walls of M. phlei and M. smegmatis; thus while it may play a role in disease production, it is not the most important virulence factor. Sulpholipids are a group of multiacylated trehalose sulphate derivatives, some of which have been shown to enhance the toxicity of cord factor (Goren et al., 1974) and to be potent inhibitors of phagosome-lysosome fusion (Goren et al., 1976).

The sulpholipids (Goren et al., 1974) together with phospholipids 21 probably account for the binding of neutral red dye in the ‘Neutral Red* virulence test of Middlebrook and colleagues (1947).

At the surface of the cell wall, outside the arabinogalactan-mycolate layer are found various peptides including poly-L-glutamic acid which has been shown to inhibit phagolysosomal fusion in macrophages in vitro

(Hart and Young, 1978). In addition, there are rope-like structures at the surface of the cell wall of some species, but not M. tuberculosis, consisting of species-specific mycosides, which may protect the cell against macrophage lysosomal enzymes.

1.1.2 Pathogenesis of tuberculosis

Mycobacteria give rise to chronic granulomatous diseases in which the host immune response is often responsible for the nature and course of the disease. Even though the tubercle bacillus is highly infectious for most previously uninfected individuals, only a relatively small propor­ tion will develop clinical disease. This depends on the virulence and infectious dose of the strain, route of infection and also the innate and specific host immunity.

When an infection with M. tuberculosis occurs in a host with no previous experience with mycobacteria it is referred to as a primary infection

(Youmans 1979). In man, a primary infection is usually acquired by the inhalation of infectious droplet nuclei, and thus the lung is usually the first organ involved. The bacilli lodge within the alveolus and are phagocytosed by alveolar macrophages and neutrophils. Neutrophils are short-lived, highly phagocytic cells and are present in the initial stages of the infection when they engulf tubercle bacilli until they are 22 destroyed by the bacilli. Due to their high resistance to destruction, not all the mycobacteria are killed by the macrophages and some may mul­ tiply within them. As the bacilli multiply, large numbers of monocytes are drawn into the Ghon focus thus established. Dissemination from this focus then occurs by erosion of blood vessels and haematogenous spread throughout the body. Most of the disseminated bacilli will be taken up by the resident mononuclear phagocytes of the organs and will continue to multiply within those cells. In a small proportion of persons with primary tuberculosis this leads to post-primary tuberculous pneumonia, miliary tuberculosis or meningitis which may eventually prove fatal.

However, in the majority, after a period of a few weeks, the number of bacilli dramatically decreases. Many of the macrophages die, the bacilli start to grow extracellularly, and the healing processes begin, brought about by the onset of delayed type hypersensitivity and acquired cellular immunity.

Secondary or adult tuberculosis is the disease that occurs in persons who have previously been infected. It may occur as a result of endo­ genous reactivation or by reinfection. Secondary disease differs from primary disease in that it is characterized by necrosis and localization of the lesion which are due to tuberculin hypersensitivity and acquired immunity. Spread of this disease can occur by extension to adjacent tissues due to necrosis and in the lung, erosion of a bronchus or a blood vessel can lead to bronchogenic or haematogenous spread, respec­ tively. The reasons for the breakdown in immunity that may lead to secondary tuberculosis are not known. However, factors like age, hormo­ nes, immunosuppression and viral infections are known to predispose to the disease. 23

1.1.3 The immune response to tuberculosis

There is evidence that anti tuberculosis immunity is essentially of the cell mediated (CMI) type. Immunity to tuberculosis can be passively transferred by syngeneic lymphocytes but not by serum. This transfer does not occur if the lymphocytes are exposed to anti-0 serum and complement (Lefford, 1975). North (1973) has also shown that experi­ mental tuberculosis is uncontrolled in T cell depleted mice and that an infusion of thymocytes restores immunity. Undoubtedly, antibodies are produced against a variety of proteins and polysaccharides of the tubercle bacillus (Daniel and Janicki, 1978) but antibodies have been shown to be unimportant in host protection (Reggiardo and Middlebrook,

1974).

Antibacterial cell mediated immunity is based on interactions between macrophages and T lymphocytes. It is now known that the prerequisite for induction of CMI is that the antigen be processed by macrophages

(Bloch and Nordin, 1960; Pearson and Raffel, 1971; Oppenheim and Seeger,

1976) and then presented to T cells on the surface in association with products encoded by the major histocompatibility complex (MHC), the HLA-

D (Bodmer, 1977) or the la (Shreffler and David, 1975) alloantigens in humans or mice respectively. Recently it has also been shown that

Langerhans cells (Stingl et al., 1980) and dendritic cells (Steinman and

Nussenzweig, 1980) also act as antigen-presenting cells and could thus be involved in the CMI response. In mice, it has been shown that Lyt 1

T cells are the cells that recognize antigen in association with the H-2 la determinants presented by the macrophages (Cantor et al., 1976; Farr et al., 1979). Activation of T cells which also seems to require macro­ phage-derived interleukin 1 (Mizel, 1982), leads to lymphoproliferation. 24

At the peak of lymphoproliferation, histological tubercles begin to appear, the host exhibits delayed hypersenstivity to tuberculin and becomes immune to challenge with tuberculosis (North et al., 1972).

During these events, the draining lymph nodes becomes substantially enlarged. Initially, lymphoproliferation results in actively dividing large lymphocytes or immunoblasts which leave the lymph nodes and remain in the circulation for a few days before disappearing. Subsequently, a population of long-living, non-dividing, recirculating small lymphocytes appears within the circulation and these are the cells that mediate antituberculous immunity (Lefford et al. , 1973). These sensitized lymphocytes were shown to be T cells by North (1973). This sequence of events have been elucidated in animal experiments and it is probable that a similar sequence occurs in man.

The term ’lymphokine' (LK) was first used by Dumonde et al. (1969) to describe non-antibody soluble mediators of cellular immunity generated by lymphocyte activation. It has been proposed that during a CMI response, sensitized T lymphocytes interact with antigen and release LKs which mediate macrophage activation. This hypothesis is supported by in vitro experiments in which macrophages were activated by LKs (Simon and Sheagren, 1971; Nathan et al., 1971). The first LK described and the most intensively investigated is macrophage migration inhibition factor (MIF). Subsequently, it is estimated that up to one hundred factors may be produced and that macrophages and monocytes themselves produce soluble mediators as well (Waksman, 1979). The lymphokines include motility inhibitors like MIF, leukocyte inhibiting factor

(LIF); growth inhibitors like cytotoxic factors (CF), proliferation inhibitory factor (PIF); activators like macrophage activating factor

(MAF) , skin reactive factor (SRF) and other factors like chemotaxins, 25

interferon and transfer factor. In tuberculosis, as the bacilli are

multiplying in the macrophages, the factors that either mobilize or

activate macrophages are likely to be most important. The above func­

tions of the lymphokines have been investigated in vitro but their

interactions in vivo are not known.

In addition to lymphokines, macrophages can also be activated as a

result of interactions with microorganisms (Sher et al. , 1975) and microbial products like lipopolysaccharide and other cell wall deriva­

tives like muramyl dipeptide (Ellouz et a l ., 1974). In addition to

soluble factors, contact co-operation between macrophages and T cells

forms an autocatalytic system of mutual interactions in T cell mitoge- nesis (Peters and Schimmelpfeng, 1979).

Granulomas begin to appear at sites where tubercle bacilli are located.

The granuloma consists of a focus of mononuclear cells comprising macro­ phages, lymphocytes and epithelioid cells (Rich, 1951; Lurie, 1964).

Recruitment of mononuclear cells is mediated by the lymphokines produced by the sensitized T cells (Kuhner et al., 1980; Kaufman et al., 1981).

The macrophages and lymphocytes migrate into the lesion from blood while epithelioid cells are derived from macrophages. In granulomas in man, multinucleated giant cells derived from macrophages are present in addi­ tion to the above cell types. The epithelioid cells are weakly phagocy­ tic but are active in pinocytosis and extracellular secretion of diges­ tive enzymes and may play a role in extracellular killing of the bacilli

(Spector, 1976). Giant cells are also weakly phagocytic and their role is unknown. It has been suggested that the formation of giant cells is a means of disposing of altered or effete cells (Spector, 1976). Within 26 the granuloma, the cells are tightly packed together and this allows cell-to-cell interactions (Boros, 1978).

The macrophages in a granuloma differ morphologically and histochemical- ly from normal tissue macrophages, and the term 'activated macrophage' has been coined to describe them (Mackaness, 1962). Activation of macro­ phages refer to a series of morphologic, functional and biochemical changes (Karnovsky and Lazdins, 1978; Cohn, 1978). Activated macropha­ ges display a ruffled plasma membrane, an increased capacity for adher­ ing and spreading on a substratum, an increased capacity for phagocyto­ sis and microbicidal activity, and increased content of golgi apparatus, lysosomal acid hydrolases, secondary lysosomes (Grogg and Pearce, 1952;

Blanden, 1968; Blanden et al., 1969). In addition, it was later shown that activated macrophages also secrete large quantities of neutral proteinases including plasminogen activator, a specific elastase and a specific collagenase (Gordon, 1976).

One of the most important feature of activated macrophages is the enhancement of microbicidal activity. It has been shown in vitro that activated macrophages have an increased ability to kill intracellular bacteria like L. monocytogenes (Blanden et al., 1969), protozoa like

Leishmania tropica (Titus et a l . , 1984; Oster and Nacy, 1984) and

Toxoplasma gondii (Nathan et al., 1983). Walker and Lowrie (1981), demonstrated that murine peritoneal macrophages exposed to LK killed more than 90 % of ingested M. microti in 24 hours. Previous results had shown that LK-treated macrophages could not kill mycobacteria but inhi­ bited their multiplication (Patterson and Youmans, 1970; Godal et al.,

1971). 27

In a tuberculous infection, macrophages that have been activated will

not only specifically kill or inhibit the growth of tubercle bacilli but

will also non-specifically inhibit or kill other unrelated organisms

(Mackaness, 1964; Blanden et al., 1969) as well as tumour cells (Hibbs

et al., 1972).

In tuberculosis, hypersensitivity takes the form of a delayed allergic

response termed delayed type hypersensitivity (DTH) which is definded as

an immunological state in which lymphocytes and macrophages show a

sensitivity to tubercle bacilli and their products. There is still

controversy as to the relationship of acquired immunity to DTH. In

experimental tuberculosis, the onset of immunity coincides with the

appearance of DTH, and adoptive transfer experiments have confirmed that

T cells are the mediators of DTH (North, 1973). One hypothesis is that

acquired immunity and DTH are both manifestations of the same cellular

immune response (Mackaness, 1967; Mackaness and Blanden, 1967; Collins

and Mackaness, 1970). The other hypothesis is that DTH and acquired

immunity are merely coincidental events that are unrelated. This view

is supported by demonstrations that each phenomenon can be elicited by

different mycobacterial substances, and thus are independent responses

of the host tissue (Anacker et a l ., 1969; Berthrong, 1970; Youmans,

1975).

The complex system of immunologic responsiveness include feedback

mechanisms by which immunoregulation is achieved. Without feedback,

responses to antigens would be uncontrolled and potentially damaging.

Several types of immunocompetent cells with suppressor function have

been identified in mycobacterial infections. Using a model of mice heavily infected with mycobacteria, Watson and Collins (1980) reported 28 suppression mediated by T cells. Other cell types have also been shown to mediate suppression; adherent cells in spleens of mice heavily infected with BCG have been shown to inhibit cytotoxic T cell generation

(Klimpel and Henny, 1978). Ellner (1978) has shown that a population of adherent cells, presumably monocytes or macrophages, with suppressor activity, can be isolated from the peripheral blood of anergic tubercu­ losis patients. This suppressor activity was shown to be antigen- specific and to involve complex cell-cell interactions. It has also been reported that B lymphocytes can suppress the cellular immune resp­ onses of T lymphocytes to mycobacterial antigens (Bona et al., 1976).

It has long been recognized that an occasional tuberculosis patient is tuberculin negative (Adams et al., 1959; Katz et al., 1972). Loss of tuberculin hypersensitivity following chemotherapy or chemoprophylaxis of tuberculosis has also been reported (Robinson et al., 1955; Atuk and

Hunt, 1971). There is evidence that the loss of tuberculin hypersen­ sitivity can be due to sequestration of sensitized lymphocytes in lymph­ oid organs which is brought about by their removal from the circulation by 'blocking’ agents; either antigen, specific antibody or specific immune complexes (Schlossman et al., 1971).

It has been postulated that tuberculosis, like leprosy and visceral leishmaniasis is a disease with an immune spectrum based on clinical and immunological grounds. Lenzini et al.(1977) proposed four groups: a) a polar group in which cell mediated immunity is fully active (RR); b) an unreactive polar group in which cell mediated immunity is undetected

(UU); c) an intermediate group towards the reactive pole (RI) and d) an intermediate group towards the unreactive pole (UI). 29

1.1.4 Mycobacteria-macrophage interactions

The pathogenic mycobacteria cause diseases which are characteristically

chronic in nature. There are several prerequisites for establishing

chronic infections. The first is for the infectious agent to be of

sufficiently low toxicity so that the host can survive a prolonged state

of parasitism. Mycobacteria have not been shown to produce exotoxins,

endotoxins or harmful enzymes. Although toxic factors like cord factor

(Middlebrook et a l . , 1947) and other toxic lipids such as sulpholipids

(Goren et al. , 1974) and phthiocerol dimycocerosate (Goren et al.,

1974a) may account for some of the capacity of M. tuberculosis to cause

progressive disease, mycobacteria are considered to be of low toxicity.

There is little or no sign of toxicity when large numbers of live

M. tuberculosis are injected intravenously into mice (Youmans and

Youmans, 1951).

The slow growth rate of pathogenic mycobacteria is another factor in its

success as a chronic pathogen. Another criterion is the ability of the

pathogenic mycobacteria to lie dormant for many years. Although anaero­

bic conditions inhibit growth, it has been shown that M. bovis sealed in

ampoules of veal broth incubated at 37°C could survive for as long as 30

years (Corper and Cohn, 1951).

An important prerequisite for chronic infections is an ability of the

infectious agent to avoid host defence mechanisms. Mycobacteria achieve

this by surviving in macrophages. Unlike neutrophils, macrophages are long-lived cells. Hence, persistence in macrophages would ensure survi­ val in the host for long periods. Intracellular persistence in macro­ phages could be due to bacterial subversion of the killing mechanisms of 30

macrophages or due to genetically-determined impaired killing mechanisms

of macrophages.

1.1.4.1 Microbicidal mechanisms in macrophages

In macrophages, two main bactericidal mechanisms operate; oxygen-

independent ones such as lysozyme, hydrolases, cationic proteases,

acidity; and oxygen-dependent ones such as superoxide, hydrogen peroxide

and hydroxyl radicals. It is still not certain how macrophages kill mycobacteria. Exposure to peroxide and related oxygen derivatives may occur in the phagosome after ingestion, while exposure to lysosomal enzymes requires phagosome-lysosome fusion. Live M. tuberculosis and

M. microti have been shown to inhibit phagolysosome formation and multi­ ply within phagosomes (Armstrong and Hart, 1971; Lowrie et al., 1975).

Other successful intracellular parasites have also been found to inhibit phagolysosome fusion (Jones and Hirsch, 1972; Friis, 1972; Weidner,

1975) and it was shown with Toxoplasma gondii that coating the parasites with antibody before phagocytosis led to promotion of fusion and death of the parasites (Jones, 1975). However, the results were less obvious with M. tuberculosis and M. microti. Coating M. tuberculosis with anti­ bodies promoted phagolysosome fusion with no effect on bacterial growth

(Armstrong and Hart, 1975) and coating M. microti with antibody also led to phagolysosome fusion with occasional stasis (Lowrie et al., 1979).

These results suggest that the lysosomal contents might not be that important in the killing of mycobacteria. However, there is the possi­ bility that phagolysosomal fusion might be lethal to mycobacteria in the intact animal. 31

Lysozyme is present in large quantities in macrophages and is involved

in the killing of gram-negative organisms. However, many organisms

including mycobacteria are resistant to degradation by lysozyme. It has

been suggested that lysozyme may have tuberculostatic properties (Oshiraa

et al., 1961).

Another possible microbicidal mechanism is the acidity of the macro­

phage. It has been known for a long time that phagocytes respond to the

ingestion of particles by acidifying their phagosomes. Early studies

using indicator dyes led Rous (1925, 1925a) to conclude that the intra-

vacuolar pH could be as low as 3.0. Subsequently, Sprick (1956) showed

that phagocytosis of M. tuberculosis and M. smegmatis led to a fall in

pH to 4.7-5.2. Jacques and Bainton (1978) showed that within 10 minutes

of phagocytosis, the pH of neutrophils and monocytes fell to 4.5-5.0.

Recent studies using a pH-sensitive fluorescent probe to measure intra-

vacuolar pH have shown that the pH in newly formed phagosomes in

neutrophils (Segal et al., 1981) and murine macrophages (Geisow et al.,

1981) transiently became alkaline in the first two minutes before the

acidification and that the raised pH is associated with the respiratory

burst. Segal et al. (1981) also postulated that the alkaline pH facili­

tates killing and bacteriolysis by granule proteins with alkaline pH

optima following which the pH of the vacuole is reduced to optimize the activities of hydrolases and other proteins of acidic pH optima. The acidity per se may be bactericidal for killing some organisms like

Vibrio cholerae (Looke and Rowley, 1962), but it is more likely that the acidity contributes to other killing mechnisms. It has been shown to enhance hydrogen peroxide toxicity for tubercle bacilli (Jackett et al.,

1978). Acidity also promotes the rate of superoxide reduction to hydrogen peroxide (Stossel, 1974). 32

The oxygen-dependent mechanisms have been established as important bact­ ericidal mechnisms in neutrophils (Klebanoff and Hamon, 1975). Interme­ diate products of oxidative phosphorylation like hydrogen peroxide and singlet oxygen have been implicated in bactericidal mechanisms. The first indirect evidence that hydrogen peroxide might be involved in the killing of tubercle bacilli was the isolation of isoniazid resistant mutants of M. tuberculosis which were catalase negative, susceptible to hydrogen peroxide and of low virulence for the guinea-pig (Barnett et a l . , 1953; Cohn et al., 1954; Mitchison, 1954). In the early 1960's it was noticed that a substantial proportion of clinical isolates of

M. tuberculosis in the Indian subcontinent were susceptible to peroxide and isoniazid and of low virulence in the guinea-pig but had the normal content of catalase (Subbaiah e t a l . , 1960; Mitchison et al., 1963).

Subsequently, Walker and Lowrie (1981) showed that the killing of

M. microti was due to macrophage hydrogen peroxide. This was indicted by the protective effect of exogenous catalase and the finding that phagocytosis of M. microti was accompanied by a release of hydrogen peroxide in parallel with the uptake of the bacilli. Another piece of evidence is the correlation of susceptibility of M. tuberculosis strains to hydrogen peroxide and diminished virulence for guinea pigs (Jackett et al., 1981).

Much of the hydrogen peroxide produces by mononuclear phagocytes is derived from superoxide (Rossi et al., 1979). However, in a study with strains of M. tuberculosis of different virulence in guinea-pigs there was no correlation between virulence, resistance to superoxide and content of superoxide dismutase (Jackett, et al., 1978). Thus, supero­ xide, hydroxyl radical and singlet oxygen are unlikely to have any direct role in tuberculocidal activity in macrophages. 33

Another factor that might be important within a granuloma is reduced oxygen tension. The development of infections with M. tuberculosis is impaired under conditions where oxygen availability is restricted

(Dubos, 1955; Sever and Youmans, 1957; Chandler, 1965). It is unlikely that only a single tuberculocidal mechanism operates in a macrophage.

It may be that the macrophage mycobactericidal mechanisms depend on a combination of acidity, hydrogen peroxide and low oxygen tension.

1.1.4.2 Mycobacterial evasion of macrophage killing mechanisms

Following ingestion, pathogenic mycobacteria have been found to occur in three sites within the host cell; the phagosome, the phagolysosome or the cytoplasm. Live M. tuberculosis and M. microti have been shown to multiply in the phagosome, M. lepraemurium can be found multiplying within the phagolysosomes (Hart et al., 1972) while M. leprae is belie­ ved to escape from the phagosomes into the cytoplasm (Evans and Levy,

1972). These sites may be interpreted as indicating three ways of avoiding intracellular killing.

At least four substances are known that may, in appropriate conditions, be produced by M. tuberculosis and which will inhibit phagosome-lysosome fusion in cultured mouse peritoneal macrophages. These substances are cyclic AMP (Lowrie et al., 1975; Lowrie et al., 1979); poly-<<-L-glutamic acid (Hart and Young, 1978); sulphatide (Goren et al., 1976); and ammo­ nia (Gordon et a l . , 1980). It is still not clear which, if any, of these substances is the active material inside the host cell infected with M. tuberculosis. 34

It would seem that M. lepraemurium Is indifferent to the effects of phagolysosomal contents as it may be found multiplying in phagolysosomes of macrophages in vitro (Hart et a l . , 1972) and in vivo (Brown and

Draper, 1976). It has been proposed that the surface structures of

M. lepraemurium are unaffected by lysosomal enzymes and impermeable to bactericidal chemicals due to a protective capsule of a characteristic peptidoglycolipid (Draper and Rees, 1970; Nishiura et al., 1972). Such a capsule is also produced by M. avium (Draper, 1974).

Mouse foot-pad experiments have suggested that M. leprae escapes into the cytoplasm through a phagocytic vacuole (Evans and Levy, 1972). It has also been reported that some rickettsias (Silverman and Wisseman,

1979) and Trypanosoma cruzi (Nogueira, 1974) use the same route. The mechanisms involved are uncertain but once established in the cytoplasm, the pathogen is now removed from killing mechanisms. The lack of cyto­ toxicity and extremely slow growth rate make M. leprae particularly suited for such intracellular existence.

In addition to the above mechanisms, mycobacteria could also modulate the host response by depressing cellular immune responses. The trigger­ ing of immune suppression mechanisms is probably a specific response to antigen. Immunopotentiating or adjuvant properties of mycobacteria are well known and studies have identified muramyl dipeptide (Ellouz et al.,

1974) and trehalose mycolates (Goren, 1975) as the mycobacterial cell wall components with adjuvant activity. Mycobacteria have also been reported to contain immunosuppressive substances. Ellner and Daniel

(1979) claimed that mycobacterial arabinomannan is immunosuppressive in vitro, and there are indications that mycobacterial cell wall arabinogalactan is a potent immunosuppressive agent (Kleinhenz et al., 35

1979) . Neta and Salvin (1979) have also shown that mycobacterial cells in Freund's complete adjuvant induced the formation of both suppressor adherent cells and suppressor B lymphocytes.

1.1.4.3 Genetic susceptibility

The heterogeneity of macrophages is well established. Mature macroph­ ages are more bactericidal than blood monocytes (Gemmell et_al., 1981) and sub-populations of murine peritoneal macrophages vary in their ability to restrict intracellular growth of L. monocytogenes (Harring- ton-Fowler and Wilder, 1982). Thus there are clearly populations of macrophages where intracellular bacteria can survive. In addition there is the possibility of genetic deficiencies adversely affecting the micr­ obicidal ability of macrophages. It has been shown in mice that geneti­ cally determined susceptibility to infections occur (Cheers et al.,

1978; Skamene and Kongshavn, 1979). Further studies have identified the macrophage as the cell population that expresses the phenotype of genet­ ically determined resistance to BCG infection (Stach et al., 1984). It has also been noted that the monocytes and macrophages in patients with chronic granulomatous disease are less efficient at microbicidal func­ tions (Davies et al., 1968). 36

1.2 Tuberculosis control

The annual risk of tuberculous infection which can used to study the epidemiology of tuberculosis, is estimated by using serial tuberculin conversion rates. In developed countries like the Netherlands, the annual risk of tuberculous infection since 1940 has closely followed an exponential downward trend, the risk decreasing annually by 13.8%. From

1913-1939, the risk was shown to decrease exponentially as well, with an annual decrease in risk of 5.5% (Styblo, et al., 1969). Before 1940, chemotherapy was not available, mass BCG vaccination and was not applied, and could not have contributed towards the decrease.

Hence, the exponential decrease before 1940 appears to have resulted from factors like improvements in socioeconomic conditions and isolation of infectious cases in sanatoria. The steeper decrease after 1940 has been attributed to compulsory pasteurization of milk and the introduc­ tion of effective chemotherapy (Styblo, et al., 1969).

The situation is rather different in poor developing countries. Similar tuberculin surveys have failed to show any significant reduction in the annual risk of tuberculous infection. In Uganda, the annual risk of infection (at age 10) was 2.8% in 1940, 2.6% in 1950, 2.4% in 1960 and

2.3% in 1970 (Stott et al. , 1973). In Lesotho, the annual risk of infection was 3% in 1957 and was unchanged in 1965 (Styblo, 1980). The current epidemiological trend in most poor developing countries is an annual risk of tuberculous infection of 2-5% about 20-50 times greater than in technically advanced countries (World Health Organization,

1982). 37

Methods of tuberculosis control have consisted of 1) BCG vaccination, 2)

case-finding and chemotherapy, and 3) chemoprophylaxis. The decline in

tuberculosis rates achieved in Eskimos in Alaska, Greenland and Canada

showed that it is possible through intensive antituberculous programmes

to reduce the tuberculosis problem rapidly in a community (Grzybowski

et a l . , 1976). Eskimos had extremely high rates of tuberculosis and in

Alaska and Greenland, the problem was reduced by 90% in 15 years. The

common feature of the control programmes in all three countries was

intensive case-finding followed by thorough chemotherapy. BCG was also

extensively used in Greenland, chemoprophylaxis in Alaska, and both BCG

and chemoprophylaxis in Canada. However, the greatest contribution was

intensive case-finding and treatment which resulted in extremely rapid

decrease of risk of infection.

1.2.1 BCG Vaccination

Despite enormous efforts to develop a non-viable anti-tuberculous

vaccine for clinical use, live BCG still remains the only practical

means of immunization. Previous attempts to immunize with killed whole

mycobacteria have shown that killed vaccines were inferior to viable

vaccines (Bloch and Segal, 1955). However, large doses of -killed

tubercle bacilli were shown to produce an immunity almost comparable to

that seen with live organisms (Acharya et al., 1958). In recent years,

three killed vaccines have been extensively investigated. These are the

mycobacterial ribosome vaccine (Youmans and Youmans, 1969), mycobacte­

rial cell wall vaccines (Ribi et al., 1966, 1971) and trypsin-extracted

mycobacterial antigen vaccine (Crowle, 1972). None of these vaccines

were more efficacious in immunization than live BCG and more importan­ 38

tly, none were suitable for clinical use.

Live BCG was first administered orally to a child in Paris by Weill-

Halle in 1925 with no untoward effects (cited by Collins, 1984) and the use of BCG vaccination spread across Europe and to America. Initially, it was administered orally, then by the sub-cutaneous route which was dropped in favour of the intra-dermal route. On the basis of clinical observations and uncontrolled trials, by 1945, 100 million people had been vaccinated. After World War II, eight major controlled trials have been carried out in Europe and the United States with results ranging from 0-80% protection (Barksdale and Kim, 1977). A trial in American

Indians showed 80% protection (Stein and Aronson, 1953), another trial in British school children gave 78% protection (Medical Research

Council, 1972) while a trial in Georgia showed no protection at all

(Comstock and Palmer, 1966). Several reasons have been put forward to explain the differences, among them the quality of the vaccine and partial protection due to environmental mycobacteria. The Chingleput,

South India trial was started in 1968 in an attempt to assess the influence of environmental mycobacteria on BCG vaccination. After seven and a half years follow-up, the trial had yet to show any protection

(World Health Organization, 1980, 1980a). Whatever the real reasons for the disparate results of the BCG trials, they are likely to be multifac­ torial and complicated.

The current view of BCG vaccination is that it primarily protects the individual without much effect on disease in the community. The communal effect of BCG vaccination was studied by Styblo and Meijer

(1976). They showed that the decreases in the annual risk of infection 39

in Denmark and Norway, where mass BCG was carried out, were similar to

that of the Netherlands, where mass BCG was never carried out. In

detailed comparisons of the age-related incidence in these countries,

the direct protective effect of BCG on the incidence in the individuals vaccinated could be demonstrated whereas they found no indirect effect within the community as a whole. It was also evident from the

tuberculosis control programmes in Eskimos that mass BCG applied at the

ages of 15-30 did not substantially influence the chain of transmission

(Grzybowski et al., 1976).

Another important factor about BCG vaccination is the duration of immun­

ity. It has been shown that the protective effect of BCG decreases with

time from 84% at 0-5 years to 59% at 10-15 years and down to -12% at 15-

20 years (Hart and Sutherland, 1977). This downward trend is signifi­

cant at the 1% level.

1.2.2 Case-finding and Chemotherapy

Case-finding and chemotherapy is considered as an entity, as case­

finding is a preliminary to treatment and cure. The importance of case­

finding and chemotherapy in tuberculosis control is readily seen in the

successful anti-tuberculosis programmes in Eskimos in Alaska, Greenland and Canada where the rapid decrease in risk of infection was attributed

to intensive case-finding and thorough chemotherapy (Grzybowski et al.,

1976). In Alaska, the risk of infection fell some 100 times in 25 years. Case-finding and chemotherapy is now considered the most power­ ful component of tuberculosis control (World Health Organization, 1982). 40

1.2.2.1 Case-finding

Case-finding may be either passive or active. Passive case-finding depends on patients reporting with symptoms and is essential in any health care programme. Passive case-finding tactics are: 1) detection of chronic cough (greater than 4 weeks duration) in patients attending clinics or hospitals; and 2) re-directing such symptomatic patients for direct smear examination of the sputum. Studies in the developing coun­ tries have shown this method to be highly effective provided there are adequate facilities for sputum examination. Active case-finding through mass radiography has now largely been abandoned due to the high costs, the rapidity with which new cases develop and the small contribution to the total number of cases detected (Meijer et al. , 1971). It has also been shown that active case-finding by mass tuberculin testing of unvaccinated individuals does not contribute significantly to the number of cases detected (Van Geuns et al., 1975). Active case-finding in high risk groups, such as household contacts of newly diagnosed tuberculous infections should continue to be employed.

In developing countries cases of tuberculosis are diagnosed mainly by direct smear examination of the sputum. In technically advanced coun­ tries, culture and radiography are used, enabling the detection of smear-negative disease which might account for 50-60% of all newly diagnosed cases of pulmonary tuberculosis. The introduction of culture service in developing countries could significantly improve case-finding and might increase the overall efficiency of the service by facilitating more accurate diagnosis. However, the organization of a culture service is technically complex and relatively expensive and might divert 41

resources from the direct-smear service in the poorer countries.

1.2.2.2 Chemotherapy

Ever since Koch discovered the etiological agent of tuberculosis, there have been attempts to find suitable chemotherapeutic agents. Thiaceta- zone was the first agent with any efficacy to be discovered and was extensively investigated in clinical trials in Germany (Domagk, 1950), but the toxicity associated with the large doses used discouraged wide­ spread use of the drug (Hinshaw and McDermott, 1950). Streptomycin was

the first chemotherapeutic agent effective for the treatment of tubercu­ losis in man (Schatz and Waksman, 1944; Feldman and Hinshaw, 1944).

Among the important events in the three decades following the discovery of streptomycin was the discovery that the emergence of drug resistance could be prevented by using a combination of antituberculous drugs

(Medical Research Council Investigation, 1950; Tempel et al., 1951); the introduction of isoniazid, an inexpensive, relatively non-toxic and highly effective antituberculous agent (Robitzek et al., 1952); the demonstration that similar results were obtained in patients treated in

their homes or in a sanatorium with isoniazid and para-aminosalicylic

acid for one year in Madras (Dawson et al., 1966) which brought about ambulatory treatment; the demonstration of the efficacy of intermittent

chemotherapy (Tuberculosis Chemotherapy Centre, Madras, 1964); and the

discovery of the higher sterilizing activity of rifampicin and pyrazin-

amide when combined with isoniazid, which enabled the shortening of

chemotherapy. 42

Standard regimens comprising various combinations of streptomycin, isoniazid, rifampicin, ethambutol, para-aminosalicylic acid and thiacet- azone given for 18-24 months are theoretically capable of producing a long and lasting cure in all patients. However, a review of the bacte­ rial and non-bacteriological failures of a standard regimen in 1968-1970 in Scotland showed that even in a technically advanced country, there are still practical problems in achieving a lasting cure with the standard regimens (Heffernan et al., 1976). One of the main problems of the standard regimens is non-compliance. In previously untreated patients, it has been estimated that the failure rate attributed to non- compliance is 2% under optimal conditions in technically advanced countries and as high as 50% in poor developing countries (Mitchison,

1980). Two major developments in chemotherapy had been directed at reducing this problem; fully supervised intermittent chemotherapy and short-course chemotherapy.

Short course chemotherapy is based on a combination of appropriate sterilizing drugs and was developed mainly as a consequence of a series of observations from studies on experimental chemotherapy in the mouse and guinea-pig, and in vitro studies (reviewed by Fox and Mitchison,

1975; Grosset, 1978) . Sterilizing activity is taken to mean the killing of the last few persisting organisms in a lesion during effec­ tive chemotherapy. In experimental tuberculosis, sterilizing activity is measured as the ability to prevent growth of bacilli from the organs of animals after 3-6 months chemotherapy and to prevent subsequent relapse, especially after immunosuppression with steroids. The main facts that emerged from these studies were: 1) rifampicin and 43

pyrazinamide were the two most potent sterilizing drugs, 2) rifampicin

and pyrazinamide were most effective when combined with isoniazid, 3)

the addition of streptomycin or ethambutol did not improve its sterili­

zing effect. These conclusions were consistent with findings of short-

course chemotherapy trials conducted on pulmonary tuberculosis patients

in several countries under the auspices of the British Medical Research

Council (reviewed by Fox and Mitchison, 1975).

These findings led Mitchison (1980) to hypothesize the existence of four

special bacterial populations within tuberculous lesions. The first

consists of bacilli growing relatively rapidly and is susceptible to

bactericidal drugs. This is supported by evidence of a very rapid fall

in viable counts of tubercle bacilli in patients in the first few days

after chemotherapy (Jindani et a l . , 1980). Comparisons of different

regimens in these first few days showed that with single drugs, isonia­

zid was the most bactericidal drug and that the contribution of the

other antituberculous drugs given in combination with isoniazid is

limited.

The second population consists of bacilli which are slowly metabolizing within the acid environment of the macrophages or in areas of acute

inflammation, and which are killed selectively by pyrazinamide. This is

supported by evidence that pyrazinamide is shown to be weakly bacteri­

cidal in vitro at a pH of 5.6 or less (McDermott and Tompsett, 1954;

Dickinson and Mitchison, 1970). The third population of slowly metabo­

lizing bacilli with spurts of metabolic activity would be effectively killed by rifampicin which is unique in the speed with which its bacte­ ricidal activity starts (Dickinson and Mitchison, 1981). 44

The fourth population of dormant bacilli are not known to be killed by any drug. This population has been shown to exist in experimental murine chemotherapeutic models where latent bacilli have been stimulated to grow by the administration of steroids in the post-chemotherapeutic period (McCune et al., 1956; Grumbach, 1975). The bactericidal mecha­ nisms on the first three populations during chemotherapy probably all

commence at the begining of chemotherapy (Dickinson and Mitchison,

1981). This assumption explains the delayed bactericidal activity that

becomes evident only months after an initial period of treatment with

rifampicin in experimental chemotherapy in mice (Grumbach et al., 1969)

and guinea-pigs (Dickinson and Mitchison, 1976).

Antituberculous drugs can be graded according to their ability to prevent the emergence of drug resistance, for their early bactericidal

activity or for their sterilizing activity (Mitchison, 1985). These

three functions are often unrelated, and the rating for one function

often has no relationship to the rating for another function. Isoniazid has a high activity for preventing the emergence of drug resistance and

for early bactericidal activity; rifampicin has high activity for preve­

ntion of drug resistance and sterilizing activity; and pyrazinamide has

a high sterilizing activity but is only moderately effective in preven­

ting drug resistance, and has virtually no early bactericidal activity.

In Britain, a standard 9 month daily regimen has been adopted consisting

of rifampicin and isoniazid supplemented with ethambutol for the first

three months. This has been shown to be highly effective with almost no

relapses (British Thoracic and Tuberculosis Association, 1976; British

Thoracic Association, 1980). Later studies have shown that the 45

duration of the regimens could be shortened to 6 months by the adminis­

tration of pyrazinamide in addition to isoniazid, rifampicin and either

ethambutol or streptomycin in the initial phase followed by isoniazid

and rifampicin for the duration (Singapore Tuberculosis Service/British

Medical Research Council, 1981; British Thoracic Association, 1982).

There is increasing evidence that there are a variety of highly

effective 6 month regimens (reviewed by Fox, 1985). All these regimens

have in common isoniazid, rifampicin and pyrazinamide initially and

isoniazid and rifampicin in the continuation phase. Some regimens are

daily throughout, some are administered thrice or twice weekly, and some

combine both daily and intermittent regimes. Some regimens include

ethambutol and some include streptomycin.

There have been attempts to reduce the duration of short-course regimens

to less than 6 months in several countries (reviewed by Fox, 1981,

1985). The latest results have confirmed that 6 month regimens had

similar relapse rates as 9 month regimens of 1%. However, 4.5-5 month

regimens had relapse rates of 4%, 4 month regimens of 12% and 3 month

regimens of 16% (Fox, 1985). It still remains remarkable that a large

proportion of patients are 'cured’ by only 3 months treatment.

1.2.3 Chemoprophylaxis

Chemoprophylaxis can be considered as a form of treatment for an

infection that has not yet occurred, is just beginning, or is in an asymptomatic subclinical state (Hoeprich, 1972). Chemoprophylaxis for

tuberculosis only became possible with the availability of a safe, 46

inexpensive oral drug, isoniazid.

In 1955, the United States Public Health Service reported on trials with guinea-pigs which showed that prophylactic isoniazid could convert a massive inoculation of virulent tubercle bacilli into a controlled benign tuberculous infection (Ferebee and Palmer, 1956). This was foll­ owed by 13 controlled trials in 7 countries involving nearly 100,000 participants (reviewed by Ferebee, 1970). Seven trials were conducted in the United States, one each in Greenland, Tunisia, Japan, Philip­ pines, Kenya and the Netherlands. While the results of the various trials differ, several general conclusions were reached. Tuberculin testing at the begining and the end of trials in Tunisia, Kenya and

United States indicated that isoniazid reduced the frequency of tubercu­ lin conversions but did not have much impact on established tuberculin reactivity. The degree of protection also varied from statistically insignificant differences in some of the smaller studies to excellent results in larger studies. Isoniazid produced the largest effects within the treatment year, but there were indications that the effect was still present in the post-treatment period. The results also showed that adverse reactions to isoniazid were uncommon among healthy persons but increased with age and gastrointestinal problems and that bacterial drug resistance was not a problem.

The current recommended duration of chemoprophylaxis is 12 months.

However, a trial conducted by the International Union against Tubercu­ losis Committee on Prophylaxis (1982) showed that while 52 weeks of isoniazid prevented the most tuberculosis, a 24-week regimen would decrease hepatitis by one-third and increase tuberculosis by 40%. 47

Chemoprophylaxis can prevent the development of tuberculosis in infected

individuals but its impact on the community will be minimal due to the

problems of application on a mass scale, even in technically advanced

countries.

1.3 Immunomodulation and immunodulating agents

There are numerous biological and chemical agents which have been shown to influence the immune system. The response elicted by these agents have been described by terms such as immunomodulation, immunostimula- tion, immunopotentiation, or immunorestoration. There have been reports that immunomodulating agents may synergise with chemotherapeutic agents.

An example of synergism has been the use of levamisole as an adjunct to antituberculous therapy (Yaseen et al., 1980; Singh et al., 1981).

The importance of modulating the immune response by chemical or biolo­ gical agents has recently been recognized in two areas of medicine: oncology and infectious diseases. In contrast with oncology, experi­ mental and clinical data referring to infectious diseases have been less abundant. This is not surprising as most bacterial diseases have been effectively controlled by antibiotics and synthetic antibacterial drugs.

However, it is an undeniable fact that even the most effective antibact­ erial agent needs the additional activity of the host's immune response.

This is seen most clearly in cases of primary immune deficiencies like congenital thymus aplasia and congenital agammaglobulinaemia where infections are difficult or impossible to treat even with the most effective antimicrobial agents. 48

One of the main problems with the use of immunomodulating agents has been the selection of the dose size and timing of doses which has largely been empirical. The difficulties have arisen from the unusual dose-response curve for immunotherapeutic agents which differ from that of the usual chemotherapeutic agents. Both the timing of administration and the dose have been shown to be critical in the effectiveness of BCG against tumours (Hawrylko and Mackaness, 1973). Levamisole has also been shown to have a time and dose-dependent efficacy against a variety of animal tumours (Mantovani and Spreafico, 1975; Fidler and Spitler,

1975). Davies (1983) suggested the existence of phase variations in the modulation of the immune response by various agents including BCG, endotoxin and ubiquinone. He showed that small changes in doses produced profound changes in the response and suggested that the existence of biphasic and multiphasic variations may have been undetected in many instances because the dose intervals chosen are often too large.

The development of safe immunomodulators for clinical use has become a major target for many drug companies. Immunomodulators can be divided into three categories: 1) agents derived from microorganisms, 2) agents derived from mammalian cells and 3) synthetic chemicals.

1.3.1 Agents derived from microorganisms

In the past, bacterial products have been the agents most extensively investigated. These have been shown to have potent immunomodulating activity but the presence of lipopolysaccharide (LPS) has limited the clinical use of preparations of bacterial origin. Mycobacteria and 49

their products have been known for years to possess strong immuno- adjuvant properties. Subsequently, a number of other effects have been reported including stimulation of the reticuloendothelial system (RES) and enhancement of cell-mediated immunity (Howard et al., 1959; Sher et al., 1975). BCG has been one of the first immunomodulating agents to be extensively investigated in experimental tumours (Old et al., 1959;

Mathe et al., 1969; Zbar et al., 1971) and in human cancer (reviewed by

Laucius et al., 1974; Bast et al., 1974).

Other microorganisms shown to have strong immunomodulating properties are Corynebacterium parvum and C. granulomatosum. The ability of C. parvum to strongly activate the RES was recognized over twenty years ago

(Halpern et a l . , 1964; Neveu et a l . , 1964) and has subsequently been shown to have antitumour properties (Scott, 1974; Oettgen et al., 1976), and the ability to enhance resistance to bacterial infections (Cronly-

Dillon, 1974). Killed C. parvum has been used in clinical trials in cancer patients (Israel, 1977) with the attendent problems of crude bacterial products.

Immunomodulating properties have also been attributed to components from other microorganisms including Pseudomonas aeruginosa (Mathe et al.,

1977); Brucella abortus (Glasgow et al., 1979), and Klebsiella pneumonia

(Griscelli et al., 1982).

Glucans, naturally occurring substances in bacteria, yeasts, fungi and higher plants have been known to be powerful RES stimulators and to play important roles in host defences. They have been tested for possible clinical usefulness (Mansell et al., 1976) and have been reported to be 50

effective against infections with some microorganisms (DiLuzio and

Williams, 1978; Williams et al., 1978) and against some tumours (Stewart

et a l ., 1978; DiLuzio et al., 1979). Lentinan is a completely purified

neutral polysaccharide extracted from an edible mushroom and has been

shown to have potent antitumour properties (Shiio and Yugari, 1980) and

the ability to enhance host resistance to M. tuberculosis (Usuda et al.,

1981; Kanai and Kondo, 1981).

Ubiquinones (Coenzyme Q), important participants in the oxidation-

reduction reactions of the mitochondrial respiratory chain, have been

reported to induce host-specific resistance to a number of bacterial and

viral infections (Block et al., 1978). Another microbial component is

bestatin, a metabolite dipeptide of Streptomyces olivoreticuli which has

been shown to stimulate both humoral and cellular immunity in vitro and

in vivo (Blomgren, 1980).

1.3.2 Agents derived from mammalian cells

Among the immunomodulating agents derived from mammalian cells are

thymic hormones, dialysable leukocyte extracts (transfer factor),

tuftsin and interferons which will be described separately in another section.

Many factors with thymic hormone-like activity have been isolated and described including thymosin fraction 5 (Hooper et al., 1975), thymopo­ ietin (Goldstein, 1975), serum thymic factor (Bach and Carnaud, 1976) and thymus humoral factor (Kook et al., 1975). These factors have been shown to promote T cell differentiation (Goldstein et a l ., 1978; 51

Trainin, 1974) and to promote antitumour effects in thymectomized mice

(Bach, 1977). Thymosin fraction 5 was the first well-defined thymic hormone preparation to be used clinically in patients with primary

immunodeficiency diseases (Wara et al., 1975; Barrett et al. , 1980) and advanced cancers (Chretien et al., 1978; Cohen et al., 1979).

Transfer factor (TF) prepared by a dialysis method (Lawrence, 1955) and shown to transfer delayed hypersensitivity from an immune to a non- immune person, is in fact a crude dialysable leukoycte extract (DLE) which has now been shown to contain several hundred chemical moieties

(Wilson and Fudenberg, 1983). Consequently, DLE is the current designation for such preparations and the term TF is now reserved for the components with antigen-specific activity. Crude DLE has been shown to contain non-specific immunomodulatory activity in addition to TF activity (Wilson et a l . , 1980). DLE has received widespread clinical use and the results obtained in some viral, fungal and other diseases have been striking (reviewed by Arala-Chaves et al., 1978).

Tuftsin is a hormone-like tetrapeptide which has been isolated from the

Fc portion of IgG. It has been shown to be the physiological stimulator of motility, phagocytosis and pinocytosis for all macrophages and blood neutrophils (Najjar, 1974); and has also been shown to possess anti­ tumour properties (Nishioka, 1979). 52

1.3.3 Synthetic immunomodulating agents

The first synthetic immunomodulating agent to be extensively investi­ gated was levamisole, which will be described in a later part of the literature review (section 1.5).

Another synthetic molecule that has been extensively examined is N- acetylmuramyl-L-alanyl-D-isoglutamine or muramyl dipeptide (MDP). MDP is a small molecular weight subunit of the mycobacterial cell wall peptidoglycan which can be used in place of whole mycobacteria for immunoadjuvant activity (Ellouz et al., 1974). However, the pyrogeni- city of MDP has been a major obstacle for clinical applications

(Dinarello et al . , 1978; Rotta et a l . , 1979). Subsequently, several hundred compounds containing the muramyl moiety have been synthesized and tested and some have been found to be superior to MDP as immuno- modulators. The N-butyl-ester of MDP has been shown to be apyrogenic and to enhance non-specific resistance to Klebsiella infection (Chedid et al., 1982). Lipophilic derivatives like 6-0-acyl esters of MDP have been shown to retain immunostimulating properties (Matsumoto et al.,

1981). Other lipophilic derivatives containing the mycolyl group at the end of the peptide chain have been shown to stimulate strongly adjuvant activity, delayed hypersensitivity and non-specific antibacterial resistance (Parant et al., 1980).

Several compounds containing nucleotides have also been shown to possess immunomodulating properties. The polynucleotides, polyinosinic-polycy- tidylic (poly I:C) and polyadenylic-polyuridylic (poly A:U) have been reported to be potent antiviral (Richmond and Hamilton, 1969) and 53

antitumour (Levy et a l . , 1968) agents as well as interferon inducers

(Field et al., 1967). Another important compound is methisoprinol or

isoprinosine (ISO), which appears to be effective in a wide variety of

viral diseases (Waldman and Ganguly, 1977; Laude et al., 1980). ISO

increases cell mediated immune functions in vitro (Bradshaw and Summer,

1977; Wybran et al ., 1978) and increases T cell levels in patients

(Friedman et al., 1980). NPT 15392 is a hypoxanthine analogue that has

recently been shown to possess neutrophil, T cell and NK cell stimula­

tory properties. Its structure is related to inosine and its action to

methisoprinol. It augments human T cell proliferation and suppressor

cell induction (Hadden et a l . , 1982) and also modulates a variety of

cytocidal functions (Florentin et al., 1982).

Pyran (maleic divinyl ether copolymer) was first used as a chemothera­

peutic antiviral agent against Friend virus-induced leukemia and the

protection was attributed to interferon induction (Merigan, 1967; Hirsch

et a l . , 1972). The problem with these early copolymers was extensive

toxicity which was related to the high molecular weight polymers. This

led to the development of copolymers based on the same structure but of

smaller molecular weights called maleic vinyl ethers (MVE). MVE-2 has

been shown to be a potent immunostimulator of low toxicity and has been

tested in clinical trials (Carrano et al., 1984).

Recently, a compound belonging to the 2-cyanaziridines group, azimexon, has been shown to have immunomodulating properties which include incre­ ased cellular immunity, NK cell activity, increased granulopoiesis and antitumour activity (Bicker, 1984). 54

1.4 Interferon

Interferon (IFN) was discovered by Isaacs and Lindenmann (1957) when they observed anti-viral activity in the supernatant fluids of chick allantoic membrane cultures inoculated with influenza virus. It is now known that IFN is a complex group of small proteins and glycoproteins produced by animal cells in response to a wide variety of stimuli.

Although IFNs were first recognized for their anti-viral activity, recent interest in them has centered on the discovery of a wide range of biological effects on the immune system. IFNs have been shown to regul­ ate cellular proliferation or differentiation as well as cellular morph­ ology and expression of cell surface antigens. In addition, IFNs appear to be potent immunoregulatory agents affecting both cell mediated and humoral immunity.

1.4.1 Description of the interferons

To date, three different types of IFNs have been described, IFN-alpha

(IFN-*<-or leukocyte IFN), IFN-beta (IFN-p or fibroblast IFN) and IFN- gamma (IFN-V or immune IFN). IFN-«* is mainly produced by B cells, natural killer (NK) cells and macrophages; and IFN-|3 mainly by fibro­ blasts after induction by a variety of agents. Among the inducing agents are viruses; double-stranded RNA; intracellular organisms like brucella, listeria, rickettsia, mycoplasma and chlamydia; microbial products like LPS; organic polymers such as pyran co-polymers; and several low molecular weight substances like cycloheximide, kanamycin and tilorone (Epstein, 1979). T lymphocytes are the main cells which produce IFN-K after stimulation by mitogens or specific antigens. 55

However, B cells and NK cells also produce IFN-Y after mitogenic stimulation (Epstein et al., 1974; Kirchner et al., 1979).

The IFNs differ antigenically, physically and chemically from each other. Antigenic differences between all three IFNs can be detected by polyclonal and more recently, by monoclonal antibodies (Secher & Burke,

1980; Nyari et a l . , 1981; Hochkeppel, 1982). Human IFN-o( and |2> have molecular weights ranging from 18,000 to 25,000 daltons but native human

IFN-X has a molecular weight of 50,000 daltons. Recent studies however, have suggested that it may be composed of dimers of smaller subunits of

20,000 and 25,000 daltons (Yip et al. , 1982). IFN-Y and IFN-[iare glycoproteins whereas most IFN-0C moieties do not appear to have carbohy­ drate components.

The antiviral effects of IFNs are relatively species specific but there are exceptions. Subtypes of human leukocyte IFN may have significant antiviral activity on bovine or mouse cells (Week et al., 1981). It has been shown that each IFN has a characteristic species activity profile.

Human IFN-0( has a high degree of antiviral activity in bovine and porc­ ine cultures whereas human IFN-{S is hardly active and human IFN-X is totally without activity in these cells (Rager-Zisman and Bloom, 1985).

1.4.2 Molecules and genes of the interferons

IFN-o( has been purified to homogeneity (Rubinstein et al., 1978; Berg and Heron, 1980) and human IFN-o( has been shown to consist of at least

13 different proteins (Berg and Heron, 1982). Recently, several groups have succeeded in cloning at least 12 distinct but almost homologous 56

IFN-<* proteins in Escherichia coli (Goeddel et al., 1980; Streuli et^ a l . , 1981; Rehberg et a l . , 1982). Only one human I F N - g e n e has been definitely isolated (Derynck et al., 1980) and has led to the production of a polypeptide with human IFN-{3 activity in E. coli (Derynck et al.,

1980a). However, it has been observed that there are at least five translationally active human IFN-^3 mRNAs (Sagar et al., 1982). There is evidence that only one IFN- gene consisting of 146 amino acids exists

(Gray and Goeddel, 1982). Recently, the human IFN-^ gene (Gray et al.,

1982), followed by the murine IFN-tf gene (Gray and Goeddel, 1983) were cloned in E. coli and monkey cells.

1.4.3 Mechanisms of action of the interferons

It is now known that IFNs exert their action by binding to specific cell-surface receptors probably consisting of gangliosides and, or glycoproteins (Friedman, 1967; Aguet, 1980). IFN-^and IFN-appear to share a class of receptors whereas IFN-# binds to a different receptor

(Branca and Baglioni, 1981). Binding assays using radiolabelled IFN-if have demonstrated that there are about 2400 and 12,000 high-affinity binding sites per human fibroblast and mouse macrophage respectively

(Celada et al., 1984). Anderson et al.(1983) showed that labelled human

IFN- bound to cells is rapidly internalised and degraded at 37°C.

Binding of IFNs initiates a series of metabolic modifications which involve the de novo synthesis of RNA and polypeptides. Two-dimensional gel electrophoresis has shown that all IFNs induce several common poly­ peptides but that IFN-JT induces in addition 12 distinct polypeptides

(Weil et al., 1983). 57

Among the proteins synthesized are 2'5 '-oligoadenylate synthetase and

protein kinase. 2'5'-oligoadenylate synthetase catalyzes the synthesis

of small molecules of 2'5' adenylate oligomers which activate endogenous

ribonucleases (Baglioni et a l . , 1978). The ribonucleases can degrade

both viral and probably host cell mRNAs which could lead to diminished

synthesis of viral specific proteins. Protein kinase is activated by

the presence of double-stranded RNA and mediates through phosphorylation

the inactivation of a eukaryotic protein synthesis initiation factor

eIF-2 (Lebleu et a l . , 1976). These two enzymatic activities could be

responsible for the restriction of viral replication. However it is

still not firmly established that either is responsible for anti-viral

activity. Both need double-stranded RNA for full activation and this

suggests that there are likely to be other mechanisms of action for the non-viral effects of IFNs.

There has been some evidence of the possible mechanisms of action of the non-viral effects of IFNs. Recently, Hamilton et al. (1985) demonstrat­ ed that murine peritoneal macrophages treated with recombinant IFN-Y had

2+ a five fold increase in Ca , phospholipid-dependent protein kinase act­ ivity (Protein kinase C). The kinetics of the elevation of kinase acti­ vity was identical to that required for induction of other activities by

IFN-Y, suggesting that protein kinase C may have direct functional con­ sequence in macrophage activation. Similarly, Weiel et al. (1985) used a model of depression of transferrin receptors on murine peritoneal macrophages after exposure to IFN-

Another enzyme induced by IFN-^ is indoleamine-2,3-dioxygenase which

catalyzes the breakdown of tryptophan. Since eukaryotic cells and many

intracellular parasites require tryptophan for growth, this enzyme may

play a role in inhibition of cell growth or intracellular parasite

(Pfefferkorn, 1984). Nagata et al. (1984) have shown that in a macro­

phage cell line, cyclic AMP may mediate the effects of IFN-*. They

selected mutant macrophage cell lines which were susceptible to anti­

viral effects but resistant to growth inhibiting effects of IFN-JT and

showed that the mutant cell lines were either lacking IFN-JT receptors or were mutants for adenylate cyclase.

1.4.4 Immunomodulating effects of the interferons

IFN preparations especially those containing IFN-f have long been known

to modulate the immune response (Gresser et a l ., 1972; Virelizier

et a l . , 1977; De Maeyer and De Maeyer-Guignard, 1982). All three types

of IFNs have been shown to affect antibody production, cell-mediated

immunity and other functions of the immune system. However, as most of

these earlier studies used only crude or partially purified IFNs or mixtures of IFNs it had been difficult to separate the immunoregulatory effects of IFNs from those of other biologically active molecules.

Recently, with the cloning of the IFN genes in mouse and man, it will now be possible to assess the actual effects of each of these molecules

on the immune response. 59

1.4.4.1 Effects on humoral immunity

Various types of IFNs have either suppressive or enhancing effects on

humoral B-cell responses both in vivo and in vitro, depending on the

relative timing of exposure to IFN and to the antigen. IFN-V in parti­

cular potentiates the effects of immunoglobulin secretion when added

late during an immune response in vitro (Sonnenfeld et al., 1978).

Recent reports with recombinant IFN-V confirm the effects observed

previously with partially pure preparations. Nakamura et al. (1984)

described an enhancement of antibody formation when antigen and IFN-V were administered together to mice. In another study, Leibson et al.

(1984) showed that cloned murine IFN-^T could act synergistically with

other helper factors as a T-cell Replacing Factor (TRF) in stimulating

B-cell antibody response in vitro. IFN-V has also been reported to act

as one of several B-cell maturation factors and induces surface

phenotype changes and immunoglobulin secretion in resting B cells

(Sidman et al., 1984).

1.4.4.2 Effects on cell surface membrane

The cell surface membrane plays a vital role in direct contacts between cells, recognition of molecular structures and initiation of cell activ­ ation and differentiation (Loor, 1979). All these interactions are known to be mediated through surface molecules on the cell surface memb­ rane. IFNs induce a variety of alterations in and on the cell surface

(Friedman, 1979). Knight and Korant (1977) described an increase in net negative surface charge on murine L cells and Chang et al. (1978) reported an increase in bouyant density of the plasma membranes in IFN 60

treated murine cells. Other studies have shown that IFN-(S treatment can

alter the structure and organization of the cytoskeleton of a variety of

cells (Pfeffer et al., 1980; Pfeffer et al., 1980a).

All three types of IFNs enhance the expression of Class I Major Histo­

compatibility (MHC) antigens, although IFN-J seems to be the most effi­

cient (Sonnenfeld et al., 1981; Fellous et al., 1979; Wallach et al.,

1982). There have been conflicting reports on the effect of IFN-** and

IFN- £ on the expression of MHC Class II antigens. However, IFN-if has

been shown to cause a substantial increase in these antigens on lymphoid

cells, myelo-monocytic cells, mast cells, fibroblasts, tumour cell lines

and melanoma cells (Wong et al., 1983; King and Jones, 1983; Virelizier

et a l . , 1984). Walker et al. (1984) reported that IFN-^ may induce la antigens on mouse macrophages via a discrete secondary factor.

Other changes consistently seen on human and murine cells following IFN

treatment are the expression of Fc-IgG receptors (Fridman et al., 1980;

Itoh et al., 1980; Guyre et a l ., 1981). However, these effects are quantitatively not as large as the increases in MHC Class I antigens and are furthermore only seen in certain subpopulations of cells.

The biological significance of the IFN-induced surface alterations is not yet established. However, the Class II MHC antigens are important in the recognition processes for cell interactions or cytotoxic acti­ vity. Zlotnik et a l . (1983) has shown that IFN-T treatment renders macrophage cell lines capable of antigen-presentation. Pober et al.

(1983) reported that human T lymphocytes recognized and lysed endothe­ lial and fibroblast cells with IFN-)f induced la antigens. Becker (1985) 61

showed that the IFN-V enhancement of monocyte antigen-induced and autologous proliferative responses was a consequence of the increased density of monocyte HLA-DR antigens induced on the accessory cells.

1.4.4.3 Effects on cell mediated immunity

IFN-tf appears to play a more important role in immunoregulatory function than IFN-c* or IFN-£J. It is one of the principle lymphokines regulating the activation of macrophages. It had been previously postulated that

IFN-X and MAF are the same molecules (Schultz and Chirigos, 1978;

Kleinschmidt and Schultz, 1982). Studies with recombinant I F N - h a v e yet to resolve the controversy. There have been reports of antigenic and functional similarities of MAF and IFN-fr (Schultz and Kleinschmidt,

1983; Schreiber et al . , 1983; Svedersky et al., 1984). Other studies suggest that IFN-3T and MAF are distinct molecules. It has been shown that culture supernatants from human T cell lines contain a soluble factor with MAF activity that is not abrogated by treatment with anti­ human IFN-V antibody (Andrew et al., 1984; Kleinerman et al., 1984). It has recently been hypothesized that the intact IFN-2T molecule may contain distinct domains for antiviral activity and for MAF activity

(Peters et al., 1985). This hypothesis is supported by the development of monoclonal antibodies specific for various epitopes of the recombi­ nant murine IFN-# molecule that differentially inhibit the antiviral and

MAF activities (Schreiber et al., 1985).

The availability of recombinant IFN-if and the development of monoclonal antibodies to IFN-^fhave enabled the unambiguous identification of IFN-k* as the active factor in lymphokines activating macrophages to produce 62

and for killing a variety of intracellular organisms in both the human and the murine system. Rothermel et al. (1983) showed that it was

IFN- X present in human concanavalin A (Con A) induced lymphokine that activated monocytes to inhibit chlamydial replication. Similarly,

Murray et al. (1983) reported that IFN-^ is the main macrophage activa­ ting molecule present in human lymphokines and that recombinant IFN-& enhances dependent and independent anti-leishmanial activity of monocytes. Nathan et al. (1983) identified IFN-if as the lymphokine that activates human macrophage oxidative metabolism and anti toxoplasmic activity. In the murine system, Murray et al. (1985) has shown that

IFN-8 activates mouse peritoneal macrophages both in vitro and in vivo for increased oxidative metabolism and anti-protozoal activity.

There is also evidence that IFN-tf can activate cells other than macro­ phages to kill intracellular organisms. Pfefferkorn (1984) reported that IFN-# blocks the growth of Toxoplasma gondii in human fibroblasts by inducing host cells to degrade tryptophan. Wiseman and Waddell

(1983) showed that factors in human lymphokines with the properties of

IFN-y’ have antirickettsial activity on Rickettsia prowazekii infected endothelial cells, fibroblasts and macrophages. There have been similar reports of the effects of IFN-& in murine cells. IFN- X has been shown to inhibit the growth of R. prowazekii in mouse fibroblasts (Turco and

Winkler, 1983). Turco et a l . (1984) reported that crude lymphokines and recombinant IFN-tf, but not a crude preparation of IFN-<* and IFN-J2, inhibited the growth of Coxiella burnetii in mouse fibroblasts.

In addition, IFN-|f has been shown to regulate natural killer cell activity (Rager-Zisman and Bloom, 1985); stimulate production of 63

interleukin-1 production (Palladino et a l . , 1983); and enhance the growth of interleukin-2 (IL-2) dependent murine cytotoxic T cell clones in the absence of IL-2 (Peters et al., 1985).

1.5 Levamisole

1.5.1 Description of levamisole

Levamisole is the levo isomer of tetramisole stereospecifically synthesized in 1966 , and available as an antihelminth for clinical use in 1968. It is a stable, white crystalline powder of molecular weight

240.75 and is very soluble in water and aqueous acidic solutions. In neutral buffers and especially alkaline solutions, the solubility drops and hydrolysis to 3-oxo-3(2-mercaptoethyl)-5-phenyl imidazoline (OMPI) occurs. Recent studies suggest that OMPI is the active compound (Van

Ginckel and DeBrabander, 1979). Levamisole is active against most nematodes but has no direct toxic effect on bacteria, viruses and fungi

(Thienpont et a l . , 1966) nor on normal and tumour cells at concentra­ tions up to 100 pg ml ^"(Sampson and Lui, 1976; Pabst and Crawford,

1974). Pharmacologically, levamisole stimulates parasympathetic and sympathetic ganglia (Van Nueten, 1972). Pharmacokinetic studies in animals (Graziani and De Martin, 1977) have shown that levamisole is rapidly absorbed from the and well distributed in the tissues with the highest tissue levels in the liver and kidneys.

Plasma half lives for the unchanged drug vary from 1-4 hours and drugs were rapidly removed from the tissues within 2-4 days. It is a drug of low toxicity with therapeutic doses in the range of 2-3 mg kg 64

1.5.2 Immunomodulating effects of levamisole

The first evidence of the immunomodulating properties of levamisole was obtained in 1971 when it was observed that levamisole increased the immunity of Brucella vaccinated mice (Renoux and Renoux, 1971). Since then, there have been a multitude of studies on levamisole, some con­ tradicting and some supporting the immunotherapeutic potential of levamisole.

The effects of levamisole on the immune system has been investigated in isolated cells, experimental animal infections, normal volunteers and patients. These studies showed that levamisole influenced cell-mediated immune reactions. In vitro and in vivo studies showed that levamisole was able to restore effector mechanisms of cell mediated immunity. The effects are most pronounced in compromised hosts with subnormal T cell or phagocyte functions (Bensa et al., 1976; Ellegaard and Boesen, 1976;

Bruley-Rosse t et a l . , 1976; Rosenthal et al., 1976). Levamisole does not usually increase an adequate immune response except at doses greatly exceeding therapeutic concentrations.

The effector phagocytic functions which may be restored by levamisole include chemotaxis (Rabson et al., 1978); phagocytosis (Molin and

Stendahl, 1977); random migration (Anderson et al.,1976); adherence and antibody and complement receptor activity (Schmidt and Douglas, 1976;

Schreiber e t a l . , 1975) of polymorphonuclear and mononuclear phagocytes.

Effector lymphocyte functions which may be restored include spontaneous, mitogen or antigen-specific proliferation (Chan and Simons, 1975; Chan 65

et a 1 ., 1976; Lewinski et al., 1977); lymphocyte counts (Moncada-

Gonzalez et a l . , 1976); antibody plaque formation (Renoux and Renoux,

1974) and migration inhibition and lymphokine production (Golding £t_

al., 1976). Levamisole does not stimulate B-cells directly. It has

been shown not to increase B-cell mitogenic proliferation (Hadden £t_

al. , 1975) nor antibody production (Flannery et al., 1975). There is

in vivo evidence that levamisole is capable of inducing maturation of T

cells. It induces thymic antigens in nude mice (Renoux and Renoux,

1977) and stimulates lymphocyte proliferation in nude and thymectomized mice (Merluzzi et al., 1976). Further evidence is provided by the

findings that levamisole restores cell-mediated immune functions in

children with primary immune deficiencies (Griscelli et al., 1978).

Levamisole has been shown to restore delayed skin sensitivity in anergic

subjects. Such observations have been made in cancer patients (Lewinski et al., 1977; Tripodi et al., 1973); aged healthy persons (Kondo et al.,

1978) ; pulmonary tuberculosis patients (Singh et al., 1981); and leprosy patients (Cardama et al., 1973).

The activity of levamisole in restoring cell-mediated functions indicates a potential use in diseases where hypofunction of T cell, polymorphonuclear cells or macrophages occurs. Levamisole has been tested in large numbers of diseases with suspected immune imbalance and some have responded well, others partially or not at all. Responses to levamisole has been mainly seen in chronic or recurrent diseases

(reviewed by Symoens and Rosenthal, 1977), some cancers (reviewed by

Amery and Verhaegen, 1978) and primary immune deficiencies (Griscelli et al, 1978). 66

1.5.3 Mechanisms of action of levamisole

The mechanisms of action of levamisole are rather unclear at present.

There have been several proposals. One possibility is that levamisole increases cyclic GMP or reduces cyclic AMP (Hadden et al., 1975; Hogan and Hill, 1978) in lymphocytes and phagocytes which would promote proliferative and secretory functions as well as receptor reactivity.

Levamisole contains an imidazole ring and it has.been suggested that this moiety is responsible for immune regulation by altering the levels of cyclic nucleotides. In vitro studies have shown that levamisole and imidazole exerted virtually identical effects, however, none of the in vivo effects of levamisole could be reproduced by imidazole (Renoux and

Renoux, 1977a).

Another possible mechanism could be through the activity of the thiol

OMPI metabolite which appears after the hydrolysis of levamisole.

Thiols like all other antioxidants could be important for maintenance of the cellular redox potential. Levamisole and OMPI have been shown to restore cell functions by the inhibition of peroxidase formation (Ander­ son et a l . , 1981). Levamisole also shares with all other antioxidants the ability to stimulate DNA synthesis (Van Wauwe and Goosens, 1979).

Another aspect is the indirect thymomimetic activity of a molecule mediated by a serum factor which is produced by responders but not non­ responders to levamisole. The serum factor has been shown to mimic thymic hormone; to be neither a complement factor nor a levamisole meta­ bolite; and animals that do not respond to levamisole respond to serum 67

factor (Symoens et al., 1979). Different mechanisms might operate depending on the conditions of the experiment, whether it was performed in vivo or in vitro or whether the immune response of the host was normal or impaired.

1.6 Targeting of drugs

An important feature of successful drugs is the ability to act predomin­ antly on the target cell thus minimizing the effects on the rest of the organism. Antibacterial agents are a good example. Most of them owe their unique selectivity to their ability to interfere with some meta­ bolic pathway peculiar to the target bacteria and not shared by the human host. The use of carriers has been proposed to improve the selec­ tivity of drugs. Carriers could deliver the drug directly to the target organ, tissue or cells. They might also favourably alter the pharmaco­ kinetics of the drug like clearance and metabolism enabling the reduc­ tion of therapeutic dosages to acceptable toxicity levels. It is also conceivable that carriers might prolong the intervals between admins- tration of the drug.

1.6.1 Description of drug carriers

A number of drug carriers have been investigated and they can be broadly divided into two categories, natural products and synthetic systems.

Natural carriers are extracted from animals or plants and either selec­ tively bind to receptors on potential targets, for example antibodies; or migrate to specific targets, for example, neutrophils attracted to 68

inflammation sites. Most of the studies with natural carriers involved

the use of antibodies in the treatment of experimental cancers. Poly­

clonal antibodies raised against the whole target cells or surface anti­

gens and coupled with a wide variety of cytotoxic drugs have been quite

selective in experimental systems in killing cells in vivo and in vitro, but were not as successful as expected in clinical use (Everall et al.,

1977; Ghose et a l ., 1977). The recent advent of monoclonal antibodies has given new impetus to antibody-mediated targetting (Thorpe et al.,

1982).

Synthetic carriers include polymers, albumin beads, acrylic microsphe­ res, magnetic particles and liposomes. Of all the synthetic carriers, liposomes have been most extensively investigated.

1.6.2 Liposomes

■1.6.2.1 Types of liposomes

A liposome is a minute spherical vesicle composed of phospholipid bilayers that enclose a volume. There are four main types of vesicles.

Multilamellar vesicles (MLV), originally described by Bangham et al.

(1965), are composed of many concentric lamellar membranes and range in diameter from 0.1 to 10 jum. Small unilamellar vesicles (SUV) with one membrane and one cavity of diameter 25-50 nm, were initially described by Papahadjopoulos and Miller (1967). Large unilamellar vesicles (LUV) are similar to the SUV except for a larger diameter of 0.2-1.0 jam (Papa- hadjopoulos et a l ., 1975; Deamer and Bangham, 1976). Reverse phase evaporation vesicles (REV) are large unilamellar liposomes of diameter 69

0.2-0.8 Aim formed by evaporation of the organic phase from an emulsion of phospholipids and aqueous buffer (Szoka and Papahadjopoulos, 1978).

1.6.2.2 In vivo interactions of liposomes

All liposomes can entrap hydrophilic substances in the aqueous compart­ ments and lipophilic substances in the phosholipid membranes of the vesicles. The use of liposomes as carriers is based on their inter­ action with cells. It has now become apparent that liposomes can associate with cells in a variety of ways. Endocytosis has been shown to occur (Weissman et al., 1975; Poste and Papahad jopoulos, 1976), and is followed by lysosomal fusion leading to disruption and release of the entrapped agents which then either act within the lysosomal vacuole or diffuse into other cell compartments. Liposomes have also been shown to fuse with the plasma membrane of the cell and introduce their contents directly into the cytosol (Poste and Papahad jopoulos, 1976). Another mechanism that has been proposed is that liposomes adsorb onto the cellular membrane without subsequent interiorization, with the release of agents by diffusion from the liposomes (Poste, 1980). It is quite likely that more than one mechanism occurs simultaneously and that parameters like cell type, liposomal lipid composition, size, surface charge and other experimental conditions will favour one mechanism over another.

After intravenous injection, liposomes are rapidly cleared from the plasma and found mainly in mononuclear phagocytes of the reticuloendo­

thelial system in the liver, spleen and circulating monocytes (Finkel- stein and Weissmann, 1981). This natural fate of the liposomes has 70

created difficulties when the drugs are to be targeted to other cell types like solid tumours. However, this passive localization of lipo­ somes can be exploited in diseases where the mononuclear phagocyte plays an active and important role in host defence like with intracellular parasites. The blockade of RE phagocytic activity produced by an intra­ venous injection of liposomes is transient and reversed fully within 24 hours (Abra and Hunt, 1982; Ellens et al., 1982).

1.6.2.3 Therapeutic applications of liposomes

Among the substances that have been encapsulated in liposomes are enzymes (Gregoriadis and Buckland, 1973; Tyrell et al., 1976); anti­ tumour drugs (Kaye and Richardson, 1979; Mayhew et.. al. ,1978) , chelating agents (Rahman et al.,1973; Young et al.,1979); steroids (Shaw et al.,

1976; De Silva et a l ., 1979); antibiotics (Desiderio and Gordon-Camp- bell, 1983; Lopez-Berestein et al., 1983); lymphokines (Fidler, 1980;

Fidler et al., 1982) and immunomodulators (Fidler et al., 1981; Philips et al., 1985).

The feasibility of exploiting passive targeting of liposomes as a means of activating macrophage-mediated host defence mechanisms has been inve­ stigated in several laboratories. Fidler, Poste and their colleagues have investigated the in vitro and in vivo effects of lymphokines encap­ sulated in liposomes (Poste et al., 1979; Fidler, 1980; Fogler et al.,

1980). They demonstrated that liposomes containing MAF activated macrophages to be tumoricidal in experimental murine metastatic cancer.

Fidler et al. (1980) showed that the formulation of the liposomes could influence the in vivo distribution after intravenous administration. 71

They demonstrated that the proportion of liposomes trapped in the lungs

of mice could be increased by using liposomes containing phosphatidyl­

choline (PC) and phosphatidylserine (PS) in a molar ratio of PC:PS =

7:3. However, even though more liposomes of this formulation are

delivered to the lungs, the majority of them are still to be found in

the liver and spleen (Poste et al., 1982). Reed et al. (1984) also

showed that lymphokine encapsulated in liposomes enhanced the in vivo

inhibition of the proliferation of L. donovani chagasi in mice.

Liposomes containing immunomodulating agents have been shown to be

highly effective in stimulating macrophage-mediated host resistance.

Fidler et a l . (1981) demonstrated the eradication of spontaneous meta-

stases and the activation of alveolar macrophages by liposomes con­

taining MDP. Fraser-Smith et al. (1983) reported the protective effect of an analog of MDP encapsulated in liposomes against an experimental

Candida albicans infection.

In addition to their role in host defence, mononuclear phagocytes are also important sites of replication for many parasites. A number of viruses, bacteria, fungi and protozoa multiply within these cells. Such intracellular infections are often difficult to eradicate and liposomal encapsulation of therapeutic agents could resolve the problem. It could provide a more efficient drug delivery system directly to the target cells, enabling a reduction of dosages thereby reducing potential toxi­ city effects. This has been shown in experimental therapy of leishma­ niasis with antimonials (Alving et al.,1978; New et al., 1978). These studies showed that the doses of liposome-encapsulated drug required to inhibit parasitic growth and cure infected animals are about 1/100 to 72

1/800 of those required of the free drug. Lopez-Berestein et al. (1983)

demonstrated that encapsulation of amphotericin B in liposomes improved

the therapeutic index in a model of experimental murine candidiasis. In a study on chemotherapy of murine cryptococcosis, Graybill et al. (1982) showed that liposome-associated amphotericin B had only 1/17 the toxi­ city of the free drug. The enhanced effects of liposomal amphotericin B in cryptococcosis was mainly due to the ability to deliver larger doses without increased toxicity. When the liposomal dose was reduced to a dose that could be safely administered as a free drug, liposomes did not convey any therapeutic advantages. Experimental murine histoplasmosis was also more effectively treated with the higher amphotericin B concen­ tration that could be achieved with liposomes (Taylor et al., 1982). 73

II. INTRODUCTION AND OBJECTIVES OF THE EXPERIMENTAL WORK

1.7 Introduction and objectives

After taking into consideration the impact of current tuberculosis control measures on the epidemiology of tuberculosis, there is universal agreement that the most powerful component in tuberculosis control is case-finding and chemotherapy. In addition to their direct and imme­ diate effect of reducing suffering and mortality, case-finding and chemotherapy also eliminate sources of infection. BCG vaccination can prevent tuberculosis in uninfected individuals but does not contribute significantly to the reduction in overall risk of infection in the community. Isoniazid prophylaxis can prevent the development of tuberc­ ulosis in infected individuals but is also unlikely to have much impact on the community as it is cannot be easily implemented on a mass scale.

At present there seems little prospect for any improved vaccines and BCG remains the only vaccine. It is possible that with the application of genetic engineering a more efficacious vaccine will be engineered (Young et a l . , 1985). However, it is likely to be decades before any new vaccine has an impact on the disease as it will have to be tested for safety and then properly evaluated for its ability to produce long lasting protection in lengthy controlled trials. Furthermore, there is an inevitable time lag between vaccination in childhood and its efficacy in preventing adult disease 30-60 years later.

Immunological research may be helpful in two ways: 1) improving case­ finding by new diagnostic techniques, and 2) improving chemotherapy by 74

using immunomodulating agents. Considering first new diagnostic

methods, development of better serological tests could lead to improved

diagnosis of childhood tuberculosis, non-pulmonary tuberculosis and

smear-negative cases of active pulmonary tuberculosis. The isolation of

pure antigens or more specific tuberculins would also be of value for

skin testing and other in vitro immunological tests. Turning to

therapy, it is important to learn whether chemotherapy might be improved

by simultaneous treatment with immunotherapeutic agents. The second

question is the subject of the present thesis. Two immunomodulating

agents, levamisole and interferon-gamma, were examined for their effects

on experimental murine tuberculosis; and for interferon, for possible

synergism between the immunomodulating agent and the chemotherapeutic

drugs, isoniazid and rifampicin.

Levamisole has been shown to have immunotherapeutic potential and has

been tested in a large number of diseases with suspected immune imbalance. There have been claims of the clinical benefits of levami­ sole as an adjunct to chemotherapy on tuberculosis patients. In a study with 73 cases of advanced pulmonary tuberculosis (Yaseen et al., 1980) it was reported that the addition of levamisole to a standard antituber­ culous regimen improved the speed of sputum conversion and the radiolo­ gical findings. They also reported increases in T cell counts and restoration of dinitrochlorobenzene (DNCB) sensitization. A trial of levamisole as an adjunct in the treatment of 50 newly diagnosed pulmo­ nary tuberculosis patients with mild immunodepression (Singh et al.,

1981) showed improved radiological clearing in the levamisole-treated group but no difference in sputum clearance rate. In the levamisole- treated group, 48% reacted to DNCB compared to 18% in the control group. 75

In another study of 100 pulmonary tuberculosis patients, Singh et al.

(1983) reported that levamisole given in conjunction with antituber­ culous drugs caused a significant difference in sputum conversion time, subjective improvement, weight gain and radiological improvement.

However, another study on the effect of levamisole in combination with chemotherapy on 15 patients with miliary tuberculosis (Singh et al.,

1983a) showed only a restoration of Mantoux and DNCB reactivity, with no clinical improvement.

A clinical trial on the effect of levamisole on the response to chemo­ therapy of pulmonary tuberculosis patients in Kenya and Zambia was being organized under the auspices of the Medical Research Council at the start of the work described here. The double-blind trial consisted of three regimens: chemotherapy with streptomycin, isoniazid, rifampicin and pyrazinamide daily for two months followed by daily thiacetazone and isoniazid for 4 months; chemotherapy supplemented with levamisole once a week for one month; and chemotherapy with levamisole for two months.

Levamisole was given at a dose of 150 mg for patients weighing 50 kg or more and 100 mg for patients less than 50 kg in weight. The existence of this major clinical study was an important reason for investigating the efficacy of levamisole in experimental murine tuberculosis.

The effects of levamisole were assessed on an infection of M. microti in mice. As M. microti is non-pathogenic for humans, all manipulations and assays could be carried out with comparative ease. In addition to moni­ toring the effect of levamisole on the growth of M. microti, the effects on T lymphocyte function and subsets were also examined to monitor the effects on the immune system. The work with levamisole was not carried 76

to completion in that no experiments were done with levamisole in combi­

nation with antituberculous drugs. The reasons for this was because

levamisole appeared to have no effect on the M. microti model and in

part because recombinant interferon-gamma became available and was given

priority for further study.

Recent evidence has implicated interferon-gamma as the important factor

in lymphokine preparations which activates macrophages for antitumour

and antimicrobial action (Pace et a l . , 1983; Rothermel et al. , 1983;

Murray et al., 1983; Nathan et al., 1983). Recombinant interferon-gamma

was examined for its effects on the growth of M. tuberculosis in the

lungs and spleens of mice. The model of an acute infection with the

virulent strain H37Rv of M. tuberculosis was chosen as it was considered

to be a more relevant model than an infection with M. microti. Lipo­

somes have been shown to be effective in delivering drugs or immunomodu-

lating agents in both in vivo and in vitro systems. Fidler (1980) had

shown that MAF encapsulated in negatively-charged MLV liposomes composed

of PC and PS (PC:PS =7:3 mole ratio) was effective in activating

alveolar macrophages for tumouricidal activity in murine lungs. It was

decided to administer interferon-gamma in liposomes formulated in a

similar way on M. tuberculosis infection in mice.

The effects of interferon-gamma in combination with isoniazid and with rifampicin were examined with M. tuberculosis infection in mice. Ison­ iazid was chosen because it is an important component of all chemothera­ peutic regimens, and for its high bactericidal activity while rifampicin was chosen for its high sterilizing activity. 77

The mononuclear phagocyte plays an important role in both the afferent and the efferent arms of the immune response to tuberculosis. Tubercle bacilli multiply in macrophages in susceptible hosts and it has been demonstrated that macrophages can be activated in vitro by lymphokines to kill M. microti (Walker and Lowrie, 1981). The effects of IFN-Y on

the growth of L. monocytogenes and M. microti in murine peritoneal macrophages were examined. The listericidal assay was chosen because

L. monocytogenes is an intracellular pathogen with a rapid growth rate and this facilitated the development of the microbicidal assays. The choice of M. microti enabled the in vitro assays to be performed with comparative ease. In addition, the in vitro activity of IFN-Y in combination with isoniazid and with rifampicin on M. microti infection in macrophages was also investigated. 78

CHAPTER 2

MATERIALS AND METHODS

2.1 Media

2.1.1 Bacteriological Culture Media and supplements

All media were obtained from Difco Laboratories (P.0. Box 14B, Central avenue, West Molesey, Surrey, UK) unless specified otherwise.

Middlebrook 7H11 Oleic acid-albumin Agar was reconstituted from the dehydrated product in distilled water containing 0.5% glycerol. After sterilization, the media was supplemented with 10% Oleic Albumin Dext­ rose Complex (OADC).

Selective 7H11 Agar (Mitchison et a l ., 1973) was prepared by the addition of antibiotics to the reconstituted Middlebrook 7H11 medium above. The following antibiotics at their respective final concentra­ tions were added after autoclaving :

1. Polymyxin B , 200 units ml ^

2. Carbenicillin , 100 }ig ml ^

3. Trimethoprim , 20 jig ml ^

4. Amphotericin B , 10 jig ml ^

Dubos Oleic Agar Base was prepared by reconstituting the dehydrated medium with distilled water and adding 10% OADC after sterilization.

Middlebrook 7H11 agar, Selective 7H11 agar and Dubos Oleic agar supple­ mented with 5% lysed horse blood were prepared as described above and 79

supplemented with 5% lysed horse blood after sterilization.

Dubos Broth (2X) was prepared double strengthed from Dubos Broth Base

(Difco Laboratories). It was used either in this form or with the

addition of 10% ADC.

Middlebrook 7H9 Broth was reconstituted from the dehydrated medium with

distilled water. It was supplemented with 10% ADC.

Middlebrook ADC Enrichment and Middlebrook OADC Enrichment were either

obtained from Difco Laboratories or were prepared in the laboratory from

the ingredients.

Tryptic Soy Agar Blood Base (TSA) and Tryptic Soy Broth (TSB) were

prepared from the dehydrated products.

Horse blood was obtained from Tissue Culture Services Ltd. (2 Perth

Estate, Slough, Berkshire SL1 4XX, UK) and hemolysed by repeated freez­

ing and thawing.

2.1.2 Tissue Culture Media and supplements

All tissue culture media and supplements were obtained from Gibco Europe

Ltd. (Unit 4, Cowley Mill Trading Estate, Uxbridge UB8 2YG) unless

specified otherwise.

Tissue culture media RPMI 1640 and RPMI 1640 containing 25 mM HEPES and

L-Glutamine. 80

Medium 199 containing Earle’s Salts, 25 mM HEPES Buffer and L-Glutamine.

Hanks’ Balanced Salts Solution without Phenol Red (HBSS).

L-glutamine was obtained as a lyophilised powder.

Foetal Calf Serum and Horse Serum (Mycoplasma-free). Both sera were

heat-inactivated at 56°C for 30 minutes before being used.

Antibiotic-antimycotic was obtained as a lyophilised powder containing

penicillin (10,000 units ml ^), streptomycin (10,000 meg ml and

amphotericin B(25 meg ml ^).

Bovine Embryo Extract (50% in Earle’s Salt Solution) was obtained from

Flow Laboratories (Woodcock Hill, Harefield Road, Hertfordshire, WD3

1PQ, UK).

HEPES Buffer was obtained as a 1 Molar solution from Flow Laboratories.

Heparin sodium B.P. (1000 units ml ^) was obtained from Paines and Bryne

Ltd. (Greenford, England).

Liver Fraction L was obtained from the United States Biochemical Corpor­ ation, Cleveland, Ohio, USA.

HBSS-HEPES was prepared by the addition of 25 mmolar HEPES buffer to

HBSS. 81

Basic Maintenance Medium (BMM) was used for the culture of macrophage monolayers.

BMM contained:

Medium 199 75% (v/v)

Horse serum 20% (v/v)

Bovine Embryo Extract 4% (v/v)

Liver Fraction L (1 mg ml ^) 1% (v/v)

2.2 Chemicals and reagents

Chemicals:

Ammonium chloride, bovine serum albumin (Fraction V powder), deoxyribo­ nucleic acid, N, N-dime thylf ormamide , formaldehyde, Hoechst No. 33258

(bisBenzimide) , 2-mercaptoethanol, <*-naphthyl butyrate, pararosaniline

HC1, trypan blue, saponin ,sodium nitrite, were obtained from Sigma

Chemical Company Ltd. (Fancy Road, Poole, Dorset BH17 7NH, UK).

2-( 4 * tert.-Butylphenyl)-5-(4"-biphenylyl)-l,3,4-oxadiazole [Butyl-PBD], gelatine powder, hydrogen peroxide (3% w/v), potassium dihydrogen ortho­ phosphate, di-sodium hydrogen orthophosphate, tris(hydroxymethyl)methyl- amine were obtained from BDH Chemicals Ltd. (Broom Road, Poole BH12 4NN,

UK) .

Dulbecco’s phosphate buffered saline tablets were from Oxoid Ltd. (Wade

Road, Basingstoke, Hants. RG24 OPW, UK).

Ficoll 400 was obtained from Pharmacia (Great Britain) Ltd. (Prince

Regent Road, Hounslow, Middlesex TW3 1NE, UK). 82

Hypaque sodium (sodium ) was from Sterling Research Labora­

tories (Onslow Street, Guildford, Surrey, UK).

Toluene, Hydrochloric acid, methanol and acetone (analytical grade) were

from May and Baker Ltd. (Liverpool Road, Barton Moss, Manchester M30

7RT, UK).

Immunological reagents:

Concanavalin A was obtained as a powder from Sigma Chemical Company.

Phy t ohaemagglutinin (Reagent grade) was obtained from Wellcome Diagnos­

tics (Temple Hill, Dartford DAI 5AH, UK) as a lyophilised pellet of 45

mg which was then reconstituted in 5 ml sterile water.

Tuberculin Purified Protein Derivative (PPD) was obtained from Evans

Medical Ltd. (Greenford, Middlesex, UK) and used a concentration of 2 mg ml ^«

3 [methyl- H] Thymidine was obtained as an aqueous solution containing 25

Ci/mmol from Amersham International pic (Lincoln Place, Aylesbury,

Buckinghamshire HP20 2TP, UK).

Histochemical reagents:

3,31 Diaminobenzidine tetrahydrochloride, Fast Red TR, levamisole hydro­ chloride, Naphthol AS-MX were obtained from Sigma Chemical Company.

Liposomal reagents:

L-o<-"phosphatidylcholine (Type V-E) from frozen egg yolk in chloroform- 83

methanol solution and L-(X-phosphatidyl-L-serine from bovine brain in

chloroform-methanol solution were obtained from Sigma Chemical Company.

Staining reagents:

Cold AFB stain LMR 22, Methylene blue, Haematoxylin (Harris), Methyl

Green, Methylene blue, D.P.X mounting medium and Apathy's mounting

medium were all obtained from Raymond A. Lamb (6 Sunbeam Road, London

NW10 6JL).

Diff-Quik was obtained from American Hospital Supply, UK Ltd.(Didcot,

Oxfordshire OX 117 NP, UK).

2.3 Bacteria

2.3.1 Bacterial Strains

The bacteria used were the virulent strain, H37Rv of M. tuberculosis

obtained from Trudeau Mycobacterial Culture Collection (TMC 102),

Trudeau Institute, Saranac Lake, New York; M. microti strain 0V 254,

pathogenic for voles, obtained originally from Dr.R.J.W. Rees and

L. monocytogenes strain NCTC 9373.

2.3.2. Animal passage and maintenance

Prior to their use in the in vivo experiments, both M. tuberculosis and

M. microti were passaged through animals. L. monocytogenes was also mouse passaged. This was to counter the risk of attenuation following serial transfer on laboratory media. 84

M. tuberculosis:

M. tuberculosis H37Rv was recovered from the spleen of a guinea-pig that had been infected intravenously with the bacilli. A portion of the spleen was homogenised in 2% Bovine albumin serum in 0.85% sodium chloride (2% BSA) and plated on Middlebrook 7H11 agar plates. After two weeks incubation at 37°C , the growth was scraped into Middlebrook 7H9 broth containing 10% Albumin Dextrose Complex (ADC) and reincubated for

7 days at 37°C. The M. tuberculosis suspension was dispensed aseptic- ally into 4 ml amounts in sterile Bijou bottles and frozen at -70°C.

This suspension was used as the infecting inoculum in all subsequent experiments.

M. microti:

M. microti was injected intravenously into the tail-vein of CFLP mice and the mice were sacrificed after 12 days and the spleens removed.

The spleens were homogenized aseptically in 2% BSA and 0.1 ml of the spleen homogenate was inoculated into 100 ml quantities of Dubos broth supplemented with 10% ADC and incubated at 37°C for 4 weeks. This broth was used as the inoculum for the next serial passage and the process was repeated. After the third passage, the broths were pooled and frozen in

4 ml aliquots in sterile Bijou bottles at -70°C. This was used as the inoculum for all subsequent experimental M. microti infections in mice.

L. monocytogenes:

L. monocytogenes was passaged through BALB/c mice. The mice were injected intraperitoneally with a broth culture of L. monocytogenes.

After two days the mice were sacrificed and the spleens removed asepti­ cally. The spleens were homogenized in 2% BSA and the homogenate plated 85

onto Tryptic Soy Agar (TSA) plates. Colonies from the plates were used

to inoculate Tryptic Soy Broth (TSB) and incubated at 37°C for 8 hours.

The culture obtained was frozen at -70°C in 1 ml amounts in sterile

ampoules (Nunc cryotubes, Nunc UK Ltd., 16 Salter Street, Stafford ST16

2JU, UK). This culture was used to seed other broths for all subsequent

in vitro experiments.

M. microti for in vitro experiments:

A culture of M. microti was serially transferred at weekly intervals in

Dubos broth medium. This was used as the infecting inoculum in the in

vitro experiments with the macrophage monolayers.

2.4 Animals

2.4.1 Mouse strains

Two different strains of mice were used. Specific-pathogen free CFLP mice were obtained from Interfauna UK (Abbots Ripton Road, Wyton,

Huntingdon, UK) and specific-pathogen free BALB/c mice were obtained

from the Imperial Cancer Research Fund (ICRF).

2.4.2 Random allocation and husbandry of mice

In the in vivo animal models, the mice were allocated into the different

treatment groups by selection based on a table of random permutations of

20 numbers (Fisher and Yates, 1963). 86

The mice were kept in cages and fed on a pelleted food diet (Labsure

PRD, Labsure Company, Poole, Dorset, UK) and water ad libitum. The

cages of mice infected with M. tuberculosis were kept within a negative-

pressure isolator (Vickers Medical Isolator, Vickers Instruments, Haxby

Rd., York Y03 7SD, UK).

2.5 Immunomodulating and Chemotherapeutic Agents

2.5.1 Agents Used

Levamisole hydrochloride was kindly presented by Janssen Pharmaceutical

Ltd. (Janssen House, Marlow, Bucks SL7 1ET, UK).

Recombinant murine gamma interferon derived from Escherichia coli was generously given by Genentech Inc. San Francisco, United States of

America. The lot number was 1551/43 with a specific activity of 7.2 x

106 U/mg.

Isonicotinic acid hydrazide (Isoniazid) was obtained from Sigma Chemical

Company, Poole, Dorset, United Kingdom.

Rifampicin (Rimactane A.S Batch No. 1935410) was a gift from Ciba

Laboratories, Horsham, West Sussex, UK.

2.5.2 In vivo administration

Levamisole was dissolved in sterile distilled water and the required dose was adminstered to the mice in 0.2 ml aliquots by oral gavage with 87

the aid of a syringe and a blunted needle.

Recombinant gamma interferon was diluted in Phosphate Buffered Saline

(PBS) containing 1 mg ml ^ of homologous mouse serum immediately before

use. The required doses of interferon was intravenously injected

through the tail vein in 0.1 ml aliquots. IFN-# was also administered

intravenously in liposomes (see section 2.5.2.1).

Isoniazid was dissolved in sterile distilled water and administered to

the mice by oral gavage in 0.2 ml amounts.

Rifampicin was suspended in 0.2% Methyl Cellulose containing 0.05% Tween

80. The suspending fluid was sterilized by autoclaving at 115°C for 10

minutes. The required dose of rifampicin was administered in 0.2 ml

aliquots by oral gavage.

2.5.2.1- Preparation of liposomes

IFN-8 was administered intravenously in multilamellar vesicles (MLV)

composed of phosphatidylcholine (PC) and phosphatidylserine (PS) in a

PC:PS ratio of 7:3 moles. The required dose of IFN-)f was delivered in

2.5 jumole of lipid contained in a final volume of 0.2 ml per mouse.

The liposomes were prepared by modifications of procedures described

elsewhere (Fidler, 1980). The required amounts of PC and PS were

dispensed into a 50 ml round bottomed flask, thoroughly mixed and evaporated to dryness with the aid of a vacuum rotary evaporator

(Corning Type 349/2, Corning Ltd., Staffordshire ST15 0BG, England) at 88

room temperature in a high vacuum. The lipids were left under vacuum for 30 minutes to ensure the complete removal of all the chloroform and methanol. The lipid film was then hydrated with the IFN-^ dilution or diluent for 30 minutes at room temperature before being vortexed for 5 minutes. The resultant liposomal preparation was then injected in the tail-vein in 0.2 ml aliquots. Liposomes were prepared and used within 2 hours. No attempt was made to separate free IFN-& from liposomal IFN-& because of the uncertainty about the stability of recombinant IFN-^.

2.5.3 In vitro administration of agents

Recombinant gamma interferon was diluted in BMM just prior to being used.

Isoniazid was dissolved in pyrogen-free water to obtain a stock solution of 100 ^g ml K The final dilution of the drug (1 pg ml was made in

BMM.

Rifampicin was dissolved in N/100 Hydrochloric acid and immediately diluted in pyrogen-free water to a stock solution of 500 pg ml The final dilution of 10 jig ml ^ was made in BMM.

2.6 Preparation of infective inocula

2.6.1 Enumeration of total number of bacilli

The total number of bacilli in a suspension was determined by using a

Helber counting chamber (Thoma, Webber, England). The Helber chamber is

1/50 mm deep and has a ruled area of large squares enclosing small 89

squares whose sides measure 1/5 and 1/20 mm. Dilutions of the suspen­

sion were made in saline or HBSS and the final count was calculated from

the dilution factor and the number of organisms counted per small square by the following formula:

Number of bacilli per ml = Dilution Factor x 2n x 10^, where n is

the mean number of organisms per small square.

2.6.2 Infective inocula for in vivo experiments

M.microti : The frozen vials of bacilli were quickly thawed in a 37°C waterbath. The bacilli were pooled into Universal containers and sonicated with a sonicating probe (Rinco Ultrasonics UK Ltd., P.O.Box

217, London W5 1BL) at a setting of 70 amplitude % for 15 seconds and with the probe 1 cm below the surface. The mice were injected with 0.2 ml of this inoculum in disposable 2 ml syringes (Monoject, Sherwood

Medical, United Kingdom) with 16 x 0.5 mm sterile disposable needles

(Monoject, UK).

M. tuberculosis : The frozen vials of bacilli were rapidly thawed in a

37°C waterbath. The bacilli were pooled into sterile Universal contai­ ners and diluted 1:1 by the addition of an equal volume of sterile 0.1% gelatine saline. The suspension was then sonicated (see above) and immediately used for infecting the animals as detailed above.

2.6.3 Infective inocula for in vitro experiments

L. monocytogenes : The infective inocula were prepared fresh for each experiment. A frozen vial of passaged bacilli was rapidly thawed in a 90

37°C waterbath and 100 pi of the thawed suspension was inoculated into

15 ml of TSB and incubated at 37°C for 8 hours. The resultant log phase culture was used to infect the monolayers. The suspension was centrifuged at 1000 g (MSE Chilspin 2, MSE Scientific Instrument, Manor

Royal, Crawley, West Sussex) for 15 minutes after which the supernatant was decanted and the bacilli resuspended in HBSS-HEPES . Dilutions were then made and the total count was determined in a Helber chamber

(section 2.6.1). The inoculum was diluted to a concentration lOOx that of the final concentration needed. 100 of the suspension was opson­ ized by incubating with 25 ul of sterile normal mouse serum at 37°C for

30 minutes. At the end of the opsonization period, the required concen­ tration of the infective inoculum was obtained by diluting lOOx with

Medium 199.

M. microti : A culture of M. microti serially transferred in Dubos broth was used for the in vitro experiments. A 6 day culture of the strain was centrifuged for 15 minutes at 1000 g (MSE Chilspin 2). The supernatant was decanted and the pellet gently resuspended in HBSS-

HEPES. The centrifugation step was repeated and the bacteria resuspended in HBSS-HEPES. The bacterial suspension was then sonicated and the total count determined in a Helber chamber. The required infective inoculum was then obtained by diluting in Medium 199 contain­ ing 10% heat-inactivated FCS. 91

2.7 Investigation of the T lymphocyte response

2.7.1 Proliferative response of splenic T lymphocytes

2.7.1.1 Preparation of splenic lymphocytes

Spleens from three mice were aseptically removed and gently teased apart

with forceps over a wire mesh in a sterile 20 mm plastic petri dish

filled with media (RPMI 1640 with 20% Foetal Calf Serum). The resulting

suspension was layered gently over Ficoll-Hypaque (65 g Ficoll and 100 g

Hypaque in 1000 ml water) in a universal container. The universal was

centrifuged at 900 g at 4°C and the band of cells at the interface was

collected. These cells were washed twice with media by centrifugation

at 150 g at 4°C and the cell pellet was resuspended in the final medium

of RPMI 1640 supplemented with 20% Foetal calf serum, 2mM L-glutamine,

5 x 10 2-mercaptoethanol (2-ME) and 1% Antibiotic-antimycotic mixture

(see section 2.1.2). The lymphocytes were diluted in 0.2% Trypan blue

and enumerated in an improved' Neubauer haemocytometer (section 2.9.2)

2.7.1.2 Antigens and mitogens

The mitogens used were phytohaeraagglutinin (PHA) and concanavalin A (Con

A). PHA was used at a dilution of 1/20 (stock solution of 9 mg ml ^)

and Con A at a concentration of 10 ;ug ml The specific antigen used was Tuberculin Purified Protein Derivative (PPD) at the concentrations

r —A _ O — O — 1 ™ 1 1x10 , 1x10 , 1x10 , 1x10 , and 1x10 ;ug ml 92

2.7«1.3 Lymphocyte Proliferation test

The proliferative responses of the T lymphocytes were determined by a micro method using terasaki plates (O'Brien et al., 1979). The splenic lymphocytes were suspended at various cell concentrations in the final medium of RPMI 1640 supplemented with 20% Foetal calf serum, 2mM L-glut- amine, 5x10 2-ME and 1% Antibiotic-antimycotic mixture. Cell concen­ trations of 1x10^ and 3x10^ per well were used for the PHA and Con A 5 6 6 assays and 5x10 , 1x10 and 3x10 for the PPD assay.

2 pi of the antigen/mitogen in various concentrations were first distri­ buted to the microterasaki plates (Nunc UK Ltd.) followed by 20 pi aliquots of the cell suspension dilutions. These aliquots were dispensed by the use of a programmable microdispenser (Microlab P,

Hamilton Company, USA). The plates were then inverted and the cells cultured on hanging menisci in humidified chambers at 37°C with 5% CO2 •

Each assay was set up in triplicates.

Cells stimulated with PHA and Con A were incubated for 3 days and the 3 PPD assays for 6 days. Before harvest, 0.5 Ci of [ H] thymidine were added to each well. Plates were then reincubated for 4 hours after which the cells were harvested using a precut filter block onto filter discs and were washed sequentially for 30 seconds in PBS, 5% Trichloro­ acetic acid, and methanol.

The discs were removed into plastic insertion vials and allowed to dry.

They were then dissolved in 1 ml of scintillating fluid (Toluene 3 containing 0.5% butyl PBD. Incorporation of [ H] thymidine was counted 93

in a liquid scintillation counter (SL 4000, Intertechnique, France) and the results were expressed as mean counts per minute (cpm) + s.e. for triplicate cultures.

2.7.2 Immunoenzymatic staining of splenic T lymphocyte subsets

Splenic T lymphocytes were isolated and cytospin preparations made for immunoperoxidase and immunoalkaline phosphatase staining.

2.7.2.1 Antibodies and sera

Monoclonal antibodies:

1. Mouse anti-mouse Thy 1.2

2. Mouse anti-mouse Lyt-1

3. Mouse anti-mouse Lyt-2

The monoclonal antibodies were obtained as 0.5 mg purified immunoglo­ bulin in 0.5 ml buffered saline from Becton Dickinson, Laboratory Impex

Ltd. (Impex House, Lion Road, Twickenham, Middlesex TW1 4JF, UK).

Other antibodies:

Peroxidase-conjugated rabbit anti-mouse IgG (Sigma Chemical Company)

Alkaline phosphatase-conjugated rabbit anti-mouse IgG (Sigma)

Serum:

Normal mouse serum (Sigma) 94

2.7.2.2 Cytospin preparations

Splenic lymphocytes were prepared as described in section 2.7.1.1. The lymphocyte suspension was counted (see section 2.9.2) and resuspended at a concentration of 1x10^ cells ml ^ . 100 }il of the cell suspension together with 25 ^pl of FCS was spun down on clean glass slides in a cytocentrifuge (Shandon Cytospin 2, Shandon Southern Products Ltd.,

Astmoor, Cheshire, UK) at 500 rpm for 5 minutes.

The cell spots were allowed to dry at room temperature overnight, indi­ vidually wrapped in aluminium foil and stored at -20°C until stained.

It had been shown by Moir et al., (1983) that slides treated in such a manner retained antigenic activity even after several months of storage.

2.7.2.3 Preparation of buffers, fixative and substrates

Buffers:

5x Concentrated buffer consisted of 2g Na^HPO^ and 10 g KH^PO^ in 60 mis of distilled water.

Tris HC1 buffer (pH 7.6) and tris HCl buffer (pH 8.2) were prepared by dissolving Tris base (0.05 M) and Tris base (0.1 M) in distilled water respectively and adjusting the pH with concentrated HCl.

Tris-buffered saline (TBS) was prepared by adding Tris HCl buffer (pH

7.6, 0.05 M) to normal saline (0.15M). TBS was used to dilute all reagents unless stated otherwise. 95

Fixative:

Buffered formol acetone (BFA) was made fresh each time.

It consisted of: 8 ml of 5x Concentrated buffer

138 ml of distilled water

33.2 ml of formalin

60 ml of acetone

Substrates:

Peroxidase substrate. Immediately before staining, 3,3' diaminobenzi- dine tetrahydrochloride was dissolved in TBS at a final concentration of

0.6 mg ml ^ and hydrogen peroxide (10 volumes, 3% w/v) added to give a final concentration of 0.01 %.

Alkaline phosphatase substrate. 2 mg Naphthol AS-MX was dissolved in

200 >il N,N-*dimethylformamide in a glass universal and 9.8 ml of Tris HC1 buffer (pH 8.2, 0.1M) was added. Immediately before staining, Fast Red

TR was dissolved in this solution at a final concentration of 1 mg ml ^ together with levamisole (final concentration 1 mM). The substrate was filtered through Whatman No.l filter paper directly onto the cell spots.

2.7.2.4 Immunoenzymatic staining technique

The slides were removed from the freezer and allowed to thaw at room temperature. The cell spots were stained by an indirect immunoperoxi- dase technique and an indirect immunoalkaline phosphatase procedure

(Moir et al., 1983). The cell spots were fixed for 30 seconds in BFA , rinsed in distilled water and then in TBS. They were then stained by either technique. 96

Indirect Immunoperoxidase procedure:

The slides were flooded with the optimal dilution of the monoclonal antibody and incubated for 60 minutes at room temperature in a humidified chamber to prevent drying of the cell spots. The slide was then washed with TBS and incubated at room temperature for 30 minutes with peroxidase-conjugated rabbit anti-mouse IgG (1/50 dilution) with normal mouse serum at a final concentration of 1/20 (to block cross­ reactivity against mouse IgG). The slide was again washed in TBS and the reaction developed with diaminobenzidine-H^O^ substrate. The devel­ opment of the reaction was monitored with a light microscope (x200 magnification) and the reaction, was stopped by washing in tap water.

The slide was then counterstained very briefly with dilute haematoxylin

(1/6 dilution) and mounted in DPX.

Indirect immunoalkaline phosphatase procedure:

The technique was essentially similar to the procedure for the immuno­ peroxidase staining as described above except that the second incubation was carried out with alkaline phosphatase-conjugated rabbit anti-mouse

IgG and the reaction was developed with alkaline phosphatase substrate.

The substrate was filtered onto the slide and the reaction monitored microscopically. The reaction was then stopped by rinsing in tap water.

The slide was briefly counterstained with dilute haematoxylin and mounted in Apathy’s mountant. 97

2.8 Assessment of growth of mycobacteria in vivo and in vitro

2.8.1 Homogenization of organs

The mouse was killed by cervical dislocation, immersed briefly in 2%

Clearsol and pinned ventral aspect upwards on a cork dissecting board.

The skin was opened up and deflected to either side. The abdominal musculature was swabbed with 70% alcohol and allowed to dry. The abdominal wall was then incised with sterile scissors right to the top to enable easy removal of the lungs. The spleen and lungs were removed aseptically with sterile instruments into labelled sterile precision- bore homogeniser tubes made of hard glass with a diameter of 16 mm.

The organs were ground with a motor-driven homogeniser using PTFE grinders and glass-homogeniser tubes. The PTFE grinder and the homo­ geniser tubes were separately autoclaved and assembled together just before homogenization. All the organs were ground in a final volume of

5 ml of a diluent, 2% bovine serum albumin in 0.85 % sodium chloride

(BSA). The diluent served the purpose of protecting the bacilli from the toxic substances released by grinding or autolysis of the tissues

(Pierce et al., 1953). For the same reason, all further dilutions were made in 0.1% BSA. During homogenization, the tube was pushed up and pulled down the revolving PTFE grinder about 10 times for spleens and about 20 times for lungs to ensure complete disruption of the tissues.

Homogenates were kept at 4°C until dilutions were made to determine the number of viable mycobacteria. 98

2.8.2 Enumeration of viable M. tuberculosis and M. microti

The numbers of viable bacilli present in the infective inocula and the

homogenates of organs were determined by plating appropriate dilutions

for surface plate counts on suitable media.

The culture media used was Middlebrook 7H11 Oleic acid-albumin agar or

Selective 7H11 agar for M. tuberculosis and Dubos Oleic Base Agar for

M. microti. All media were incubated at 37°C for 24 hours before use to

check for sterility and to ensure that the surface was sufficiently dry.

Appropriate ten-fold dilutions of the homogenates were made in 0.1% BSA

and 100 pi aliquots were pipetted on 1/3 segments of the agar plate in

duplicate. The inocula were allowed to dry into the agar and the plates

were then packed in polythene bags and incubated at 37°C.

7H11 and Selective 7H11 plates were incubated for 3-4 weeks before the

number of colonies were counted. Plates of M. tuberculosis were exposed

to formalin vapour overnight before being enumerated. Dubos Oleic Base

Agar plates were sealed by parafilm before being packed into polythene

bags and were enumerated after 4 weeks incubation at 37°C.

2.8.3 Enumeration of L. monocytogenes and M. microti in macrophage monolayers

The number of viable bacteria per macrophage monolayer was determined either by scraping the macrophages from the tissue culture wells or by lysing the monolayer with 1% saponin and then performing a viable count. 99

After the monolayers had been harvested from the tissue culture wells by either method, the cell lysates were placed Into sterile tubes and submitted to a brief sonication step to ensure that all the cells were lysed and that the bacilli were not clumped.

The determination of the viable count was accomplished by a method similar to the one described above. L. monocytogenes was grown on TSA plates and M. microti on Dubos Oleic Base agar. The TSA plates were enumerated after overnight incubation at 37°C.

2.9 In vitro techniques used in the macrophage microbicidal assays

2.9.1 Preparation of monolayers

The mice were sacrificed by cervical dislocation, pinned ventral aspect upwards on a cork board and the abdominal region liberally swabbed with

70% alcohol. The skin was incised and deflected to the left side of the body. 3 ml of ice-cold RPMI 1640 containing 10 units of heparin per ml were injected slowly into the peritoneal cavity with a 21 gauge needle.

The lavage fluid was carefully injected by first going obliquely through the abdominal wall musculature before penetrating the peritoneum. This ensured that there was no leakage. The peritoneal cavity was gently massaged for a few minutes and then the lavage fluid was removed by drawing back into the syringe. The peritoneal cells from several mice were pooled, kept on ice and were centrifuged at 4°C at 150 g (MSE

Chilspin 2). The cells were resuspended in Medium 199 containing 10% heat-inactivated FCS. The cell suspension was then enumerated (see section 2.9.2), diluted to the required concentration and aliquoted in 100

500 pi or 200 ;ul amounts Into either 16 mm tissue culture multidishes

(Linbro Space Saver, Flow Laboratories) or Lab-Tek 8-Chambered Tissue

culture slides (Miles Scientific Laboratories, Stoke Poges, Slough SL

4LV, UK) respectively. The cell suspension was incubated at 37°C in a

CO2 incubator (5% CO^) for 2-3 hours to enable macrophages to adhere.

At the end of this time, the plates were gently agitated, the superna­

tants removed and the monolayers were washed twice with 500 ;ul amounts

of warm Medium 199 containing 5% FCS to ensure the removal of all non­

adherent cells. The monolayers were then overlaid with 500 ;ul amounts

of BMM and reincubated.

2.9.2 Enumeration of cell suspensions

The enumeration of cells (macrophages, lymphocytes) in a suspension was

achieved by adding 25 til of the cell suspension to 225 til of tris-

ammonium chloride and 250 til of 0.2% Trypan Blue (1/20 dilution). The

tris-ammonium chloride lyses any erythrocytes present and the trypan

blue distinguishes dead from living cells by dye exclusion. After

thorough mixing, the dilution was transferred to an improved Neubauer haemacytoraeter chamber and counted microscopically by phase contrast.

The improved Neubauer chamber consists of 25 large squares, each divided 3 into 16 smaller squares. The large square has a volume of 0.004 mm ,

and the number of cells are calculated from the formula: 4 No. of cells per ml= Dilution factor x n x 10 , where n is the

number of cells in 25 large squares 101

2.9.3 Characterization of macrophage monolayers

Characterization of the monolayers were performed on those set up in the

8-Chamber tissue culture slides. At required intervals, some monolayers

were differentially stained by a modified Wright's stain (Diff-Quik) and

for non-specific esterase (Koski et al., 1976).

Diff-Quik Staining:

Reagents : Diff-Quik solutions I and II and fixative

Procedure : The supernatant was aspirated and the monolayers washed by

warm HBSS-HEPES. The chamber component was removed and the slide was

immersed in fixative for 15 seconds followed by 15 seconds in Diff-Quik

I and 30 seconds in Diff-Quik II. It was then thoroughly washed, air

dried, mounted in DPX and examined under a light microscope.

Non-specific esterase staining:

Reagents:

1. Fixative (pH6.6) containing 20 mg Na2HP0^, 100 mg KI^PO^, 45 ml

acetone, 25 ml formaldehyde (30%) and 30 ml distilled water.

2. Pararosaniline solution containing 1 g pararosaniline HC1 in 25

ml 2N HC1.

3. 4% Sodium nitrite (freshly prepared)

4. M/15 Sorenson's phosphate buffer (pH 6.3)

5. c^-Naphthyl butyrate solution containing 1 g o<-Naphthyl butyrate

in 50 ml dimethyl formamide.

6. 0.5% methyl green counterstain.

Procedure:

The monolayers were washed with warm HBSS-HEPES, the chamber component 102

discarded and the slide was immersed in fixative for 30 seconds, rinsed thoroughly in distilled water and air-dried for 30 minutes. Hexazotiza- tion of pararosaniline was carried out by adding 1 ml of filtered para- rosaniline solution to 1 ml of sodium nitrite and leaving the mixture for 1 minute. The reaction mixture was then prepared by adding in sequence 44.5 ml M/15 phosphate buffer, 0.25 ml hexazotized pararos­ aniline and 3 ml cX-naphthyl butyrate solution. After immersion of the slide in this mixture for 45 minutes at 37°C, it was rinsed thoroughly in distilled water, counterstained briefly in methyl green and rinsed in distilled water. It was then air-dried, mounted in DPX and examined mi croscopi cally.

2.9.4 Harvesting of bacteria from the monolayers

Two methods were used to harvest bacteria from the adherent macrophage monolayers :

1. HBSS-HEPES was added to the tissue culture vessel and the monolayer was physically scraped off the well with the aid of a specially designed autoclavable PTFE-tipped rod. This procedure was repeated to ensure that all the cells were scraped off. The process was also monitored with an inverted microscope. This method allowed the DNA content and hence the number of cells in the monolayer to be determined.

2. 1% saponin was added to the culture chamber which was then reincu­ bated for 20 minutes at 37°C. At the end of this time, the cells were easily removed by gently scraping with the blunted ends of sterile 1 ml serological pipettes. HBSS-HEPES was then added and the process repeated. The complete removal of all the cells was confirmed by 103

observation under an inverted microscope.

2.9.5 Analysis of DNA

The analysis of DNA content of cell lysates was determined for some of the samples and used as a means of quantitating of the number of cells in the monolayer. DNA analysis was carried out by a simple and rapid assay based on the enhancement of fluorescence seen when bisbenzimid- azole (Hoechst Compound 33258) binds to DNA (Labarca and Paigen, 1980).

The assay can detect as little as 10 ng of DNA. The number of macro­ phages can be established from the conversion factor that 1x10^ murine peritoneal macrophages has 7.5 jug of DNA (Lowrie et al., 1979a).

Reagents:

1. 0.05M phosphate buffer pH 7.4 made 2M in NaCl

2. 0.10M phosphate buffer pH 7.4 made 4M in NaCl

3. Hoechst compound 33258, stock solution in DMSO, used at 5 ;ig

ml ^ with 0.05M phosphate buffer

4. DNA standards prepared in 5 mM NaOH

Procedure:

The monolayers were scraped into 500 jul of HBSS-HEPES and 500 jul of

0.10M phosphate buffer and 250 jul of H33258 (5 jug ml ^) were added.

Standards were set up with 0-20 jug ml ^ of DNA in HBSS-HEPES in exactly the same way. The mixture was vortexed and left for 3 hours for penetration into the cells. Fluorescence was read with a Perkin Elmer

1000M Fluorimeter with an excitation filter of 365 nm and an emmission wavelenght of 456 nm. Results were read off a DNA standard curve. 104

2.10 In vivo models In the assessment of immunomodulating agents

2.10.1 Effect of levamisole on M. microti infection in CFLP mice

The model of an infection of M. microti in CFLP mice was used to assess

the effects of levamisole in vivo. The effects of levamisole on the

infection of M. microti was monitored in two ways : firstly, the effect

on the growth of M. microti in the lungs and spleens of the infected

animals by performing viable counts (see section 2.8.2); and secondly,

the effect on the splenic T lymphocytes of uninfected and infected mice

as measured by the proliferative response of T lymphocytes (section

2.7.1) and immunoenzymatic staining of subsets of T lymphocytes (see

section 2.7.2).

2.10.2 Effect of interferon-gamma on M. tuberculosis infection in mice

The model of an acute infection of M. tuberculosis H37Rv was used to assess the effects of the IFN-Y on the growth of M. tuberculosis in the whole animal. In all the experiments, mice were infected with animal- passaged M. tuberculosis (section 2.6.2).

The mice were infected by intravenous injection through a tail vein by the use of a specially designed mouse restrainer. The size of the infective dose was decided by a preliminary experiment in which diffe­ rent dilutions of the passaged H37Rv were used to infect mice and the outcome of the infection was then monitored by performing viable counts of the bacilli in the lungs and the spleens (see sections 2.8.1 and

2 .8 .2). 105

The effects of size of dose of IFN-tf was investigated. This was done by randomly allocating mice into different groups receiving different doses of IFN-Jf. Several schedules of administration of IFN-& were also tested to determine the optimal schedule. The feasibility of using MLV lipo­ somes as drug delivery vehicles was also investigated. Different dilu­ tions of IFN-fr were encapsulated within liposomes (see section 2.5.2.1) and were intravenously administered to the mice.

Assessment of the effect of IFN-'fc was achieved by performing viable counts of M. tuberculosis in the lungs and the spleens of the animals at selected time points after infection. The number of viable bacilli present in the inoculum was determined in each experiment by performing the viable count at infection. The uptake of bacilli by the organs in each experiment was determined by performing viable counts of the lungs and spleens of 4-6 mice an hour after intravenous infection.

2.10.3 Effect of interferon-gamma in combination with isoniazid and with rifampicin on M. tuberculosis infection in mice

A series of experiments were performed to determine the effect of IFN-Jf in combination with antibacterial agents of tuberculosis. A model of infection with M. tuberculosis was used.

In the first set of experiments, mice were randomly allocated into groups that received either IFN-tf, isoniazid, isoniazid and IFN-V or were untreated. IFN-ft was given at a dose of 2000 units per mouse while isoniazid was given at a daily dose of 25 mg kg In one experiment,

IFN-J was given both before and after infection, and isoniazid was 106

started at Day+1. In another experiment, IFN-# was either given both before and after infection or only 5 days after infection. The effects of each treatment were assessed by determining the viable counts of

M. tuberculosis in lungs and spleens.

In another experiment, mice were allocated into groups which received either IFN-tf , rifampicin, IFN-Jf and rifampicin or were untreated. Two doses of IFN-if at 2000 units per mouse were given at Days -2 and +1.

Rifampicin was given at 25 rag kg ^ per day with the first dose on Day 0.

The efficacy of each treatment group was ascertained by the growth of

M. tuberculosis in the lungs and the spleens.

2.11 In vitro assessment of the effects of interferon-gamma

The in-vi t ro assessment of the effects of interferon-gamma were all performed on murine resident peritoneal macrophages. Monolayers were established in 16 mm tissue culture multi-well plates and 8-chamber tissue culture slides (see section 2.9.1). In all of the experiments, triplicate wells were used and the experiments were performed at least twice unless stated otherwise.

2.11.1 Effect of saponin on the viable count of L. monocytogenes and

M. microti

In some experiments, saponin was used to lyse the macrophage monolayers and it was thus important to determine if saponin had any effect on the viable counts obtained. 107

To investigate the effect of saponin on L. monocytogenes and M. microti, experiments were performed to compare the viable counts of bacterial suspensions left in room temperature in distilled water, 1% saponin or

2.5% saponin for at least two hours. The suspensions were then briefly sonicated (M. microti only) and dilutions made in distilled water and viable counts performed (see sections 2.8.2 and 2.8.3).

2.11.2 Listericidal assay

The listericidal assay used was based on the method of Harrington-Fowler e t a l . (1981) with several modifications. One major modification was that no antibiotics were used at any stage of the assay.

The macrophage monolayers were prepared in 16 mm Linbro wells (section

2.9.1). In this assay, the monolayers were pre-incubated with various concentrations of IFN-X in 3MM or with BMM for either 24 or 48 hours prior to infection. Treated and untreated (control) monolayers were infected with L. monocytogenes at a bacteria:macrophage ratio of 1:10.

The infective inoculum was prepared as described in section 2.6.3. The wells were washed twice with warm HBSS-HEPES and the inoculum in a volume of 500 jjl was added to each well and the plates incubated at 37°C to allow phagocytosis to occur. After 40 minutes, the supernatant was aspirated and the monolayers washed five times with warm HBSS-HEPES to remove non-attached bacteria. The last rinse was cultured to give an indication of the level of viable bacteria In the supernatant at the begining of the experiment. The replicate monolayers which were to represent the number of cell-associated bacteria after phagocytosis (T^) were then lysed while the rest of the monolayers were reincubated with 108

Medium 199 containing 10% heat-inactivated FCS. These monolayers were

incubated at 37°C with 5% CO^ for a further four hours, with the media

being changed every half hour. At the end of four hours, all the

monolayers (T^) were lysed with 1% saponin (see section 2.9.4) and the

lysates diluted and plated on TSA (section 2.8.3) to determine the

viable count. The experiments were done with duplicate monolayers.

2.11.3 Effect of interferon-gamma on growth of M. microti in vitro

In these experiments, macrophage monolayers were either pre-incubated

with IFN-X before and after infection with M. microti or were infected

and then treated with IFN- .

2.11.3.1 Effect of previous exposure of monolayers to interferon-gamma

In these series of experiments, monolayers were previously exposed to

various concentrations of IFN-K for either 48 or 72 hours. Monolayers

in 16 mm Linbro wells were infected with M. microti at a macrophage:

bacteria ratio of 1:1. The infective inocula were prepared as in

section 2.6.3. The monolayers were washed twice with warm HBSS-HEPES

and infected with the required number of bacilli in a volume of 500 pi per well. The wells were then incubated at 37°C with 5% CO2 to allow

phagocytosis to occur. Phagocytosis periods of 15, 30, 60, 90 and 120 minutes were used in the different experiments.

After the phagocytosis period, the wells were washed four times with warm HBSS-HEPES to remove non-attached bacteria. This was checked with

an inverted phase-contrast microscope and the last rinses were cultured 109

to determine the level of viable bacteria in the supernatant at the end

of the phagocytosis period. The monolayers representing (immediately

after phagocytosis) were then lysed and the other monolayers were

reincubated at 37°C with 5% CO^ for a further 24, 48 or 72 hours with

either BMM and IFN-S or BMM alone. The medium was changed every day by

removing the old medium and adding fresh BMM with and without IFN-^. In

these series of experiments, both the scraping method and the saponin method were used to disrupt the monolayers and release the bacteria

(section 2.9.4). The viable counts of the M. microti were determined as

described in section 2.8.3. The number of cells per monolayer was

determined by assaying the DNA content of the cell lysates for those

experiments where the scraping method was used (section 2.9.5).

2.11.3.2 Effect of interferon-gamma on the phagocytosis of M. microti

Some experiments were performed to investigate the effect of previous

exposure of monolayers to interferon-gamma on the uptake of bacilli.

The monolayers were set up in 8-chambered tissue culture slides. After

infection with a bacteriarmacrophage ratio of 10:1 and a phagocytic

period of 2 hours, some of the wells were assessed for the viable count

and the others were stained for acid-fast bacilli.

Viable mycobacterial counts were obtained by lysing the macrophages with

1% saponin and plating dilutions of the lysates on Dubos Oleic Base agar plates (see section 2.8.3).

Acid-fast bacillary counts were obtained by staining the monolayers with a cold Ziehl-Nielsen method. The monolayers were washed once with HBSS- 110

HEPES and fixed with methanol for 5 minutes. The chambers were then

removed and they were rinsed with HBSS-HEPES, air-dried and stained for

5 minutes with Cold AFB stain LMR 22. This was followed by decolouriza-

tion with acid-alcohol and counterstaining with 2% methylene blue. The

slides were then mounted with D.P.X. Mounting Medium and enumerated

under oil-immersion. Macrophages were enumerated in a random way by

utilizing the vernier scales on the microscope stage to scan fields at

regular intervals enabling all sections of the monolayer to be examined.

300 macrophages were counted per monolayer and scored as the number of macrophages containing no acid-fast bacilli, the number with 1-2

bacilli, the number with 3-5 bacilli and the number with more than 5

bacilli. For the acid-fast counts, the experiments were done with 6

replicates and for the viable counts, triplicate wells were used.

2.11.3.3 Effect of interferon-gamma added after infection of monolayers with M. microti

In this series of experiments, monolayers were first established for 48 hours in 16 mm Linbro wells and then infected with M. microti at a bacteria:macrophage ratio of 1:1 for 120 minutes. At the end of the phagocytosis period, the monolayers were thoroughly washed 4 times with

HBSS-HEPES to remove all extracellular bacteria. The Tq monolayers were then lysed to determine the number of bacilli phagocytosed and the remaining monolayers were reincubated with various doses of IFN-JT at

37°C with 5% CC^* At various time points after infection (24, 48 and 72 hours) the monolayers were assessed for the number of viable mycobact­ eria (see section 2.8.3). Media was removed and fresh BMM with and without IFN-^ was added daily. In some experiments, the medium that was Ill

removed was pooled and cultured for the number of viable bacteria.

2.11.3.4 Effect of interferon-gamma in combination with isoniazid and with rifampicin on the growth of M. microti in macrophages

A series of experiments were carried out to investigate the effect of interferon-gamma in combination with either isoniazid or rifampicin on the growth of M. microti in macrophage monolayers.

In these experiments, the monolayers were established in 16 mm Linbro wells for 48 hours before infection. A macrophage:bacteria ratio of 1:1 was used. Phagocytosis was allowed to proceed for 90 minutes and the wells were thoroughly washed four times to remove non-attached bacteria and the last rinses were cultured to give an indication of the number of viable bacteria in the supernatant before the start of the experiment.

The Tq monolayers were then scraped off the wells to determine the number of M. microti taken up by the macrophages (section 2.8.3) and the remainder of the monolayers were reincubated at 37°C in 5% CO^*

The dose of IFN-K used was 100 units ml ^. Isoniazid was used at 1 jig ml ^ and rifampicin at 10 jjg ml The media containing the antibact­ erial agents were prepared as described in section 2.5.3, and were replaced every 24 hours with fresh media. In these experiments, the media that was removed was pooled and cultured to give an estimate of the viable bacteria in the supernatants at all the time points. At 24,

48 and 72 hours after infection, the monolayers were lysed with saponin and the number of viable mycobacteria was determined (section 2.8.3). 112

2.12 Statistical analysis of the data

Standard error of the mean (s.e.m.) was used to measure the precision of the means of the different experimental groups.

The analysis of variance (ANOVA) is a technique for performing multiple simultaneous comparisons. The analysis of variance was used to evaluate the results obtained in both the in vivo and the in vitro experiments.

Two-way and three-way analyses were used, depending of the number of variables in the experiments.

Analysis of variance was carried out by log^ transformation of the data which were then entered into a statistical package computer programme

(Mini tab) for the two-way analysis or GLIM (General Linear Interactive

Modelling) for the 3-way analysis. The variance ratio F was calculated and referred to F ratio tables (Fisher and Yates, 1963) for the prob­ ability of it being significant. 113

CHAPTER 3

THE IMMUNOMODULATING EFFECT OF LEVAMISOLE

ON M. MICROTI INFECTION IN MICE

This Chapter reports the effects of levamisole on a M. microti infection

in mice. The immunomodulation by levamisole is assessed by monitoring

the growth of the bacillus in organs of the mice and the effect on

splenic T lymphocyte subsets and proliferation.

3.1 Preliminary experiments

3.1.1 Choice of media for growth of M. microti

A variety of media and conditions were tested to select the best media

and conditions for performing viable counts with M. microti. The media

tested were:

1) Middlebrook 7H11 agar with and without 0.5% glycerol

2) Middlebrook 7H11 agar with and without 5% lysed horse blood

3) Dubos Oleic Base agar with and without 5% lysed horse blood

4) Selective 7H11 agar with and without 5% lysed horse blood

The media were prepared as described in section 2.1.1 and dried for 24 hours before use. A culture of M. microti was incubated at 37°C for 6

days in Dubos broth. The suspension was sonicated (section 2.6.2) and

10-fold dilutions in saline were made. 100 jil of the dilutions were plated in duplicate on all the different media (see section 2.8.2). The media were then incubated under a variety of conditions. The conditions 114

tested Included sealing with parafilm, incubation in air or with 5% CC^ at 37°C. Before incubation, all the plates that were incubated in air were bagged in polyethylene envelopes. The plates were examined at weekly intervals and counts were made when the colonies were large enough. The results are shown in Table 3.1.

TABLE 3.1

VIABLE COUNTS OF M. MICROTI ON VARIOUS MEDIA UNDER DIFFERENT CONDITIONS

Media L o g ^ c^u *** microti per ml* for condition: tested Air Air

5% C02i sealed unsealed

7H11 agar 7.95 (5w) 6.82 (8w) 6.78 (8w)

7H11 agar + 5% blood 8.34 (3w) 8.32 (5w) CT

Selective 7H11 agar 8.20 (4w) 8.04 (5w) 8.00 (6w)

Selective 7H11 agar + 5% blood 8.26 (4w) 8.18 (5w) CT

7Hllg agar 7.60 (5w) 6.20 (8w) 6.35 (8w)

7Hllg agar + 5% blood 8.18 (4w) 8.26 (5w) 8.26 (6w)

Selective 7Hllg agar 8.15 (4w) 6.83 (8w) 6.53 (8w)

Selective 7Hllg agar + 5% blood 8.23 (4w) 8.20 (5w) 8.20 (6w)

Dubos oleic agar 8.23 (3w) 8.32 (4w) 8.28 (5w)

Dubos oleic agar + 5% blood CT 8.32 (4w) 8.32 (4w)

* Figures in parentheses indicates the length of incubation in weeks

7H11 agar refers to Middlebrook 7H11 agar prepared without glycerol

7Hllg refers to Middlebrook 7H11 agar prepared with 0.5% glycerol

CT = contaminated plates 115

The results showed that Dubos Oleic base agar with and without 5% horse blood supported the growth of M. microti equally well and that counts could be made within 4 weeks on plates sealed with parafilm. The results of the viable counts obtained using Middlebrook 7H11 agar or

Selective Middlebrook 7H11 agar with or without glycerol were variable.

Some plates gave very low counts even after prolonged incubation. The low counts observed on 7H11 and selective 7H11 agar were abrogated by the addition of 5% lysed horse blood. The addition of lysed horse blood also hastened the growth rate on 7H11 plates as did incubation with 5%

CC>2; however both these conditions increased the contamination rate.

0.5% glycerol had no effect on the growth of this strain.

After taking into consideration all the above results, it was decided to perform viable counts of M. microti with Dubos Oleic base agar and to seal culture plates with parafilm and incubate for 4 weeks at 37°C.

3.1.2 Course of infection of M. microti in CFLP mice

The course of infection of M. microti was investigated in CFLP mice. A volume of 0.2 ml of the inoculum of M. microti that had been mouse- passaged (see sections 2.3.2 and 2.6.2) was injected intravenously into each of 54 female CFLP mice weighing 18-20 g at the begining of the experiment. From viable counts set up at infection, the infective dose was 1.4 x 10^ cfu of M. microti. The infection was monitored over 106 days by performing viable counts of the lungs and spleens; 5 mice were sacrificed at each time point. The viable count results (Figure 3.1) showed that the mouse-passaged M. microti was capable of multiplying in

CFLP mice. In the lung, there was an initial decrease in the counts . irt i te ug (•) ad h sles A o 5 mice. of (A) spleens the and ) • ( microti in lungs the M. i 31 rwho . irt n ug ad pen o FP ie Mice Growth were infected microti with mice.M.CFLPof of spleens and 3.1in lungs Fig 1.4 x 10® viable organisms per mouse on Day 0. Each point represents the mean ± s.e cfu the means.e ± Each point represents 10® x mouse 0. Day viableon per organisms 1.4 Mean log. viable mycobacteria per organ asatr infection after Days 117

followed by a period of exponential growth for 4-5 weeks after which there was a period of a very gradual increase till Day 106. The mice sacrificed on Day 106 were apparently healthy but upon post-mortem, the lungs were full of necrotic lesions. The counts in the spleen gradually increased until Day 48 and then began to decrease. Overall, the results suggest the emergence of appreciable immunity in the 4th and 5th weeks of infection which substantially reduced bacillary growth. The outcome was eventually fatal after approximately four months (results not shown).

From these results it was decided to infect CFLP mice with this infec­ tive dose size and allow the infection to proceed for 10 days, when in vivo growth was most rapid in the lungs and spleen, before commencing levamisole treatment.

3.2 The effect of levamisole on M. microti infection in CFLP mice

150 female CFLP mice, 20-22 g at the begining of the experiment, were randomly allocated into six groups. Three groups were infected with mouse-passaged M. microti (see section 2.6.3) by intravenous tail vein inoculation. One group consisted of the untreated controls, another received 2.5 mg kg ^ of levamisole twice a week and the third group received 25 mg kg ^ twice weekly. The remaining three groups of mice were kept as uninfected controls, one group being the uninfected untreated controls and the other two groups receiving 2.5 and 25 mg kg ^ of levamisole twice weekly respectively. Levamisole was administered by oral gavage twice weekly for 5 weeks with the first dose on Day +10.

The effect of levamisole on the growth of M. microti was monitored by 118

performing organ counts on Days +3, +7, +15, +22, +29, +36 and +43. The effect on splenic T lymphocytes was assessed by the proliferative res­ ponse to PPD, PHA and Con A before treatment started, half-way through and at the end of treatment. The subsets of splenic T-cells were also determined at the same time points by immunoenzymatic staining. In the immunological tests, comparison was also made with the uninfected controls.

3.2.1 The effect of levamisole on the growth of M. microti

4 mice were sacrificed for each time point. Viable counts set up at infection showed that the infective dose was 2.4 x 10^ cfu per mouse.

The results of the viable counts in the lungs and spleens are shown in

Figure 3.2 and Figure 3.3 respectively. For clarity, the standard errors are not depicted in the Figures but full details are shown in

Tables 3A.1 and 3A.2 in Appendix 1. It was evident from the Figures that there was not much difference between the growth of M. microti in the untreated and the levamisole treated groups in both lungs and spleens. This was confirmed by analysis of variance of the results

(Table 3.2) which showed that only time (Days) had a highly significant effect on the growth of M. microti in both lungs and spleens. i. . Efc flvmsl ngot fM mcoi inlungslevamisole ongrowthofM. microti Effect Fig. 3.2 fCL ie Mc ee infected2.4xwere Mice with CFLPmice.of e os nDy0ad rae ih0m/g •, . gk (♦), 2.5mg/kg (•), treatedand0on0perDay with mouse mg/kg r2 gk () eaioe wc ekyfo a 1. Each twicefrom levamisoleDay +10.weekly (A) or25 mg/kg point represents the mean logio cfu per lung of 4 the lunglogio cfuofmice.pointperrepresentsmean Mean log ^viable mycobacteria per lung 4 3 5 6 7 8 0 r I i I I1 7 5 2 9 36 29 22 15 7 3 I I I I I I I I as fe infection after Days ---- 119 i i I I 1 ---- 106 i viable bacilli viable U 1 --- 1 H3 * A

Control 25mg/kg 2.5mg/kg 2.5mg/kg

of CFLP mice. Mice were infected with 2.Ainfectedwere with x Mice CFLPmice.of Fig. 3.3 Effect of levamisole on growth of M. microti in spleenslevamisoleongrowthinof M. microti Effect 3.3Fig. e os nDy0ad rae ih0m/g •, . gk (♦), 2.5mg/kg (•), treated0and0on with mg/kgDay permouse r2 gk () eaioe wc ekyfo a.1. Each from twice levamisoleweeklyDay.+10. (A) 25ormg/kg point represents the mean logio cfu per spleem theperlogiocfuAof mice.point represents mean Mean log ^viable mycobacteria per spleen 3 1 2 2 3 43 36 29 22 15 7 3 0 i i i i i i i I I I II II II I L L asatr infection after Days LLLLLL ----- 120 1 ----- 106 L=levamisole viable bacilliviable

1 ----- LL 1

121

TABLE 3.2

ANALYSIS OF VARIANCE OF EFFECT OF LEVAMISOLE AND TIME ON GROWTH OF M. MICROTI IN CFLP MICE

Source of LUNG SPLEEN variation DF MS F MS F

LMS 2 0.010 0.1 0.046 0.9 *** Days 4 13.450 140 0.772 8.9

LMS x Days 8 0.188 1.9 0.059 0.6

Replicate Error 45 0.096 0.090

LMS = levamisole *** pCO.OOl

3.2.2 The effect of levamisole on proliferative responses of splenic

T lymphocytes

The proliferative responses of splenic T lymphocytes from uninfected and infected mice that were treated with 0, 2.5 or 25 mg kg ^ levamisole were tested before treatment (Day +9), after 2 weeks treatment (Day +24) and after 5 weeks treatment (Day +45).

Different cell concentrations were investigated but invariably, the concentration of 1 x 10^ cells per ml gave the lowest background counts and hence only proliferative responses of this cell concentration were reported.

The results of the proliferative responses on Day +9 to PHA, Con A and

PPD together with their respective controls are shown in Table 3.3. 122

TABLE 3.3

PROLIFERATIVE RESPONSES OF SPLENIC T CELLS OF UNINFECTED AND INFECTED MICE BEFORE LEVAMISOLE TREATMENT

T cells Uptake of [3H] Thymidine + s.e. (cpm) cultured with: Uninfected mice Infected mice

No mitogen 394 + 68 1689 + 490

PHA 65331 + 9010 118086 + 19480

Con A 12190 + 3600 31715 + 9800

No antigen 50 ± 26 544 + 304 * PPD 337 + 200 51103 + 9333

PPD 0.001 pg ml

The above results showed that 9 days after Infection, there was an increase in proliferative response to PHA, Con A as well as a substantial increase in antigen-specific response to PPD when compared with the uninfected controls.

The proliferative responses to PHA and Con A after two weeks of levamisole treatment are shown in Table 3.4. 123

TABLE 3.4

PROLIFERATIVE RESPONSES OF SPLENIC T CELLS OF UNINFECTED AND INFECTED MICE AFTER TWO WEEKS OF LEVAMISOLE TREATMENT

3 T cells from Uptake of [ H]-Thymidine + s.e. (cpm)

group: Control ^ PHA CON A

Uninfected

0 mgkg LMS 1061 + 241 73809 + 18330 16106 + 1341

2.5 mgkg 1 LMS 18962 + 5542 100693 + 6100 28004 + 2771

25 mgkg 1 LMS 21796 + 4492 137111 + 8320 53260 + 7203

Infected

0 mgkg 1 LMS 244 + 36 124227 + 15620 23416 + 6842

2.5 mgkg 1 LMS 1238 + 222 69186 + 6790 10809 + 3800

25 mgkg 1 LMS 1725 + 329 36038 + 2520 34437 + 5900

LMS = levamisole

These results showed several evident trends. In the uninfected controls, treatment with levamisole increased the background prolife­ ration of control cultures by 18-20 fold with a concomitant increase in mitogen-stimulated cultures. However, in the infected cultures, the background proliferation of unstimulated cultures from treated mice were just a little greater than those from untreated mice and considerably less than in the uninfected mice. The response to PHA of the infected and untreated mice was much greater than those of treated mice. Hence, it appeared that levamisole elevated the PHA response in uninfected mice but decreased the response in the infected mice. The response to Con A in the uninfected mice was also different to the response in the infected mice. Like the PHA response, levamisole in uninfected mice 124

elevated the Con A response; but in infected mice the lower dose of levamisole decreased, while the higher dose increased the response. The

PPD response was very variable (results not shown) with very high back­ ground counts for the unstimulated cultures.

The proliferative responses to PHA and Con A after 5 weeks of levamisole treatment are shown in Table 3.5.

TABLE 3.5

PROLIFERATIVE RESPONSES OF SPLENIC T CELLS OF UNINFECTED AND INFECTED MICE AFTER FIVE WEEKS OF LEVAMISOLE TREATMENT

T cells from Uptake of [^H]-Thymidine + s.e. (cpm)

group : Control PHA CON A

Uninfected -1 0 mgkg LMS 942 + 90 64879 + 3371 5219 + 2131 -1 2.5 mgkg LMS 7864 + 1124 108035 + 7076 9353 + 890 -1 25 mgkg LMS 5222 + 1247 104348 + 5721 16170 + 3600

Infected , -1 0 mgkg LMS 92 + 51 61755 + 6195 3376 + 790 -1 2.5 mgkg LMS 1296 + 412 46050 + 8642 6883 + 1645 -1 25 mgkg LMS 1754 + 153 57137 + 12018 7998 + 3500

LMS = levamisole

The proliferative response to PHA after 5 weeks levamisole still showed a similar trend. Levamisole increased the PHA response in uninfected mice but decreased the response in infected mice. The background pro­ liferation of unstimulated cultures were still high but not as high as 125

after 2 weeks levamisole treatment. The response to Con A had decreased

compared to the earlier time points but levamisole still elevated the

response in the uninfected mice and also the infected mice. PPD res­

ponses were again very variable with very high background counts

(results not shown).

3.2.3 The effect of levamisole on splenic T cell subsets

Preliminary results had shown that the optimal dilution of the mono­

clonal antibodies was 1 in 500. The monoclonals were stored at 4°C and diluted just prior to use. Both immunoenzymatic staining methods gave

similar results but the immunoalkaline phosphatase method facilitated enumeration as the cells were stained red against a bluish background

(Figure 3.4).

The monoclonal antibodies enabled the following determinations to be made: a) total T cell count using the anti-Thy 1,2 antibody; b) helper T cell phenotype count using the anti-Lyt 1 antibody; and 3) suppressor

/cytotoxic T cell phenotype using the anti-Lyt 2 antibody.

The splenic T cell subsets were determined on Day +9 (pre-treatment),

Day +24 (after 2 weeks treatment) and on Day +45 (after 5 weeks treatment). In all the results, the number of T cells stained by a particular antibody was calculated as a % of the total number of spleen cells counted. Differential staining (Diff-Quik staining) of the spleen cell suspensions had shown that they comprise 85% lymphocytes, 10% macrophages and 5% polymorphonuclear cells. 126

Fig. 3.4 Immunoalkaline phosphatase staining of cytocentrifuged murine splenic T cells with monoclonal Thy 1.2 antibody. Thy 1.2+ T cells are stained red (Naphthol AS-MX/Fast Red substrate, light haematoxylin counterstain). Magnification x400. 127

The results of the total number of T cells stained by the anti-Thy 1,2 antibody are shown in Table 3.6.

TABLE 3.6

SPLENIC THY 1,2+ T CELLS IN INFECTED AND UNINFECTED MICE BEFORE AND AFTER LEVAMISOLE TREATMENT

Spleen cells Thy 1,2+ T cells (% total spleen cells + s.e.) on:

from: Day 9 Day 24 Day 45

Uninfected

0 mgkg 1 26.2 + 2.3 26.5 + 1.6 28.0 + 2

2.5 mgkg 1 3 2 . 2 + 3 34.0 + 2.2

25 mgkg 1 18.4 + 4.3 38.7 + 2.3

Infected

0 mgkg”1 25.A + 2.2 2 1 . 3 + 2 25.4 + 1.9

2.5 mgkg 1 2 1 . 0 + 1 23.9 + 3.1

25 mgkg 1 20.4 + 1 27.4 + 3.7

These findings showed that after 2 weeks treatment with 2.5 mg kg 1 levamisole there was an increase in the number of total T cells in the uninfected mice but had no effect on the number of T cells in the infected mice. The 25 mg kg 1 dose caused a decrease in T cells in the infected mice and a slight decrease in the uninfected mice. After 5 weeks of levamisole, the T cell numbers in uninfected mice were still elevated but there were hardly any differences between the treated and untreated infected mice. 128

Table 3.7 shows the results of the cells staining with anti-Lyt 1

antibodies.

TABLE 3.7

SPLENIC LYT 1+ T CELLS IN UNINFECTED AND INFECTED MICE BEFORE ANJ) AFTER LEVAMISOLE TREATMENT

Spleen cells Lyt 1+ T cells (% total spleen cells + s.e.) on:

from Day 9 Day24 Day 45

Uninfected

0 mgkg 20.1 + 1.5 18.4 + 1.6 16.3 + 1

2.5 mgkg 18.4 +1 . 5 22.6 + 1.6

25 mgkg 10.3 + 1 . 2 32.0 + 3.4

Infected

0 mgkg 17.3 + 1.7 17.0 + 1.2 15.9 + 1 - 5

2.5 mgkg 18.2 + 2.1 18.4 + 1 . 9

25 mgkg 12.5 + 0.9 15.9 + 1.4

These results showed that in the uninfected mice, treatment with 2.5 mg kg ^ did not change the number of helper (Lyt 1+) T cells but the higher dose caused a decrease after 2 weeks and an increase after 5 weeks of treatment. In the infected mice, there was not much difference in the number of helper T cells between treated and untreated mice.

The number of cells stained by the Lyt 2+ antibodies are shown in Table

3.8. 129

TABLE 3.8

SPLENIC LYT 2+ T CELLS IN INFECTED AND UNINFECTED MICE BEFORE AND AFTER LEVAMISOLE TREATMENT

Spleen cells Lyt 2+ cells as % total spleen cells + s.e. on:

from: Day 9 Day 24 Day 45

Uninfected

0 mgkg 1 8.9 + 1.4 7.4 + 1 7.1 + 0.7

2.5 mgkg 1 4.1 + 0.5 10.2 + 0.3

25 mgkg 1 6.1 + 0.7 15.7 + 1.1

Infected

0 mgkg 1 5.2 + 0.7 3.5 + 0.5

2.5 mgkg 1 9.9 + 1.0 5.4 + 1 . 5

25 mgkg 1 5.6 + 0.6 3.2 + 0.8

Approximately 10% of the total spleen cells have the Lyt 2+ phenotype in both the uninfected and infected untreated mice at the begining of the infection. As the infection progresses, the number of Lyt 2+ cells decreases slightly in the infected mice and the 2.5 mg kg ^ levamisole dose seemed to cause a slight elevation of the number of cells. In the uninfected mice, treatment with levamisole seemed to increase the number of Lyt 2+ cells on Day 45 after an initial decrease on Day 24. 130

3.3 Discussion

M. microti has been reported to grow slower than M. tuberculosis and to be inhibited by glycerin in primary culture (Wells, 1946). It had also been observed (Leach and Wells, 1956) that the addition of 5% whole blood and adequate retention of moisture improved the recovery of freeze-dried M. microti. Later, a method for obtaining viable counts of

M. microti in 21 days was reported (Sharp, 1973) using oleic-albumin agar containing 5% defibrinated horse blood and a tight seal. It was decided to perform some preliminary experiments with a variety of media and conditions to determine the best system. Those experiments showed that reproducible results could be obtained in 4 weeks by using Dubos

Oleic Base agar supplemented with 10 % OADC and sealing the plates with parafilm before enclosing in polyethylene bags. Under these circum­ stances, growth was only slightly slower than that of M. tuberculosis.

The standard oral dose of levamisole is 150 mg twice weekly in adult humans. The choice of 2.5 mg kg ^ levamisole twice weekly was based on the clinical therapeutic dose given in a clinical study of the effect of levamisole on the response to chemotherapy of pulmonary tuberculosis in

Kenya and Zambia (Kenyan/Zambian/British Medical Research Council

Collaborative Study, in preparation). The 25 mg kg ^ dose was selected to represent a higher dose. At these doses, levamisole did not have any effect on the growth of M. microti in the lungs or spleens. This was evident from the growth curves and confirmed statistically.

Shepherd et al. (1977) also reported that levamisole had no effect on the mouse footpad growth curve. In a study of the effect of levamisole 131

on experimental murine infection with M. tuberculosis (Chumak and

Kostromin, 1981) the authors reported that levamisole (12 mg kg \twice weekly) did not increase the survival rate nor improved the extent of disease when given on its own. However, when levamisole was given in conjunction with isoniazid, there was an increase in survival rate and increased lymphoid proliferation in the spleen and thymus. In another study, there was a similar finding that levamisole at 2.5 and 25 mg kg ^

(thrice weekly) in mice and guinea-pigs respectively, was effective in decreasing the extent of disease in combination with isoniazid but not on its own (Alexandrova and Zabolotnykh, 1981). However, in both these reports, the actual growth of M. tuberculosis was not monitored.

It has been reported that responses to levamisole are not always predictable due to the presence of responders and non-responders in every animal species (Symoens et al., 1979). In mice, the responsive­ ness to levamisole seems to be related to their ability to produce a serum factor (Renoux, 1978). In a series of experiments Renoux and colleagues (1979) showed that host factors like genetics, sex and age of mice can lead to differences in response to levamisole. A recent report showed that the fat composition of a mouse diet modified the effects of levamisole on growth and spread of a murine tumour (Boeryd and Hallgren,

1985). That could be one of the reasons for the conflicting reports that have appeared in the literature about the immunotherapeutic potential of levamisole.

Although there have been a few reports in the literature about the effects of levamisole in conjunction with chemotherapy on clinical cases of pulmonary tuberculosis, the evidence is not very convincing (see 132

section 1.7). Preliminary results of a trial of levamisole in combina­

tion with chemotherapy on tuberculosis patients in Zambia and Kenya have

shown no clinical benefit of levamisole (Kenyan/Zambian/British Medical

Research Council Collaborative Study, in preparation).

Gatner and Anderson (1982) examined immune parameters, sputum bacterio­

logy and radiological evidence in seven newly diagnosed pulmonary tuber­

culosis patients given a single oral dose of levamisole (150mg) once a week in addition to standard antituberculous therapy for 3 months. The

humoral parameters (serum immunoglobulin, salivary IgA, C-reative prot­

ein, ^-antitrypsin levels) were initially elevated but returned to normal. Among the CMI parameters, neutrophil chemotactic responses

increased within 2 hours after ingestion but was not sustained; lympho­

cyte proliferation to PHA and PPD was also augmented 2 hours after

ingestion and was sustained. No evidence of improved sputum conversion

time or significant radiological improvement was observed. It was noted

that while immune function parameters were restored during levamisole therapy, a gradual return of these indices to normal generally occurred during standard therapy.

The proliferative responses to PHA and Con A after levamisole treatment were surprisingly different in uninfected and infected mice. Levamisole elevated the PHA response in the uninfected mice but had the reverse effect in the infected mice both after 2 weeks and 5 weeks treatment.

The effects on Con A response were not so clearly defined but generally, levamisole increased the response to Con A regardless of infection. The other noticeable effect of levamisole was the elevation of the back­ ground proliferation of control cultures especially after 2 weeks. The 133

response to PPD was not clear due to the presence of a very high and variable background proliferation on Days 24 and 45. The reasons for the variable background proliferation counts of control cultures in the

PPD assay were not known. The mi croteraski method uses only a small number of cells. In the PPD assay, there are relatively fewer antigen- reactive cells compared to reative cells in mitogenic assays and the microterasaki method may have been too insensitive.

The response to PHA correlated with the increase in total splenic T cells seen in uninfected but not infected mice. Generally, the total T cell and T cell subsets were not very different after levamisole treatment in infected mice although there were some slight differences in the uninfected mice. The significance of these observations is unknown. It would seem that infection with M. microti radically affects the proliferative response of splenic T cell to PHA. These findings confirm the earlier observations that the effect of levamisole to a large extent depended on host factors and that infection with M. microti altered the host and consequently altered the proliferative responses.

Ultimately though, despite the differences seen in the immunological tests, levamisole did not affect the in vivo growth of M. microti.

Levamisole has been shown to augment the responsiveness of T cells to mitogens and antigens. Levamisole has been shown to be weakly mitogenic for mouse spleen cells (Merluzzi et al., 1975) and Renoux (1978) reported an increase in DNA synthesis of murine splenocytes after in vivo treatment with levamisole. This was consistent with the findings reported in this Chapter of increased background proliferative responses of unstimulated splenocytes. 134

In an In vitro study Chan e t a l . (1976) showed that preincubation of lymphocytes from tuberculosis patients with levamisole induced an enhanced proliferative response to PHA. Gatner (1981) subsequently reported that a single 150 mg dose of levamisole augmented in vitro PHA and PPD-induced lymphocyte proliferation of tuberculin-positive control subjects.

As levamisole did not have any significant effect on the growth of

M. microti in organs of mice, no attempts were made to investigate the effect of levamisole in combination with antituberculous drugs. At the time this decision was taken, preliminary results of the clincal trials in Kenya and Zambia were begining to show a lack of clinical effect of levamisole, and recombinant IFN-)( also became available. 135

CHAPTER 4

THE IMMUNOMODULATING EFFECT OF INTERFERON-GAMMA

ON M. TUBERCULOSIS INFECTION IN MICE

The immunomodulating effect of Interferon-gamma was investigated in an

acute infection of M. tuberculosis H37Rv in mice. The experiments were

performed with two strains of mice. The initial experiments were

performed on CFLP mice, an outbred strain, but most of the results

reported in this chapter were experiments done on BALB/c mice. The

reason for the switch from the outbred strain to an inbred strain was

due to preliminary results which had shown that the effects of IFN-V

were not very large. In addition, it was noted that there were diffe­

rences in the growth rate of M. tuberculosis in untreated controls

between some experiments with the CFLP mice. It is well known that the

cleanliness of mice affects the host resistance and even though the CFLP

mice were obtained from a specific-pathogen free colony, there was

uncertainty about their cleanliness. Thus, when a source of BALB/c mice

from a reputable specific-pathogen free colony became available, most of

the later experiments were performed on these mice. As each experiment had its own control, there was no problem with correlation of results

from both sets of mice. The untreated controls showed that both

strains reacted very similarly to infection with M. tuberculosis.

A range of dose sizes were tested as well as the efficacy of liposomal carriage of IFN-J^. The schedule of administration of IFN-tf was also investigated. The results of some preliminary experiments on the in vivo model are also reported. 136

4.1 Preliminary experiments

4.1.1 Sonicatlon of M. tuberculosis

The hydrophobic nature of the mycobacterial species means that they

readily clump. The introduction of surfactants like Tween 80 into

culture media (Dubos, 1945) has helped to resolve the problem but it has

been shown that suspensions of mycobacteria grown in liquid media with

Tween 80 are still mainly in small clumps, each containing 4-20

organisms (Fenner, 1949). Ultrasonication has been used as a means of

dispersing clumps in mycobacterial suspensions (Blanden et al., 1969).

Some experiments were carried out to determine the optimal conditions

for obtaining a dispersed suspension with a sonicating probe (Rinco

Ultrasonics UK Ltd.).

12 ml samples of a suspension of H37Rv in 0.1% gelatine saline were placed in universals and the probe was used at an amplitude of 70% with

the probe 1 cm below the surface of the suspension. In one experiment, viable counts were performed after sonication periods of 0, 5, 10, 15,

30, 45 and 60 seconds in non-continuous bursts of 5 seconds. In another experiment, total counts were carried out after the same sonication periods. In addition to enumeration, the total counts were also scored as the number of single bacilli, the number of clumps of 2, 2-5 and >5 bacilli. The results of different lengths of sonication on the viable counts are shown in Figure 4.1.

It can be seen that the viable counts increased dramatically after 5 seconds of sonication to its maximum after 30 seconds. Even 60 seconds ahpitrpeet the tuberculosisrepresentspointnumberatofcfuof M.Each aiu tm onsatr sonication. timepointsaftervarious Fig. 4.1 Effect of sonicationofofmycobacteria. Effectoncounts viable Fig. 4.1 Viable mycobacteria (x10 )/ ml Time in seconds 137

138

did not seem to affect the viability of the mycobacteria. The results of different sonication lengths on the total counts are shown in Table

4.1.

TABLE 4.1 TOTAL COUNTS OF M. TUBERCULOSIS AFTER SONICATION

Sonication Total count % Bacteria in aggregates of : time (secs) (orgs /ml) one two 2-5 >5

0 1.0 x 107 46 6 27 21

5 3.6 x 107 65 10 18 7

10 3.8 x 107 73 17 7 3

15 4.4 x 107 80 13 6 1

30 5.2 x 107 81 10 8 1

45 6.2 x 107 80 14 6 0

60 6.1 X 107 83 14 3 0

Before sonication, 27 % of the bacilli were aggregated as clumps of 2-5 and 21% as clumps of >5 bacilli. These aggregations dropped consider­ ably after sonication but even after 60 minutes sonication, there were still 3.6% of the bacilli in small clumps of 2-5.

It was decided that a sonication period of 15 seconds at a setting of

70% amplitudes and with the probe 1 cm below the surface of the suspen­ sion was to be adopted in all the experiments. 139

4.2 The effect of infective dose size of M. tuberculosis on the growth of bacilli In the lungs and spleens of CFLP mice

In this experiment, different infective doses of M. tuberculosis were used to determine an optimal inoculum size. Different dilutions of the animal-passaged H37Rv inoculum stored at -70°C were injected into CFLP mice. 72 mice were divided randomly into 3 groups. Viable counts performed at infection showed that one group received 5 x 10^ cfu per 5 4 mouse, another 5 x 10 cfu and the last group 5 x 10 cfu. The growth of the bacilli in the mice was monitored by performing viable counts of the bacilli in the lungs and the spleens at days 1, 8, 15, 22, 29 and 43 after infection. Four mice were sacrificed at each time point.

The results are shown in Figures 4.2 and 4.3. The details of the results are shown in Table 4A.1 in Appendix 1. The mice that received 5 x 10^ cfu per mouse had enlarged spleens and necrotic lesions in the lungs by day 15 and began to die after day 20. None of the mice in the 4 other two groups died and the mice in the group that received 5 x 10 cfu per mouse were still apparently healthy at day 43. However upon post-mortem, the lungs showed evidence of necrotic lesions.

From these results, it was decided that an infective dose size mid-way between 5 x 10^ and 5 x 10^ cfu per mouse was to be used in all subsequent experments. This infective dose size was achieved by diluting the original frozen inoculum 1 in 2. 1 40

1 cn . c 3 i- * E « .0 OJ > o cn o

Fig. 4.2 The effect of infective dose size on the growth of M. tuberculosis in lungs of CFLP mice. Mice were infected with 5 x 106 (#), 5 x 105 (A) or 5 x 10^ ( ♦ ) cfu per mouse. Each point represents the mean + s.e. of 4 mice. 141

c

n i i i i i 01 8 15 22 29 43 Days after infection

Fig. 4.3 The effect of infective dose size on the growth of M. tuberculosis in spleens of CFLP mice. Mice were infected with 5 x 106 (#), 5 x 105 (A) or 5 x 10^ (♦) cfu per mouse. Each point represents the mean + s.e. of 4 mice. 142

4.3 The effect of dose size of interferon-gamma on the growth of

M. tuberculosis In BALB/c mice

65 male BALB/c mice, 18-20 g in weight, were randomly allocated into 4 groups. 20 mice were used as controls and the other groups of 15 mice each received either 200, 1000 or 5000 Units of interferon-gamma per mouse. The control group was given PBS. Treatment was started 2 days prior to infection (Day-2) and each group received a dose every third day, that is, on Days +1, +4 and +7.

5 mice were sacrificed per treatment group for each time point. In this experiment, the viable count of the M. tuberculosis was assessed in the lungs and spleens on Days +3, +5 and +9 after infection.

Viable counts performed at infection showed that the inoculum per mouse was 1.26 x 10^ cfu. The results of the mean viable counts in the lungs are shown in Figure 4.4. For clarity, the s.e. of the means are not depicted but are shown in Table 4A.2 in Appendix 1. The mean viable count of M. tuberculosis in the lungs an hour after infection was 4.78 x

10** cfu. Over the next 3-5 days, there was a fall in viable counts in all the groups followed by an increase after Day+5. On Day+9, the mean viable count was 1.3 x 10^ cfu in the untreated controls and consider­ ably lower in the groups treated with IFN-V . There was hardly any difference between the groups given 1000 or 5000 units IFN-V but both these groups had a lower cfu than the group given 200 units. 143

The corresponding results of the spleen counts are shown in Figure 4.5 with the full details in Table 4A.3 (Appendix 1). The mean viable count

an hour after infection was 1.43 x 10^ cfu. There was hardly any diffe­

rence in the growth curves of the untreated controls and the group that

received 200 units of IFN-S. The growth curves of the groups that

received 1000 or 5000 units were very similar and there was an obvious

inhibition of bacillary growth.

The viable count results of all the groups were then examined statistic­ ally by analysis of variance (AN0VA). The results of the AN0VA of both

lungs and spleens are shown in Table 4.2.

TABLE 4.2

ANALYSIS OF VARIANCE OF EFFECTS OF INTERFERON-GAMMA DOSE SIZE AND TIME ON GROWTH OF M. TUBERCULOSIS IN BALB/C MICE

Source of Lung Spleen variation DF MS F MS F

** IFN-8 3 0.291 2.6 0.104 10.8 *** *** Days 2 2.244 20.0 6.960 718

Days x IFN-# 6 0.068 0.4 0.004 0.4

Replicate error 48 0.112 0.009

*** p < 0.001 ** p<0.01

The IFN-y dose effect was highly significant in the spleens (p<0.01) but did not attain statistical significance at the 5% level in the lungs. 144

U) c 3 L. o CL 0 u

.2il 0) ■*-> u (0 JD O 200u U 5000u E o 10OOu Si to

Days after infection

Fig. 4.4 Effect of dose size of interferon gamma on the growth of M. tuberculosis in lungs of BALB/c mice. Mice were infected with 1.3 x 106 cfu M. tuberculosis per mouse on Day 0 and treated with 0 u (•), 200 u (O ), 1000 u (♦) or 5000 u (A ) IFN-Jf per mouse on Days -2, +1, +4 and +7. Each point represents the mean + s.e. cfu per lung of 5 mice. Mean log^viable mycobacteria per spleen + s.e. cfu per spleen of spleen5 permice. cfus.e. + os nDy 2 1 + n 7 Ec on ersns therepresentspoint Each mean andon +4 Days -2,+1, +7. mouse with 0 u (•), 200 u 200 (•), 0u with treated tuberculosisonand0per cfumouseDay M.1.3106 x with fM tbruoi i pen fBL/ ie Mc ee infectedwere Mice spleensBALB/cof in tuberculosismice. ofM. i. . Efc fds sizetheinterferon-gammaofgrowthdoseonof Effect Fig. 4.5 as fe infection after Days (O ), ), (O 00u( o 00u A IFN-Yper (♦ ) (A) or50001000uu 145

146

This was probably due to the higher mean square of the replicate error

in the lungs compared to the spleen. As there was no difference between

dosage with 1000 or 5000 units of IFN-* , most of the subsequent experi­ ments were performed with doses of 1000 or 2000 Units IFN-V per mouse

given at 3 day intervals, with the first dose given 2 days prior to

infection.

4.4 The effect of the administration of interferon-gamma in liposomes

on the growth of M. tuberculosis in mice

Two sets of experiments were performed on the effect of the administra­

tion of IFN-y in liposomes on the growth of M. tuberculosis in mice.

One experiment was carried out in CFLP mice with dose sizes of 1000 and

20000 units of IFN-X. The other experiment was performed in BALB/c mice

with a single dose size of 1000 units.

4.4.1 The effect of the interferon-gamma when administered in liposomes

on M. tuberculosis infection in CFLP mice

In the first experiment, 62 female CFLP mice, 18-20 g in weight, were

randomly allocated into 5 groups. All the mice were infected intra­

venously with 1.4 x 10^ cfu of M. tuberculosis per mouse. The control

group was treated with PBS while the other groups were given 1000 units

of IFN-V, 1000 units of IFN-* in liposomes, 20000 units of IFN-V or

20000 units of IFN-tf in liposomes. The treatments were given at 3 day

intervals with the first dose 2 days prior to infection and subsequent

doses on Days +1, +4 and +7. The IFN-# dilutions and liposomes were made as described in section 2.5.2.1. Due to the unknown stability of 147

the recombinant IFN-& no attempts were made to separate the free IFN-&

from the encapsulated IFN-Jf.

The growth of the bacilli was assessed by sacrificing 6 mice per group

on Days +5 and +9 post-infection. The uptake of the inoculum was deter­

mined by performing viable counts on organs of 6 mice an hour after 4 5 infection. The uptake was 7.1 x 10 cfu in the lungs and 3.6 x 10 cfu

in the spleen.

The results of the viable counts of M. tuberculosis in the lungs are

shown in Figure 4.6 and the viable counts in the spleens are represented

by Figure 4.7. The mean viable counts + s.e. of the lungs and spleens

are shown in Tables 4A.4 and 4A.5 respectively (Appendix 1).

The Figures clearly show that both 1000 and 20000 units of IFN-V had a

significant effect on the growth of M. tuberculosis in both lungs and

spleens of the mice. The viable counts of the untreated controls, mice given 1000 units IFN-^ and mice given 1000 units IFN-tf in liposomes were analysed separately by a 2-way AN0VA. Orthogonal linear contrast showed

that 1000 units IFN- £ had a highly significant effect in the lungs (F ratio=28.5, p < 0.001) and in the spleen (F ratio=72.2, p<0.001) when compared with the untreated controls. There was no difference between

the results of treatment with 1000 units IFN-* and 1000 units IFN-V in liposomes in both organs. The administration of IFN-if in liposomes does not seem to have increased the effect in the spleens. However, in the lungs, the shape of the growth curve of the group that had been treated with 1000 units IFN-tf in liposomes suggests that encapsulation in liposomes results in a prolongation of the effect of IFN-*' while the Mean log^viable mycobacteria per lung f nefrngma n iooe. ie ee netd ih . x 0 ibe ail on bacilli viable 10 x 1.4 with infected Mice were liposomes. in interferon-gamma of en o^ f pr ug f mice. 6 of lung per cfu log^ mean Day 0 and treated with 0 u ( • ) , 1000 u (A ), 20000 u (■ ) IFN-Y , or 1000 u (A ), ), (A 1000 u or IFN-Y, ) (■ 20000 u ), (A 1000 u , ) • ( u 0 with administration treated and 0 miceafter Day CFLP of lungs in tuberculosis M. of counts Viable 4.6 Fig 00 u □ INYi lpsmso Dy -, 1 + n +. ah on rpeet the represents point Each +7. +4and +1, -2, Days liposomes.on IFN-Yin (□) 20000 u asatr infection after Days Mean loginviable mycobacteria per spleen f nefrngma n iooe. ie ee netd ih . x 0 val bcli on bacilli 106 viable x 1.4 with infected Mice were liposomes. in interferon-gamma of en og f pr pen f mice. 6 of spleen per efu g^ lo mean a 0 n tetd ih u 1000 u , ) • ( u 0 with administration treated after mice and 0 CFLP Day of spleens in tuberculosis M. of counts Viable 4.7 Fig 00 u □ INyi lpsms n as 2 +, 4ad 7 Ec pit ersns the represents point Each +7. +4and +1, -2, Days on liposomes in IFN-y (□) 20000 u as fe infection after Days (A), (A), 00 u ■) F-f o 10 u (A), 1000 u or IFN-lf, ) (■ 20000 u 150

curve of mice treated with 20000 units in liposomes suggests a decrease in the effect.

Statistical evaluation of the lung and spleen counts of mice receiving

IFN-Y or IFN-Y encapsulated in liposomes was performed by a three-way analysis of variance. The results of the analysis are shown in Table

4.3.

TABLE 4.3

3-WAY ANALYSIS OF VARIANCE ON THE EFFECT OF INTERFERON-Y DOSE SIZE, LIPOSOMES AND TIME ON M. TUBERCULOSIS INFECTION IN CFLP MICE

Source of LUNG SPLEEN variation DF MS F MS F

** IFN-Y 1 1.728 9.6 0.108 2.8

Liposomes 1 0.530 3.0 0.000 0.0 *** * Days 1 2.980 16.6 0.242 6.2

IFN-Y x Days 1 0.150 0.8 0.053 1.4 * Liposomes x Days 1 0.319 1.8 0.256 6.6

IFN-Y x Liposomes 1 0.154 0.9 0.029 0.7

Days x IFN-Y x Liposomes 1 0.146 0.8 0.000 0.0

Replicate error 40 0.179 0.039

*** p<0.001 * p < 0.05

In the 3-way analysis of variance, the sources of variation are broken down into main effects, first-order and second-order interactions. In this experiment, the main effects are IFN-Y , liposomes and time (days).

The analyses showed that among the main effects; the dose size of IFN-Y 151

was highly significant (p<0.01) in the lung but not significant in the spleen; time had a significant effect in the lungs and spleens; and liposomes did not have a significant effect on either organ. There was a significant first-order interaction of time and liposomes in the spleens.

A.4.2 The effect of the administration of interferon-gamma in liposomes on M. tuberculosis infection in BALB/c mice

The previous experiment had shown that the administration of IFN-Y in liposomes did not have a statistically significant additional effect to that of IFN-Y even though the shape of the growth curve in the lungs of the mice given 1000 units IFN-Y in liposomes seems to indicate a prolon­ gation effect. It was decided to examine the effect of empty liposomes on the growth of M. tuberculosis in mice.

In this experiment, an additional control group receiving PBS in lipo­ somes was included and a single dose size of IFN-* was tested. 65 female BALB/c mice, 18-20 g in weight, were randomly allocated into 4 groups. One control group was given PBS used for diluting IFN-^ ; the other control group received PBS encapsulated in liposomes and the other two groups were given 1000 units of IFN- V either in PBS or encapsulated in liposomes.

4 mice were sacrificed at each time point and the lungs and spleens cultured to determine the number of viable M. tuberculosis. Viable counts performed at infection showed that the number of organisms in the inoculum was 7.53 x 10^ cfu per mouse. At one hour after infection, 152

k 4 there were 7.16 x 10 cfu in the lungs and 9.23 x 10 cfu in the spleen.

The results of the viable counts of the bacilli in the lungs of the various groups are shown in Figure 4.8. Similarly, the results of the viable counts in spleens are shown in Figure 4.9. For clarity, the standard errors of the means are not depicted in the graphs, but the full details of the mean counts in the lungs and spleens are shown in

Tables 4A.6 and 4A.7 respectively (Appendix 1).

In the lungs, there was hardly any difference between the bacillary growth curves of the groups that had been treated with PBS or PBS encapsulated in liposomes. However, there was a lowering of the lung viable counts in the groups of mice that had received either 1000 units of IFN-tf or 1000 units encapsulated within liposomes. The viable count results of the spleen (Figure 4.9) show a definite trend. There was a slight lowering of viable counts in the group that had received PBS in liposomes. Both groups that had been given 1000 units IFN-fc' showed lower spleen counts than the PBS control, with the liposomal IFN-^T group showing slightly lower counts than the IFN-fc group.

Analysis of variance of the lung and spleen counts was performed by a three-way analysis of variance. The results of the AN0VA are shown in

Table 4.4. ieatramnsrto fINYi iooe. iewr infected Micewere IFN-Yinliposomes.of afteradministration mice ihPS •, B nlpsms O, 00uINY() r 1000 or (♦) 1000IFN-Y u liposomes (O), in PBS (•), PBS with organisms per treated1050viableand7.5xmouseon Day with u IFN-Jr in liposomes ( O ) on Days -2, +1, +4 and +7. Each point Each on-2,IFN-JrDays and+1,u +4 +7. ) O inliposomes ( ersns thelung logiocfuperofmean represents4 mice. i. 8 ibecut oftuberculosislungscountsM. the BALB/cinof Viable .8Fig. A Mean loginviable mycobacteria per lung as fe infection after Days 153 PBS PBS,L 1000u/L lOOOu

154

Days after infection

Fig. 4.9 Viable counts of M. tuberculosis in the spleens of BALB/c mice after administration of IFN-V in liposomes. Mice were infected with 7.5 x 105 viable organisms per mouse on Day 0 and treated with PBS (•), PBS in liposomes (O), 1000 u IFN-y (♦) or 1000 u IFN-Y" in liposomes (O) on Days -2, +1, +4 and +7. Each point represents the mean loglO cfu per spleen of 4 mice. 155

TABLE 4.4

THREE-WAY ANALYSIS OF VARIANCE OF THE EFFECT OF INTERFERON-GAMMA, LIPOSOMES AND TIME ON M. TUBERCULOSIS INFECTION IN BALB/C MICE

Source of LUNG SPLEEN variation DF MS F MS F

** *** IFN-Y 1 0.004 11.3 0.125 35.3 *** Liposomes 1 0.004 0.1 0.125 13.9 *** *** Days 2 4.530 108 7.272 808 * IFN-V x Days 2 0.044 1.0 0.054 6.0

IFN-^ x Liposomes 1 0.007 0.2 0.001 0.1

Days x Liposomes 2 0.034 0.8 0.008 0.9

IFN-y x Days x Liposomes 2 0.026 0.6 0.001 0.1

Replicate error 48 0.042 0.009

*** p < 0.001 ** p<0.01 * p < 0.05

In the lungs, the effect of IFN-Jf and time were highly significant, and

the effect of liposomes was not significant. In the spleen, all the main effects were highly significant. A synergistic effect of liposomal

encapsulation and IFN-Y would have resulted in a significant interaction

of liposomes x IFN-tf. However, the sum of squares for this interaction was very small and not significant, which suggests that liposomes per se had an effect irrespective of IFN-Jf. 156

4.5 The effect of pretreatment of mice with interferon-gamma on

M. tuberculosis infection in BALB/c mice

It was evident from the results of the earlier experiments that the effects of IFN-fr was established very early in the infection and did not seem to increase with further administration of IFN-V. This observation was confirmed by analysis of variance which showed a significant effect of IFN-X and a significant effect of time but did not reveal any signi­ ficant interactions of IFN-lf and days. This absence of significant interactions implies that there was no amplification of the effects of

IFN-lf with further doses of IFN-JT. Examination of the growth curves of

M. tuberculosis in the treated and untreated groups in lungs and spleens generally show parallel growth curves which again suggest that the effects occurred early and did not increase with time. Hence, it would appear that the preinfection dose was of paramount importance.

This experiment was designed to examine the effect of preinfection trea­ tment of mice with several multiplicities of doses as well as schedules of administration of IFN-K.

60 female BALB/c mice, 18-20 g in weight, were randomly allocated into 6 groups. One group was used as the control; 3 groups received a single dose of IFN-Jf given on Day-3, Day-2 or Day-1; one group received two doses of IFN-f on Days -2 and -1 and the last group received three doses on Days -3, -2 and -1. IFN-& was used at 2000 units per mouse. The mice were pretreated accordingly and were then infected intravenously with the same inoculum level. Four mice were sacrificed per time point and viable counts performed 1 hour after infection, and on Days +1 and 157

+2 post-infection. The effect of pretreatment with IFN-V on the lodgement of the bacilli in the lungs and spleens after one hour was also assessed. 6 mice of the control group and another receiving 3 doses of IFN-Y were sacrificed 1 hour after infection to determine if

IFN-# had affected the uptake of bacilli in lungs and spleens.

Viable counts performed at infection showed that the inoculum per mouse was 4.12 x 105 cfu. The results of the viable counts in the organs of mice 1 hour after infection are shown in Table 4.5. When the values were tested by the analysis of variance they gave F ratios of 0.5 and

2.1 for the lung and the spleen respectively indicating that pretreat­ ment with IFN-tf did not have any significant effect on the lodgement of the organisms in the lungs and the spleen an hour after intravenous infection.

TABLE 4.5

THE EFFECT OF INTERFERON-V PRETREATMENT ON THE UPTAKE OF M. TUBERCULOSIS IN ORGANS OF BALB/C MICE

cfu per organ after IFN-V treatment: Log10 Mouse Lung Spleen Number 0 u 2000 u 0 u 2000 u

1 4.972 5.031 4.703 4.826

2 4.982 5.061 4.794 4.699

3 5.054 4.932 4.854 4.697

4 5.042 5.068 4.885 4.512

5 5.081 4.943 4.798 4.787

6 4.988 4.952 4.648 4.663

Mean 5.020 4.998 4.780 4.697 + s.e + 0.02 + 0.02 + 0.04 + 0.04 ** —— 158

The results of the effects of the different regimens on growth of the

M. tuberculosis in the lungs are graphed in Figure 4.10 (details of the

mean viable counts + s.e. are in Table 4A.8 in Appendix 1). In the

first two days after infection, the lung viable counts were decreasing

and there were no apparent differences between the treated groups and

the untreated control.

The viable counts of M. tuberculosis in the spleen of the treatment

groups are shown in Figure 4.11 with full details given in Table 4A.9

(Appendix 1). The growth curves of the bacilli in the different experi­

mental groups show a definite trend. All the groups that had been given

IFN-tf showed a slower growth of the bacilli than the control. It was

also clear that there was a difference in the growth curve depending on

the day of administration of IFN-V. These results showed that the admi­

nistration of a single dose of IFN-f on Day-1 was more effective than on

Day-2, with Day-3 being the least effective. It was also obvious that a

single dose at Day-1 was as effective as two doses at Days -2 and -1 or

three doses at Days -3, -2 and -1.

A two-way analysis of variance was performed on both the lung and spleen

counts (Table 4.6). In the lungs, the effect of IFN-^ was shown to be

statistically insignificant. This was probably due to the high value of

the mean square of the replicate error. In the spleen, it was confirmed

statistically that the pretreatment dose(s) of 2000 units of IFN-V per mouse given solely before infection had a highly significant effect on

the growth of M. tuberculosis.- As the results of this experiment had

shown that administration of the preinfection dose on Day-1 was more mice after pretreatment with IFN-)f.pretreatmentwith after mice tMice pretreatedwithwere iewr infected105onorganisms4.1x Day 0. Each viable with wereMice n oeo 00uINJ nDy3 □, a- () Dy (O); Dayl (O), 2000IFN-JTof Day-2udose one (□), on Day-3 on rpeet thelogio lungcfuper4 representsof mice.point mean i. .0 ibecut fM tuberculosislungsofBALB/cofcountsM.in Viable Fig. 4.10 w oe o 00u() tredsso 00u(A o nrae (#). oruntreated A) ( threedosesof2000u (A); 2000 ofutwodoses Mean log 1Qviable mycobacteria per lung as fe infection after Days 159 en o1 f pr pen f mice. 4 of spleen per log1Qmean cfu Day-3(D), IFN-Yon 2000 u of dose one with pretreated Mice were interferon-gamma. with nrae ()ad netd ih . x 0 f o Dy . ah on rpeet the represents point Each 0. Day on cfu 10 x 4.1 with infected (#).and untreated a- ( Dy1 O) to oe o 20 u F- A; he dss f 00 INYA; or IFN~Y(A); 2000 u of doses three IFN-Y(A); 2000 u of doses two ); (O Day-1 , ) (0 Day-2 Fig 4.11 Viable counts of M. tuberculosis in lungs of BALB/c mice after pretreatment pretreatment mice after BALB/c of lungs in tuberculosis M. of counts Viable 4.11 Fig Mean Iogin viable bacilli per spleen as fe infection after Days 5 161

effective than on Day-3 or Day-2 (Figure 4.11), in some of the later

experiments combining IFN-Xand chemotherapy, the preinfection dose was

given one day prior to infection.

TABLE 4.6

ANALYSIS OF VARIANCE OF THE EFFECTS OF INTERFERON-GAMMA PRETREATMENT ON M. TUBERCULOSIS INFECTION IN BALB/C MICE

Source of LUNG SPLEEN variation DF MS F MS F *** IFN-* 5 0.041 1.92 0.054 5.4 *** Days 1 0.999 46.7 0.025 2.5

IFN-)f x Days 5 0.012 0.6 0.009 0.89

Replicate error 36 0.360 0.010

*** p < 0.001

4.6 Discussion

In all the in vivo experiments involving both M. microti (Chapter 3) and

M. tuberculosis, the infective inocula were prepared in large quantities

and stored at -70°C. It had been shown by Grover et al. (1967) that an

inoculum of M. tuberculosis preserved at -70°C retained its viability

and virulence for guinea-pigs over a period of one year. Subsequently

it was shown that there was no decrease in viability over a three year

period of storage at 70°C of several strains of mycobacteria (Kim and

Kubica, 1972, 1973). The use of low temperature preservation of the infective inocula was adopted in view of the following advantages: it avoided the risk of attenuation following serial transfer on laboratory media ; it eliminated the need to periodically passage a strain through 162

an animal to regain virulence and it enabled a reproducible and precise

level of infection from one experiment to another.

Throughout these experiments, an infective dose size of approximately

5 x 10^ to 1 x 10^ viable M. tuberculosis H37Rv per mouse was used.

This infective dose size was chosen after preliminary experiments

comparing various doses. At this dose, the H37Rv proliferated in the

spleen from the first day while in the lung, there was an initial lag

period of about 3 days where the counts decreased before exponential

growth begins around the fifth day.

The initial investigations with recombinant interferon-gamma were con­

cerned with trying to determine an optimal dose size, schedule and

frequency of administration. The IFN-Ywas obtained at a concentration of 2.5 mg ml ^ with a specific activity of 7.2 x 10^ antiviral units per mg. Due to the lack of long-term stability data, the concentrated IFN-'JJ was stored at 4°C and diluted just prior to use. Dilutions were made in

PBS containing 1 mg ml ^ of homologous mouse serum albumin.

A range of dose sizes from 200 units to 20,000 units per mouse were

tested. With the exception of 200 units which had virtually no effect,

the other doses tested had a significant inhibitory effect on the growth of M. tuberculosis in both lungs and spleens of CFLP and BALB/c mice.

However, no significant differences could be detected between the effects of 1000, 2000, 5000 units. The inhibitory effect of IFN-Y was evident from the growth curves of the mycobacteria in the organs and was confirmed statistically by analysis of variance. There was a suggestion from the results of one experiment (4.4.1) that 20,000 units IFN-Y in 163

liposomes had an adverse effect (Fig 4.6). However, this was only evident in the lungs and not the spleen. Analysis of variance of the results of that experiment (Table 4.3) showed a significant effect of dose size of IFN-Y in the lungs but not the spleens.

The schedule of administration most commonly used for these experiments was at three day intervals with the first dose two days prior to infect­ ion. Results with this schedule suggested that the effects of IFN-Y occurred early and did not seem to be potentiated by further doses.

This led to an investigation of the preinfection doses of IFN- Jr. The results showed that IFN-tf given a day before infection was the most effective preinfection dose and also that multiple preinfection doses did not confer any extra benefits.

There has been two reports of the effects of recombinant murine IFN-V derived from E. coli on intracellular pathogens in vivo. Kiderlan et a l . (1984) showed that IFN-y was capable of protecting mice from a systemic or local infection with L. monocytogenes. The IFN- was given intravenously at a dose of 1 x 10^ or 4 x 10^ units a day before and on the day of infection in the systemic infection model. In the local infection model, protection was observed with subcutaneous foot-pad 3 injection of a minimal dose of 10 units. The other report described the effect of IFN-5f on murine toxoplasmosis (McCabe et al., 1984). They concluded that recombinant murine IFN-y had a significant activity against toxoplasma and that the activity appeared to be associated with enhanced antibody response and activated macrophages. They used doses 3 of 5 x 10 units per mouse injected intra-peritoneally and various schedules varying from two doses given just before and after infection 164

to doses every other day. There were some similarities between these these results and the results reported in this Chapter. In both these reports, IFN-fc was given before and after infection and there did not seem to be a difference between the higher doses used. Similarly, results of this Chapter showed no detectable differences between doses ranging from 1000 to 5,000 units per mouse.

The pharmacokinetics of recombinant murine IFN-V in vivo are not known.

A recent clinical study of cancer patients treated intravenously and intramuscularly with recombinant human IFN-V (Kurzrock, 1985) has shown the pharmacokinetics of recombinant IFN-V in man. After intravenous administration, IFN-V was cleared exponentially from the serum with a half-life of 25-35 minutes which was independent of the dose. Intra­ muscular injection gave a half-life varying from 227-462 minutes indep­ endently of dose size. This study has confirmed the short half-life of

IFN-Jf in man. Hence, if liposomes could prolong the half-life of IFN- in vivo, encapsulation in liposomes should increase the efficacy of

IFN-Jf. MDP has been shown to have a very short half-life. After paren­ teral administration, 50% of water-soluble MDP is excreted in the urine within 30 minutes (Parant et al., 1979). Fidler et al. (1980) have also demonstrated that LKs like MAF have a very short half-life due to rapid binding to serum proteins. The enhancement of activity by liposomes has been shown in studies with lymphokines (MAFs) and immunomodulators like analogues of MDP where encapsulation in MLV liposomes had conferred a greater tumoricidal effect on experimental murine tumours (Fidler, 1980;

Fidler et al., 1981; Fidler et al., 1982). 165

Another reason for using liposomes Is to exploit the natural fate of

liposomes to ’target’ passively to cells of the reticuloendothelial

system. Consequently, experiments were performed to investigate whether

encapsulation of IFN-Y in MLV liposomes would increase its effects.

The two sets of experiments where IFN-Y was administered either in PBS

or encapsulated in liposomes reaffirmed that IFN-Y of 1000 Units dose

size per mouse had a highly significant inhibitory effect on the growth

of M. tuberculosis in both the lungs and spleens. The effect of admini­

stration of IFN-Y in MLV liposomes was not so clearly defined after

considering both sets of results. In the first set of experiments

(A.4.1), the appearance of the growth curves in the lungs (Fig. 4.6)

suggested that liposomes prolonged the effect of 1000 units of IFN-Y but

had an adverse effect on 20,000 units IFN-Y. However, statistical

analysis (Table 4.3) did not reveal any effect of encapsulation in

liposomes on the efficacy of IFN-Y.

In the second set of experiments, a 3-way analysis of variance (Table

4.4) showed that in spleens, liposomes per se had a significant

inhibitory effect. However, there was no significant interaction of

liposomes and IFN-Y which would have been expected if liposomes enhanced

the effects of IFN-Y. The control group receiving liposomes with PBS did show an effect on the mycobacterial growth in the spleen as shown by

comparison of the growth curves (Fig. 4.9). This would suggest that encapsulation of IFN-Y in liposomes did not really have any additional effect over IFN-Y administered in PBS. The reason for the lack of any effect of liposomes and PBS seen in the lungs (Fig. 4.8) is probably related to the greater uptake of liposomes by spleens than lungs. 166

Despite the specific formulation of the liposomes used in these experi­ ments, it is known that the major proportion of liposomes is still taken up by the liver and the spleen (Poste et al., 1982). A study on experi­ mental chemotherapy of murine tuberculosis (Vladimirsdy and Ladigina,

1982) showed that liposomal encapsulation of streptomycin decreased the bacillary counts in the spleen but not the lungs.

This finding was a bit surprising in view of reports of enhanced in vivo tumoricidal, antiviral and microbicidal activity after encapsulation in

liposomes. There have been reports of increased activity after lipo­ somal encapsulation of anti leishmanial drugs like antimonials (Alving et a l . , 1978) in experimental visceral leishmaniasis; and antifungal drugs like amphotericin B in experimental Candida albicans infections

(Fraser-Smi th et a l ., 1983; Lopez-Berestein et al., 1983). This seems to be due to the lowered toxicity of the drugs when encapsulated within liposomes thus enabling higher doses to be given. There have also been reports of the enhancement of the antiviral activity of liposome- encapsulated ribavirin against Rift Valley Fever virus infection in mice

(Kende et al. , 1985) and of protection of mice against systemic herpes simplex type 2 infection by liposome encapsulated muramyl tripeptide

(Koff et al., 1985).

One possible explanation for the lack of enhancement of IFN-X activity reported in this Chapter could be related to the mode of activity of

IFN-V. It has been shown that IFN-V reacts with cell surface receptors on murine macrophages in a specific way which initiates macrophage activation for tumoricidal activity (Celada et al., 1984). Eppstein et a l . (1985) has also reported that the antiviral activity of liposome 167

encapsulated murine IFN-Y in an in vitro system is mediated by leakage

of IFN-Jf from the liposomes and subsequent interaction with a cell membrane receptor. The assumption that interaction of IFN-Y with cell-

surface receptors is essential for macrophage activation could explain

the lack of enhancement by liposomes cited in this thesis. IFN-V

encapsulated in liposomes internalized by macrophages would not have

been allowed to bind with the cell-surface receptors, thereby prevent­

ing the initiation of macrophage activation. It has also been reported

that the in vivo effect of IFN-Y in inhibiting herpes simplex virus-2 murine infections is unchanged by encapsulation in liposomes

(unpublished results cited in Eppstein et al. 1985).

There is a further possible reason for the failure to detect any enhancement of effects on the in vivo growth of M. tuberculosis in mice.

It could be that the lowest dose size of IFN-Y tested (1000 units) had already maximally activated macrophages thus masking any additional effects of liposomes. On retrospective consideration, a much lower dose of IFN-Y should have been used for encapsulation in liposomes.

Recently it has also been documented that an inhibition instead of an enhancement of effect can been obtained in vitro with liposomes.

Gilbreath et al. (1985) reported a differential inhibition of macrophage microbicidal activity by liposomes in an in vitro infection of murine resident peritoneal macrophages with L. tropica. They found that LKs induced an enhanced microbicidal activity against the protozoan amastigotes but that liposomes abrogated that enhancement. The effect was differential and depended on the type of liposome involved. MLV liposomes composed of PC and PS (molar ratio, 7:3) with and without LK, 168

completely abrogated the LK-induced enhancement of killing but not tumour cytotoxicity. In a subsequent publication (Gilbreath et al.,

1985a) concluded that the inhibition of LK-enhanced killing was probably due to the interference of liposomes with one of the early stages of macrophage activation. 169

CHAPTER 5

THE IMMUNOMODULATING EFFECT OF INTERFERON-GAMMA IN COMBINATION WITH

ISONIAZID AND WITH RIFAMPICIN ON MURINE M. TUBERCULOSIS INFECTION

The next stage of the experiments involved the investigation of the

effects of IFN-X in combination with antituberculous drugs. The effects

of IFN-X in combination with isoniazid and with rifampicin were examined

in an acute infection with M. tuberculosis in BALB/c mice.

5.1 Assessment of interferon-gamma and isoniazid alone and in

combination on M. tuberculosis infection in BALB/c mice.

5.1.1 The effect of interferon-gamma and isoniazid when interferon-

gamma is administered before and after infection of BALB/c mice with

M. tuberculosis

In this experiment, 110 male BALB/c mice, 18-20 g in weight, were

randomly allocated into 4 groups. One group was given 2000 units of

IFN-Y every three days with the first dose 2 days prior to infection;

the second group was given a daily dose of isoniazid (25 mg kg

starting a day after infection; the third group received both IFN-X" and isoniazid in the same schedule and dosages, and the last group comprised untreated controls.

All mice were infected intravenously with a 1:1 dilution of the frozen inoculum. Viable counts set up at infection showed that the infective dose was 6.6 X 10^ cfu per mouse. The number of viable organisms in both organs were determined on Days +1, +2, +3, +4, +6 and +9 post 170

infection. Four mice were sacrificed per time point.

The results of the mean viable counts in the lungs are shown in Figure

5.1. For clarity, the s.e. of the means are not depicted on the Figure but are shown in Table 5A.1 in Appendix 1. The mean viable count of 4 M. tuberculosis in the lungs was 7.2 x 10 one hour after infection.

Over the next three days there was a fall in viable counts in the lungs.

This was a consistent finding in all the experiments performed using this protocol of animal infection. In this experiment, the mean count 3 of the controls fell to reach 2.6 x 10 cfu on Day +3. After Day +3 the mean viable lung counts started increasing exponentially until they 4 reached 4.5 x 10 on Day +9. In the group that had been given IFN-fc there was an initial sharper decrease in mean viable counts over the 3 first two days post infection to 1.87 x 10 cfu. This levelled off over 4 Days +2 and +3 before increasing to 2.03 x 10 on Day +9. This was reflected in the growth curve of the IFN-tf treated group which was para­ llel to that of the untreated controls after Day +4. Thus, it seemed that IFN-tf treatment resulted in an early decrease in mean viable counts in the lungs, and this effect did not increase further with time. This was consistent with the results obtained in the earlier experiments where different dose sizes of IFN-V were investigated (Chapter 4, Fig.

4.4 and Fig. 4.6).

Isoniazid was first given one day post infection and the effects were evident by Day +3. From Day +4 however, the decrease in mean viable counts became progressively larger reflecting an increasing effect with o time until by Day +9 the mean count was only 3.13 x 10 cfu. The group of mice that had received both isoniazid and IFN-if also showed a similar Mean log ^viable mycobacteria per lung 3.0“ 2.0J 2.5“ 3. 5“ 4.0“ 4.5“ 5.0“ 7pu al snai fo a+ () o eeutetd (#). untreated or were (■); fromDay+1 isoniaziddaily plus +7 IFN-Jf from Day +1; ♦) ( Days -2,onisoniazid daily +1,+4, +7; Each point represents the mean loglO cfu per lung ofloglOcfu4 thelung per mean mice.represents point Each BALB/c mice when IFN-lfBALB/cwhen mice after andadministeredbefore was tuberculosis inlungsoftheofgrowth M.on combination in isoniazidaloneandandinterferon-gamma of Effect 5.1Fig. IFN-y was given atIFN-y given was e os adamnsee F-T() on-DaysIFN-JTand+1,-2,administered+4 and per mouse (A) infection. Mice were infected with 6.6 x 105 viable organisms6.6 infected105x with viable were Mice infection.

2000 u per mouse and isoniazidat25andpermouse u mg/kg. as fe infection after Days 171

172

trend except that there was a greater decrease in mean viable counts

during the first day after adminstration of IFN-X. Thereafter the

curves were virtually parallel to each other. Overall, there was a more

rapid decrease in mean viable counts in this group as compared with the

isoniazid group.

Figure 5.2 depicts the results of the mean viable counts in the spleens

of the various experimental groups. The means + s.e. of this experiment

are given in Table 5A.2 in Appendix 1. The Figure shows that the mean 4 viable count in the spleens was 7.7 x 10 cfu at 1 hour after infection

and rose rapidly to 3.65 x 10^ by Day +9. In the group of mice treated

with 2000 units of IFN-K there was an initial period of 2 days where

there was hardly any increase in the mean viable count but after that

period, the mean viable count started increasing in an exponential

manner forming a growth curve parallel to that of the control. Again

this was consistent with the results obtained in the lungs in this

experiment as well as in the previous experiments described in Chapter

4. As the isoniazid was only given a day after infection, its bacteri­

cidal effect was only expected to become evident on or after Day +2. By

Day +2, the mean viable count in the spleen had already been reduced.

This effect increased with time (and doses) until by Day +9 the mean 3 viable counts in the spleen were reduced to 3.78 x 10 cfu. The same

trend was seen in the mean viable spleen counts of the group of mice

that had received both isoniazid and IFN-V, except that there was no

initial rise in the spleen count over the first day post infection.

Hence, there was an initial period of stasis followed by a fall in the

counts. Thereafter, the growth curve was parallel to that of the group on isoniazid alone. Overall, there was a more rapid decrease in mean 7.0 “ 1

3 .0 J |------1------1 I I I I 01 2 31(56789 Days after infection

Fig. 5.2 Effect of interferon-gamma and isoniazid alone and in combination on the growth of M. tuberculosis in spleens of BALB/c mice when IFN-Y was administered before and after infection. Mice were infected with 6.6 x 105 viable organisms per mouse and administered IFN-Y (A) on Days -2 , + 1 , +4 and + 7; daily isoniazid (♦) from Day+l; IFN-Y on Days -2, +1 , +4, +7 plus daily isoniazid from Day+l (■); or were untreated (•). IFN-Y was given at 2000 u per mouse and isoniazid at 25 mg/kg. Each point represents the mean logio cfu per spleen of A mice. 174

viable spleen counts in this group when compared with the group receiv­

ing isoniazid alone.

Analysis of variance of both sets of results was performed by a 3-way

ANOVA (Table 5.1). The main effects are isoniazid (H), interferon (IFN)

and time (Days). In both organs, the main effects were all highly

significant. The first-order interactions showed a highly significant

interaction of isoniazid and days which could be explained by referring

to Fig. 5.1 and Fig. 5.2 which showed as might be expected, that the

counts for mice treated with isoniazid decreased from the untreated mice

increasingly as the experiment proceededi There was no interaction of

isoniazid and IFN which showed that statistically, the effects of

isoniazid and IFN were independent and not additive.

TABLE 5.1 THREE-WAY ANALYSIS OF VARIANCE OF THE EFFECTS OF INTERFERON-V, TIME AND ISONIAZID ON THE GROWTH OF M. TUBERCULOSIS IN BALB/C MICE

Source of LUNG SPLEEN variation DF MS F MS F

*** *** Isoniazid(H) 1 9.600 241 31.380 3826 *** *** IFN-S 1 0.820 20.6 0.340 41.5 *** *** Days 1 0.260 6.6 0.070 8.8

H x IFN-V 1 0.000 0.0 0.000 0.0 *** *** H x Days 1 2.960 74.6 5.168 630

IFN-Jr x Days 1 0.020 0.6 0.007 0.8

H x IFN-* x Days 1 0.020 0.5 0.013 1.5

Replicate error 60 0.040 0.008

*** p < 0.001 175

5»1.2 The effect of interferon-gamma and isoniazld when interferon-

gamma was administered both before and after infection, and only after

Infection of BALB/c mice with M. tuberculosis

In this experiment, the effect of the timing of administration of IFN-Jf

alone and in combination with isoniazid on M. tuberculosis infection was

examined. In the first part of the experiment, IFN-V was given on Day-1

and Day+1 and daily isoniazid was started on Day 0. In the second part,

daily isoniazid was started on Day 0 and IFN-tf was given on Day+5 and

Day+7.

96 male BALB/c mice, were randomly allocated into 6 groups. All mice

were infected intravenously with 3 x 10^ cfu of M. tuberculosis per

mouse on Day 0. The first group was given IFN-V on Day-1 and Day+1; the

second, IFN-Jf on Day+5 and Day+7; the third, daily isoniazid starting

from Day 0; the fourth, IFN-V on Day-1 and Day+1 and daily isoniazid on

Day 0 and Day+1; the fifth, daily isoniazid from Day 0 and IFN-V on

Day+5 and Day+7. The sixth group was the untreated controls. IFN-V was

used at a dose of 2000 units per mouse and isoniazid at 25 mg kg ^ . The

number of viable organisms in lungs and spleens were determined on Days

0, +1, +2, +5, +7, +8 and +9 after infection. Four mice were sacrificed

per group for each time point.

The results of the mean viable counts in the lungs and spleens on Days

0, +1 and +2 are shown in Figures 5.3 and 5.4 respectively. The mean viable count + s.e. are detailed in Tables 5A.3 and 5A.4 in Appendix 1. Mean log^viable mycobacteria per lung Each point represents the mean logio cfu per lung of4mice.lung thecfu logio per represents mean point Each 1ad+, al snai () rmDy0 IFN-JT' from Day 0, (♦) isoniazid daily and-1 +1, -1onDays mice in the first two days after infection, when IFN-)finfection, firsttwoafter the when daysin mice given was n 1 ls snai fo a () o eeutetd (•). oruntreated were (A), from isoniazidDay 0 plusand +1 ibeognssprmueo a n ie F-f( ) onDays andIFN-lfongiven 0per Day organismsmouse viable (O IFN-/ was given at 2000 u per mouse and isoniazid at 25atisoniazidmg/kgmg.and2000 per givenatumouse IFN-/ was before and after infection. Mice were infected with 3 x 1053 x infected with were Mice infection.after beforeand in combination on the growth of M. tuberculosis in lungs of BALB/cof tuberculosis inlungs theM. of ongrowth incombination Fig. 5.3 Effect of interferon-gamma and isoniazid alone andaloneisoniazidand ofinterferon-gamma Effect Fig. 5.3 as fe infection after Days 176

, IFN-ygivenat isoniazid2000 at25andperwas umouse mg/kg. Mean log10viable mycobacteria per spleen a gvnbfr n fe ifcin Mc ee infected Micewere with givenbefore infection.and afterwas Each point representspoint theEach spleenloglOper ofcfu4 mean mice. as-,+ pu al snai rmDy0 A, orwere untreated. daily plusfrom (A), Daysisoniazid-1,Day 0 +1 3organisms105xviable pergivenIFN-ifmouse,on and0 Day (O) nDy 1 n 1 diyioizd ♦ fo a , IFN-if from0,Day on and(♦) Days isoniazid daily -1 +1, on ABcmc i the inBALB/c firstinfection, twomice after daysIFN-lf when incombinationon. the spleens in tuberculosisgrowthofM. of ofinterferon-gammaisoniazid Effectalone andand Fig. 5.4 177 + IFN +H Control H IFN

178

The mean count in the lungs one hour after infection was 2.6 x 10 cfu.

As seen in all the previous experiments, there was a fall in viable counts over the first two days after infection. There was no obvious effect of isoniazid on the growth of M. tuberculosis in the lungs in the first two days. The mice that had been given IFN-Y and IFN-Y together with isoniazid showed an initial sharper decrease in viable counts than the untreated and isoniazid alone groups.

4 In the spleens, the viable count one hour after infection was 1.5 x 10 cfu. Over the next two days there was an increase in viable count in the untreated controls to reach 1.2 x 10^ cfu. The mean viable counts in the group treated with IFN-Y was lower than that of the untreated controls. On Dayf 1, there was no obvious difference between the counts of the groups that had been given IFN-Y, isoniazid, or isoniazid plus

IFN-&. However, all three treatment groups had a lower mean spleen count than the untreated controls. By Day+2, the mean counts in the isoniazid groups were evidently lower than the group given IFN-Jf alone, but there was no difference between the isoniazid and the isoniazid plus

IFN-Y group.

Statistical analysis of the results was performed by a 3-way analysis of variance. The results are shown in Table 5.2. 179

TABLE 5.2

THREE-WAY ANALYSIS OF VARIANCE OF THE EFFECTS OF TIME, ISONIAZID AND INTERFERON-8 GIVEN BOTH BEFORE AND AFTER INFECTION OF BALB/C MICE WITH M. TUBERCULOSIS

Source of LUNG SPLEEN variation DF MS F MS F

* ** IFN-Y 1 0.671 7.2 0.076 9.5 *** Isoniazid (H) 1 0.075 0.8 0.316 39.5

Days 1 0.116 1.2 0.0004 0.05

Days x IFN-Y 1 0.014 0.2 O'. 003 0.4 * Days x H 1 0.248 2.7 0.042 5.3 ** IFN-Y x H 1 0.022 0.2 0.070 8.8

Days x H x IFN-Y 1 0.001 0.01 0.004 0.5

Replicate error 24 0.093 0.008

*** p < 0.001 ** p < 0.01 * p < 0.05

Among the main effects, IFN-Y and isoniazid were highly significant in

the spleen and IFN-Y was significant at the 5% level in the lung. These results confirmed that IFN-Y given a day before and a day after infec­

tion significantly lowered the mean viable count in the spleen and lungs in the first two days of infection. The effect of isoniazid was highly significant in the spleen but not in the lung in the first two days after infection. There was a significant first-order interaction between isoniazid and IFN-JT in the spleen. However, this was not due to an additive effect between isoniazid and IFN-Y. Figure 5.4 showed that even though IFN-Y decreased the counts it did not increase the rate at which isoniazid killed the bacilli. 180

The results of the second part of the experiment investigating the

effect of IFN-)| administered on Day+5 and Day+7 alone and in combination with isoniazid are shown in Figures 5.5 and 5.6.

Figure 5.5 shows that mean counts of M. tuberculosis in the lungs from

Day 0 to Day+9 with the full details in Table 5A.5 in Appendix 1. In 4 the untreated controls, the mean count was 2.6 x 10 cfu per lung one 3 3 hour after infection and remained in the range of 6.0 x 10 to 8.8 x 10

cfu until Day+5. The mean viable count then rose steadily to reach 2.5 4 x 10 cfu on Day+9. In the group given IFN-Y on Days +5 and +7, there was a decrease in the mean count on Day+7 followed by a steady increase

parallel to the controls. Isoniazid rapidly caused a decrease in the 2 mean viable counts to reach 1.6 x 10 cfu per lung on Day+9. The addition of IFN-& on Day+5 and +7 to the isoniazid treatment did not appear to have had much effect on the counts.

The corresponding mean viable counts in the spleens are shown in Figure

5.6 and the full details in Table 5A.6 in Appendix 1. A similar trend was seen in the spleen. The mean viable count in the untreated controls

rose steadily to reach 3.0 x 10^ cfu per spleen on Day+9. Administra­

tion of IFN-Y on Day+5 and Day+7 caused a slight decrease in the mean count when compared with the controls. Isoniazid given daily caused the 4 mean viable count to decrease rapidly to reach 4.1 x 10 cfu on Day+9.

The addition of IFN-Y on Day+5 and +7 to the isoniazid treatment did not cause any further decrease in viable counts. Days after infection Fig 5. 5 Effect of interferon-gamma and isoniazid alone and in combination on growth of M. tuberculosis in lungs of BALB/c mice when IFN-Y was .administered on Days +5 and +7 and isoniazid from Day 0. Mice were infected with 3 x 103 viable organisms per mouse on Day 0 and given IFN-Y (O) on Days +5 and +7, daily isoniazid (♦ ) from Day 0, daily isoniazid from Day 0 plus IFN-Y on Days +5 and +7 (A ), or were untreated ( • ) . IFN-Y was given at 2000 units per mouse and isoniazid at 25 mg/kg. Each point represents the mean log.^ cfu per lung of 4 mice. 6.5 -| Control ^-OlFN 0c) _0> a 6.0- V.

u ra n 5. 0 _ uo >N E XJ ro U.5-

O) o a.o- ac3

Days after infection

Fig 5.6 Effect of interferon-gamma and isoniazid alone and in combination on growth of M. tuberculosis in spleens of BALB/c mice when IFN-lf was administered on Days +5 and +7 and isoniazid from Day 0. Mice were infected with 3 x 105 viable organisms per mouse on Day 0 and given IFN-y (O) on Days +5 and +7, daily isoniazid (4 ) from Day 0, daily isoniazid from Day 0 plus IFN-T on Days +5 and +7 (A), or were untreated ( •). IFN-X was given at 2000 units per mouse and isoniazid at 25 mg/kg. Each point represents the mean log-jQ cfu per spleen of 4 mice. 183

The lung and spleen counts on Days +5, +7, +8 and +9 were examined

statistically by a three-way analysis of variance (Table 5.3).

TABLE 5.3

THREE-WAY ANALYSIS OF VARIANCE OF THE EFFECTS OF TIME, ISONIAZID AND INTERFERON-GIVEN AFTER INFECTION OF BALB/C MICE WITH M. TUBERCULOSIS

Source of LUNG SPLEEN variation DF MS F MS F

IFN-* 1 0.490 2.9 0.020 2.5 *** *** Isoniazid (H) 1 30.37 178 73.4 9175 * Days 2 0.085 0.5 0.035 4.3

Days x IFN-V 2 0.073 0.4 0.005 0.6 *** *** Days x H 2 1.750 10.2 0.391 49

H x IFN-V 1 0.140 0.8 0.016 2

H x Days x IFN-^ 2 0.010 0.06 0.009 1

Replicate error 36 0.171 0.008

*** p < 0.001 * p < 0.05

These results showed that among the main effects, isoniazid was highly

significant in both lungs and spleens, but IFN-V did not have any signi­

ficant effect in either organ. There was a significant first-order

interaction between isoniazid and time in both the lungs and the spleen which was obviously due to the fact that the counts for mice treated with isoniazid decreased from the untreated mice increasingly as the experiment progressed (Figures 5.5 and 5.6). Thus, it appears that

IFN-V administered on Day+5 and Day+7 did not have any significant effect on the growth of M. tuberculosis in both organs. This was indep­ endent of either time or isoniazid. 184

5.2 The effect of Interferon-gamma and rifamplcin alone and In combination on M. tuberculosis Infection In BALB/c mice

In this experiment, 70 male BALB/c mice, 18-20 g in weight, were

randomly allocated into 4 groups. The first group was given 2000 units

of IFN-^ per mouse on Days -2 and +1; the second group was given a daily

dose of rifampicin at 25 mg kg with the first dose 20 minutes after

infection; the third group received both IFN-V and rifampicin in the

same dosages and schedules and the last group consisted of the untreated

controls. All mice were infected intravenously with 4.8 x 10^ cfu

M. tuberculosis per mouse. The mean viable counts were monitored on

Days +1, +2 and +3 with 4 mice per time point.

The results of the mean viable counts in the lungs are shown in Figure

5.7 with the details of the means + s.e. in Table 5A.7 (see Appendix 1).

As the mean counts were only monitored over the first three days post

infection, Figure 5.7 depicts decreasing counts only. The mean viable 4 counts of the control untreated group decreased from 5.1 x 10 to 3 2.8 x 10 cfu per lung on Day +3. The group that had been treated with

IFN-tf showed a sharper decrease over the first day and then a decrease parallel to that of the untreated group. However, the rifampicin group

showed a much greater decrease over the first day which progressively 2 became larger, to a count of 4.5 x 10 cfu. The group that had been

treated with both IFN-V and rifampicin showed an almost identical decrease in mean viable counts to the rifampicin group.

Figure 5.8 depicts the results of the mean viable counts of the spleens of the various experimental groups. The corresponding means and 185

Days after infection

Fig. 5.7 Effect of interferon-gamma and rifampicin alone and in combination on the growth of M. tuberculosis in lungs of BALB/c mice. Mice were infected with 4.8 x 105 viable organisms per mouse and given IFN-Jf ( A ) on Days -2 and +1, daily rifampicin (♦) from Day 0, IFN-/ on Days -2 and +1 plus daily rifampicin from Day 0 ( ■) or were untreated (•). IFN-)f was given at 2000 u per mouse and rifampicin at 25 mg/kg. Each point represents the mean logio cfu per lung of 4 mice. 186

Fig. 5.8 Effect of interferon-gamma and rifampicin alone and in combination on the growth of M. tuberculosis in spleens of BALB/c mice. Mice were infected with 4.8 x 105-viable organisms per mouse and given IFN-JT (A) on Days -2 and +1, daily rifampicin from Day 0 (♦), IFN-V on Days -2 and +1 plus daily rifampicin from Day 0 (■), or were untreated (•). IFN-if was given at 2000 u per mouse and rifampicin at 25 mg/kg. Each point represents the mean logio cfu per spleen of 4 mice. 187

standard errors are shown in Table 5A.8 in Appendix 1. The mean viable 4 5 spleen count rises from 6.2 x 10 to 1.7 x 10 cfu per spleen in the untreated control group. In the group treated with IFN-Y , the mean viable count showed a much slower increase over the first day but then gradually increased to reach 1.1 x 10^ cfu. This again reflects the generally observed effects of IFN-Y in the previous experiments. As seen with the mean lung counts, rifampicin was remarkably efficacious in lowering the mean count. Again, the effect became progressively larger 4 and within three days the mean count was reduced to 1.6 x 10 cfu.

There was hardly any difference between the group receiving both IFN-& and rifampicin and the rifampicin group.

This experiment showed that treatment of mice with IFN-tf resulted in a considerable reduction in mean viable counts in both lungs and spleens.

However, the combination of IFN-Y and rifampicin did not appear to increase the bactericidal activity of rifampicin.

Analysis of variance of both sets of results was performed by a 3-way

ANOVA. The results of this analysis are shown in Table 5.4. The main effects in this ANOVA are rifampicin (Rf), interferon (IFN) and time

(Day). All the main effects were significant in both lungs and spleen.

There was a significant first-order interaction of rifampicin and time in both organs showing that as the experiment progressed, the viable counts of mice treated with rifampicin decreased from the untreated mice increasingly. There was a significant interaction of rifampicin and

IFN-t in the spleen. This can be explained by referring to Figure 5.8 which shows that even though IFN-Y reduced the viable count it did not increase the rate at which rifampicin killed the tubercle bacilli. 188

TABLE 5.4

THREE-WAY ANALYSIS OF VARIANCE OF THE EFFECTS OF INTERFERON-GAMMA, RIFAMPICIN AND TIME ON M. TUBERCULOSIS INFECTION IN BALB/C MICE

Source of LUNG SPLEEN variation DF MS F MS F

*** *** Rifampicin (Rf) 1 3.033 132 4.075 680 * *** IFN-Y 1 0.121 5.3 0.121 20 *** *** Days 2 2.025 88 0.220 37 ** Rf x IFN-Y 1 0.024 1.0 0.079 13.2

IFN-Y x Days 2 0.006 0.3 0.009 1.5 ** *** Rf x Days 2 0.225 9.8 0.550 92

Rf x Days x IFN-Y 2 0.007 0.3 0.007 1.2

Replicate error 36 0.023 0.006

*** p< 0.001 ** p < 0.01 * p < 0.05

5.3 Discussion

When two antibacterial agents are used in combination, the resulting effect usually falls into one of four categories-synergism, addition, indifference and antagonism (Hami11 o n - M i H e r , 1985). The term

’synergism’ has been used where the combined activity exceeds the sum of the separate activities. ’Addition’ has been used to describe the phenomenon where the combined activity gives an effect greater than that of the single most active component, while the term ’indifference' has been used where individual drugs in combination showed no increased or decreased activity. Where the combination results in a total effect smaller than that produced by the more active component, the term 189

'antagonism* has been used.

The effect of the addition of IFN-& to isoniazid was examined with various combinations of both agents. In the first experiment (see

5.1.1), IFN-& was administered every three days with the first dose on

Day-2 and isoniazid was given daily from Day+1. The results of this experiment (Figures 5.1 and 5.2) showed that there was a suggestion of an 'addition' effect of IFN-lf on the bactercidal activity of isoniazid.

However, statistical analysis revealed no significant effect of IFN-V on isoniazid activity in both organs.

In the second experiment (5.1.2), isoniazid was given daily from Day 0 and IFN-tf was either given on Days -1, +1 or on Days +5, +7. When IFN-fc was given both before and after infection, there was again no statis­ tically significant effect of IFN-& on the bactericidal activity of isoniazid. IFN-ft administered on Day+5 and Day+7 to mice did not have a significant effect on bacillary growth in both organs. Similarly, IFN-# given on Day+5 and Day+7 to a group of mice treated with isoniazid from

DayO did not enhance the bactericidal effect of isoniazid. The appearance of the growth curves (Figs. 5.5 and 5.6) suggest that the effects of IFN-^f and isoniazid are 'indifferent' when IFN-& is administered on Day+5 and +7. Statistical analysis confirmed that

IFN-fc given after infection did not have any significant effect on the bactericidal activity of isoniazid.

The results of the combination of rifampicin with IFN-)( also did not / show any evidence of enhancement of bactericidal activity of rifampicin by IFN-Jf. In this experiment, IFN-if was administered before infection 190

(Day-2) and rifamplcin on Day 0. The growth curves (Figures 5.7 and

5.8) suggest that the effects of IFN-& and rifampicin are ’indifferent* and this was confirmed statistically.

The experiments reported in this Chapter showed that IFN-tf had a significant effect on the growth of M. tuberculosis in the organs of mice when it was administered before infection but not if it was administered five days after infection. Also, there was no significant effect of IFN-lf on the activity of isoniaziti irrespective of whether it was given before or after isoniazid treatment. Similarly, IFN-^f did not increase the bactericidal effect of rifampicin when it was given before rifampicin.

It has been observed that immunomodulating agents must be administered very early in the course of infection if they are to be effective

(Bicker et a l . , 1979; Kokoshis et al., 1978; Block et al., 1978). The best results have generally been obtained if a compound is administered at least 24 hours before infection. The results presented in this

Chapter and the previous Chapter are consistent with these findings.

There have been reports of synergistic effects of combinations of immunomodulators and chemotherapeutic agents. In some of these reports, the timing and mode of administration of both agents largely affected the outcome of the combination. In one such report, Connell et al.

(1985) demonstrated the synergistic effect of combinations of acyclovir and recombinant interferon-alpha against a lethal murine infection of

Herpes simplex virus Type I which depended on the mode and timing of adminstration of both agents. Simultaneous parenteral administration of 191

both agents were only synergistic at certain dose combinations whereas

sequential administration of parenteral IFN followed by oral acyclovir was always synergistic. Another example of the importance of sequence

of administration was shown by Scott (1979) who noted synergism in a

murine tumoricidal model if C . parvum was given before cyclophosphamide

but not in the reverse order. However, the results described in this

Chapter of the combination of IFN-^ and isoniazid did not seem to show

any dependence on sequence of administration of either agent.

Antibiotics are most effective in the presence of an adequate host

immune response (Weinstein and Dalton, 1968). The importance of the

immune response in controlling infections is emphasized by the increase

in the number of infections, especially opportunistic infections, that

occur in immunocompromised patients (Bodey, 1975; Atkinson et al., 1974;

Kreger et al., 1980). It has been reported that several antibiotics are

immunosuppressive. The immunosuppressive properties of chloramphenicol

have been documented (Weisberger et al., 1964; Weisberger et al., 1966)

and have been attributed to inhibition of protein synthesis. Similarly,

the tetracyclines also inhibit protein synthesis and have also been

shown to be immunosuppressive (Munster et al., 1977).

There have been conflicting reports about the immunosuppressive

properties of rifampicin. It has been cited that rifampicin caused

immunosuppression of cellular and humoral responses in animal studies

(Paunescu, 1970; Grassi and Pozzi, 1972; Dajani et al., 1973). Simil­

arly, depressed circulating T cells, suppressed in vitro PHA prolifera­

tive responses and diminished PPD skin reactivity responses have been

detected in tuberculosis patients receiving rifampicin (Mukerjee et al., 192

1973; Ruben et a l . , 1974; Gupta et al., 1975). However, Humber et al.

(1980) have shown no effect of rifampicin on humoral or cellular responses In tuberculosis patients and contacts in a controlled double­ blind study. Miller (1978) also reported that long-term therapy (mean duration 12.7 months) with rifampicin in tuberculosis patients did not suppress the secondary antibody response to influenza vaccination.

Goldstein et al. (1976) noted that rifampicin therapy of 4-24 months in tuberculosis patients caused a depression of PHA but not Con A or pokeweed mitogen proliferative responses, and no alteration of in vivo and in vitro responses to PPD. They concluded that since a favourable therapeutic outcome was achieved, clinically, rifampicin is not significantly immunosuppressive.

It is well established that depressed cell mediated immune responses occur in some tuberculosis patients (Humber et al., 1980). There is the possibility that the use of an immunomodulating agent in combination with antituberculous drugs could increase the the efficacy of the antituberculous drugs by modulating the immune response. Bicker et al.

(1979) used murine models of experimental bacterial infections to inves­ tigate the effects of azimexon (see section 1 .3.3), chloramphenicol and cyclophosphamide. They showed that administration of cyclophosphamide alone increased the mortality among mice infected with C. albicans and

Ps. aeruginosa, but the addition of azimexon reduced the mortality.

Azimexon on its own had no significant effect on the survival rate in acute E. coli infection, however, a synergistic effect of azimexon and chloramphenicol, a bacteriostatic antibiotic, was demonstrated.

Gillisen (1985 ) also explored the possibility of improving the 193

efficiency of antibiotics in the immunocompromised host by concomitant administration of immunomodulators. Azimexon and immunoferon (a glucopeptide) were each given to mice infected with Staphylococcus aureus and immunosuppressed by cyclophosphamide or cortisone. The results showed that low doses of azimexon stimulated the immune response while higher doses were inhibitory; in contrast, low doses of immunoferon inhibited the response while higher doses stimulated it.

These variable responses illustrate the difficulty in achieving the optimal dose with immunomodulators. 194

CHAPTER 6

IN VITRO ASSESSMENT OF THE EFFECTS OF INTERFERON-GAMMA

6.1 Preliminary experiments

6.1.1 Characterization of monolayers

The monolayers were stained by a modified Wright's stain and for non­ specific esterase (see section 2.9.3). Both staining methods confirmed that the monolayers were composed of macrophages (Fig. 6.1A and 6.IB).

6.1.2 The effect of saponin on the viable counts of L. monocytogenes and M. microti

L. monocytogenes:

A suspension of L. monocytogenes containing approximately 5 x 10^ organisms ml ^ was diluted 1 in 10 in duplicate with either distilled water or 2.5 % saponin (w/v in distilled water). All the tubes were incubated at 37°C for 20 minutes and left at room temperature for 2 hours before being sampled for the number of viable organisms. The results of the viable counts are shown in Table 6.1.

The results showed that saponin did not grossly affect the viability of

L. monocytogenes under the conditions tested. On the contrary, the counts of the bacteria suspended in saponin were slightly higher than in distilled water. 195

Fig. 6.1A Wright’s (Diff-Quik) staining of macrophage monolayers established in tissue culture 8-chambered slide for 2 days. Magnification xAOO.

Fig. 6.IB Non-specific esterase staining of macrophage monolayers established in tissue culture 8-chambered slide for 2 days. Magnification x400. 196

TABLE 6.1

THE EFFECT OF SAPONIN ON VIABLE COUNTS OF L. MONOCYTOGENES

Suspended in : Viable listeria per ml

Distilled water A .45 x 105

Distilled water 2.95 x 105

2.5 % saponin 7.65 x 105

2.5 % saponin 4.25 x 105

M. microti:

In this experiment, a suspension of M. microti containing approximately

2 x 10 ^ organisms ml * was used. 100 )il of this bacterial suspension was dispensed into duplicate tubes containing 900 jil of either distilled water, 1 % saponin or 2.5 % saponin. All the tubes were then incubated at 37°C for 20 minutes, left at room temperature for 2 hours after which they were briefly sonicated and sampled for the number of viable myco­ bacteria (see section 2.8.2). The results are shown in Table 6.2.

These results showed that neither 1 % nor 2.5 % saponin had any gross effect on the viable counts of M. microti after 2 hours when compared with the controls. As observed in the experiment with L. monocytogenes, the viable counts obtained after suspension in saponin solutions were slightly higher. This was probably due to the surfactant activity of saponin which could have prevented the M. microti from reclumping after sonication. 197

TABLE 6.2

THE EFFECT OF SAPONIN ON VIABLE COUNTS OF M. MICROTI

Bacteria suspended in: Viable mycobacteria per ml

Distilled water 1.06 X 105

Distilled water 9.6 X 104

1 % saponin 1.47 X io5

1 % saponin 1.59 X 105 Ul

2.5 % saponin 1.37 X I—* o

2.5 % saponin 1.53 X io5

6.2 Listericidal Assay

6.2.1 The effect of dose size of interferon-gamma on the listericidal

activity of peritoneal macrophages after previous exposure for 24 hours

Duplicate monolayers in 16 mm Linbro wells were preincubated for 24

hours with either BMM or BMM containing 10, 100 or 1000 units of IFN- ml The monolayers were then infected with L. monocytogenes at a

bacteria:macrophage ratio of 1:10. The viable counts immediately after

the period allowed for phagocytosis (T q ) and four hours later (T^) of

the monolayers are depicted in Figure 6.2. From the Figure, the control 2 monolayers showed an increase in viable count from 9.4 x 10 to 5.8 x 3 10 cfu in 4 hours, a 617 % increase of the Tq count. Treatment with 10

units resulted in a slight increase in viable count to 134 % of the base

count. 100 units caused a decrease in the T^ counts to 96 % of the Tq

count while 1000 units resulted in a decrease to 70 %. Monolayers that or. ahmnlyrws infected monolayer Each waswith hours. activity of peritoneal macrophages after prior exposure forafterexposureofperitonealpriormacrophages activity 2 Fig. for Viable listeria per monolayer monolayers after infectionafter monolayers 40 6.2 minutes and each bar represents theeachs.eandbarcfuof mean minutes + feto oe sizeofinterferon-gammaofdoselistericidalonEffect nt Itreo / ml / Interferon Units To 198 ( Q ) andafter 2.4 x 104 4 hours cfu listeriacfu T 4 (

24 0>-

199

had been treated with IFN-Y appeared to show a dose-related listericidal activity. Analysis of variance was performed on the viable counts of the monolayers that had been treated with 10, 100 or 100 units of IFN-Y.

The results (Table 6.3) showed that linear regression of dose

(IFN-)f concentration) on time was highly significant confirming the existence of dose-related listericidal activity.

TABLE 6.3

ANALYSIS OF VARIANCE OF THE EFFECT OF INTERFERON-GAMMA DOSE SIZE AND TIME ON LISTERICIDAL ACTIVITY

Source of variation DF MS F

Time (Hours) 1 0.0005 0.3 * Dose 2 0.0152 7.6 * Time x Dose 2 0.0207 10.4 ** Linear dose regression 1 0.0414 20.7

Replicate error 6 0.0020

** p < 0.01 * p < 0.05 200

6.2.2 The effect of varying the dose size and pretreatment period of

interferon-gamma on listericidal activity of peritoneal macrophages

In this experiment, the monolayers were pretreated with 0, 100, 1000

units IFN-Y ml ^ for 24 hours or 0, 10, 100, 1000 units IFN-Jf ml ^ for

48 hours. The results are shown in Figure 6.3. As was seen in the

previous experiment, both 100 and 1000 units of IFN-# activated the

macrophages to be listericidal after previous exposure for 24 or 48

hours. Pretreatment with 10 units IFN-* for 48 hours did not activate

for listericidal activity. The % decrease of viable count over 4 hours

for both treated groups was slightly greater in the 24 hour than 48 hour

group. This difference in the magnitude of the response seemed to be

related to the ability of the untreated monolayers in the 48 hour

pretreatment group to allow extensive proliferation of the bacilli. In

the 48 hour pretreatment monolayers the % increase in viable count over

4 hours was 817 % compared to an increase of only 332 % in the 24 hour monolayers.

Thus, pretreatment of the monolayers for 48 hours did not result in an

increased magnitude of listericidal activity which was probably directly

related to the decreased microbicidal activity of macrophages the longer

it has been cultured in vitro. 201

li 3x10 4 p 2x10 A A 4 A i. 1x10 □ T o • H r , , J2 A o A c 5x1 03 o A E ^ A u 0) a JT. to T A / £ A A l/J / 1x1 03 A / ?- _a> * 1 A / y A ✓ A y .2 / > A y ✓ / c A / y (0 / y

Fig* 6.3 Effect of varying the dose size and pretreatment period of interferon-gamma on listericidal activity of macrophages. Monolayers were preincubated with IFN- for either 24 or 48 hours and infected with 3.3 x 104 cfu listeria per monolayer. Each bar represents the mean + s.e cfu of 2 monolayers after infection TO ( □ ) and after 4 hours T4 ([23) • 202

6*3 In vitro assessment of effects of interferon-gamma on the growth of M. microti

The effects of IFN-V on the growth of mycobacteria in macrophages was investigated. M. microti was a natural choice as it is virulent for mice and has the advantage of ease of handling, being a non-human pathogen. In the first experiments, various doses of IFN-V were added to macrophage monolayers for various periods before and at daily intervals after infection with M. microti. The next set of experiments assessed the effects of various doses of IFN-V when added after infection. Finally, the effects of IFN-V in combination with isoniazid and with rifampicin were investigated in the latter model.

6.3.1 Treatment of macrophage monolayers with interferon-gamma before and after infection

6.3.1.1 The effect of dose size of interferon-gamma added both before and after infection on the growth of M. microti in macrophages

Duplicate monolayers in 16 mm Linbro wells each containing 5 x 10 macrophages were preincubated for 72 hours with either BMM or BMM containing 10, 100 or 1000 units of IFN-V ml The monolayers were infected with 4.4 x 10^ viable M. microti for 60 minutes. The number of viable mycobacteria per monolayer was determined by disruption of monolayers by scraping (section 2.9.4) and performing viable counts.

There was no significant loss of cells from the control or IFN-V treated monolayers as assessed by their DNA content. This was confirmed by 203

daily microscopic examination of the monolayers. Figure 6.4A shows a monolayer that had been been incubated with BMM for 3 days and Figure

6.4B shows a monolayer that had been incubated with BMM plus 100 units

IFN-X per ml for 3 days with daily medium changes. The mean viable counts were determined 1 hour, 1 day and 3 days after infection.

The results of the mean viable mycobacterial counts are shown in Figure

6.5. The treated monolayers appeared to show a dose related response to

previous exposure to IFN-Y, 1000 units having the greatest effect. The bactericidal effects of IFN-Y were evident 1 hour after infection and

did not appear to increase with time. The number of viable bacteria detected in the supernatants after the phagocytosis period and the last

rinses was approximately 5% of the monolayer-associated counts.

Throughout the three days the number of bacteria in the supernatants did not rise above 2% of the monolayer-associated counts.

Figure 6.6 shows the results of an experiment which differed only in the length of time allowed for exposure to IFN-Y and for phagocytosis. The wells which had been previously exposed to 0, 100 or 1000 units IFN-Y for 48 hours were infected with 3.6 x 10^ cfu each and incubated at 37°C for 2 hours. At the end of 2 hours there were considerably more bacilli in these monolayers than in the previous experiment. As seen earlier,

the bactericidal effect of IFN-X was evident after the phagocytosis period and did not increase further with time. However, there were differences between Figures 6.5 and 6.6. In Figure 6.6, there were hardly any differences between the monolayers treated with 100 or 1000 units of IFN-Y except immediately after the phagocytosis period. 204

Fig. 6.4A Macrophage monolayer after 3 days incubation in BMM (with daily medium changes). Magnification x250.

Fig. 6.4B Macrophage monolayer after 3 days incubation in BMM plus 100 u IFN-tf per ml (with daily medium changes). Magnification x250. M.microti in macrophagesafter prior exposure for 72 hours. Monolayerswere infected with 4.4 x105 viable organisms each i . . EffectFig.of 6.5dose size of interferon-gamma on growth of 0 □, or IFfHf1000 100 Eachper(□), u upoint(O) ml.represents themean s.e. + cfu of for 60 minutesafter prior exposure to ) 0 ! u ( , Mean viable mycobacteria per monolayer 2 monolayers. 205 10 u(A), Control lOOOu IFN 1OOu IFN O IFNlOu

M. microti in macrophages after prior exposure for 48 hours. Eachpoint represents the mean 4-s.e. cfu of 3 monolayers. Monolayers wereexposed to or 100 1000 0u u (□), (•), u(^)IFN-X' Fig. before infection with3.6 x105 viable organisms each for Mean viable mycobacteria per monolayer 6.6 Effectof dose size of interferon-gamma on growth of 206 2 hours.

Control 0 u100 1 000 u

207

6.3»1.2 The effect of previous exposure to interferon-gamma on the phagocytosis of M. microti

The previous set of experiments had shown that immediately after the period allowed for phagocytosis, the mean viable counts in treated monolayers were considerably lower than counts in untreated monolayers.

In order to exclude the possibility that this was due to a decrease in uptake of organisms during phagocytosis, an experiment was performed in which both viable counts and counts of stained bacilli were done on macrophages previously exposed to IFN-Y.

This experiment was performed in tissue culture 8-chambered slides with approximately 2 x 10~* macrophages per well. The monolayers were preincubated with BMM or BMM containing 100 or 500 units IFN-Y ml ^ for

48 hours. Infection was achieved with 2 x 10 cfu per well and phagocy­ tosis allowed to proceed for 2 hours. The higher infectivity ratio was chosen to yield countable numbers of bacilli in the stained monolayers.

Mean viable counts mmediately after phagocytosis (T^) and 24 hours later

(T24) are shown in Table 6.4. The viable count results showed the same trend as in previous experiments with the difference in mean viable count between treated and untreated groups apparent immediately after the period allowed for phagocytosis. 208

TABLE 6.4

THE EFFECT OF PREVIOUS EXPOSURE TO INTERFERON-GAMMA ON VIABLE COUNTS OF M. MICROTI IN MONOLAYERS IN 8-CHAMBERED SLIDES

4 IFN-* Mean + s.e.(xlO ) cfu per monolayer

units per ml T T 0 *24

0 9.5 + 0.9 19.4 + 0.9

100 6.7 + 0.9 12.1 + 0.9

500 6.6 + 0.7 12.5 + 0.5

The monolayers were stained for acid-fast bacilli (AFB) immediately after phagocytosis. In each monolayer, 300 macrophages were scored as the number of macrophages containing no AFB, 1-2 AFB, 3-5 AFB and > 5

AFB. There were hardly any macrophages containing >5 AFB and conse­ quently this catergory was excluded from the results. The distribution of uptake of AFB by the monolayers as assessed by staining is depicted in Figure 6.7. The figure showed clearly that there was no detectable difference in uptake of M. microti between the untreated and treated monolayers. From the bacillary counts, the percentage of macrophages infected, the mean number of AFB per macrophage and the mean number of

AFB per infected macrophage were also calculated (Table 6.5). These indices of infection also confirmed that there were no detectable differences between the untreated and treated groups. The results showed that the reduced viable counts immediately after the period allowed for phagocytosis was not due to reduced uptake of bacilli. bar represents the mean s.e.+ number of macrophages containing macrophagesafter pretreatment with interferon-gamma. Distribution Fig. Each 6.7of uptake of acid-fast bacilli by 0 Number of macrophages (□), , ) □ ( 1-2 ), □ or 3-5 (JHJJ) ( bacilli per macrophage. nt Itreo pr ml per Interferon Units 209

210

TABLE 6.5 THE EFFECT OF PREVIOUS EXPOSURE TO INTERFERON-GAMMA ON COUNTS OF ACID-FAST BACILLI IN MACROPHAGE MONOLAYERS

Index of Treatment with IFN-Y ( units/ml) infection 0 100 500

% macrophages infected 28.7 33.3 27.0

Mean AFB per macrophage 0.48 0.62 0.48

Mean AFB per infected macrophage 1.68 1.87 1.50

6.3.1.3 The effect of previous exposure of macrophages to interferon- gamma on viable intracellular M. microti at intervals during the phagocytosis period.

The previous experiments had shown that previous exposure of macrophages to IFN-Y for 48 or 7 2 hours caused a decrease in the number of viable intracellular M. microti immediately after a phagocytosis period of 1 or

2 hours and that this was not due to a decrease in bacillary uptake.

In order to investigate the events occurring during the period allowed for phagocytosis, the number of viable intracellular M. microti was determined at intervals during this period. Monolayers in 16 mm Linbro wells were pretreated for 48 hours with BMM or BMM containing 100 units

IFN-Y ml ^ and were then infected with 3.1 x 10^ cfu M. microti per well. The monolayers were lysed with 1 % saponin after periods of 15,

30, 60 and 120 minutes. One set of wells that had been infected for 120 minutes were overlaid with fresh media and reincubated for a further 24 hours before being lysed. 211

The results are shown in Figure 6.8 . At 15 minutes after infection,

there was not much difference between the number of viable intracellular

M. microti in the untreated and treated monolayers. There were 1.54 3 3 x 10 cfu per untreated monolayer and 1.25 x 10 cfu per treated mono-

layer. However, by 30 minutes after infection, there was a substantial

difference between the treated and untreated monolayers. At 30 minutes

after infection, the mean count of M. microti in the control monolayers

was 6.1 x 10 cfu compared with a mean of 3.6 x 10 in the IFN-tf treated

monolayers. The increase in the number of viable bacilli in both sets

of monolayers were approximately parallel from 30-120 minutes after

infection. The results of the mean viable counts of both sets of

monolayers at the end of the 120 minute phagocytosis period and 24 hours

later were consistent with the previous results (Figures 6.5 and 6.6).

In the next experiment, monolayers in Linbro wells were prepared and

overlaid with either BMM or BMM containing 100 units IFN-X ml The

monolayers were all infected with 3.6 x 10^ cfu M. microti per well for

15, 30 or 60 minutes. At each of these time points, after thorough

washing to remove unattached bacilli, half the monolayers were lysed

w‘ith 1 % saponin to determine the number of cfu while the other half

were overlaid with fresh BMM or BMM containing 100 units IFN-V ml * and

reincubated. After 24 hours, the mean number of viable M. microti per

monolayer was determined.

The results are shown in Figure 6.9. In this experiment, even at 15

minutes after infection there was a difference between the number of

viable bacilli in the treated and untreated monolayers. The mean viable

count in the untreated monolayers 15 minutes after infection was extracellularbacilli from the well. viable intracellular M.microti at intervals during the phagocytosis per ml for 48 hours before infection with3.1 x 105 viable bacilli eidad ■ ■)period representsand (■ the period after the removal of e el Eachper point well.represents the mean s.e. + cfu of 3 monolayers. (- — -) represents the events occurring during the phagocytosis Fig. e id Monolayersperiod. were exposed to 0 (u or IFN-lf 100 •) u (O) Mean viable mycobacteria per monolayer x0 I “ 3x10 x0 - 1x10 5x103- 5x102_ IxlO3- 6.8 U H Effectof previous exposure to interferon-gamma on I I I I 1 3 60 30 15 0 ----- minutes ie fe infection after Time 1 - 212 ~T~ 120 n --- hours 24

1

and ( were exposed ) or IFN-K to100 0 peru u (O (#) ml for 48hours bacilli from the well. beforeinfection with3.6 x 105 viable bacilli Each per well. viable intracellular M.microti at intervals during the phagocytosis represents the events occurring during the phagocytosis period on rpeet h en+se cuo ooaes ) point represents the mean s.e.+ cfu of 3 monolayers. periodand their subsequent fate 24 Monolayershours later. EffectFig.of 6.9previous exposure to interferon-gamma on Mean viable mycobacteria per monolayer x0 - - 6x10H x0 “ 5x10 x0 — 4x10 3x1 3x10 2x10 _ _ 2x10 x0-- 1x10 5X102- ---- °J —i i i— i— °3J u 4 4 4_ 4 4 ) represents the period after the removal of extracellular 1 3 60 30 15 0 minutes ----- ie fe infection after Time r 213 hours "1 24 0 u (15m) u 100 0 u (30m) u 100 u (15m) u 0 30m) ( u 0 0 u (60m) u 100

214

3 3 8.5 x 10 cfu and increased slightly to 9.1 x 10 after 24 hours.

However, in the treated monolayers, the count after 15 minutes was 4.7 3 3 x 10 cfu and decreased slightly to 4.0 x 10 . At 30 minutes after 4 infection the mean count of untreated monolayers was 1.4 x 10 cfu and 4 increased to 1.7 x 10 cfu. In the treated monolayers, the mean counts 3 were 6.1 x 10 at 30 minutes after infection and increased slightly to 3 6.9 x 10 cfu after 24 hours. However, at 60 minutes after infection, 4 the mean viable count in the untreated monolayers was 3.2 x 10 and 4 increased substantially to 4.7 x 10 cfu after 24 hours. The correspon- 4 4 ding counts in the treated monolayers were 1.4 x 10 and 1.7 x 10 cfu.

Both experiments showed that as early as 15 to 30 minutes after infection, differences in the number of viable intracellular bacilli were evident between untreated and treated monolayers. The previous experiment had shown that IFN-V did not reduce bacillary uptake and no significant loss of macrophages from monolayers was seen in any experi­ ment. All the earlier results indicate that macrophages were activated to be bactericidal and the results from both these experiments showed that the bactericidal activity occurs as early as 15-30 minutes after infection. 215

6.3.2 The effect of interferon-gamma added after M. microti infection

of peritoneal macrophages

In these experiments, triplicate monolayers in 16 mm Linbro wells were

established for 48 hours before infection with M. microti. The

monolayers were infected with 5.6 x 10^ viable organisms and phagocy­

tosis allowed to proceed for 2 hours. monolayers were disrupted by

scraping and the remaining monolayers were overlaid with BMM or BMM

containing either 100 or 1000 units IFN-Y ml The mean viable counts

were performed after 1 , 2 and 3 days.

The results are shown in Figure 6.10. In the control untreated

monolayers, the viable count increased from 5 x 10 immediately after

infection to 1.5 x 10^ on Day +3. There was hardly any difference

between all monolayers at Day +1 but by Day +2, both the treated mono-

layers showed an inhibition of growth of M. microti. The monolayers

treated with either 100 or 1000 units of IFN-Y showed fairly similar

viable counts. The results showed that IFN-Y added after infection of macrophages with M . microti had a bacteriostatic effect on the mycobacteria. The bacteriostatic effect was not seen immediately, but was reflected in the viable counts one day later, and there was no

apparent difference between monolayers that had been treated with 100 or

1000 units IFN-Y. After phagocytosis and all the washes the number of

M. microti detected in the supernatants of the monolayers represented only approximately 1 % of the number of monolayer-associated bacteria.

Throughout the 3 days of infection, the number of bacteria present in

the supernatants did not rise above 1 % of the monolayer-associated viable counts. macrophages with M. Monolayers microti. were each infected with •, 0 □, or 1001000 IFN-V Each u(□), 0 u(•), upoint per (A) ml. i . .0 EffectFig.of 6.10interferon-gamma addedafter infection of represents the mean s.e. + of 3 monolayers. x105 5.6viable bacilli for 2 hours before the addition of Mean viable mycobacteria per monolayer x0 J r “J 1x10 ----- as fe infection after Days 1 1 ------

1 2 ----- Control

217

Analysis of variance of all the results confirmed that IFN-X treatment of the monolayers had a highly significant inhibitory effect on the growth of M. microti (F ratio = 11.4; p<0.001).

6.3.3 The effect of interferon-gamma and isoniazid alone and in combination on the growth of M. microti in vitro

Monolayers in 16 mm Linbro wells were established for 48 hours and infected with 2.4 x 10^ viable M. microti per well for 90 minutes. The

Tq monolayers were disrupted by scraping and the remaining monolayers were overlaid with BMM, BMM with IFN-y, BMM with isoniazid or BMM with

IFN-V plus isoniazid. IFN-V' was used at 100 units ml ^ and isoniazid at

1 p g ml Viable counts were determined on Days +1, +2 and +3 by lysing with 1% saponin. The results are shown in Figure 6.11.

4 After phagocytosis, the mean count in the monolayers was 1.3 x 10 cfu per monolayer. The treated monolayers showed a distinct inhibition of bacillary proliferation after Day +1 when compared to the untreated monolayers. The isoniazid treated monolayers inhibited the growth of

M. microti by Day +1 and the inhibition became greater by Day +2. The monolayers that had been treated with IFN-X and isoniazid showed a similar inhibition to the isoniazid treated ones.

A three-way analysis of variance was performed on all the results. The results of the analysis are shown in Table 6.6 and showed a significant interaction of isoniazid and IFN-X. The interaction was present because the counts with IFN-V alone were less than those for the control, whereas those with IFN-Y and isoniazid were marginally higher than those were each infected with 2.4 x105 viable bacilli for 90 minutes. l snai ( ). isoniazidml (A Eachpoint s.e. represents cfu of 3 replicates the mean of in combination on growth of M. microti in Monolayersmacrophages. untreatedmonolayers or after (•), the addition of 100 IFN-ifu i* .1 EffectFig* of 6.11 interferon-gamma and isoniazid alone and (O), Mean viable mycobacteria per monolayer x0 — 2x10 x0 — 3x10 5x10 x0 — 4x10 IxlO4— 5x 103__ 4 4 4 Pr isoniazidor 100 (♦), u IFN-V plus 1 pgper Per 1 4 _ 1 3 2 1 0 as fe infection after Days 218

219

with isoniazid alone. However, although the findings suggest slight

'antagonism' between IFN-V and isoniazid, the difference between the counts at 1 , 2 and 3 days for IFN-Y and isoniazid alone do not attain statistical significance.

TABLE 6.6 THREE-WAY ANALYSIS OF VARIANCE OF THE IN VITRO EFFECTS OF TIME, ISONIAZID AND INTERFERON-t ON M. MICROTI INFECTION OF MACROPHAGES

Source of variation DF MS F

IFN-y 1 0.007 2.6 *** Isoniazid (H) 1 0.332 123 *** Days 2 0.059 22 Days x IFN-df 2 0.004 1.5

Days x H 2 0.010 3.6*

** H x IFN-JT 1 0.028 10.4

Days x H x IFN-* 2 0.008 3.0

Replicate error 24 0.0027

*** p < 0.001 ** p < 0.01 * p <0.05

As in the previous experiments, the number of M. microti present in the supernatants did not exceed 5% of the monolayer-associated count and after Day +1, was less than 1%. Phase contrast microscopy did not show any significant loss of macrophages in any monolayer throughout the duration of the experiment. 220

6.3.A The effect of rifampicin and interferon-gamma alone and in combination on the in vitro growth of M» microti

48 hour monolayers in 16 mm Linbro wells were infected with 2.2 x 10^ cfu of M. microti per well. After 90 minutes phagocytosis, the Tq monolayers were disrupted by scraping while the remaining monolayers were overlaid with BMM, BMM with IFN-Y, BMM with rifampicin, or BMM with

IFN-Y and rifampicin. IFN-Y was used at 100 units ml ^ and rifampicin at 10 jjg ml Viable counts were performed on Days +1, +2 and +3 by disrupting the monolayers with 1 % saponin. The mean viable counts are shown in Figure 6.12.

4 After phagocytosis, there were 1.3 x 10 viable mycobacteria in the monolayers. In the untreated monolayers, the bacilli proliferated to 4 reach 3.9 x 10 cfu per monolayer by Day+3. As was seen in the previous results, the IFN-Y treated monolayers showed an inhibition of the pro­ liferation after Day+1. The ri f ampicin-treated monolayers showed a 3 dramatic decrease in the mean counts to 3 x 10 cfu by Day+1, and 1 x 3 10 cfu by Day+3. The monolayers that had been treated with both rifampicin and IFN-Yshowed a similar inhibition to the monolayers treated with rifampicin alone, but to a lesser extent.

A three-way analysis of variance was performed on all the above results

(Table 6.7). The results of the analysis showed a highly significant interaction of IFN-Y and rifampicin. The interaction was because the counts for mice treated with IFN-Y alone were less than those for control mice, whereas counts with IFN-Y and rifampicin were slightly higher than those with rifampicin alone. Even though these findings ml rifampicinml (A). were each infected with 2.2 x 105 viable bacilli for 90 minutes. Eachpoint represents the mean s.e. + cfuof 3 replicates of untreated ormonolayers after the (•), addition of 100 uIFN-JT i* .2 Effect ofFig*interferon-gamma 6.12 and rifampicin alone and incombination on growth of M. microti in macrophages. Monolayers O, 10 pg/mlrifampicin(O), or100 uIFN-£(♦), plus 10 pgper Mean viable mycobacteria per monolayer as fe infection after Days 221

222

suggest 'antagonism' between IFN-Y and rifampicin, the difference between the counts at 1, 2 and 3 days for IFN-V plus rifampicin and rifampicin alone was not statistically significant.

As in the previous experiments, the number of viable bacteria in the supernatants did not exceed 5% of the monolayer-associated count after phagocytosis and was less than 2% after the first day. Phase contrast microscopy did not show any significant loss of macrophages in any monolayer throughout the duration of the experiment.

TABLE 6.7 THREE-WAY ANALYSIS OF THE IN VITRO EFFECTS OF TIME, RIFAMPICIN AND INTERFERON-* ON THE GROWTH OF M. MICROTI IN MACROPHAGES

Source of variation DF MS F

ifn-3 1 0.000 0 *** Rifampicin (R) 1 5.733 940 ** Days 2 0.045 7.4

Days x IFN-Y 2 0.000 0 *** Days x R 2 0.211 35 ** R x IFN-Y 1 0.062 10

R x Days x IFN-Y 2 0.016 2.6

Replicate error 24 0.006

*** p< 0.001 ** p < 0.01 223

6.4 Discussion

All the experiments in this Chapter were performed on murine resident

peritoneal cells. In the L. monocytogenes and M. microti assays, a low

bacteria : macrophage ratio was used. This ensured that extracellular

bacteria were easily removed after phagocytosis and that the assays were

not compromised by extracellular events. In all of the experiments, the

extracellular component only accounted for about 1-5 % of the monolayer-

associated bacteria at the end of phagocytosis and after the first day

they rarely accounted for more than 1%. The low numbers of bacteria in

the supernatants meant that there was no need to resort to the use of

antibiotics at any time. This was important because it had been shown

that antibiotics influence the results of intracellular killing assays

(Hart, 197A; Cole and Brostoff, 1975; Biroum-Noerjasin, 1977).

The listericidal assays established that IFN-Y activated macrophages for

listericidal activity. Preincubation of macrophages for 24 hours was

sufficient for activation and 48 hours did not seem to increase the

magnitude of the listericidal activity. However, this was probably due

to the decreased microbicidal activity of macrophages the longer it has

been cultured in vitro. Nathan et al. (1983) had observed that the

effects of IFN-Y were less dramatic when added to freshly isolated monocytes and that this was due to the release of copious ^2^2' ^he

activation of macrophages to be listericidal by IFN-Y was shown to be

significantly dose-related. 10 units IFN-Y ml ^ was insufficient and a

dose between 10-100 units ml ^ was required for activation with 1000 units giving the biggest response. 224

The experiments with M. microti established that IFN-V could activate

macrophages for antimycobacterial activity. The effect of IFN-tf was

investigated when it was added both before and after infection and also

when it was added only after infection.

IFN-X added 48 or 72 hours before infection caused a decrease in the

viable monolayer-associated counts of M. microti. This bactericidal

effect was evident as early as an hour after infection and did not

increase with further doses of IFN-V. There were several explanations

for the phenomenon. It could have been due to a loss of macrophages in

the treated monolayers, a decrease in uptake of bacilli by treated cells

or a rapid kill of bacilli occurring during phagocytosis. The monitor­

ing of macrophage numbers by phase-contrast microscopy and DNA analysis

confirmed that there was no significant loss of macrophages in the

treated monolayers. The experiment where both viable counts and acid-

fast bacilli staining were performed showed that the reduction in viable

counts in the treated monolayers was not accompanied by a similar

reduction in acid-fast bacillary counts, indicating that there was no

reduced uptake of bacilli. All the results suggest that the most likely

explanation for the phenomenon was rapid kill of bacilli occurring

during phagocytosis.

In an attempt to elucidate the events during the period allowed for

phagocytosis, viable counts were performed at short intervals after

infection. The results showed that the bactericidal effects of previous

exposure to IFN-V was seen as early as 15-30 minutes after infection.

These results suggest that the bactericidal activity of macrophages

occurs simultaneously with phagocytosis. Lefford and Runft (1984) have 225

recently reported the rapid killing of 90% of an inoculum of BCG or

M. smegmatis in vivo in 1 hour by peritoneal macrophages in BCG infected mice.

Early bactericidal activity was also observed in all the listericidal

assays. Phagocytosis periods of 40 minutes were employed and without

exception, treated monolayers contained fewer monolayer-associated

bacteria than control monolayers. It has been estimated (Davies, 1983a)

that L. monocytogenes could be killed by activated rat peritoneal macro­

phages approximately 3 minutes after the bacterium makes contact with

the cell. In a study of the listericidal activity of activated human macrophages, Biroum-Noerjasin (1977) reported that the bactericidal

activity started early and was of short duration. He showed that 15 minutes after in vitro infection, activated macrophages had a faster and

greater killing capacity than unactivated macrophages. Blanden (1968)

also reported killing of S. typhimurium by murine peritoneal macrophages within minutes of phagocytosis. This is not totally unexpected because

it has been demonstrated that completion of phagocytosis (Hirsch, 1962),

stimulation of oxidative metabolism (Cohen et al., 1981; Smith et al.,

1980) , and the formation of digestive vacuoles (Zucker-Franklin et al.,

1964; De Heer et al., 1980) occur within minutes of phagocyte-particle

contact.

The addition of IFN-JT after infection demonstrated that IFN-2T could activate macrophages for antimycobacterial activity without the need for previous exposure before infection. There were differences in the response when compared to the pretreatment experiments. The major difference was that when IFN-V’ was added after infection, a bacterio­ 226

static instead of a bactericidal effect was seen. Another difference

was the delay of the onset of the IFN-V effect. There was no effect 24

hours after addition of IFN-Y, but by 48 hours, there was an obvious

difference in viable counts between the control and IFN-y treated

monolayers.

The minimal dose of IFN- that could activate macrophages for antilist-

erial and antimycobacterial activity was between 10-100 unit ml

This is in accord with the doses shown to be effective in several other

murine studies. Murray et al . (1985) reported that preincubation of

murine peritoneal macrophages with 100 units ml ^ recombinant IFN- )(

caused an almost complete inhibition of T. gondii and that 65-75% of

intracellular L. donovani were killed by preincubation with 10-100 units

per ml. They also found that extending the pretreatment period to 72

hours or increasing the concentrations to above 100 units did not

further enhance antiprotozoal activity. In another study, Wirth et al.

(1985) found that preincubation with 125 units ml ^ recombinant IFN-Jf

enhanced the phagocytosis and killing of Trypanosoma cruzi in mouse

peritoneal macrophages as well as a macrophage-like cell line P388D1.

They showed that preincubation of at least 24 hours is needed for the

enhancement of phagocytosis and suggested that IFN-jT triggered time-

dependent cellular events leading to enhanced phagocytosis.

Recombinant IFN-& has also been shown to activate human monocytes

in vitro. Murray et al. (1983) showed that treatment for 72 hours with 300 units ml ^ IFN-V primed monocytes to generate 7-8 fold more

H2O2 and enhanced ant i lei shmanial activity. Similarly, Nathan et al.

(1983) established that the incubation of monocytes with 100-300 units 227

ml * IFN-V for 2-3 days resulted in increased peroxide generation and toxoplasmacidal activity.

There have been conflicting reports about the role of LPS in enhancement of tumoricidal activity by IFN-^f. Pace et al. (1985) found that IFN-^f could not activate peritoneal mouse macrophages to kill P815 cells unless LPS was present as a second signal. However Varesio et al.

(1984) found significant levels of cytolytic activity induced by the same source of recombinant IFN-X in the absence of LPS. Subsequently, both sets of investigators collaborated in some experiments to unravel the controversies and concluded that the strain of mouse and assay conditions influences the role of LPS in IFN-& activation for tumoricidal activity (Pace et al., 1985).

The level of LPS contamination of the IFN-2T used in the results reported in Chapter 6 was examined by testing the levels of endotoxin in the media used in some experiments. BMM with and without IFN-V was tested for endotoxin by the limulus lysate assay (assays were performed by J.

McConnell, Bacteriology Department, RPMS). Most of the samples tested revealed no presence of endotoxin (<0.1 ng ml ^) and the highest level recorded was 10 ng ml The amount of endotoxin that has been shown to influence activation of macrophages for tumoricidal activity by MAF is

500 ng ml ^ (Svedersky et al., 1984). It was thus rather unlikely that

LPS played a role in the antilisterial and antimycobacterial activities reported in this Chapter.

Although the growth curves of M. microti suggested that IFN-X might have been slightly 'antagonistic* to isoniazid and to rifampicin, this effect 228

was not statistically significant. Hence, the effect of the combination

of IFN-Jf with isoniazid and rifampicin should be considered as one of

'indifference'. The suggestion of slight 'antagonism' observed was not

totally unexpected. Both isoniazid and rifampicin are bactericidal

agents and it is known that bactericidal agents can be adversely

affected by agents or conditions that slow down bacterial growth.

Lowrie et al. (1979a) have demonstrated that the activity of penicillin,

a bactericidal agent, on S. typhimurium within macrophages was directly

related to the bacterial growth rate as well as the penicillin concen­

tration. Similarly, Dickinson and Mitchison (1981) have shown that when

the growth rate of M. tuberculosis was reduced uniformly by lowering the

incubation temperature or the pH of the culture medium, the bactericidal

activities of rifampicin and isoniazid were decreased to a similar

extent.

There was a distinct difference in the activity of isoniazid and

rifampicin in this in vitro system. Unlike rifampicin which immediately

decreased the number of M. microti, isoniazid permitted the mycobacteria

to grow within the macrophages for the first day. This was not due to

resistance of M. microti to isoniazid as in vitro sensitivity tests had

shown that, like most strains of M. tuberculosis, the minimal inhibitory

concentration was 0.1 ^g ml This difference in early bactericidal

activity between isoniazid and rifampicin had been previously observed

in in vitro studies on these drugs. An increase in viable counts of a

culture of M. tuberculosis occurred in the first day after exposure to 1

}ig ml * isoniazid (Dickinson and Mitchison, 1966). In contrast, the

bactericidal action of 0.2 jig ml ^ rifampicin was evident after periods

of exposure as short as 2 and 6 hours (Dickinson and Mitchison, 1970a). 229

CHAPTER 7

GENERAL DISCUSSION

In the four decades since the discovery of streptomycin, remarkable advances have been made in the chemotherapy of tuberculosis. However, despite all of these, there is still room for improvement. Shortening the duration of treatment and the use of fully supervised intermittent regimens have helped to overcome some, but not all of the problems of chemotherapy. In technically advanced countries, the most serious problem with short-course chemotherapy is non-compliance (Addington,

1979). It is thus desirable to try to shorten regimens further.

The situation is rather different in poor developing countries. Despite all the improvements made in short-course regimens, their prohibitive cost and the lack of organizational facilities in the Health Services have limited the applications of short-course chemotherapy in most developing countries. Chemotherapy in poor developing countries still largely rely on standard regimens of isoniazid with streptomycin and thiacetazone given for 12-24 months. Even though such regimens have been shown to ensure a long and lasting cure, in actual practice, it falls short of the ideal (New Delhi Tuberculosis Centre, 1977; East

Af ri ca/Bri t i sh Medical Research Council Tanzania Tuberculosis Survey,

1977; Second East Africa/British Research Council Kenya Tuberculosis

Survey, 1979). The main reasons are the lack of treatment centres making it difficult for patients to obtain the drugs, and non-compliance

Non-compliance is a particularly large problem in developing countries because of the long duration of chemotherapy. 230

Immunomodulating agents might be of value in two ways. First, by

increasing the speed with which organisms are killed by antituberculous drugs and second, by killing persisting organisms remaining dormant in

the lesions after completion of chemotherapy. In either case, the result of effective immunomodulating activity would be to shorten the

duration of chemotherapy necessary to achieve a stable cure.

Although there have been a few reports of levamisole in conjunction with

chemotherapy augmenting the clinical response to tuberculosis, the evidence is not very convincing (see section 1.7). The findings reported in this thesis showed that levamisole did not have any signifi­

cant effect on the growth of M. microti in mice. The preliminary results of clinical trials in Zambia and Kenya also showed no evidence of augmentation of response when levamisole was added to the chemothera­ peutic regimen (Kenyan/Zambian/British Medical Research Council Collabo­ rative Study, in preparation). It would appear unlikely that levamisole will have a role in the therapy of tuberculosis.

The results obtained in the in vivo experiments with murine tuberculosis in Chapter 4 showed that 1000 units of recombinant IFN-V administered intravenously every three days with the first dose one or two days prior to infection, had an inhibitory effect on the growth of M. tuberculosis.

However, even though the results were statistically significant, the effects were not very large. An increase in dose size, multiple daily dosage before infection and encapsulation in MLV liposomes did not increase the effect.

The results of the experiments described in Chapter 5 did not show any 231

significant *synerglstlc1 effect of IFN-Y with either Isoniazld or rifampicin. Although there was a suggestion of an Addition* effect of

IFN-'tf with isoniazid In one experiment, this was not shown to be

statistically significant. The effects observed in the combination of

IFN-Y and isoniazid and also with rifampicin in vivo can be described as

’indifferent'. This was apparent irrespective of the timing of adminis­ tration of IFN-V.

The experiments in Chapter 6 showed that IFN-Y could activate murine peritoneal macrophages in vitro for microbicidal activity. Listericidal activity could be achieved by incubating macrophages with IFN-Y for 24 hours prior to infection. The assays with M. microti revealed that exposure of macrophage monolayers to IFN-Y for 2 days prior to infection resulted in a bactericidal effect that occurred mainly in the first

15-30 minutes after infection. However, the addition of IFN-Y to macrophages which had already been infected with M. microti resulted in a bacteriostatic effect on bacillary growth that was delayed by 24 hours.

As was seen in the in vivo experiments, the addition of IFN-Y in vitro did not increase the bactericidal activities of isoniazid or rifampicin.

Although the growth curves of M. microti in macrophages obtained in the in vitro experiments suggested that IFN-Y might have been slightly

’antagonistic’ to the bactericidal activities of isoniazid and rifampi­ cin, this effect was not statistically significant. Hence the in vitro effects of IFN-Y in combination with isoniazid and with rifampicin can be considered as one of ’indifference’. From the results obtained in both the in vivo and in vitro experiments, it would seem that it 232

is unlikely that IFN-8 will have a major impact on the chemotherapy of active tuberculosis, that is to say, in the early stages of drug treatment.

The effect of IFN-}f on established tuberculosis was only assessed in one experiment (see section 5.1.2, Figures 5.5 and 5.6). The results showed that IFN-V did not have any significant effect on the growth of

M. tuberculosis when given 5 days after infection. This was consistent with the findings of others that immunomodulating agents were most effective when administered shortly before infection. Similarly, when

IFN-tf was given to mice that had been treated with isoniazid for 5 days, the effect of IFN-)( was ’indifferent' to the bactericidal activity of isoniazid. Mitchison and Selkon (1956) had shown that BCG vaccination of guinea-pigs increased the bactericidal activity of isoniazid in an

’additive' manner. The results that they obtained suggested that the immune response in the guinea-pigs is primarily bactericidal. However,

Rees and Hart (1960) had shown that in chronic murine tuberculosis, the immune response was primarily bacteriostatic. It would be of consider­ able interest to assess the effects of IFN-tf in combination with isoniazid and with rifampicin in a model of established tuberculosis in mice where its modification of the immune response might influence the outcome of the interactions of IFN-V and antituberculous drugs.

In the past, there was considerable interest in the use of corticoste­ roids in the treatment of tuberculosis. Steroids cause immunosuppres­ sion which might allow dormant bacilli to multiply, thereby becoming susceptible to the bactericidal action of antituberculosis drugs. There have been conflicting reports on the role of corticosteroids in the 233

treatment of tuberculosis. Steroids accelerate experimental disease in steroid-sensitive animals such as mice (Hart and Rees, 1950; Batten and

McCune, 1957), albino-rats (Michael et al., 1950) and rabbits (Bunn and

Drobeck, 1952) and have been administered in the post-chemotherapeutic period to assess the relapse rates in murine experimental chemotherapy

(McCune et al., 1966; Grumbach, 1975). Batten and McCune (1957a) reported that steroids given simultaneously with antituberculous drugs did not enhance nor diminish drug activity. It was noted that steroids did not enhance the sterilizing ability of the combination of pyrazin- amide and isoniazid.

Early controlled clinical trials on the influence of corticosteroids on treatment of pulmonary tuberculosis (Horne, 1960; United States Public

Health Service Tuberculosis Therapy Trials, 1960; British Tuberculosis

Association, 1961) suggested that steroids increased the rate of sputum conversion slightly in the early months and hastened the rate of radiographic improvement. In a review of the literature, Horne (1966) concluded that except for the moribund patient, there is no indication for the use of corticosteroids in routine treatment of pulmonary tuberculosis. The role of steroids in short-course chemotherapy for pulmonary tuberculosis was recently examined in a trial in India

(Tuberculosis Research Centre, 1983). The results showed that predniso­ lone did not influence the speed of sputum conversion. The overall relapse rates were low but there was little difference between the prednisolone group and the controls. It appears unlikely that there will be a role for steroids in reducing even further the duration of short-course chemotherapy. 234

Immunomodulating agents could possibly have a role In the elimination of dormant organisms. Kanal and Kondo (1981) Investigated the ability of an immunomodulator, lentinan (see section 1.3.1) to influence the relapse rate in experimental murine chemotherapy. Mice that had been infected with M. tuberculosis were treated with a combination of either streptomycin, isoniazid and rifampicin or ethambutol, isoniazid and rifampicin for five months. This was followed by lentinan treatment for one month, one month without treatment and then another month of lentinan. The relapse rates were then assessed over the following four months by the number of organs with organisms detectable by culture.

Among the 20 control mice, 11 lungs and 14 spleens were culture-positive compared with 4 positive lungs and 10 positive spleens among 20 mice treated with lentinan. It would seem that lentinan treatment in the post-chemotherapeutic period had reduced the relapse rate in the lungs but not in the spleens.

The problem of dormant tubercle bacilli persisting in lesions is of considerable importance in the control of tuberculosis. An agent effective against the last remaining bacilli in tuberculous lesions would have a direct and immediate effect on the chain of transmission of

M. tuberculosis and would have important epidemiological consequences.

Chemoprophylaxis with isoniazid has been impractical on a massive scale because of the duration of treatment needed. However, an immunomodula- ting agent which acts by stimulating the immune response could be effective against dormant bacilli without the need for extended treatment. The effectiveness of IFN-V on dormant tubercle bacilli when administered after chemotherapy should be explored. This role of IFN-'K could be examined in a post-chemotherapeutic murine model similar to one 235

described by the Cornell group (McCune et al. , 1956; McCune et al.,

1966) or by Kanai and Kondo (1981).

Immunotherapy in tuberculosis has also been attempted with transfer factor. Whitcomb and Rocklin (1973) administered transfer factor to a patient with progressive primary tuberculosis refractory to standard chemotherapy and in whom a defect in cell mediated immune response to tuberculosis was shown. The administration of transfer factor restored immunologic reactivity accompanied by a dramatic clinical improvement.

Rubinstein and colleagues (1977) also successfully used transfer factor to treat a patient with progressive tuberculosis refractory to eight drugs. Clinical recovery was accompanied by recovery of cell-mediated immune function. Transfer factor has also been used to treat a patient with disseminated non-reactive infection with M. kansasii (Daniel et al. , 1975). The patient had failed to respond to 5 weeks of appro­ priate chemotherapy and manifested cutaneous anergy and depressed lymphoproliferative responses. Treatment with transfer factor resulted in dramatic clinical recovery and restoration of DTH and lymphoproli- ferative response to PHA. IFN-X might conceivably have a role in the treatment of infections caused by other mycobacteria like M. avium which are refractory to standard antituberculous drugs.

Usuda and colleagues (1981) have used lentinan with limited success in three patients excreting drug-resistant tubercle bacilli for 10 years.

There was an improvement in phagocytic ability of neutrophils and lymphoproliferative responses in all 3 patients, but only 1 patient ceased bacillary excretion. There is a possibility that other immunomo- dulating agents like IFN-X might prove more effective in treatment of 236

drug-resistant tuberculosis.

The encapsulation of IFN-Y in MLV liposomes did not confer any enhance­ ment of the effect of IFN-Y. The reasons for the lack of enhancement have been discussed in Section 4.6. There are many advantages of using liposomes as drug carriers. They are made from natural constituents and thus are biodegradable and relatively non-toxic. Liposomes are also easily made from a variety of components which lead to a wide variety of biological properties. The sizes of liposomes can also be altered by using different methods of preparation and encapsulation is achieved without the need for chemical modification as no bond-formation is needed. However, there are also several disadvantages in the use of liposomes as drug carriers. Their biodegradability results in a relati­ vely short half-life compared to other synthetic carriers. Another disadvantage is the natural fate of the liposomes to target mainly to macrophages of the liver and spleen, although this might not be a disadvantage in certain applications. Thus, liposomes could be very effective in delivering drugs or immunomodulating agents in diseases caused by intracellular parasites, in neoplastic disorders of histiocytes and lysosomal storage diseases.

Much of the interest in using liposomes has centred on the possibility of ’targeting' these structures to specific cell types by incorporating ligands into the liposomal membrane that would selectively bind to specific cell surface determinants. Leserman et al. (1981) demonstrated a method of covalent coupling of monoclonal antibodies to liposomes that enabled a specific delivery of methotrexate to target murine cells.

There is the exciting possibility to targeting immunomodulators and 237

antituberculous drugs specifically to macrophages harbouring tubercle

bacilli by incorporating monoclonal antibodies to M. tuberculosis into

the liposomal membrane. It would seem that the area of liposome

targeting offers considerable clinical potential.

There is also the possibility that combinations of immunomodulators will

be more effective than single agents. An example is the combination of

C. parvum and levamisole which were effective together but not alone in murine neoplastic models (Anaclerio et al. , 1977). As in the other

reports of combination of agents, the timing of administration of both

levamisole and C. parvum was crucial to the synergy. Brehmer et al.

(1981) demonstrated the synergism of MDP and TDM (trehalose dimycolate)

on resistance of mice against aerogenic infection with M. tuberculosis

and influenza-A virus. MDP was ineffective on its own but when combined

with TDM, it completely protected against influenza-A and inhibited the

growth of M. tuberculosis in the lungs.

One of the major problems with immunotherapy has been that most of the

potent agents are derived from bacteria and other microorganisms. These

natural agents have been largely macromolecular in nature and chemically

not defined. A large number have been pyrogenic and consequently have

been excluded from clinical use. Some of these problems have been

resolved by unravelling the chemical nature of potent agents and

chemical synthesis of analogous molecules. The synthesis of MDP and its

analogues is a good example. Unfortunately, the synthetic molecule is

often not biologically active. In the case of MDP, there have been

hundreds of derivatives with different biological properties. Other

synthetic derivatives of natural agents are bestatin and tuftsin. There 238

is the possibility that by protein engineering, derivatives of agents could be produced that are biologically more active than the original agents. New forms of interferons have been engineered by recombinant

DNA technology. One example is the artificially engineered IFN-beta ser which differs from natural IFN-beta by substitution of serine for cysteine in position 17 (Khosrovi, 1983). This engineered product has biological properties similar to the native product but has superior specific activity and stability. Other examples are 'hybrid' IFNs formed by joining the N-terminal and C-terminal halves of different IFN- alpha subtypes (Streuli et al., 1980; Week et al., 1981; Rehberg et al.,

1982). The 'hybrid' molecules were shown to have biological properties different from their parent molecules.

One other problem is that immunomodulating agents must be administered very early in the course of infection if they are to be effective. This means that in clinical practice they might be of greater value in prophylaxis of opportunistic infections in patients at risk than in the treatment of already established infections. There is difficulty in selecting the dosage, frequency and intervals between administrations.

Part of the difficulty is because the dose-response curve for immuno- therapeutic agents is different from that of other chemotherapeutic agents. It has been shown that supraoptimal doses can produce paradoxical effects. Another problem with immunomodulating agents is that in addition to the desired effect on the immune system there could be other undesirable effects like hypersensitivity or aggravation of autoimmune responses.

The ideal immunomodulating agent should have a defined chemical corapo- 239

sltion to eliminate batch to batch variation and be devoid of carcino­ genic or antigenic activity. A reasonable amount of toxicological and pharmacological evaluations should have been performed to delineate the range of doses and to predict the side-effects. It should also have been thoroughly tested for the whole range of immunological effects to avoid the risk of undesirable immunological reactivity. A more complete understanding of the cellular mechanisms involved in the immune response should enable the development of immumomodulators into an area of safe and efficacious clinical potential. The concept of immumomodulating agents, while still in its infancy, appears to offer considerable therapeutic potential. 240

REFERENCES

ABRA RM & HUNT CA (1982). Liposome disposition in vivo IV. The interac­ tion of sequential doses of liposomes having different diameters. Res Comm Pathol 3j3, 17-31.

ACHARYA K, SULLIVAN RM & ROBSON JM (1958). Immunity in experimental tuberculosis. Am Rev Tuberc Pulm Dis _78, 203-225.

ADAMS JM, KALAJAN VA, MORK BO, ROSENBLATT M, R0THR0CK WJ & O ’LOUGHLIN BJ (1959). Reversal of tuberculin reaction in early tuberculosis. Chest Dis 35, 348-356.

ADDINGTON WW (1979). Patient compliance: The most serious remaining problem in the control of tuberculosis in the United States. Chest 76, 741S-743S.

125 AGUET M (1980). High-affinity binding of I-labelled mouse interferon to a specific cell surface receptor. Nature 284, 459-461.

ALEXANDROVA AE & ZABOLOTNYKH NV (1981). Levamisole treatment of experi­ mental tuberculosis (Russian). Probl Tuberk 10, 55-58.

ALVING CR, STECK EA, CHAPMAN W L , WAITS VB, HENDRICKS LD, SWARTZ EM & HANSON WL (1978). Therapy of leishmania: superior efficacies of liposome-encapsulated drugs. Proc Natl Acad Sci USA 7_5, 2959-2963.

AMERY WK & VERHAEGEN H (1978). Effects of levamisole treatment in cancer patients. J Rheumatol _5 (Suppl 4), 123-134.

ANACKER RL, BARCLAY WR, BREHMER W, GOODE G, LIST RH, RIBI E & TARMANIA DF (1969). Effectiveness of cell walls of Mycobacterium bovis strain BCG administered by various routes and indifferent adjuvants in protec­ ting mice against airborne infection with Mycobacterium tuberculosis strain H37Rv. Am Rev Respir Dis _99, 242-248.

ANACLERIO A, CONTI G, MORAS ML, BARALE C & SPREAFICO F (1977). Effect of the combination of Corynebacterium parvum and levamisole on murine tumours. Eur J Cancer 13, 1451-1453. 241

ANDERSON R, GLOVER A, KOORNHOF HJ & RABSON AR (1976). In vitro stimula­ tion of neutrophil motility by levamisole: maintenance of cyclic GMP levels in chemotactically stimulated levamisole-treated neutrophils. J Immunol 117, 428-432.

ANDERSON R, OOSTHUIZEN R & GRABOW G (1981). Prevention of peroxidase mediated inhibition of neutrophil motility and lymphocyte transformation by levamisole, OMPI, sodium aurothiomalate, indomethacin and tolmetin in vitro. Int J Immunopharmacol 123-132.

ANDERSON P, YIP YK & VILCHEK J (1983). Human interferon-gamma is internalized and degraded by cultured fibroblasts. J Biol Chem 258, 6497-6502.

ANDREW PW, REES ADM, SCOGING A, DOBSON N, MATTHEWS R, WHITTAL TJ, COATES ARM & LOWRIE DB (1984). Secretion of a macrophage-activating factor distinct from interferon-gamma by human T cell clones. Eur J Immunol 14, 962-964.

ARALA-CHAVES MP , HORMANHEIMO M, GOUST JM & FUDENBERG HH (1978). Biological and clinical aspects of transfer factor. In: Immunological engineering (ed DW Jirsch) pp 35-82. MTP Press Ltd., Lancaster UK.

ARMSTRONG JA & HART PD'A (1971). Response of cultural macrophages to Mycobacterium tuberculosis, with observations on fusion of lysosomes with phagosomes. J Exp Med 134, 713-740.

ARMSTRONG JA & HART PD'A (1975). Phagosome-lysosome interactions in cultured macrophages infected with virulent tubercle bacilli. Reversal of the usual nonfusion pattern and observations on bacterial survival. J Exp Med 142, 1-16.

ATKINSON K, KAY HE & MCELWAIN TJ (1974). Septicaemia in the neutropenic patient. Br Med J 3i, 244-247.

ATUK NO & HUNT EH (1971). Serial tuberculin testing and isoniazid therapy in general hospital employees. JAMA 218, 1795-1798. 242

BACH JF (1977). Thymic hormones: Biochemistry and biological and clinical activities. Annu Rev Pharmacol Toxicol L7, 281-291.

BACH JF & CARNAUD C (1976). Thymic factors. Prog Allergy 21.» 342-408.

BAGLIONI C, MINKS MA & MARONEY PA (1978). Interferon action may be mediated by activation of a nuclease by pppA 2 'p5 'A2 'p5f A. Nature 273, 684-687.

BANGHAM AD, STANDISH MM & WATKINS JC (1965). Diffusion of univalent ions across the lamellae of swollen phospholipids. J Mol Biol JJ3, 238- 252.

BARKSDALE L & KIM KS (1977). Mycobacterium. Bacteriol Rev 41, 217-372.

BARNETT M, BUSHBY SRM & MITCHISON DA (1953). Tubercle bacilli resistant to isoniazid: virulence and response to treatment with isoniazid in guinea-pigs and mice. Br J Exp Pathol _34_, 568-581.

BARRETT D J , WARA DW, AMMANN AJ & COWAN MJ (1980). Thymosin therapy in DiGeorge syndrome. J Pediat _97, 66-71.

BAST RC, ZBAR B, BORSOS T & RAPP HJ (1974). BCG and cancer. N Eng J Med 290, 1413-1420.

BATTEN JC & MCCUNE RM (1957). The influence of corticotrophin and certain corticosteroids on populations of Mycobacterium tuberculosis in tissues of mice. Br J Exp Pathol ^3, 413-423.

BATTEN JC & MCCUNE RM (1957a). The influence of corticotrophin and cortisone with antituberculous drugs on populations of Mycobacterium tuberculosis in tissues of mice. Br J Exp Pathol _38, 424-437.

BECKER S (1985). Interferon gamma accelerates immune proliferation via its effect on monocyte HLA-DR expression. Cell Immunol 91^, 301-307.

BENSA JCL, FAURE J, MARTIN H, S0TT0 JJ & SCHAERER R (1976). Levamisole in angio-immunoblastic lymphadenopathy. Lancet 1, 1081. 243

BERG K & HERON I (1980). The complete purification of human leukocyte Interferon. Scand J Immunol 489-502.

BERG K & HERON I (1982). Human leukocyte interferon comprises a continuum of 13 interferon species. In: Human lymphokines (eds A Khan & NO Hill) pp 397-416. Academic Press, New York.

BERTHRONG M (1970). The macrophage in tuberculosis. Adv Tuberc Res 17, 1-24.

BICKER UF (1984). Immunopharmacological properties of 2-Cyanaziridine derivatives. In_: Immune modulation agents and their mechanisms (eds RL Fenichel & MA Chirigos)pp 447-473. Marcel Dekker Inc. New York & Basel.

BICKER U, ZIEGLER AE & HEBOLD G (1979). Investigations in mice on the potentiation of resistance to infections by a new immunostimulant compound. J Infect Dis 139, 389-395.

BIROUM-NOERJASIN (1977). Listericidal activity of non-stimulated and stimulated human macrophages in vitro. Clin Exp Immunol 28^, 138-145.

BLANDEN RV (1968). Modification of macrophage function. J Reticuloendo Soc _5, 179-202.

BLANDEN R V , LEFFORD MJ & MACKANESS GB (1969). The host response to Calmette-Guerin bacillus infection in mice. J Exp Med 129, 1079-1107.

BLOCH H & N0RDIN AA (1960). Production of tuberculin sensitivity. Nature 187, 434-435.

BLOCH H & SEGAL W (1954). Viability and multiplication of vaccines in immunization against tuberculosis. Am Rev Tuberc Pulm Dis 228-248.

BLOCK LH, GE0RG0P0UL0S A, MAYER P & DREWS J (1978). Non-specific resistance to bacterial infection. Enhancement by ubiquinone-8. J Exp Med 14H, 1228-1240.

BLOMGREN H (1980). BestatimA new immunomodulator. In vivo and in vitro studies in man. (Abstr.) Int J Immunopharmacol 2^, 166. 244

BODEY GP (1975). Infections in cancer patients. Cancer Treat Rev 89-128.

BODMER WF (1977). The HLA system. In: Human genetics (eds S Armendares & R Lisker). pp 295-307. Excerpta Medica, Amsterdam.

BOERYD B & HALLGREN B (1985). The fat composition of a mouse diet modifies the effects of levamisole on growth and spread of a murine tumour. Acta Path Microbiol Immunol Scand Sect A 92>_y 99-103.

BONA C, AUDIBERT F, JUY D & CHEDID L (1976). Cell suppression in PPD- induced blast specific response of human peripheral blood lymphocytes. Clin Exp Immunol 26_, 258-266.

BOROS DL (1978). Granulomatous inflammations. Prog Allergy^, 183-267.

BRADSHAW LJ & SUMMER HL (1977). In vitro studies on cell-mediated immunity in patients treated with inosiplex for herpes virus infection. Ann NY Acad Sci 248, 190-196.

BRANCA AA & BAGLIONI C (1981). Evidence that types I and II interferons have different receptors. Nature 294, 768-770.

BREHMER W, MASIHI KN, LANGE W, RIBI E & SCHWARTZMAN S (1981). Non­ specific immunostimulation against aerogenic infection of tuberculosis and influenza in mice by synthetic muramyl dipeptide and trehalose dimy- colate (P3). JEn: Immunomodulation by microbial products and related synthetic compounds (eds Y Yamamura, S Kotani, I Azuma, A Koda & T Shiba) pp 233-236. Excerpta Medica, Amsterdam.

BRITISH THORACIC ASSOCIATION (1980). Short-course chemotherapy in pulmonary tuberculosis. Lancet JL, 1182-1183.

BRITISH THORACIC ASSOCIATION (1982). A controlled trial of six months chemotherapy in pulmonary tuberculosis: results up to 24 months following the end of chemotherapy. Am Rev Respir Dis 126, 460-460.

BRITISH THORACIC AND TUBERCULOSIS ASSOCIATION (1976). Short-course chemotherapy in pulmonary tuberculosis. Lancet 2_> 1102-1104. 245

BRITISH TUBERCULOSIS ASSOCIATION (1961). A trial of corticotrophin and prednisolone with chemotherapy in pulmonary tuberculosis. Tubercle 42, 391-412.

BROWN CA & DRAPER P (1976). Growth of Mycobacterium leprae in the mouse bone marrow: an ultrastructural study. Infect Immun JL3, 1199-1204.

BRULEY-ROSSET M, FLORENTIN I & MATHE G (1976). In vivo and in vitro macrophage activation by systemic adjuvants. Agents Action 251-255.

BUNN P & DROBECK B (1952). Use of rabbit-eye as a tissue to study tuberculosis. III. Effect of adrenal hormones upon course of disease and upon certain immunological responses. Am Rev Respir Dis ^ 6, 175-187.

CANTOR H, SHEN FW & BOYSE EA (1976). Separation of helper T cells from suppressor T cells expressing different Ly components. II Activation by antigen: after immunization, antigen-specific suppressor and helper activities are mediated by distinct T cell subclones. J Exp Med 143, 1391-1401.

CARDAMA JE, GATTI JC, BALINA LM, CABRERA HN & FLIESS EL (1973). Study of the immunostimulating action of levamisole in leprosy. Int J Lepr U, 567-568.

CARRANO RA, IULIUCCI JD, LUCE JK, PAGE JA & IMONDI AR (1984). MVE-2. Development of an immunoadjuvant for cancer treatment. _In: Immune modulation agents and their mechanisms (eds RL Fenichel & MA Chirigos) pp 243-260. Marcel Dekker, New York & Basel.

CELADA A, GRAY PW, RINDERKNETCHT E & SCHREIBER RD (1984). Evidence for a gamma-interferon receptor that regulates macrophage tumouricidal activity. J Exp Med 160, 55-74.

CHAN SH, LEE SK & SIMONS MJ (1976). Levamisole augmentation of lymphocyte hyporesponsiveness to phytohaemagglutinin in patients with pulmonary tuberculosis. Proc Soc Exp Biol Med 151, 716-719.

CHAN SH & SIMONS MJ (1975). Levamisole and lymphocyte responsiveness. Lancet 1, 1246-1247. 246

CHANDLER PJ, ALLISON MJ , MARGOLIS G & GERSZTEN E (1965). The effects of intermittent hyperbaric oxygen therapy on the development of tubercu­ losis in the rabbit. Am Rev Respir Dis jU, 855-860.

CHANG EH, JAY FT & FRIEDMAN RT (1978). Physical, morphological and biochemical alterations in the membrane of AKR mouse cells after interferon treatment. Proc Natl Acad Sci USA 7_5, 1859-1863.

CHEDID LA, PARANT MA, AUDI BERT FM, RIVEAU G J, PARANT FJ, LEDERER E, CHOAY JP & LEFRANCIER PL (1982). Biological activity of a new synthetic muramyl peptide adjuvant devoid of pyrogenicity. Infect Immun 35, 417- 424.

CHEERS C, MCKENZIE FC, PAVLOV H, WAID C & YORK J (1978). Resistance and susceptibility to bacterial infection: course of listeriosis in resistant and susceptible mice. Infect Immun JL9_, 763-770.

CHRETIEN PB, LIPSON SD, MAKUCH R, KENADY DE, COHEN MH & MINNA JD (1978). Thymosin in cancer patients: In vitro effects and correlations with clinical response to thymosin immunotherapy. Cancer Treat Rep 62^, 1787- 1790.

CHUMAK AA & K0STR0MIN AP (1981). Effect of levamisole on the immuno­ logical reactivity of mice infected with tuberculosis (Russian). Probl Tuberk _10, 58-62.

COHEN MH, CHRETIEN PB, IHDE DC, BYRON E, FOSSIECK MD, MAKUCH R, BUNN PA, JOHNSTON AV, SHACKNEY SE, MATTHEWS M J , LIPSON SD, KENADY DE & MINNA JD (1979). Thymosin fraction V and intensive combination chemotherapy. Prolonging the survival of patients with small-cell lung cancer. JAMA 241, 1813-1815.

COHEN MS, RYAN JL & ROOT RK (1981). The oxidative metabolism of thioglycollate-elicited mouse peritoneal macrophages: The relationship between oxygen, superoxide and hydrogen peroxide and the effect of monolayer formation. J Immunol 127, 1007-1011. 247

COHN ML, KOVITZ C, ODA U & MIDDLEBROOK G (1954). Studies on isoniazid and tubercle bacilli. II. The growth requirements, catalase activities, and pathogenic properties of isoniazid-resistant mutants. Am Rev Tuberc 70, 641-664.

COHN ZA (1978). The activation of mononuclear phagocytes: Fact, fancy and future. J Immunol 121, 813-816.

COLE P & BR0ST0FF J (1975). Intracellular killing is due to antibio­ tics. Nature 256, 515-517.

COLLINS FM (1984). Tuberculosis. Tn: Bacterial vaccines (ed R Germai- nier) pp 373-418. Academic Press, London.

COLLINS FM & MACKANESS GB (1970). The relationship of delayed hypersen­ sitivity to acquired antituberculous immunity. I. Tuberculin sensitivity and resistance to reinfection in BCG-vaccinated mice. Cell Immunol JL, 253-265.

COMSTOCK GW & PALMER CE (1966). Long-term results of BCG vaccination in the Southern United States. Am Rev Respir Dis 171-183.

CONNELL EV, CERRUTI RL & TROWN PW (1985). Synergistic activity of combinations of recombinant alpha interferon and acyclovir, administered concomitantly and in sequence, against a lethal Herpes simplex virus Type 1 infection in mice. Antimicrob Agents Chemother 28, 1-4.

CORPER HJ & COHN ML (1943). Studies with the vole (meadow mouse) acid- fast bacillus. Virulence, pathogenicity and specific allergic sensitiz­ ing and immunizing properties. Am J Clin Pathol L3, 18-26.

CORPER HJ & COHN ML (1951). The viability and virulence of old cultures of tubercle bacilli. (Studies on 30-year-old broth cultures maintained at 37°C). Tubercle 32, 232-237.

CRONLY-DILLON S (1974). The effect of preadministration of Corynebacte- rium parvum on the protection afforded by heat-killed and acetone-killed vaccines against experimental mouse typhoid. J Hyg 72, 13-18. 248

CROWLE AJ (1972). Trypsin-extracted immunizing antigen of the tubercle bacillus. A practical vaccine? Adv Tuberc Res 1_8, 31-102.

DA JAN I B, KASIK JE & THOMPSON JS (1973). Effect of rifampicin on the immune response in guinea-pigs. Antimicrob Agents Chemother 3^ 451-455.

DANIEL TM & JANICKI BW (1978). Mycobacterial antigens: A review of the isolation, chemistry and immunological properties. Microbiol Rev 42, 84-113.

DANIEL TM, POWELL AE & BEARD NS (1975). Disseminated non-reactive infection with Mycobacterium kansasii treated with specific transfer factor. Clin Res Z3, 444A.

DAVIES M (1983). Phase variations in the modulation of the immune response. Immunol Today 4_, 103-106.

DAVIES WA (1983a). Kinetics of killing Listeria monocytogenes by macrophages. Rapid killing accompanying phagocytosis. J Reticuloendo Soc 34, 131-141.

DAVIES WC, HUBER H, DOUGLAS SD & FUDENBURG HH (1968). A defect in circulating mononuclear phagocytes in chronic granulomatous disease of childhood. J Immunol 101, 1093-1095.

DAWSON JJY, DEVADATTA S, FOX W, RADHAKRISHNA S, RAMAKRISHNAN CV, SOMADU- NDARAN PR, STOTT H, TRIPATHY SP & VELU S (1966). A five year study of patients with pulmonary tuberculosis in a concurrent comparison of home and sanatorium treatment for one year with isoniazid and para-aminosali- cyclic acid. Bull WHO 3., 533-551.

DEAMER D & BANGHAM AD (1976). Large volume liposomes by an ether vaporization method. Biochim Biophys Acta 443, 629-634.

DE HEER E, KERSTEN MC, VAN DER MEER C, LINNEMANS WA & WILLERS JM (1980). Electron microscopic observations on the interaction of L. monocytogenes and peritoneal macrophages of normal mice. Lab Invest 43, 449-455. 249

DE MAEYER E & DE MAEYER-GUIGNARD J (1982). Immunomodulating properties of interferons. Phil Trans R Soc Lond B 299, 77-90.

DERYNCK R, CONTENT J, DE CLERCK E, VOLEKAERT G, TAVERNIER J, DEVOS R & FIERS W (1980). Isolation and structure of a human fibroblast inter­ feron gene. Nature 285, 542-547.

DERYNCK R, REMAUT E, SAMAN E, STANSSENS P, DE CLERCK E, CONTENT J & FIERS W (1980a). Expression of human fibroblast interferon gene in E. coli. Nature 287, 193-197.

DESIDERIO JV & GORDON-CAMPBELL S (1983). Liposome encapsulated cepha- lothin in the treatment of experimental murine salmonellosis. J Reticu- loendo Soc 279-287.

DE SILVA M, PAGE THOMAS DP, HAZELMAN BL & WRAIGHT P (1979). Liposomes in arthritis: a new approach. Lancet 1320-1322.

DICKINSON JM & MITCHISON DA (1966). In vitro studies on the choice of drugs for intermittent chemotherapy of tuberculosis. Tubercle 4_7, 370- 380.

DICKINSON JM & MITCHISON DA (1970). Observations in vitro on the suita­ bility of pyrazinamide for intermittent chemotherapy of tuberculosis. Tubercle 5^, 389-396.

DICKINSON JM & MITCHISON DA (1970a). Suitability of rifampicin for intermittent administration in the treatment of tuberculosis. Tubercle 51, 82-94.

DICKINSON JM & MITCHISON DA (1976). Bactericidal activity in vitro and in the guinea-pig of isoniazid, rifampicin and ethambutol. Tubercle 57, 251-258.

DICKINSON JM & MITCHISON DA (1981). Experimental models to explain the high sterilizing activity of rifampicin in the chemotherapy of tuberculosis. Am Rev Respir Dis 123, 367-371. 250

DILUZIO NR, MCNAMEE R B , WILLIAMS DL, GILBERT KM & SPANGERS MA (1979). Glucan induced inhibition of tumour growth and enhancement of survival in a variety of transplantable and spontaneous murine tumour models. Adv Exp Med Biol 121A, 269-290.

DILUZIO NR & WILLIAMS DL (1978). Protective effect of glucan against systemic Staphylococcus aureus septicaemia in normal and leukemic mice. Infect Immun 20^y 804-810.

DINARELLO CA, ELIN RJ, CHEDID L & WOLFF SM (1978). The pyrogenicity of the synthetic adjuvant muramyl dipeptide and two structural analogues. J Infect Dis 138, 760-767.

D0MAGK G (1950). Investigations on the antituberculous activity of the thiosemicarbazones in vitro and in vivo. Am Rev Tuberc 6^, 8-19.

DRAPER P (1974). The mycoside capsule of Mycobacterium avium 357. J Gen Microbiol J53, 431-433.

DRAPER P & REES RJW (1970). Electron-transparent zone of mycobacteria may be a defence mechanism. Nature 228, 860-861.

DUB0S RJ (1945). Rapid and submerged growth of mycobacteria in liquid media. Proc Soc Exp Biol Med _58, 361-362.

DUBOS RJ (1955). Properties and structures of tubercle bacilli concerned in their pathogenicity. Symp Soc Gen Microbiol 103-125.

DUMONDE DC, WOLSTENCROFT RA, PANAYI GS, MATTHEW M, MORLEY J & HOWSON WT (1969). 'Lymphokines': non-antibody mediators of cellular immunity generated by lymphocyte activation. Nature 224, 38-42.

EAST AFRICA/BRITISH MEDICAL RESEARCH COUNCIL TANZANIA TUBERCULOSIS SURVEY FOLLOW-UP (1977). Tuberculosis in Tanzania: a follow-up of a national sampling survey of drug resistance and other factors. Tubercle 58, 55-78.

ELLEGAARD J & B0ESEN AM (1976). Restoration of defective cellular immunity by levamisole in a patient with immunoblastic lymphadenopathy. Scand J Haematol 17, 36-43. 251

ELLENS H, MAYHEW E & RUSTUM YM (1982). Reversible depression of the reticuloendothelial system by liposomes. Biochem Biophys Acta 714, 479- 485.

ELLNER JJ (1978). Suppressor adherent cells in human tuberculosis. J Immunol 121, 2573-2579.

ELLNER JJ & DANIEL TM (1979). Immunosuppression by mycobacterial arabinomannan. Clin Exp Immunol ^_5, 250-257.

ELLOUZ F, ADAM A, CIORBARU R & LEDERER E (1974). Minimal structural requirements for adjuvant activity of bacterial peptidoglycan derivatives. Biochem Biophys Res Commun 59, 1317-1325.

EPPSTEIN DA, MARSH YV, VAN DER PAS M, FELGNER PL & SCHREIBER AB (1985). Biological activity of liposome-encapsulated murine interferon gamma is mediated by a cell membrane receptor. Proc Natl Acad Sci USA jJ2^ 3688- 3692.

EPSTEIN LB (1979). The comparative biology of immune and classical interferons. _In: Biology of the lymphokines (eds S Cohen, E Pick and JJ Oppenheim) pp 443. Academic Press, New York.

EPSTEIN LB, KRETH HW & HERZEN LA (1974). Fluorescence-activated sorting of human T and B lymphocytes. II. Identification of the cell type respo­ nsible for interferon production and all proliferation in response to mitogens. Cell Immunol ^^2, 407-421.

EVANS MJ & LEVY L (1972). Ultrastructural changes in cells of the mouse foot-pad infected with Mycobacterium leprae. Infect Immun j), 238-247.

EVERALL JD, DOWD P, DAVIES DAL, O ’NEILL GJ & ROWLAND GF (1977). Treat­ ment of melanoma by passive humoral immunology using antibody drug synergism. Lancet _1, 1105-1106.

FARR AG, KIELY JM & UNANUE ER (1979). Macrophage-T cell interactions involving Listeria monocytogenes -role of the H-2 complex. J Immunol 122, 2395-2404. 252

FELDMAN WH & HINSHAW HC (1944). Effects of streptomycin on experimental tuberculosis in guinea-pigs: A preliminary report. Proc Staff Meetings Mayo Clinic J_9, 593-599.

FELLOUS M, KAMOUN M, GRESSER I & BONO R (1979). Enhanced expression of HLA antigens and ^ 2”raicro8l°bulin on interferon-treated human lymphoid cells. Eur J Immunol 9^, 446-449.

FENNER F (1949). The enumeration of viable tubercle bacilli in cultures and infected tissues. Ann NY Acad Sci S2, 751-764.

FEREBEE SH (1970). Controlled chemoprophylaxis trials in tuberculosis: A general review. Adv Tuberc Res _1_7, 28-106.

FEREBEE SH & PALMER CE (1956). Prevention of experimental tuberculosis with isoniazid. Am Rev Tuberc Pulm Dis 7_3, 1-18.

FIDLER IJ (1980). Therapy of spontaneous metastases by intravenous injection of liposomes containing lymphokines. Science 208, 1469-1471.

FIDLER IJ, BARNES ZL, FOGLER WE, KIRSH R, BUGELSKI P & POSTE G (1982). Involvement of macrophages in the eradication of established metastases following intravenous injection of liposomes containing macrophage activators. Cancer Res 4_2, 496-501.

FIDLER IJ, RAZ A, FOGLER WE, KIRSH R, BUGELSKI P & POSTE G (1980). Design of liposomes to improve delivery of macrophage-augmenting agents to alveolar macrophages. Cancer Res 4460-4466.

FIDLER IJ, S0NE S, FOGLER WE & BARNES ZL (1981). Eradication of sponta­ neous metastases and activation of alveolar macrophages by intravenous injection of liposomes containing muramyl dipeptide. Proc Natl Acad Sci USA _78, 1680-1684.

FIDLER IJ & SPITLER LW (1975). Effect of levamisole on in vitro and in vivo murine host response to syngeneic transplantable tumour. J Natl Cancer Inst 55, 1107-1112. 253

FIELD AK, TYRELL AA, LAMPSON GP & HILLEMAN MR (1967). Inducers of Interferon and host resistance. II. Multistranded synthetic polynucleo­ tide complexes. Proc Natl Acad Sci USA _58, 1001-1010.

FINKELSTEIN MC & WEISSMANN G (1981). Targeting of liposomes. In: Liposomes: from physical structure to therapeutic applications (ed CG Knight) pp443-464. Elsevier/North Holland Biomedical Press.

FISHER RA & YATES F (1963). Statistical tables for biological, agricul­ tural and medical research (6th edition). Oliver & Boyd, London.

FLANNERY GR, ROLLAND JM & NAIRN RC (1975). Levamisole. Lancet _1, 750- 751.

FLORENTIN I, TAYLOR E, DAVIGNY M, MATHE G & HADDEN J (1982). Kinetic studies of the immunopharmacologic effects of NPT 15392 in mice. Int J Immunopharmacol 4_, 225-233.

FOGLER WE, RAZ A & FIDLER IJ (1980). In situ activation of murine macrophages by liposomes containing lymphokines. Cell Immunol _53, 214- 219.

FOX W (1979). The chemotherapy of pulmonary tuberculosis: A review. Chest 76, 785S-795S.

FOX W (1981). Whither short-course chemotherapy? Br J Dis Chest 75, 331-357.

FOX W (1985). Short-course chemotherapy for pulmonary tuberculosis and some problems of its programme application with particular reference to India. Bull Int Union Tuberc 6(D, 40-49.

FOX W & MITCHIS0N DA (1975). Short-course chemotherapy for pulmonary tuberculosis. Am Rev Respir Dis 111, 325-353.

FRASER-SMITH EB, EPPSTEIN DA, LARSEN MA & MATTHEWS TR (1983). Protec­ tive effect of a muramyl dipeptide analog encapsulated in or mixed with liposomes against Candida albicans infection. Infect Immun 39, 172-178. 254

FRIDMAN WH, GRESSER I, BANDU M T , AGUET M & NEAUPORT- SAUTES C (1980). Interferon enhances the expression of Fc-gamma receptors. J Immunol 124, 2436-2441.

FRIEDMAN RM (1967). Interferon binding: The first step in establishment of antiviral activity. Science 156, 1760-1761.

FRIEDMAN RM (1979). Interferons: Interactions with cell surfaces. In: Interferon 1 (ed I Gresser) pp 53-74. Academic Press, New York.

FRIEDMAN H, COLE R & MORIN A (1980). Increased active T cells after isoprinosine. Immunological evidence for a viral etiology certain malignant tumour. Int J Immunopharmacol 2_> 153-164.

FRIIS RR (1972). Interaction of L cells and Chlamydia psittaci: entry of the parasite and host responses to its development. J Bact 110, 706-721.

GATNER EMS (1981). Levamisole augmentation of PPD-induced lymphocyte proliferation. S Afr Med J 59_, 109-110.

GATNER EMS & ANDERSON R (1982). Immune responses and immunostimulation in tuberculosis therapy. S Afr Med J 6^, 707-710.

GEISOW M J , HART PD'A & YOUNG MR (1981). Temporal changes of lysosome and phagosome pH during phagolysosome formation in macrophages: studies by fluorescence spectroscopy. J Cell Biol 8#, 645-652.

GEMMELL CG, PETERSON PK, REGELMAN W, SCHMELING D, HOIDAL JR & QUIE PG (1981). Phagocytosis and killing of Streptococcus pyogenes by human alveolar macrophages. Infect Immun 32^, 1298-1300.

GHOSH T, NORVELL ST, GUCLU A, BODURTHA A, TAI J & MACDONALD AS (1977). Immunochemotherapy of malignant melanoma with chlorambucil-bound antime­ lanoma globulins: Preliminary results in patients with disseminated disease. J Natl Cancer Inst _58, 845-852.

GILBREATH MJ, NACY CA, HOOVER DL, ALVING CR, SWARTZ GM & MELTZER MS (1985a). Macrophage activation for microbicidal activity against Leishmania major: inhibition of lymphokine activation by phosphatidyl- choline-phosphatidylserine liposomes. J Immunol 134, 3420-3425. 255

GILBREATH MJ, SWARTZ CM, ALVING CR, NACY CA, HOOVER DL & MELTZER MS (1985). Differential inhibition of macrophage microbicidal activity by liposomes. Infect Immun 47, 567-569.

GILLISEN G (1985). Antimicrobial chemotherapy in immunocompromised host. Cancer Treat Symp 1_, 37-43.

GLASGOW LA, CRANE JL, SCHLENPNER CJ, KERN ER, YOUNGNER JS & FEINGOLD DS (1979). Enhancement of resistance to murine osteogenic sarcoma in vivo by an extract of Brucella abortus (Bru-Pel): Association with activation of reticuloendothelial system macrophages. Infect Immun 23, 19-26.

GODAL T, REES RJW & LAMVIK JO (1971). Lymphocyte-mediated modification of blood-derived macrophage function in vitro; inhibition of growth of intracellular mycobacteria with lymphokines. Clin Exp Immunol 8^, 625- 637.

GOEDDEL DV, YELVERTON E, ULRICH A, HEYNECKER HL, MIOZZARI G, HOLMES W, SEEBURG PH, DULL T, MAY L, STEBBING N, CREA R, MAEDA S, MCCANDLISS R, SLOMA A, TABOR JM, GROSS M, FAMILETTI PC & PESTKA S (1980). Human leukocyte interferon produced by E. coli is biologically active. Nature 287, 411-416.

GOLDING H, GOLDING B, JACOBSON R, LOMNITZER R, K00RNH0F HJ & RABSON AR (1976). In vitro reversal of cellular unresponsiveness induced by levamisole. Clin Exp Immunol 26_, 295-301.

GOLDSTEIN AL, THURMAN BG, LOW TL, ROSSIO JL & TRIVERS GE (1978). Hormo­ nal influences on the reticuloendothelial system: current status of the role of thymosin in the regulation and modulation of immunity. J Retic- uloindo Soc 23, 253-266.

GOLDSTEIN G (1975). The isolation of thymopoietin (thymin). Ann NY Acad Sci 249, 177-185.

GOLDSTEIN R A , ANG U H , FOELLMER JW & JANICKI BW (1976). Rifampin and cell-mediated immune responses in tuberculosis. Am Rev Resp Dis 113, 197-202. 256

GORDON AH, HART P ’D & YOUNG MR (1980). Ammonia inhibits phagosome - lysosome fusion in macrophages. Nature 286, 79-80.

GORDON S (1976). Macrophage neutral proteinases and chronic inflamma­ tion. Ann NY Acad Sci 278, 176-189.

GOREN MB (1975). Cord factor revisited: A tribute to the late Dr Hubert Bloch. Tubercle 56^ 65-71.

GOREN MB, BROKL 0 & SCHAEFER WB (1974). Lipids of putative relevance to virulence in Mycobacterium tuberculosis: correlation of virulence with elaboration of sulfatides and strongly acidic lipids. Infect Immun 9_, 142-149.

GOREN MB, BROKL 0 & SCHAEFER WB (1974a). Lipids of putative relevance to virulence in Mycobacterium tuberculosis: phthiocerol dimycocerosate and the attenuation indicator lipid. Infect Immun 9^, 150-158.

GOREN MB, HART P'D, YOUNG MR & ARMSTRONG JA (1976). Prevention of phagosome-lysosome fusion in cultured macrophages by sulfatides of Mycobacterium tuberculosis. Proc Natl Acad Sci USA 7_3, 2510-2514.

GRASSI GG & POZZI E (1972). Effect of rifampicin on delayed hypersensitivity reactions. J Infec Pis 126, 542-544.

GRAY PW & GOEDDEL DV (1982). Structure of the human immune interferon gene. 298, 859-863.

GRAY PW & GOEDDEL DV (1983). Cloning and expression of murine immune interferon cDNA. Proc Natl Acad Sci USA j$0, 5842-5846.

GRAY PW, LEUNG DW, PENNICA D, YELVERTON E, NAJARIAN R, SIMONSEN CC, DERYNCK R, SHERWOOD PJ, WALLACE DM, BERGER SL, LEVINSON AD & GOEDDEL DV (1982). Expression of human immune interferon cDNA in Escherichia coli and monkey cells. Nature 295, 503-508.

GRAYBILL JR, CRAVIN PC, TAYLOR RL, WILLIAMS DM & MAGEE WE (1982). Treatment of murine cryptococcosis with liposome-associated amphotericin B. J Infec Dis 145, 748-752. 257

GRAZIANI G & DE MARTIN GL (1977). Pharmacokinetic studies on levami- sole. Absorption, distribution, excretion and metabolism of levamisole

in animals. A review. Drugs Exp Clin Res 2 > 221-233.

GREGORIADIS G & BUCKLAND RA (1973). Enzyme containing liposomes allev­ iate a model for storage disease. Nature 244, 170-172.

GRESSER I, MAURY CH & BROUTY-BOYE D (1972). Mechanism of the antitumour effect of interferon in mice. Nature 239, 167-168.

GRISCELLI C, GROSPIERRE B, MONTREUIL J, FOURNET B, BRUVIER G, LANG JM, MARCHIANI C, ZALISZ R & EDELSTEIN R (1982). Immunomodulation by glyco­ protein fractions isolated from Klebsiella pneumoniae. In: Immunomodula­ tion by microbial products and related synthetic compounds, (eds Y Yama- mura, S Kotani, I Azuma, A Koda & T Shiba) pp 261-265. Excerpta medica, Amsterdam-Oxford-Princeton.

GRISCELLI C, PRIEUR AM & DA GUILLARD F (1978). Levamisole therapy in congenital immunodeficiencies. Ln: Immune modulation and control of neoplasia by adjuvant therapy (ed MA Chirigos) pp 165-170. Raven Press, New York.

GROGG E & PEARCE AGE (1952). The enzymic and lipid histochemistry of experimental tuberculosis. Br J Exp Pathol 33^, 567-576.

GROSSET J (1978). The sterilizing value of rifampicin and pyrazinamide in experimental short course chemotherapy. Tubercle _5^9, 287-297.

GROVER AA, KIM HH, WIEGESHAUS EH & SMITH DW (1967). Host-parasite relationships in experimental airborne tuberculosis. II. Reproducible infection by means of an inoculum preserved at -70°C. J Bacteriol 94, 832-835.

GRUMBACH F (1975). La duree optimale de l'antibiotherapie par l'assoc- iation isoniazide et rifampicin dans la tuberculose experimentale de la souris. Etude de la phase post-therapeutique epreuve de la cortisone. Rev Fr Mai Resp 3, 625-634. 258

GRUMBACH F, CANETTI G & LE LIRZIN M (1969). Rifampicin in daily and intermittent treatment of experimental murine tuberculosis, with emphasis on late results. Tubercle fiO, 280-293.

GRZYBOWSKI S, STYBLO K & DORKEN E (1976). Tuberculosis in eskimos. Tubercle _57, S1-S58.

GUPTA S, GRIECO MH & SIEGEL I (1975). Suppression of T-lymphocyte rosettes by rifampsn. Studies in normals and patients with tuberculosis. Ann Intern Med 82^, 484-488.

GUYRE PM, CRABTREE G R , BODWELL JE & MUNCK A (1981). MLC-condi tioned media stimulate an increase in Fc receptors on human macrophages. J Immunol 126, 666-668.

HADDEN JW, COFFEY RG, HADDEN EM, LOPEZ-CORRALES E & SUNSHINE GH (1975). Effect of levamisole and imidazole on lymphocyte proliferation and cyclic nucleotide levels. Cell Immunol ^0, 98-103.

HADDEN JW, HADDEN EM, SPIRA T, SETTINERI R, SIMON L & GINER-SOROLLA A (1982). Effects of NPT 15392 in vitro on human leukocyte functions. Int J Immunopharmacol 4_, 235-242.

HALPERN BN, PREVOT AR, BIOZZI G, STIFFEL C, M0UT0N D, MORARD JC, B0UTHI- LLIER Y & DECREUSEFOND C (1964). Stimulation de l’activite phagocytair du systeme reticuloendothelial provoquee par Corynebacterium parvum. J Reticuloendo Soc _1, 77-96.

HAMILTON TA, BECTON DL, SOMERS SD, GRAY PW & ADAMS DO (1985). Inter­ feron gamma modulates protein kinase activity in murine macrophages. J Biol Chem 260, 1378-1381.

HAMILTON-MILLER JMT ( 1985). Rationalization of terminology and methodology in the study of antibiotic interaction. J Antimicrob Chemo L5> 655-658.

HARRINGTON-FOWLER L, HENSON PM & WILDER MS (1981). Fate of Listeria monocytogenes in resident and activated macrophages. Infect Immun 33, 11-16. 259

HARRINGTON-FOWLER L & WILDER MS (1982). Fate of Listeria monocytogenes in murine peritoneal macrophage subpopulations. Infect Immun 3>5, 124- 132.

HART P'D (1974). Critical approach to technique of assessment of antibacterial effects of activated macrophages. Ln: Activation of macrophages (eds WH Wagner & H Hahn) pp 131-136. Excerpta medica & American Elsevier, Amsterdam, New York.

HART P'D, ARMSTRONG JA, BROWN CA & DRAPER P (1972). ULtrastructural study of the behavoir of macrophages toward parasitic mycobacteria. Infect Immun 5_, 803-807.

HART P'D & REES RJW (1950). Enhancing effect of cortisone on tubercu­ losis in the mouse. Lancet 2, 391-395.

HART P'D & SUTHERLAND I (1977). BCG and vole bacillus vaccines in the prevention of tuberculosis in adolescence and early adult life. Final report to the Medical Research Council. Br Med J 2^, 293-295.

HART P'D & YOUNG MR (1978). Manipulations of the phagosome-lysosome fusion response in cultured macrophages. Enhancement of fusion by chloroquine and other amines. Exp Cell Res 114, 486-490.

HAWRYLKO E & MACKANESS GB (1973). Immunopotentiation with BCG III. Modulation of the response to a tumour specific antigen. J Natl Cancer Inst 1677-1682.

HEFFERNAN JF, NUNN AJ, PETO J et al.. (1976). A two-year follow up of a national sample survey of new cases of respiratory tuberculosis notified in 1968. Tubercle 57^, 161-175.

HIBBS JB, LAMBERT LH & REMINGTON JS (1972). Possible role of macrophage mediated non-specific cytotoxicity in tumour resistance. Nature New Biology 235, 48-50.

HINSHAW HC & MCDERMOTT W (1950). Thiosemicarbazone therapy of tubercu­ losis in humans. Am Rev Tuberc 61, 145-157. 260

HIRSCH JG (1962). Cinemicrophotographic observations on granule lysis in polymorphonuclear leucocytes during phagocytosis. J Exp Med 116, 827-833.

HIRSCH MS, BLACK PH, WOOD ML & MONACO AP (1972). Effects of pyran copolymer on oncogenic virus infections in immunosuppressed hosts. J Immunol 108, 1312-1318.

HOCHKEPPEL HK (1982). Monoclonal antibody against human interferon- gamma. Nature 296, 258.

HOEPRICH PD (1972). Chemoprophylaxis of infectious diseases. _In: Infec­ tious diseases: a guide to the understanding and management of infec­ tious processes (ed PD Hoeprich) pp 207-222. Harper & Row, New York.

HOGAN NA & HILL HR (1978). Enhancement of neutrophil ehemotaxis and alteration of levels of cellular cyclic nucleotides by levamisole. J Infect Dis 138, 437-444.

HOOPER JA, MCDANIEL MC, THURMAN GB, COHEN GH, SCHULOF RS & GOLDSTEIN AL (1975). Purification and properties of bovine thymosin. Ann NY Acad Sci 249, 125-144.

HORNE NW (1960). Prednisolone in treatment of pulmonary tuberculosis: A controlled trial. Final report to the Research Committee of the Tuberculosis Society of Scotland. Br Med J 2_> 1751-1756.

HORNE NW (1966). A critical evaluation of corticosteroids in tuberculosis. Adv Tuberc Res JL5, 1-54.

HOWARD JG, BIOZZI G, HALPERN BN, STIFFEL C & M0UT0N D (1959). The effect of Mycobacterium tuberculosis (BCG) infection on the resistance of mice to bacterial endotoxin and Salmonella enteritidis infection. Br J Exp Pathol 4U, 281-290.

HUMBER DP, NSAZUMUHIRE H, ALUOCH JA, WEBSTER ADB, ABER VR, MITCHISON DA, GIRLING DJ & NUNN AJ (1980). Controlled double-blind study of the effect of rifampin on humoral and cellular immune responses in patients with pulmonary tuberculosis and in tuberculosis contacts. Am Rev Respir Dis 122, 425-436. 261

INTERNATIONAL UNION AGAINST TUBERCULOSIS COMMITTEE ON PROPHYLAXIS (1982) Efficacy of various durations of isoniazid preventive therapy for tuberculosis: five years of follow-up in the IUAT trial. Bull WHO 60, 555-564.

ISAACS A & LINDENMANN J (1957). Virus interference. I The interferon. Proc Roy Soc Lond (B) 147, 258-267.

ISRAEL L (1977). Immunochemotherapy with Corynebacterium parvum in disseminated cancer. Ann NY Acad Sci 277, 241-251.

ITOH K, INOUE M, KATAOKA S & KUMAGAI K (1980). Differential effect of interferon expression of IgG and IgM Fc receptors on human lymphocytes. J Immunol 124, 2589-2595.

JACKETT PS, ABER VR & LOWRIE DB (1978). Virulence and resistance to superoxide, low pH and hydrogen peroxide among strains of Mycobacterium tuberculosis. J Gen Microbiol 104, 37-45.

JACKETT PS, ABER VR, MITCHISON DA & LOWRIE DB (1981). The contribution of hydrogen peroxide resistance to virulence of Mycobacterium tubercu­ losis during the first six days after intravenous infection of normal and BCG-vaccinated guinea-pigs. Br J Exp Pathol 6J2, 34-40.

JACQUES YV & BAINTON DF (1978). Changes in pH within the phagocytic vacoules of human neutrophils and monocytes. Lab Invest 3J9, 179-185.

JINDANI A, ABER VR, EDWARDS EA & MITCHISON DA (1980). The early bactericidal activity of drugs in patients with pulmonary tuberculosis. Am Rev Respir Dis 121, 939-949.

JONES TC (1975). Phagosome-lysosome interaction with Toxoplasma. In: Mononuclear phagocytes in immunity, infection and pathology (ed R van Furth) pp 595-605. Blackwell. Oxford.

JONES TC & HIRSCH JG (1972). The interaction between Toxoplasma gondii and mammalian cells. II. The absence of lysosomal fusion with phagocytic vacuoles containing living parasites. J Exp Med 136, 1173-1194. 262

KANAI K & KONDO E (1981). Immunomodulating activity of lentinan as demonstrated by frequency limitation effect on post-chemotherapy relapse in experimental mouse tuberculosis. Tn: Manipulation of host defence mechanisms (eds T Aoki, I Urushizaki & E Tsubura) pp 65-75. Excerpta Medica, Amsterdam.

KARNOVSKY ML & LAZDINS JK (1978). Biochemical criteria for activated macrophages. J Immunol 121, 809-813.

KATZ J, KUNOFSKY S & KRASNITZ A (1972). Variation in sensitivity to tuberculin. Am Rev Respir Dis 106, 202-212.

KAYE SB & RICHARDSON VJ (1979). Potential of liposomes as drug carriers in cancer chemotherapy: a review. Cancer Chemother Pharmacol 3^, 81-85.

KAUFMAN SHE, SIMON MM & HAHN H (1981). Macrophage activation in immunity to facultative intracellular bacteria: which T cell subset(s) is(are) involved? Ln: Heterogeneity of mononuclear phagocytes (eds 0 Forster & M Landy) pp 464-468. Academic Press, London.

KEERS RY (1978). Tuberculosis in antiquity. Tn: Pulmonary tuberculosis. A journey down the centuries pp 1-5. Balliere Tindall, London.

KENDE M, ALVING CR, RILL WL, SWARTZ GM & CAN0NIC0 PG (1985). Enhanced efficacy of liposome-encapsulated ribavarin against Rift Valley Fever Virus infection in mice. Antimicrob Agents Chemother Z7, 903-907.

KENYAN/ZAMBIAN/BRITISH MEDICAL RESEARCH COUNCIL COLLABORATIVE STUDY. Double-blind study to explore the role of levamisole in short-course chemotherapy for pulmonary tuberculosis. (in preparation).

KH0SR0VI B (1983). The production, characterization, and testing of a modified recombinant human interferon beta. _In: Interferon:Research, Clinical application and regulatory consideration (eds KC Zoon, PD Noguichi & TY Liu) pp 89-99. Elsevier, Amsterdam.

KIDERLAN A F , KAUFMANN SHE & LOHMANN-MATTHES ML (1984). Protection of mice against the intracellular bacterium Listeria monocytogenes by recombinant immune interferon. Eur J Immunol 14, 964-967. 263

KIM TH & KUBICA GP (1972). Long term preservation and storage of mycobacteria. Appl Microbiol 24_, 311-317.

KIM TH & KUBICA GP (1973). Preservation of mycobacteria: 100% viability of suspensions stored at -70°C. Appl Microbiol .25, 956-960.

KING DP & JONES PP (1983). Induction of la and H-2 antigens on a macrophage cell line by immune interferon. J Immunol 131, 315-318.

KIRCHNER H, PETER H H , HIRT HM, ZAWATZKY R, DALUGGE H & BRADSTREET P (1979). Studies of the producer cell of interferon in human lymphocyte cultures. Immunobiol 156, 65-75.

KLEBANOFF SJ & HAMON CB ( 1975). Antimicrobial systems of mononuclear phagocytes. Iri: Mononuclear phagocytes in immunity, infection and pathology (ed R van Furth) pp 507-529. Blackwell, London.

KLEINERMAN ES, ZICHT R, SARIN RS, GALLO RC & FIDLER IJ (1984). Consti­ tutive production and release of a lymphokine with macrophage-activating factor ability distinct from gamma-interferon by a human T cell leukemia virus-positive cell line. Cancer Res 44^ 4470-4475.

KLEINHENZ ME, ELLNER JJ & DANIEL TM (1979). Immunosuppressive proper­ ties of tubercle bacillus arabinogalactan. Clin Res 27_, 638A.

KLEINSCHMIDT WJ & SCHULTZ RM (1982). Similarities of murine gamma- interferon and the lymphokine that renders macrophages cytotoxic. J Interferon Res _2, 291-299.

KLIMPEL GR & HENNY CS (1978). BCG-induced suppressor cells. I. Demons­ tration of a macrophage-like suppressor cell that inhibits cytotoxic T cell generation in vitro. J Immunol 120, 563-569.

KNIGHT E & K0RANT BD (1977). A cell surface alteration in mouse L cells induced by interferon. Biochem Biophys Res Commun lk_> 707-713.

KOCH R (1882). Die aetiologie der Tuberkulose. Berl Klin Wochenschr 19, 221- 230. 264

KOFF WC, SHOWALTER SD, HAMPAR B & FIDLER IJ (1985). Protection of mice against fatal Herpes Simplex Type 2 infection by liposomes containing muramyl tripeptide. Science 228, 495-497.

KOKOSHIS PL, WILLIAMS DL, COOK JA & DILUZIO NR (1977). Increased resistance to Staphylococcus aureus infection and enhancement in serum lysozyme activity by glucan. Science 199, 1340-1342.

KONDO M, KATO H & MASUDA M (1978). Levamisole and serum complement. N Eng J Med 298, 1146.

KOOK A I , YAKIR Y & TRAININ N ( 1975). Isolation and partial chemical characterization of THF, a thymus hormone involved in immune maturation of lymphoid cells. Cell Immunol 19_, 151-157.

KOSKI IR, POPLACK DG & BLAESE RM (1976). A non-specific esterase stain for identification of monocytes and macrophages. In: In vivo methods in cell mediated and tumour immunity (eds BR Bloom and JR David) pp 359- 362. Academic Press, New York.

KREGER BE, CRAVEN DE & MCCABE WR (1980). Gram-negative bacteremia. IV. Re-evaluation of clinical features and treatment in 612 patients. Am J Med 68, 344-355.

KUHNER AL, CANTOR H & DAVID JR (1980). Ly phenotype of lymphocytes producing murine migration inhibitory factor (MIF). J Immunol 125, 1117-1119.

KURZROCK R, ROSENBLUM MG, SHERWIN SA, RIOS A, TALPAZ M, QUESADA JR & GUTTERMAN JU (1985). Pharmacokinetics, single dose tolerance, and biological activity of recombinant gamma-interferon in cancer patients. Cancer Res 4_5, 2866-2872.

LABARCA C & PAIGEN K (1980). A simple, rapid and sensitive DNA assay procedure. Anal Biochem 102, 344-352.

LAWRENCE HS (1955). The transfer in humans of delayed skin sensitivity to streptococcal M substance and to tuberculin with disrupted leuco­ cytes. J Clin Invest 34, 219-230. 265

LAUCIUS JF, BODURTHA A J , MASTRANGELO MJ & CREECH RH (1974). Bacillus Calmette-Guerin in the treatment of neoplastic disease. J Reticuloendo Soc 16, 347—373.

LAUDE J, LE SOUND B, RANCUREL G, DOUMERC S & MOULIAS R (1980). Isopri- nosine trial in Herpes zoster. Effects on delayed cutaneous hypersensi­ tivity. (Abstr.) Int J Immunopharmacol 2^ 195.

LEACH RH & WELLS AQ (1956). Observations on the growth and the enumera­ tion of vole bacilli. Tubercle J7, 347-357.

LEBLEU B, SHAILA S, CABRER B & LENGYEL P (1976). Interferon, double- stranded RNA and protein phosphorylation. Proc Natl Acad Sci USA 73, 3107-3111.

LEFFORD MJ (1975). Transfer of adoptive immunity in tuberculosis in mice. Infect Immun 11, 1175-1181.

LEFFORD MJ, MCGREGOR DD & MACKANfcSS GB (1973). Properties of lympho­ cytes which will confer adoptive immunity to tuberculosis in rats. Immunology 25, 703-715.

LEFFORD MJ & RUNFT DL (1984). Rapid killing of mycobacteria by acti­ vated mouse peritoneal macrophages. (Abstr U26, p 91). Abstracts of the Annual Meeting of Am Soc Microbiol 1984.

LEIBSON HJ, GEFTER M, ZLOTNIK A, MARRACK P & KAPPLER JW (1984). Role of gamma interferon in antibody-producing responses. Nature 309, 799-801.

LENZINI L, R0TT0LI P & R0TT0LI L (1977). The spectrum of human tubercu­ losis. Clin Exp Immunol 27_> 230-237.

LESERMAN L D , MACHY P & BARBET J (1981). Cell-specific drug transfer from liposomes bearing monoclonal antibodies. Nature 293, 226-228.

LEVY H B , LAW LW & RABSON AS (1968). Inhibition of tumour growth by polyinosinic-polycitidylic acid. Proc Natl Acad Sci USA ^2, 357-361. 266

LEWINSKI UH, MAVLIGIT GM, GUTTERMAN JU & HERSH EM (1977). Administra­ tion of a single dose of levamisole to carcinoma patients: in vivo and in vitro enhancement of cellular immune response. In.: Control of neoplasia by modulation of the immune system (ed MA Chirigos) pp 183- 196. Raven Press, New York.

LOOKE E & ROWLEY D (1962). The lack of correlation between sensitivity of bacteria to killing by macrophages or acidic conditions. Aust J Exp Biol Med Sci AO, 315-320.

L00R F (1979). Structure and dynamics of the lymphocyte surface. _In: B and T cells in immune recognition (eds F Loor & GE Rollants) pp 153-189. Wiley Publication, London..

LOPEZ-BERESTEIN G, MEHTA R, HOPFER RL, MILLS K, KASI L, MEHTA K, FAINS- TEIN V, LUNA M, HERSH EM & JULIANA R (1983). Treatment and prophylaxis of disseminated infection due to Candida albicans in mice with liposome- encapsulated amphotericin B. J Infect Dis 147, 939-945.

LOWRIE DB, ABER VR & CARROL MEW (1979a). Division and death rates of Salmonella typhimurium inside macrophages:use of penicillin as a probe. J Gen Microbiol 110, 409-419.

LOWRIE D B , ABER VR & JACKETT PS (1979). Phagosome-lysosome fusion and cyclic adenosine 3’:5'-monophosphate in macrophages infected with Mycobacterium microti, Mycobacterium bovis BCG or Mycobacterium leprae- murium. J Gen Microbiol 110, 431-441.

LOWRIE DB, JACKETT PS & RATCLIFFE NA (1975). Mycobacterium microti may protect itself from intracellular destruction by releasing cyclic AMP into phagosomes. Nature 254, 600-602.

LURIE MB (1964). Resistance to tuberculosis: experimental studies in native and acquired defence mechanisms. Harvard University Press, Massachusett.

MACKANESS GB (1962). Cellular resistance to infection. J Exp med 116, 381-406. 267

MACKANESS GB (1964). The Immunological basis of acquired cellular resistance. J Exp med 120, 105-120.

MACKANESS GB (1967). The relationship of delayed hypersensitivity to acquired cellular resistance. Br Med Bull 23^, 52-54.

MACKANESS GB & BLANDEN RV (1967). Cellular immunity. Prog Allergy 11, 89-140.

MANSELL PWA, DILUZIO NR, MCNAMEE R, ROWDEN G & PROCTOR JW (1976). Recognition factors and non-specific macrophage activation in the treatment of neoplastic disease. Ann NY Acad Sci 277, 20-44.

MANT0VANI A & SPREAFICO F (1975). Allogeneic tumour enhancement by levamisole a new stimulatory compound: Studies in cell mediated immunity and humoral antibody response. Eur J Cancer JU, 537-544.

MATHE G, P0UILLART P & LA PEYRAQUE E (1969). Active immunotherapy of L1210 leukemia applied after the graft of tumour cells. Br J Cancer 23, 814-824.

MATHE G, FLORENTIN I, BRULEY-ROSSET M, HAYAT M & B0URUT C (1977). Heat- killed Pseudomonas aeruginosa as a systemic adjuvant in cancer immunotherapy. Biomedicine 27, 368-373.

MATSUM0T0 K, 0GAWA H, KUSUMA T, NAGASE 0, SAWAKI N, INAGE M, KUSUM0T0 S, SHIBA T & AZUMA I (1981). Stimulation of non-specific resistance to infection induced by 6-0-Acyl muramyl dipeptide analogs in mice. Infect Immun S2, 748-758.

MAYHEW E, PAPAHADJ0P0UL0S D, RUSTUM YM & CHANDRAKANT D (1978). Use of liposomes for the enhancement of the cytotoxic effects of cytosine arabinoside. Ann NY Acad Sci 308, 371-384.

MCCABE RE, LUFT BJ & REMINGTON JS (1984). Effect of murine interferon gamma on murine toxoplasmosis. J Infect Dis 150, 961-967.

MCCUNE RM, FELDMANN FM, LAMBERT HP & MCDERMOTT W (1966). Microbial persistence. I The capacity of tubercle bacilli to survive sterilization in mouse tissues. J Exp Med 123, 445-468. 268

MCCUNE RM, TOMPSETT R & MCDERMOTT W (1956). The fate of Mycobacterium tuberculosis in mouse tissues as determined by the microbial enumeration technique. II. The conversion of tuberculous infection to the latent state by the administration of pyrazinamide and a companion drug. J Exp Med 104, 763-802.

MCDERMOTT W & TOMPSETT R (1954). Activation of pyrazinamide and nicotinamide in acidic environments in vitro. Am Rev Tuberc Pulm Dis 70, 748-754.

MEDICAL RESEARCH COUNCIL (1956). Medical Research Council Tuberculosis Vaccines Clinical Trials Committee: BCG and vole bacillus vaccines in the prevention of tuberculosis in adolescents. First (Progress) Report. Br Med J _1, 413-427.

MEDICAL RESEARCH COUNCIL (1972). BCG and vole bacillus vaccines in the prevention of tuberculosis in adolescents. Bull WHO 46, 371-385.

MEDICAL RESEARCH COUNCIL INVESTIGATION (1950). Treatment of pulmonary tuberculosis with streptomycin and para-aminosalicylic acid. Br Med J _2, 1073-1085.

MEIJER J, BARNETT G D , KUBIK A & STYBL0 K (1971). Identification of sources of infection. Bull Int Union Tuberc 4_5, 5-50.

MERIGAN TC (1967). Induction of circulating interferon by synthetic anionic polymers of known composition. Nature 214, 416-417.

MERLUZZI VJ, BADGER AM, KAISER CW & C00PERBAND SR (1975). In vitro stimulation of murine lymphoid cell cultures by levamisole. Clin Exp Immunol 22^, 486-492.

MERLUZZI VJ, KAISER CW & C00PERBAND SR (1976). Differential effects of levamisole on murine lymphoid tissues. Fed Proc _35, 334.

MICHAEL M, CUMMINGS MM & BLOOM WL (1950). Course of experimental tuber­ culosis in the albino-rat as influenced by cortisone. Proc Soc Exp Biol Med 75, 613-616. 269

MIDDLEBROOK G, DUBOS RJ & PIERCE C (1947). Virulence and morphological characteristics of mammalian tubercle bacilli. J Exp Med 86^ 175-184.

MILLER W (1978). Long-term therapy with rifampin and the secondary antibody response to killed influenza vaccine. Am Rev Respir Dis 117, 605-607.

MITCHISON DA (1954). Tubercle bacilli resistant to isoniazid. Virulence and response to treatment with isoniazid in guinea-pigs. Br Med J _1, 128-130.

MITCHISON DA (1980). Treatment of tuberculosis. The Mitchell Lecture 1979. J Roy Coll Phys Lond _14, 91-99.

MITCHISON DA (1985). Mechanisms of drug action in short-course chemo­ therapy. Bull Int Union Tuberc 6£, 34-37.

MITCHISON DA, ALLEN BW & LAMBERT RA (1973). Selective media in the isolation of tubercle bacilli from tissues. J Clin Pathol _2(3, 250-252.

MITCHISON DA & SELK0N JB (1956). The bactericidal activities of antituberculous drugs. Am Rev Tuberc Pulm Dis J74_ Suppl., 109-116.

MITCHISON DA, SELK0N JB & LLOYD J (1963). Virulence in the guinea-pig, susceptibility to hydrogen peroxide, and catalase activity of isoniazid- sensitive tubercle bacilli from South Indian and British patients. J Pathol Bacteriol j$6, 377-386.

MIZEL SB (1982). Interleukin 1 and T cell activation. Immunol Rev 63, 51-72.

M0IR DJ, GHOSH A K , ABDULAZIZ Z, KNIGHT PM & MASON DY (1983) . Immuno- enzymatic staining of haematological samples with monoclonal antibodies. Br J Haematol ^5, 395-410.

M0LIN L & STENDAHL 0 (1977). Enhancing effect of levamisole on the phagocytic activity of human neutrophil polymorphonuclear leucocytes in_ vitro. Scand J Haematol 19, 93-98. 270

MONCADA-GONZALEZ B, RODRIGUEZ-ESCOBEDO L & CASTANEDO DE ALBA JP (1976). Effect of levamisole on E-rosettes. N Eng J Med 295, 230.

MUKERJEE P, SCHULDT S & KASIK JE (1973). Effect of rifampin on cutaneous hypersensitivity to purified protein derivative in humans. Antimicrob Agents Chemother 4_, 607-611.

MUNSTER AM, L0ADH0LDT CB, LEARY AG & BARNES MA (1977). The effect of antibiotics on cell-mediated immunity. Surgery 81^, 692-695.

MURRAY HW, RUBIN BY & ROTHERMEL CD (1983). Killing of intracellular Leishmania donovani by 1ymphokine-sti mulated human mononuclear phagocytes. J Clin Invest 72, 1506-1510.

MURRAY HW, SPITALNY GL & NATHAN CF (1985). Activation of mouse perito­ neal macrophages in vitro and in vivo by interferon-gamma. J Immunol 134, 1619-1622.

NAGATA Y, ROSEN 0M, MAKMAN MH & BLOOM BR (1984). Biochemical analysis of mutants of a macrophage cell line resistant to growth inhibitory, of interferon. J Cell Biol 98_, 1342-1347.

NAJJAR VA ( 1974 ). The physiological role of gamma-globulin. In: Advances in enzymology Vol 41 (ed A Meister) pp 129-178. Wiley, London.

NAKAMURA N, MANSER T, PEARSON GDN, DALEY MJ & GEFTER ML (1984). Effect of interferon-gamma on the immune response in vivo and on gene expression in vitro. Nature 307, 381-382.

NATHAN C, KARNOVSKY ML & DAVID JR (1971). Alterations of macrophage functions by mediators from lymphocytes. J Exp Med 133, 1356-1376.

NATHAN CF, MURRAY HW, WIEBE ME & RUBIN BY (1983). Identification of interferon-gamma as the lymphokine that activates human macrophage oxidative metabolism and antimicrobial activity. J Exp Med 158, 670- 689.

NETA R & SALVIN SB (1979). Adjuvants in the induction of suppressor Infect Immun 23, 360-365. 271

NEVEU T, BRANELLEC A & BIOZZI G (1964). Proprietes adjuvantes de Corynebacterium parvum sur la production d'anticorps et sur 1'induction de 1 'hypersensibilite retardee envers les proteins conjuguees. Ann Inst Pasteur 106, 771-777.

NEW RR, CHANCE ML, THOMAS SC & PETERS W (1978). Antileishmanial activity of antimonials entrapped in liposomes. Nature 272, 55-56.

NEW DELHI TUBERCULOSIS CENTRE (1977). Annual report 1977. New Delhi Tuberculosis Centre, New Delhi.

NISHIOKA K (1979). Anti-tumour effect of physiological tetrapeptide tuftsin. Br J Cancer 39^, 342-345.

NISHIURA M, UEHIRA K, HASEGAWA T & TAKEUCHI M (1972). The morphological difference between human and murine leprosy bacilli as revealed by the freeze-etching technique. Int J Leprosy ^0, 89-90.

NOGUEIRA M (1974). The escape of Trypanosoma cruzi from the vacuolar system of macrophages. (Abstr of 14th annual meeting of the American Society for Cell Biology). J Cell Biol ^3, 246A.

NORTH RJ (1973). Importance of thymus-derived lymphocytes in cell- mediated immunity to infection. Cell Immunol _7, 166-176.

NORTH RJ, MACKANESS GB & ELLIOT RW (1972). The histogenesis of immunologically committed lymphocytes. Cell Immunol 3^, 680-694.

NYARI LJ, TAN YH & ERLICH HA (1981). Production and characterization of monoclonal antibodies to human fibroblast interferon. Tn: The Biology of the Interferon system (eds E De Maeyer, G Galasso & H Schellekens) pp 67-71. Elsevier/North Holland Biomedical Press.

O'BRIEN J, KNIGHT S, QUICK NA, MOORE EH & PLATT AS (1979). A simple technique for harvesting lymphocytes cultured in Terasaki plates. J Immunol Methods 27^, 219-223.

OETTGEN HF, PINSKY CM & DELMONTE L (1976). Treatment of cancer with immunomodulators. Corynebacterium parvum and levamisole. Med Clins N America 60, 511-537. 272

OLD LJ, CLARKE DA & BENACERRAF B (1959). Effect of Bacillus Calmette- Guerin infection on transplanted tumours in the mouse. Nature 184, 291- 292.

OPPENHEIM JJ & SEEGER RC (1976). The role of macrophages in the induction of cell-mediated immunity in vivo. In: Immunobiology of the macrophage (ed DS Nelson) pp 111-130. Academic Press, New York.

OSHIMA S, MYRVIK QN & LEAKE E (1961). The demonstration of lysozyme as a dominant tuberculostatic factor in extracts of granulomatous lungs. Br J Exp Pathol 4^2, 138-144.

OSTER CN & NACY CA (1984). Macrophage activation to kill Leishmania tropica: Kinetics of macrophage response to lymphokines that induce antimicrobial activities against amastigotes. J Immunol 132, 1494-1500.

PABST HF & CRAWFORD JA (1974). Enhancement of in vitro cellular immune response by L-tetramisole (abstr). Pediatr Res JB, 416.

PACE JL, RUSSELL SW, TORRES BA, JOHNSON HM & GRAY PW (1983). Recombi­ nant mouse gamma-interferon induces the priming step in macrophage activation for tumour cell killing. J Immunol 130, 2011-2013.

PACE JL, VARESI0 L, RUSSEL SW & BLASI E (1985). The strain of mouse and assay conditions influence whether MuIFN-V primes or activates macro­ phages for tumour cell killing. J Leuk Biol T7, 475-479.

PALLADINO MA, SVEDERSKY LP & SHEPARD HM (1983). Interleukin regulation of the immune system ’’IRIS". Ln: Interferon research, clinical applica­ tion and regulatory consideration (eds KC Zoon, PD Noguichi & TY Liu) pp 139-148. Elsevier, Amsterdam.

PAPAHADJ0P0UL0S D & MILLER N (1967). Phospholipid model membranes. I. Structural characteristics of hydrated liquid crystals. Biochim Biophys Acta 135, 624-638.

PAPAHADJOPOULOS D, VAIL WJ, JACOBSON K & POSTE G (1975). Formation by fusion of unilamellar lipid vesicles. Biochim Biophys Acta 394, 483- 491. 273

PARANT M, AUDIBERT F, CHEDID L, LEVEL M, LEFRANCIER P, CHOAY J & LEDERER E (1980). Immunostimulant activities of a lipophilic muramyl dipeptide derivative and of desmuramyl peptidolipid analogs. Infect Immun 27, 826-831.

PARANT M, PARANT F, CHEDID L, YAPO A, PETIT JF & LEDERER E (1979). Fate 14 of the synthetic immunoadjuvant, muramyl dipeptide ( C-labelled) in the mouse. Int J Immunopharmacol 35-41.

PATTERSON RJ & YOUMANS GP (1970). Demonstration in tissue culture of lymphocyte-mediated immunity to tuberculosis. Infect Immun JL_, 600-603.

PAUNESCU E (1970). In vivo and in vitro suppression of humoral and cellular immunological response by rifampicin. Nature 228, 1188-1190.

PEARSON MN & RAFFEL S (1971). Macrophage-digested antigen as inducer of delayed hypersensitivity. J Exp Med 133, 494-505.

PETERS JH & SCHIMMELPFENG L (1979). Contact co-operation in T lympho­ cyte mitogenesis: autocatalytical system of mutual interactions between macrophages and lymphocytes. In: The molecular basis of immune cell function (ed JG Kaplan) pp 660-663. Elsevier, Amsterdam.

PETERS PM, SVEDERSKY LP, REFAAT SHALABY M & PALLADINO MA (1985). Interferon gamma: more than a MAF. Lymphokine Res 4^, 265-273.

PFEFFER LM, WANG E & TAMM I (1980). Interferon effects on microfilament organization, cellular fibronectin distribution and cell motility in human fibroblasts. J Cell Biol ^5, 9-17.

PFEFFER LM, WANG E & TAMM I (1980a). Interferon inhibits the redistribution of cell surface components. J Exp Med 152, 469-474.

PFEFFERKORN ER (1984). Interferon gamma blocks the growth of Toxoplasma gondi i in human fibroblasts by inducing the host cells to degrade tryptophan. Proc Natl Acad Sci USA 81, 908-912. 274

PHILIPS NC, MORAS ML, CHEDID L, LEFRANCIER P & BERNARD JM (1985). Activation of alveolar macrophage tumour!cidal activity and eradication of experimental metastases by freeze-dried liposomes containing a new lipophilic muramyl dipeptide derivative. Cancer res 4j5, 128-134.

PIERCE CH, DUBOS RJ & SCHAEFFER WB (1953). Multiplication and survival of tubercle bacilli in the organs of mice. J Exp Med 97_, 189-205.

POBER JS, COLLINS T, GIMBRONE MA, COTRAN RS, GITLIN JD, FIERS W, CLAYBERGER C, KRENSKY AM, BURAKOFF SJ & REISS CS (1983). Lymphocytes recognise human vascular endothelial and dermal fibroblast la antigens induced by recombinant immune interferon. Nature 305, 726-729.

POSTE G (1980). The interaction of lipid vesicles (liposomes) with cultured cells and their use as carriers for drugs and macromolecules. In: Liposomes in biological systems (eds G Gregoriadis & AC Allison) pp 101-151. Wiley, New York.

POSTE G, BUCANA C, RAZ A, BUGELSKI P, KIRSH R & FIDLER IJ (1982). Analysis of the fate of systemically administered liposomes and implications for their use in drug delivery. Cancer Res 4_2, 1412-1422.

POSTE G, KIRSH R, FOGLER WE & FIDLER IJ (1979). Activation of tumouri- cidal properties in mouse macrophages by lymphokines encapsulated in liposomes. Cancer res _39^ 881-892.

POSTE G & PAPAHADJOPOULOS D (1976). Lipid vesicles as carriers for introducing materials into cultured cells: influence of vesicle lipid composition on mechanism(s) of vesicle incorporation into cells. Proc Natl Acad Sci USA 73, 1603-1607.

RABSON AR, ANDERSON R & GLOVER A (1978). Defective neutrophil movement and recurrent infection. In vitro and in vivo effects of levaraisole. Clin Exp Immunol 35, 142-149.

RAGER-ZISMAN B & BLOOM BR (1985). Interferons and natural killer cells. Br Med Bull 41, 22-27. 275

RAHMAN YE, ROSENTHAL MW & CERNY EA (1973). Intracellular plutonium: removal by liposome encapsulated chelating agent. Science 180, 300-302.

REED SG, BARRAL-NETTO M & INVERSO JA (1984). Treatment of experimental visceral leishmaniasis with lymphokine encapsulated in liposomes. J Immunol 132, 3116-3119.

REES RJW & HART P'D (1960). Analysis of the host-parasite equilibrium in chronic murine tuberculosis by total and viable bacillary counts. Br J Exp Pathol 42_, 83-88.

REGGIARD0 Z & MIDDLEBR00K G (1974). Failure of passive serum transfer of immunity against aerogenic tuberculosis in rabbits. Proc Soc Exp Biol Med 145, 173-175.

REHBERG E, KELDER B, H0AL EG & PESTKA S (1982). Specific molecular activities of recombinant and hybrid leukocyte interferons. J Biol Chem 257, 11497-11502.

RENOUX G (1978). Modulation of immunity by levamisole. Pharmac Ther 2_t 397-423.

RENOUX G & RENOUX M (1971). Effet immunostimulant d'un imidothiazole dans 1 ’ immunization des souris contre 1'infection par Brucella abortus. C R Acad Sci (D) Paris 272, 349-350.

RENOUX G & RENOUX M (1974). Modulation of immune reactivity by phenyl- imidothiazole salts in mice immunized by sheep red blood cells. J Immunol 113, 779-790.

RENOUX G & RENOUX M (1977). Thymus-like activities of sulphur deriva­ tives on T cell differentiation. J Exp Med 145, 466-471.

RENOUX G & RENOUX M (1977a). Roles of the imidazole or thiol moiety on the immunostimulant action of levamisole. In: Control of neoplasia by modulation of the immune system (ed MA Chirigos) pp 67-80. Raven Press, New York. 276

RENOUX G, RENOUX M & GUILLAUMIN JM (1979). Genetic and epigenetic control of levamlsole-induced immunostimulation. Int J Immunopharmacol l, 43-48.

RIBI E, ANACKER RL, BARCLAY WR, BREHMER W, HARRIS SC, LEIF WR & SIMMONS J (1971). Efficacy of mycobacterial cell walls as a vaccine against air borne tuberculosis in the rhesus monkey. J Infect Dis ml23, 527-538.

RIBI E, LARSON C, WICHT W, LIST R & GOODE G (1966). Effective non­ living vaccine against experimental tuberculosis in mice. J Bacteriol 91, 975-983.

RICH AR (1951). The pathogenesis of tuberculosis. Charles C Thomas, Springfield.

RICHMOND JY & HAMILTON LD (1969). Foot and mouth disease virus inhibi­ tion induced in mice by synthetic double-stranded RNA (Polyriboinosinic and polyribocytidylic acids). Proc Natl Acad Sci USA jj4_, 81-86.

ROBINSON A, MEYER M & MIDDLEBROOK G (1955). Tuberculin hypersensitivity in tuberculous infants treated with isoniazid. N Eng J Med 252, 983- 985.

ROBITZEK EH, SELIKOFF IJ & ORNSTEIN GG (1952). Chemotherapy of human tuberculosis with hydrazine derivatives of isonicotinic acid. Quart Bull Sea View Hosp J3, 27-51.

ROSENTHAL M, TRABERT U & MUELLER W (1976). The effect of levamisole on peripheral blood lymphocyte subpopulations in patients with rheumatoid arthritis and ankylosing spondylitis. Clin Exp Immunol 15_, 493-496.

ROSSI R, DRI P, BELLAVITE P, ZABUCCHI G & BERTON G (1979). Oxidative metabolism of inflammatory cells. Tn: Advances in inflammation research Vol 1 (ed G Weissmann) pp 139-155. Raven Press, New York.

ROTTA J, RYE M, MASEK K & ZAORAL M (1979). Biological activity of synthetic subunits of streptococcus peptidoglycan. I. Pyrogenic and thrombocytolytic activity. Exp Cell Biol 47, 258-268. 277

ROTHERMEL CD, RUBIN BY & MURRAY HW (1983). Gamma interferon is the factor in lymphokine that activates human macrophages to inhibit intracellular Chlamydia psittaci replication. J Immunol 131, 2542-2544.

ROUS P (1925). The relative reaction within living mammalian tissues. I. General features of vital staining with litmus. J Exp Med 4_1, 379- 397.

ROUS P (1925a). The relative reaction within living mammalian tissues. II. On the mobilization of acid material within cells, and the reaction as influenced by the cell state. J EXp Med 4J^, 399-411.

RUBEN FL, WINKELSTEIN A & FOTIADIS IG (1974). Immunological responsive­ ness of tuberculosis patients receiving rifampin. Antimicrob Agents Chemother _5, 383-387.

RUBINSTEIN A, MELAMED J & RODESCU D (1977). Transfer factor treatment in a patient with progressive tuberculosis. Clin Immunol Immunopathol 8_, 39-50.

RUBINSTEIN M, RUBINSTEIN S, FAMILLETTI PC, GROSS MS, MILLER RS, WALDMAN AA & PESTKA S (197 8). Human leukocyte interferon purified to homoge­ neity. Science 202, 1289-1290.

SAGAR AD, SEHGAL PB, SLATE L & RUDDLE FH (1982). Multiple human beta interferon genes. Proc Natl Acad Sci USA 7_6, 640-644.

SAMPSON D & LUI A (1976). The effect of levamisole on cell-mediated immunity and suppressor cell function. Cancer Res ^36, 952-955.

SCHATZ A & WAKSMAN SA (1944). Effect of streptomycin and other antibio­ tic substances upon Mycobacterium tuberculosis and other related orga­ nisms. Proc Soc Exp Biol Med 57_> 244-248.

SCHLOSSMAN SF, LEVIN HA, ROCKLIN RE & DAVID JR (1971). The compart­ mentalize t ion of antigen-reactive lymphocytes in desensitized guinea- pigs. J EXp Med L34, 741-750. 278

SCHMIDT ME & DOUGLAS SD (1976). Effects of levamisole on human monocyte function and immunoprotein receptors. Clin Immunol Immunopathol 6^, 279- 285.

SCHREIBER AD, PARSONS J & COOPER RA (1975). Effect of levamisole on the human monocyte IgG receptor (abstr.). Blood 4j>, 1018.

SCHREIBER RD, HICKS LJ, CELADA A, BUCHMEIER NA & GRAY PW (1985). Mono­ clonal antibodies to murine gamma interferon which differentially modu­ late macrophage activation and antiviral activity. J Immunol 134, 1609- 1618.

SCHREIBER R D , PACE JL, RUSSELL SW, ALTMAN A & KATZ DH (1983). Macrop­ hage activating factor produced by a T cell hybridoma: physiochemical and biosynthetic resemblance to gamma-interferon. J Immunol 131, 826- 832.

SCHULTZ RM & CHIRIGOS MA (1978). Similarities among factors that render macrophages tumouricidal in lymphokine and interferon preparations. Cancer Res 38^t 1003-1007.

SCHULTZ RM & KLEINSCHMIDT WJ (1983). Functional identity between murine gamma interferon and macrophage activating factor. Nature 305, 239-240.

SCOTT MT (1974). Corynebacterium parvum as an immunotherapeutic anti­ cancer agent. Semin Onco _1» 367-378.

SCOTT MT (1979). Analysis of the principles underlying chemoimmuno- therapy of mouse tumours. I. Treatment with cyclophosphamide followed by Corynebacterium parvum. Cancer Immunol Immunother 6^, 107-112.

SECHER DS & BURKE DC (1980). A monoclonal antibody for large scale purification of human leukocyte interferon. Nature 285, 446-450.

SECOND EAST AFRICAN/BRITISH MEDICAL RESEARCH COUNCIL KENYA TUBERCULOSIS SURVEY FOLLOW-UP (1979). Tuberculosis in Kenya: Follow-up of the second (1974) national sampling survey and a comparison with the follow-up data from the first (1964) national sampling survey. Tubercle ^0, 125-149. 279

SEGAL AW, GEISOW M, GARCIA R, HARPER A & MILLER R (1981). The respiratory burst of phagocytic cells is associated with a rise in vacuolar pH. Nature 290, 406-409.

SEVER JL & YOUMANS GP (1957). The relationship of oxygen-tension to virulence of tubercle bacilli and to acquired resistance in tuberculosis. J Infect Dis 101, 193-202.

SHARP K (1973). M. microti: a method for viable counts within 21 days of culture. Appl Microbiol Z5, 1023-1024.

SHAW IH, KNIGHT CG & DINGLE JT (1976). Liposomal retention of a modi­ fied anti-inflammatory steroid. Biochem J 158, 473-476.

SHEPARD CC, VAN LANDINGHAM R & WALKER LL (1977). Effect of levamisole on M. leprae in mice. Infect Immun Jl5, 564-567.

SHER NA, CHAPARAS SD, GREENBERG LD & BERNARD S (1975). Effects of BCG, Corynebacterium parvum and methanol-extraction residue in the reduction of mortality from Staphylococcus aureus and Candida albicans infections in immunosuppressed mice. Infect Immun JL2^, 1325-1330.

SHII0 T & YUGARI Y (1980). Antitumour effect of lentinan on syngeneic and autologous tumour-host systems, and suppression on chemical carcino­ genesis. (Abstr.) Int J Immunopharmacol 2_, 172.

SHREFFLER DC & DAVID CS (1976). The H-2 Major Histocompatibility Complex and the I Immune response region: Genetic variation, function and organization. Adv Immunol J20, 125-195.

SIDMAN CL, MARSHALL JD, SCHULTZ LD, GRAY PW & JOHNSON HM (1984). Gamma interferon is one of several direct B-cell maturing lymphokines. Nature 309, 801-804.

SILVERMAN DJ & WISEMAN CL (1979). In vitro studies of rickettsia-host cell interactions: Ultrastructural changes induced by Rickettsia ricke- 11sii infection in chicken embryo fibroblasts. Infect Immun 26, 714- 727. 280

SIMON HB & SHEAGREN JN (1971). Cellular immunity In vitro. I. Immunol ogi cal ly mediated enhancement of macrophage bactericidal activity. J Exp Med 133, 1377-1389.

SINGAPORE TUBERCULOSIS SERVICE/BRITISH MEDICAL RESEARCH COUNCIL (1981). Clinical trial of six-month and four-month regimens of chemotherapy in the treatment of pulmonary tuberculosis: The results up to 30 months. Tubercle 62, 95-102.

SINGH AN, MISHRA V, SINGH P & SHASHI R (1983). Immunostimulation of anti tubercular chemotherapy with levamisole in active pulmonary tuber­ culosis. J Assoc Phys India 3J, 405-407.

SINGH MM, JAIN AK, KUMAR R & MALAVIYA AN (1983a). Immunostimulatory effect of levamisole in miliary tuberculosis. Indian J Chest Dis Allied Sci 25, 91-95.

SINGH MM, KUMAR P, MALAVIYA AN & KUMAR R (1981). Levamisole as an adjunct in the treatment of pulmonary tuberculosis. Am Rev Respir Dis 123, 277-280.

SKAMENE E & KONGSHAVN PAL (1979). Phenotypic expression of genetically controlled resistance to Listeria monocytogenes. Infect Immun 2_5, 345- 351.

SMITH RL, HUNT NH, MERRITT J E , EVANS T & WEIDEMANN MJ (1980). Cyclic nucleotide metabolism and reactive oxygen production by macrophages. Biochem Biophys Res Commun 96, 1079-1087.

SONNENFELD G, MANDEL AD & MERIGAN TC (1978). Time and dosage dependence of immunoenhancement by murine Type II interferon preparations. Cell Immunol 40, 285-293.

SONNENFELD G, MERUEL0 D, MCDEVITT HO & MERIGAN T (1981). Effect of Type I and Type II interferons on murine thymocyte surface antigen expression: induction or selection. Cell Immunol _57, 427-439.

SPECT0R WG (1976). Epitheloid cells, giant cells and sarcoidosis. Ann NY Acad Sci 278, 3-6. 281

SPRICK MG (1956). Phagocytosis of Mycobacterium tuberculosis and Mycobacterium smegmatis stained with indicator dyes. Am Rev Respir Dis 74, 552-565.

STACH JL, GROS P, FORGET A & SKAMENE E (1984). Phenotypic expression of genetically-controlled natural resistance to Mycobacterium bovis BCG. J Immunol 132, 888-892.

STEIN SC & ARONSON JD (1953). The occurrence of pulmonary lesions in BCG-vaccinated and unvaccinated persons. Am Rev Respir Dis j>8, 695-712.

STEINMAN RM & NUSSENZWEIG MC (1980). Dendritic cells: Features and functions. Immunol Rev 53, 127-147.

STEWART CC, VALERIOTE FA & PEREZ CA (1978). Preliminary observations on the effect of glucan in combination with radiation and chemotherapy in four murine tumours. Cancer Treat Rep 62^ 1867-1872.

STINGL G, TAMAKI K & KATZ SI (1980). Origin and function of epidermal Langerhans cells. Immunol Rev S3, 149-174.

STOSSEL TP (1974). Phagocytosis. N Eng J Med 290, 774-780.

STOTT H, PATEL A, SUTHERLAND I, THORUP I, SMITH PW, KENT PW & RYKUSHIN YP (1973). The risk of tuberculous infection in Uganda, derived from the findings of national tuberculin surveys in 1958 and 1970. Tubercle j>4, 1-22.

STREULI M, HALL A, BOLL W, STEWART WE, NAGATA S & WEISSMAN C (1981). Target cell specificity of 2 species of human interferon-alpha produced in Escherichia coli and the hybrid molecules derived from them. Proc Natl Acad Sci USA 7j8, 2848-2852.

STYBL0 K (1980). Recent advances in epidemiological research in tuber­ culosis. Adv Tuberc Res J20, 1-63.

STYBL0 K & MEIJER J (1976). Impact of BCG vaccination programmes in children and young adults on the tuberculosis problem. Tubercle 57_, 17- 43. 282

STYBLO K, MEIJER J & SUTHERLAND I (1969). The transmission of tubercle bacilli. Its trend In a human population. Bull Int Union Tuberc 42^ 5- 104.

SUBBAIAH TV, MITCHISON DA & SELKON JB (1960). The susceptibility to hydrogen peroxide of Indian and British i soniazi d-sensi ti ve and isoniazid-resistant tubercle bacilli. Tubercle 4^1, 323-333.

SULA L (1958). Five years experience with a vaccine prepared from Mycobacterium tuberculosis var muris. Tubercle 39^, 10-17.

SVEDERSKY LP, BENTON CV, BERGER WH, RINDERKNECHT E, HARKINS RN & PALLA- DINO MA (1984). Biological and antigenic similarities of murine inter­ feron gamma and macrophage activating factor. J Exp Med 159, 812-827.

SYMOENS J & ROSENTHAL M (1977). A Review: Levamisole in the modulation of the immune response: The current experimental and clinical state. J Reticuloendo Soc 21^ 176-187.

SYMOENS J, ROSENTHAL M, DE BRABANDER M & GOLDSTEIN G (1979). Immunore- gulation with levamisole. Springer Semin Immunopathol 2, 49-68.

SZOKA FC & PAPAHADJOPOULOS D (1978). A new procedure for preparation of liposomes with large internal aqueous space and high capture, by reverse phase evaporation (REV). Proc Natl Acad Sci USA 7_5, 4194-4198.

TAYLOR RL, WILLIAMS DM, CRAVEN PC, GRAYBILL JR, DRUTZ DJ & MAGEE WE (1982). Amphotericin B in liposomes: a novel therapy for histoplas­ mosis. Am Rev Respir Dis 125, 610-611.

TEMPEL CW, HUGHES F J , MARDIS RE, TOWBIN MN & DYE WE (1951). Combined intermittent regimens employing streptomycin and para-aminosalicylic acid in the treatment of pulmonary tuberculosis. Am Rev Tuberc Pulm Dis 63, 295-311.

THIENPONT D, VANPARYS D F J , RAEYMAEKERS AHM, VANDENBERK J, DEMOEN PJA, ALLEWIJN FTN, MARSB00M RPH, NIEMEGEERS CJE, SCHELLENKENS KHL & JANSSEN PAJ (1966). Tetramisole (R8299)j a new, potent broad spectrum antihel- mintic. Nature 209, 1084-1086. 283

THORPE PE, EDWARDS DC, DAVIES AJS & ROSS WCJ (1982). Monoclonal anti­ body-toxin conjugates: aiming the magic bullet. In: Monoclonal antibo­ dies in clinical medicine (eds AJ Me Michael & JW Fabre) pp 167-201. Academic Press, London.

TITUS RG, KELSO A & LOUIS JA (1984). Intracellular destruction of Leishmania tropica by macrophages activated with macrophage activating factor/interferon. Clin Exp Immunol 5_5, 157-165.

TRAININ N (1974). Thymic hormones and the immune response. Physiol Rev 54, 272-315.

TRIPODI D, PARKS LC & BRUGMANS J (1973). Drug-induced restorationm of cutaneous delayed hypersensitivity in anergic patients with cancer. N Eng J Med 289, 354-357.

TUBERCULOSIS CHEMOTHERAPY CENTRE, MADRAS (1964). A concurrent compari­ son of intermittent (twice-weekly) isoniazid plus streptomycin and daily isoniazid plus para-aminosalicyclic acid in the domiciliary treatment of pulmonary tuberculosis. Bull WHO 31, 247-271.

TUBERCULOSIS RESEARCH CENTRE, MADRAS (1983). Study of chemotherapy regimens of 5 and 7 months duration and the role of corticosteroids in the treatment of sputum-positive patients with pulmonary tuberculosis in South India. Tubercle ^4, 73-91.

TURCO J, THOMPSON HA & WINKLER (1984). Interferon-gamma inhibits growth of Coxiella burnetii in mouse fibroblasts. Infect Immun 4j>, 781-783.

TURCO J & WINKLER HH (1983). Cloned mouse interferon-gamma inhibits the growth of Rickettsia prowazeki in cultured mouse fibroblasts. J Exp Med 158, 2159-2164.

TYRELL DA, RYMAN BE, KEETON BR & DUBOWITZ V (1976). Use of liposomes in treating Type II glycogenosis. Br Med J 1_, 88.

UNITED STATES PUBLIC HEALTH SERVICE TUBERCULOSIS THERAPY TRIALS (1960). Preliminary observations from a controlled trial of prednisolone in the treatment of pulmonary tuberculosis. Am Rev Respir Dis 91, 598-600. 284

USUDA Y, I MAI K, TAKAHASHI S, ABE S, MIYAKOSHI H, WATANBE K & AOKI T (1981). Drug-resistant pulmonary tuberculosis treated with lentinan. I n : Manipulation of host defence mechanisms (eds T Aoki, I Urushizaki & E Tsubura) pp 50-64. Excerpta Medica, Amsterdam.

VAN GEUNS HA, MEIJER J & STYBLO K (1975). The yield from mass tubercu­ lin testing of unvaccinated children and adolescents. Bull Int Union Tuberc j^, 82-89.

VAN GINCKEL R & DE BRADANDER M (1979). The influence of a levamisole metabolite (DL-2-oxo-3- [2-rae reap to-ethyl ]-5-phenylimidazolidine) on carbon clearance in mice. J Reticuloendo Soc Z5, 125-131.

VAN NUETEN JM (1972). Pharmacological aspects of tetramisole. In; Comparative biochemistry of parasites (ed H Van Den Bossche) pp 101-115. Academic Press, New York.

VAN WAUWE J & G00SENS J (1979). Stimulation of mouse thymocytes by concanavalin A: effect of levamisole and antioxidants. Ann Immunol (Paris) 130C, 756.

VARESIO L, BLASI E, THURMAN GB, TALMADGE JE, WILTROUT RA & HERBERMAN RB (1984). Potent activation of mouse macrophages by recombinant interfe­ ron-gamma. Cancer Res 4^4, 4465-4469.

VIRELIZIER JL, ALLISON AC & DE MAEYER E (1977). Production by mixed lymphocyte cultures of a Type II interferon able to protect macrophages against virus infections. Infect Immun 17, 282-285.

VIRELIZIER JL, PEREZ N, ARENZANA-SEISDEDOS R & DEVOS R (1984). Pure interferon gamma enhances Class II HLA antigens on human monocyte lines. Eur J Immunol J_4, 106-108.

VLADIMIRSKY MA & LADIGINA GA (1982). Antibacterial activity of liposome entrapped streptomycin in mice infected with Mycobacterium tuberculosis. Biomedicine 36, 375-377.

WAKSMAN BH (1979). Overview: biology of the lymphokines. In_: Biology of the lymphokines (eds E Pick, S Cohen & JJ Oppenheim) pp 585-616 . Academic Press, New York. 285

WALDMAN RH & GANGULY R (1977). ISO in rhino virus infections. Thera­ peutic efficacy of inosiplex (isoprinosine) in rhino virus infection. Ann NY Acad Sci 284, 153-160.

WALKER EB, MAINO V, SANCHEZ-LANIER M, WARNER N & STEWART C (1984). Murine gamma interferon activates the release of a macrophage-derived la inducing factor that transfers la inductive capacity. J Exp Med 159, 1532-1547.

WALKER L & LOWRIE DB (1981). Killing of Mycobacterium microti by immunologically activated macrophages. Nature 293, 69-70.

WALLACH D, FELLOUS M & REVEL M (1982). Preferential effect of gamma interferon on the synthesis of HLA antigens and their mRNAs in human cells. Nature 299, 833-836.

WARA DW, GOLDSTEIN AL, DOYLE N & AMMAN AJ (1975). Thymosin activity in patients with cellular immunodeficiency. N Eng J Med 292, 70-74.

WATSON SR & COLLINS FM (1980). Development of suppressor T cells in mice heavily infected with mycobacteria. Immunology 39, 367-373.

WECK PK, APPERSON S, MAY L & STEBBING N (1981). Comparison of the antiviral activities of various cloned human interferon-alpha subtypes in mammalian cell cultures. J Gen Virol jT7, 233-237.

WECK PK, APPERSON S, STEBBING N, GRAY PW, LEUNG D, SHEPARD HM & GOEDDEL DV (1981). Antiviral activities of hybrids of two major human leukocyte interferons. Nucleic Acids Res j), 6153-9156.

WEIDNER E (1975). Interactions between Encephalitozoon cuniculi and macrophages. Parasitophorous vacoule growth and the absence of lysosomal fusion. Zeitschrift fur Parasitenkunde 47, 1-9.

WEIEL JE, ADAMS DO & HAMILTON TA (1985). Biochemical models of gamma- interferon action: altered expression of transferrin receptors on murine peritoneal macrophages after treatment in vitro with PMA or A23187. J Immunol 134, 293-298. 286

WEIL J, EPSTEIN CJ, EPSTEIN LB, SEDMAK J J , SABRAM JL & GROSSBERG SE (1983). A unique set of polypeptides is induced by gamma-interferon in addition to those induced in common with alpha and beta interferons. Nature 301, 437-439.

WEINSTEIN LW & DALTON AC (1968). Host determinants of response to anti­ microbial agents. N Eng J Med 279, 580-588.

WEISBERGER AS, DANIEL TM & HOFFMAN A (1964). Suppression of antibody synthesis and prolongation of homograft survival by chloramphenicol. J Exp Med 120, 183-196.

WEISBERGER AS, MOORE RD & SCHOENBERG MD (1966). Modification of experi­ mental immune nephritis by chloramphenicol. J Lab Clin Med 67_, 58-69.

WEISSMAN G, BLOOMGARDEN D, KAPLAN R, COHEN C, HOFFSTEIN S, COLLINS T, GOTTLEIB A & NAGLE D (1975). A general method for the introduction of enzymes by means of immunoglobulin-coated liposomes into lysosomes of deficient cells. Proc Natl Acad Sci USA 72, 88-92.

WELLS AQ (1946). The murine type of tubercle bacillus (the vole acid- fast bacillus). Br Med Res Counc Spec Rep Ser No 259.

WELLS AQ (1957). The vole bacillus vaccine. Adv Tuberc Res 8^, 61-78.

WHITCOMB ME & ROCKLIN RE (1973). Transfer factor therapy in a patient with progressive primary tuberculosis. Ann Intern med _79> 161-166.

WILLIAMS DL, CORK JA, HOFFMANN EO & DILUZIO NR (1978). Protective effect of glucan in experimentally induced Candidiasis. J Reticulendo Soc 23, 479-490.

WILSON GB & FUDENBERG HH (1983). Is controversy about ’transfer factor therapy* nearing an end? Immunol Today 4_, 157-161.

WILSON GB, FUDENBERG HH, JOHNSON HT & SMITH CL (1980). Effects of dialysable leukocyte extracts with transfer factor activity on leukocyte migration in vitro. IV. Two distinct effects of DLE on leukocyte migra­ tion can be produced by prostaglandin. Clin Immunol Immunopathol 16, 90-102. 287

WIRTH JJ, KIERSZENBAUM F, SONNENFELD G & ZLOTNIK A (1985). Enhancing effects of gamma interferon on phagocytic cell association with and killing of Trypanosoma cruzi. Infect Immun 49^ 61-66.

WISEMAN CL & WADDELL A (1983). Interferonlike factors from antigen and mitogen stimulated human leukocytes with antirickettsial and cytolytic actions on Rickettsia prowazeki. J Exp Med 157, 1780-1793.

WONG GHW, CLARK-LEWIS I, MCKIMM-BRESCHKIN JL, HARRIS AW & SCHRADER JW (1983). Interferon gamma induces enhanced expression of la and H-2 antigens on B lymphoid, macrophage and myeloid cell lines. J Immunol 131, 788-793.

WORLD HEALTH ORGANIZATION (1980). Vaccination against tuberculosis. WHO/Technical Report Series No. 651.

WORLD HEALTH ORGANIZATION (1980a). BCG Vaccination policies. WHO/Tech­ nical Report Series No. 652.

WORLD HEALTH ORGANIZATION (1982). Tuberculosis control. WHO/Technical Report Series No. 671.

WYBRAN J, GOVAERTS A & APPELB00M T (1978). Inosiplex, a stimulating agent for normal human T cells and human leukocytes. J Immunol 121, 1184-1189.

YASEEN NY, THEWAINI A J , AL-TAWIL NG & JAZRAWI FY (1980). Trial of immunopotentiation by levamisole in patients with pulmonary tubercu­ losis. J Infect ^2, 125-136.

YIP YK, BARROWCLOUGH B, URBAN C & VILCHEK J (1982). The molecular weight of human gamma interferon is similar to that of other human interferons. Science 215, 411-413.

YOUMANS AS & YOUMANS GP (1969). Factors affecting immunogenic activities of mycobacterial ribosomal and ribonucleic acid preparations. J Bacteriol 99, 42-50. 288

YOUMANS GP (1975). Relation between delayed hypersensitivity and immunity in tuberculosis. Am Rev Respir Dis 111, 109-118.

YOUMANS GP (1979). Pathogenesis of tuberculosis. In: Tuberculosis pp 317-326. WB Saunders Company, London.

YOUMANS GP & YOUMANS AS (1951). The relation between the size of the infecting dose of tubercle bacilli and the survival time of mice. Am Rev Tuberc J)4_, 534-540.

YOUNG RA, BLOOM BR, GROSSKINSKY CM, IYANYI J, THOMAS D & DAVIS RW (1985). Dissection of Mycobacterium tuberculosis antigens using recombinant DNA. Proc Natl Acad Sci USA jS2^ 2583-2587.

YOUNG SP, BAKER E & HUEHNS ER (1979). Liposome entrapped desferriox- amine and iron transporting ionophores: a new approach to iron chelation therapy. Br J Pharm 41, 357-363.

ZBAR B, BERNSTEIN ID & RAPP HJ (1971). Suppression of tumour growth at the site of infection with living Bacillus Calmette-Guerin. J Natl Cancer Inst 4b, 831-839.

ZLOTNIK A, SHIMONKEVITZ R P , GEFTER M C , KAPPLER F & MARRACK P (1983). Characterization of the gamma interferon mediated induction of antigen presenting ability on P388D1 cells. J Immunol 131, 2814-2820.

ZUCKER-FRANKLIN D & HIRSCH JG (1964). Electron microscope studies on the degranulation of rabbit peritoneal leukocytes during phagocytosis. J Exp Med 120, 569-575. 289

APPENDIX 1

TABLE 3A.1 EFFECT OF LEVAMISOLE ON THE GROWTH OF M. MICROTI IN LUNGS OF CFLP MICE

Days Log viable mycobacteria per lung after after treatment with levamisole (mg/kg)* infection 0 2.5 25

3 3.904 + 0.08 7 3.886 + 0.21 15 4.667 + 0.10 4.668 + 0.14 4.316 + 0.09 22 5.376 + 0.13 5.482 + 0.13 5.220 + 0.09 29 5.993 + 0.25 5.842 + 0.11 6.342 + 0.26 36 6.822 + 0.08 6.620 + 0.15 6.604 + 0.23 43 7.031 + 0.09 7.141 + 0.06 7.466 + 0.12 * Mean + s.e. of 4 mice

TABLE 3A.2 EFFECT OF LEVAMISOLE ON THE GROWTH OF M. MICROTI IN SPLEENS OF CFLP MICE

Days Log viable mycobacteria per spleen after after treatment with levamisole (mg/kg)* infection 0 2.5 25

3 5.447 + 0.17 7 5.267 + 0.08 15 5.606 + 0.13 5.610 + 0.08 5.607 + 0.16 22 6.004 +0.10 6.107 + 0.12 5.867 + 0.06 29 6.348 + 0.06 6.132 + 0.13 6.484 + 0.11 36 6.095 + 0.20 5.936 + 0.22 6.102 + 0.14 43 5.885 + 0.16 5.906 + 0.19 6.106 + 0.22

Mean + s.e. of 4 mice 290

TABLE 4A.1 EFFECT OF SIZE OF INFECTIVE DOSE ON GROWTH OF M. TUBERCULOSIS IN MICE

Infective dose Days after viable mycobacteria/organ Lo8io. (orgs/mouse) infection Lung Spleen

5 x 106 1 5.323 + 0.04 6.316 + 0.08 8 6.188 + 0.05 6.924 + 0.04 15 8.023 + 0.04 7.138 + 0.04 22 9.155 + 0.27 6.962 + 0.24

5 x 105 1 3.926 + 0.09 5.247 + 0.05 8 5.074 + 0.10 6.303 + 0.15 15 6.814 + 0.09 6.376 + 0.15 22 7.874 + 0.26 6.460 + 0.09 29 7.847 + 0.08 5.986 + 0.24 4 5 x 10 1 3.074 + 0.07 4.132 + 0.12 8 3.873 + 0.09 5.484 + 0.05 15 5.954 + 0.08 6.228 + 0.09 22 6.564 + 0.13 5.793 + 0.10 29 6.698 + 0.05 5.438 + 0.16 43 6.906 -f 0.39 5.138 + 0.21

* Mean + s.e. of 4 mice

TABLE 4A.2 EFFECT OF SIZE OF DOSE OF INTERFERON-GAMMA ON THE GROWTH OF M. TUBERCULOSIS IN LUNGS OF INFECTED MICE

Log viable mycobacteria per lung* after treatment Days after witn interferon-gamma (units /mouse) infection 0 200 1000 5000

0 5.679 + 0.04 3 4.470 + 0.04 4.323 + 0.12 4.216 + 0.24 4.087 + 0.17 5 4.232 + 0.05 4.456 + 0.09 4.166 + 0.21 4.104 + 0.21 9 5.116 + 0.12 4.806 + 0.14 4.696 + 0.08 4.729 + 0.13

* Mean + s.e. of 5 mice 291

TABLE 4A.3 EFFECT OF DOSE SIZE OF INTERFERON-GAMMA ON THE GROWTH OF M. TUBERCULOSIS IN SPLEENS OF INFECTED MICE

Log._ viable mycobacteria per spleen after treatment Days after witn interferon-gamma (units/mouse) infection 0 200 1000 5000

0 5.154 + 0.05 3 5.552+0.04 5.514 + 0.04 5.367 + 0.05 5.377 + 0.03 5 5.887 + 0.04 5.914 + 0.04 5.776 + 0.04 5.703 + 0.02 9 6.674 + 0.05 6.648 + 0.06 6.560 + 0.04 6.549 + 0.05

* Mean + s.e. of 5 mice

TABLE 4A.4 EFFECT OF ADMINISTRATION OF INTERFERON-GAMMA IN LIPOSOMES ON GROWTH OF M. TUBERCULOSIS IN LUNGS OF CFLP MICE

L o g ^ viable mycobacteria per lung* Days after infection Treatment group 0 (1 hour) 5 9

Control 4.974 + 0.15 5.605 + 0.13

1000 u 4.243 + 0.15 4.903 + 0.10

1000 u, liposomes 4.614 + 0.22 4.727 + 0.14 5.551 + 0.12 20.000 u 4.508 + 0.24 5.172 + 0.12

20.000 u, liposomes 4.885 + 0.26 5.442 + 0.12

* Mean + s.e. of 6 mice 292

TABLE 4A.5 EFFECT OF ADMINISTRATION OF INTERFERON-GAMMA IN LIPOSOMES ON GROWTH OF M. TUBERCULOSIS IN SPLEENS OF CFLP MICE

L°gio viable mycobacteria per spleen* Days after infection Treatment group 0 (1 hour) 5 9

Control 4.851 + 0.06 5.822 + 0.08 6.316 + 0.07

1000 u 5.286 + 0.06 5.931 + 0.06

1000 u, liposomes 5.377 + 0.08 5.730 + 0.04

20.000 u 5.397 + 0.13 5.910 + 0.07

20.000 u, liposomes 5.589 + 0.06 5.808 + 0.04

* Mean + s.e. of 6 mice

TABLE 4A.6 EFFECT OF ADMINISTRATION OF INTERFERON-GAMMA IN LIPOSOMES ON GROWTH OF M. TUBERCULOSIS IN LUNGS OF BALB/C MICE

viable mycobacteria per lung* ment group Days after PBS PBS 1000 u 1000 u infection liposomes liposomes

0 4.855 + 0.08

3 3.506 + 0.1 3.558 + 0.15 3.377 + 0.09 3.540 + 0.13

6 3.678 + 0.05 3.712 + 0.12 3.511 + 0.05 3.372 + 0.08

9 4.441 + 0.04 4.474 + 0.14 4.268 + 0.12 4.237 + 0.08

* Mean + s.e. of 4 mice TABLE 4A.7 EFFECT OF ADMINISTRATION OF INTERFERON-GAMMA IN LIPOSOMES ON GROWTH OF M. TUBERCULOSIS IN SPLEENS OF BALB/C MICE

viable mycobacteria per lung* Tr tliment group Days after PBS PBS 1000 u 1000 u infection liposomes liposomes

0 4.965 + 0.06 3 5.239 + 0.03 5.206 + 0.03 5.172 + 0.05 5.111 + 0.05 6 5.867 + 0.07 5.779 + 0.05 5.792 + 0.04 5.672 + 0.07 9 6.586 + 0.03 6.455 + 0.03 6.312 + 0.06 6.199 + 0.05

* Mean + s.e. of 4 mice

TABLE 4A.8 EFFECT OF PRETREATMENT WITH INTERFERON-GAMMA ON THE GROWTH OF M. TUBERCULOSIS IN THE LUNGS OF BALB/C MICE

Treatment Mean log viable mycobacteria per lung* on: group Day 0 (I hr) Day 1 Day 2

Control 5.020+0.02 3.783 + 0.04 3.512 + 0.04 2000 u, Day -3 3.836 + 0.02 3.661 + 0.06 2000 u, Day -2 3.718 + 0.03 3.416 + 0.04 2000 u, Day -1 3.768 + 0.07 3.530 + 0.09 2000 u x 2, Day -2,-1 3.728 + 0.04 3.365 + 0.07 2000 u x 3, Day -3,-2,-1 3.841 + 0.10 3.458 + 0.06

* Mean + s.e. of 4 mice 294

TABLE 4A.9 EFFECT OF PRETREATMENT WITH INTERFERON-GAMMA ON THE GROWTH OF M. TUBERCULOSIS IN THE SPLEENS OF BALB/C MICE

Treatment Mean log viable mycobacteria per spleen* on: group Day 0 hr) Day 1 Day 2

Control 4.780+0.04 4.966 + 0.04 5.108 + 0.02 2000 u, Day -3 4.923 + 0.03 4.980 + 0.03 2000 u, Day -2 4.845 + 0.05 4.933 + 0.03 2000 u, Day -1 4.876 + 0.05 4.823 + 0.04 2000 u x 2, Day -2,-1 4.831 + 0.05 4.853 + 0.04 2000 u x 3, Day -3,-2,-1 4.813 + 0.02 4.832 + 0.06

* Mean + s.e. of 4 mice

TABLE 5A.1 THE EFFECTS OF INTERFERON-GAMMA AND ISONIAZID ALONE AND IN COMBINATION ON THE GROWTH OF M. TUBERCULOSIS IN LUNGS OF BALB/C MICE

Mean log.^ viable mycobacteria per lung* after treatment with: Days after Untreated IFN-tf + infection controls IFN-V Isoniazid isoniazid

0 (1 hr) 4..858 + 0.09

1 3.983 + 0.12 3.865 + 0.2 2 3.584 + 0.13 3.263 + 0.10 3.656 + 0.11 3.379 + 0.16 3 3.419 + 0.15 3.273 + 0.06 3.318 + 0.09 3.226 + 0.02 4 3.562 + 0.10 3.364 + 0.13 3.065 + 0.06 2.854 + 0.05 6 3.673 + 0.09 3.634 + 0.06 2.854 + 0.09 2.624 + 0.10 9 4.651 + 0.12 4.308 + 0.10 2.496 + 0.06 2.344 + 0.05

* Mean + s.e. of 4 mice 295

TABLE 5A.2 THE EFFECT OF INTERFERON-GAMMA AND ISONIAZID ALONE AND IN COMBINATION ON THE GROWTH OF M. TUBERCULOSIS IN SPLEENS OF BALB/C MICE

Mean log^ viable mycobacteria per spleen* after treatment with: Days ------:------after Untreated IFN-Y + infection control IFN-Y Isoniazid isoniazid

0 (1 hr) 4.887 + 0.04

1 5.071 + 0.04 4.930 + 0.06 2 5.043 + 0.03 4.937 + 0.02 4.938 + 0.05 4.886 + 0.04 3 5.196 + 0.05 5.062 + 0.03 4.721 + 0.05 4.593 + 0.06 4 5.352 + 0.03 5.274+0.06 4.471 + 0.07 4.264 + 0.06 6 5.999 + 0.05 5.892 + 0.02 4.128 + 0.04 4.014 + 0.04 9 6.562 + 0.07 6.282 + 0.02 3.578 + 0.04 3.478 + 0.03

* Mean + s.e. of 4 mice

TABLE 5A.3 THE EFFECT OF INTERFERON-GAMMA AND ISONIAZID ALONE AND IN COMBINATION ON THE GROWTH OF M. TUBERCULOSIS IN LUNGS OF BALB/C MICE IN THE FIRST TWO DAYS AFTER INFECTION WHEN IFN-Y WAS GIVEN BEFORE AND AFTER INFECTION

Mean log viable mycobacteria per lung* after treatment with: Days after Untreated i fn-Y + infection control IFN-Y Isoniazid isoniazid

0 (1 hr) 4.416 + C1.10

1 3.860 + 0.06 3.651 + 0.02 3.979 + 0.16 3.691 + 0.06

2 3.944 + 0.21 3.678 + 0.10 3.737 + 0.25 3.340 + 0.18

* Mean + s.e. of 4 mice 296

TABLE 5A.4 THE EFFECT OF INTERFERON-GAMMA AND ISONIAZID ALONE AND IN COMBINATION ON THE GROWTH OF M. TUBERCULOSIS IN SPLEENS OF BALB/C MICE IN THE FIRST TWO DAYS AFTER INFECTION WHEN IFN-V WAS GIVEN BEFORE AND AFTER INFECTION

Mean log.^ viable mycobacteria per spleen* after treatment with: Days after Untreated IFN-Y + infection control IFN-X Isoniazid isoniazid

0 (1 hr) 4.171 + 0.06

1 5.013 + 0.03 4.824 + 0.03 4.813 + 0.09 4.770 + 0.05

2 5.094 + 0.02 4.900 + 0.01 4.708 + 0.03 4.744 + 0.06

* Mean + s.e. of 4 mice

TABLE 5A.5 THE EFFECTS OF INTERFERON-GAMMA AND ISONIAZID ALONE AND IN COMBINATION ON THE GROWTH OF M. TUBERCULOSIS IN LUNGS OF BALB/C MICE WHEN IFN-y WAS ADMINISTERED ON DAYS +5 AND +7.

Mean log^Q viable mycobacteria per lung* after treatment with: Days after Untreated ifn-Y + infection controls IFN-X Isoniazid isoniazid

0 (1 hr) 4.416 + 0.10

1 3.860 + 0.06 3.979 + 0.16 2 3.944 + 0.21 3.737 + 0.25 5 3.778 + 0.20 3.100 + 0.20 7 3.977 + 0.06 3.777 + 0.18 2.906 + 0.33 2.666 + 0.10 8 4.275 + 0.02 4.157 + 0.17 2.445 + 0.27 2.534 + 0.28 9 4.402 + 0.13 4.152 + 0.30 2.206 + 0.34 2.076 + 0.18

* Mean + s.e. of 4 mice 297

TABLE 5A.6 THE EFFECT OF INTERFERON-GAMMA AND ISONIAZID ALONE AND IN COMBINATION ON THE GROWTH OF M. TUBERCULOSIS IN SPLEENS OF BALB/C MICE WHEN IFN-tf WAS ADMINISTERED ON DAYS +5 AND +7

Mean log.^ viable mycobacteria per spleen* after treatment with: Days after Untreated IFN-Y + infection control iFN-y Isoniazid isoniazid

0 (1 hr) 4.171 + 0.06

1 5.013 + 0.03 4.813 + 0.09 2 5.094 + 0.02 4.708 + 0.03 5 5.692 + 0.03 4.123 + 0.03 7 6.016 + 0.03 6.014 + 0.05 3.864 + 0.04 3.855 + 0.02 8 6.221 + 0.07 6.129 + 0.03 3.717 + 0.03 3.670 + 0.04 9 6.480 + 0.04 6.328 + 0.05 3.610 + 0.07 3.639 + 0.05

* Mean + s.e. of 4 mice

TABLE 5A.7 THE EFFECT OF INTERFERON-GAMMA AND RIFAMPICIN ALONE AND IN COMBINATION ON GROWTH OF M. TUBERCULOSIS IN LUNGS OF BALB/C MICE

Mean log^ viable mycobacteria per lung* after treatment with: Days after Untreated IFN-V + i nfection controls IFN-Y Ri fampicin rifampicin

0 (1 hr) 4.707 + 0.06

1 3.898 + 0.04 3.769 + 0.05 3.548 + 0.1 3.563 + 0.09

2 3.617 + 0.03 3.487 +_0.1 3.141 + 0.13 3.085 + 0.11

3 3.447 + 0.07 3.245 + 0.04 2.654 + 0.04 2.585 + 0.09

* Mean + s.e. of 4 mice 298

TABLE 5A.8 THE EFFECT OF INTERFERON-GAMMA AND RIFAMPICIN ALONE AND IN COMBINATION ON GROWTH OF M. TUBERCULOSIS IN SPLEENS OF BALB/C MICE

Mean log^Q viable mycobacteria per spleen* after treatment with: Days after Untreated IFN-JT + infection controls IFN-# Rifampicin rifampicin

0 (1 hr) 4.792 + 0.005

1 5.877 + 0.04 4.930 + 0.05 4.760 + 0.02 4.751 + 0.03

2 5.060 + 0.05 4.913 + 0.02 4.491 + 0.02 4.473 + 0.03

3 5.242 + 0.06 5.029 + 0.06 4.218 + 0.08 4.147 + 0.01

* Mean + s.e. of 4 mice