You are on page 1of 258

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/227236827

Pathogenesis of Mycoplasma pneumoniae infections: Adaptive immunity,


innate immunity, cell biology, and virulence factors

Chapter · January 1970


DOI: 10.1007/978-3-7643-7563-8_9

CITATIONS READS

12 375

7 authors, including:

Jerry William Simecka Norbert Suttorp


University of North Texas HSC at Fort Worth Charité Universitätsmedizin Berlin
110 PUBLICATIONS   3,366 CITATIONS    786 PUBLICATIONS   16,030 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Antimicrobial therapy and Biofilms View project

PROGRESS View project

All content following this page was uploaded by Jerry William Simecka on 26 May 2014.

The user has requested enhancement of the downloaded file.


Birkhäuser Advances in Infectious Diseases

BAID

Series Editors

Axel Schmidt Manfred H. Wolff


University Witten/Herdecke University Witten/Herdecke
Faculty of Medicine Faculty of Biosciences
Alfred-Herrhausen-Str. 50 Stockumer Str. 10
58448 Witten 58448 Witten
Germany Germany

Stefan H.E. Kaufmann


Max-Planck-Institut für Infektionsbiologie
Department of Immunology
Schumannstrasse 21/22
10117 Berlin
Germany
Community-Acquired
Pneumonia

Edited by N. Suttorp, T. Welte and R. Marre

Birkhäuser Verlag
· ·
Basel Boston Berlin
Editors

Norbert Suttorp Reinhard Marre


Charité – University Medicine Berlin University of Ulm
Dpt. of Internal Medicine Medical Microbiology and Hygiene
Augustenburger Platz 1 Robert-Koch-Str. 8
D-13353 Berlin D-89081 Ulm
Germany Germany

Tobias Welte
Medizinische Hochschule Hannover
Carl-Neuberg-Str. 1
D-30625 Hannover
Germany

A CIP catalogue record for this book is available from the library of Congress,
Washington, DC, USA

Bibliographic information published by Die Deutsche Bibliothek


Die Deutsche Bibliothek lists this publication in the Deutsche Nationalbibliografie;
detailed bibliographic data is available in the internet at http://dnb.ddb.de

ISBN 3-7643-7562-0 Birkhäuser Verlag, Basel - Boston - Berlin

The publisher and editor can give no guarantee for the information on drug dosage and
administration contained in this publication. The respective user must check its accuracy by
consulting other sources of reference in each individual case.
The use of registered names, trademarks etc. in this publication, even if not identified as such,
does not imply that they are exempt from the relevant protective laws and regulations or free
for general use.
This work is subject to copyright. All rights are reserved, whether the whole or part of the
material is concerned, specifically the rights of translation, reprinting, re-use of illustrations,
recitation, broadcasting, reproduction on microfilms or in other ways, and storage in data
banks. For any kind of use, permission of the copyright owner must be obtained.

© 2007 Birkhäuser Verlag, P.O. Box 133, CH-4010 Basel, Switzerland


Part of Springer Science+Business Media
Printed on acid-free paper produced from chlorine-free pulp. TFC '
Cover illustration: Colored transmission electron micrograph of Mycoplasma pneumoniae
(green) demonstrating flask-shaped morphology. The background shows a chest x-ray from
a patient. With the friendly permission of Kristen L. Hoek, Vanderbilt University Medical
Center, Nashville, and Matthias Krüll.
Printed in Germany
ISBN-10: 3-7643-7562-0 e-ISBN-10: 3-7643-7563-9
ISBN-13: 978-3-7643-7562-1 e-ISBN-13: 978-3-7643-7563-8
987654321 www. birkhauser.ch
Contents

List of contributors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . vii

Preface ................................................................... ix

Tobias Welte
Diagnosis and treatment of community acquired pneumonia –
the German perspective . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1

Reinhard Marre
Detection of respiratory bacterial pathogens ........................... 15

Walter Hampl and Thomas Mertens


Viral pathogens and epidemiology, detection, therapy
and resistance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27

Mathias W.R. Pletz, Lesley McGee and Tobias Welte


Resistance in Streptococcus pneumoniae . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57

Hans-Dieter Klenk
Influenza . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 73

Matthias Krüll and Norbert Suttorp


Pathogenesis of Chlamydophila pneumoniae infections –
epidemiology, immunity, cell biology, virulence factors . . . . . . . . . . . . . . . . . 83

Dina M. Bitar, Marina Santic, Yousef Abu Kwaik


and Maëlle Molmeret
Legionnaires’ disease and its agent Legionella pneumophila . . . . . . . . . . . 111

Sven Hammerschmidt, Gavin K. Paterson, Simone Bergmann


and Timothy J. Mitchell
Pathogenesis of Streptococcus pneumoniae infections:
adaptive immunity, innate immunity, cell biology, virulence factors . . . . 139
vi Contents

Ken B. Waites, Jerry W. Simecka, Deborah F. Talkington


and T. Prescott Atkinson
Pathogenesis of Mycoplasma pneumoniae infections:
adaptive immunity, innate immunity, cell biology,
and virulence factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 183

Pablo D. Becker and Carlos A. Guzmán


Community-acquired pneumonia: paving the way towards
new vaccination concepts . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 201

Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 247
List of contributors

Yousef Abu Kwaik, Department of Microbiology and Immunology, Room


MS-410, University of Louisville, Louisville, KY 40209, USA
T. Prescott Atkinson, Department of Pediatrics, University of Alabama at
Birmingham, Birmingham, AL 35249, USA
Pablo D. Becker, Department of Vaccinology, Helmholtz Centre
for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig,
Germany; e-mail: pablo-daniel.becker@helmholtz-hzi.de
Simone Bergmann, Research Center for Infectious Diseases, University of
Würzburg, Röntgenring 11, D-97070 Würzburg, Germany
Dina M. Bitar, Department of Microbiology and Department of Medical
Microbiology and Immunology, Faculty of Medicine, Al-Quds University,
Jerusalem, 19356, Israel; e-mail: dbitar@med.alquds.edu
Carlos A. Guzmán, Department of Vaccinology, Helmholtz Cen-
tre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig,
Germany; e-mail: carlos.guzman@helmholtz-hzi.de
Sven Hammerschmidt, Research Center for Infectious Diseases, University
of Würzburg, Röntgenring 11, D-97070 Würzburg, Germany;
e-mail: s.hammerschmidt@mail.uni-wuerzburg.de
Walter Hampl, Institute for Virology, University Clinic of Ulm, Albert-Ein-
stein-Allee 11, 89081 Ulm, Germany;
e-mail: walter.hampl@uniklinik-ulm.de
Hans-Dieter Klenk, Institut für Virologie, Philipps-Universität Marburg,
Hans-Meerwein-Str. 3, 35043 Marburg, Germany;
e-mail: klenk@staff.uni-marburg.de
Matthias Krüll, Dept. Internal Medicine/Infectious Diseases and Pulmonary
Medicine, Charité, Universitätsmedizin Berlin, Augustenburgerplatz 1,
13353 Berlin, Germany; e-mail: Matthias.kruell@charite.de
Reinhard Marre, University of Ulm, Medical Microbiology & Hygiene, Rob-
ert-Koch-Str. 8, 89081 Ulm, Germany;
e-mail: reinhard.marre@uniklinik-ulm.de
Lesley McGee, Hubert Department of Global Health, Emory University,
1518 Clifton Road, Atlanta, GA, 30322, USA;
e-mail: lmcgee@sph.emory.edu
Thomas Mertens, Institute for Virology, University Clinic of Ulm, Albert-
Einstein-Allee 11, 89081 Ulm, Germany;
e-mail: thomas.mertens@uniklinik-ulm.de
viii List of contributors

Timothy J. Mitchell, Division of Infection and Immunity, Institute of Bio-


medical and Life Science, Joseph Black Building, University of Glasgow
G12–8QQ, UK
Maëlle Molmeret, Department of Microbiology and Immunology, Room
MS-410, University of Louisville, Louisville, KY 40209, USA
Gavin K. Paterson, Division of Infection and Immunity, Institute of Bio-
medical and Life Science, Joseph Black Building, University of Glasgow
G12–8QQ, UK
Mathias W.R. Pletz, Department of Respiratory Medicine, Hannover Medi-
cal School, Carl-Neuberg-Str.1, Hannover, 30625, Germany;
e-mail: pletz.mathias@mh-hannover.de
Marina Santic, Department of Microbiology and Parasitology, Medical Fac-
ulty, University of Rijeka, Croatia
Jerry W. Simecka, Department of Molecular Biology and Immunology, Uni-
versity of North Texas Health Science Center, Fort Worth, TX 76107,
USA
Deborah F. Talkington, Division of Foodborne, Bacterial and Mycotic Dis-
eases, Centers for Disease Control and Prevention, Atlanta, GA 30333,
USA
Ken B. Waites, Department of Pathology, WP 230, University of Alabama at
Birmingham, 619 19th St. South, Birmingham, AL 35249, USA;
e-mail: waites@path.uab.edu
Tobias Welte, Department of Pulmonary Medicine, Carl-Neuberg-Str. 1,
30625 Hannover, Germany; e-mail: welte.tobias@mh-hannover.de
Preface

Community-acquired pneumonia is a disease of high morbidity and mor-


tality. Demographic changes in industrialised countries with a growing
population of elderly persons will add to its significance. In the last years
much progress in the field of community-acquired pneumonia has been
achieved. Vaccination programs against influenza and Streptococcus pneu-
moniae have been established. Risk-adjusted management of patients with
community-acquired pneumonia allows to identify patients in need of hos-
pitalisation and intensive care and helps to choose an effective antibiotic
therapy. “New” pathogens such as C. pneumoniae, Legionella pneumophila,
Chlamydia-like organisms, the human coronavirus or the avian influenza
virus have been detected. In spite of all progress, clinical diagnosis of com-
munity-acquired pneumonia is by no means trivial; detection of respiratory
pathogens often fails or gives inconclusive results and duration and choice
of antibiotics still is a matter of debate.
Moreover, many patients progress from uncomplicated pneumonia to
severe pneumonia and even to pneumonia-related septic shock despite
adequate antibiotic therapy. Therefore, besides new antibiotics we definitely
need a non-antibiotic approach and a better understanding of what deter-
mines individual immune responses to pneumonia is crucial. Fundamental
molecular and cellular pathologic characteristics of disease must be linked
with clinical aspects of infection.
The present book is intended to bridge the gap between basic science,
clinical research and patient management and to crosslink patient care with
biology and microbiology. It gives a state of the art information on differ-
ent aspects of community-acquired pneumonia and allows the reader to
get data on recent developments in community-acquired pneumonia. The
editors Norbert Suttorp, Tobias Welte and Reinhard Marre themselves,
representing clinical medicine, clinical research, microbiology as well as cell
biology, hope that this book will help to manage patients with community-
acquired pneumonia and to identify promising areas of research.

Berlin/Hannover/Ulm, August 2006 Norbert Suttorp


Tobias Welte
Reinhard Marre
Community-Acquired Pneumonia 1
ed. by N. Suttorp, T. Welte and R. Marre
© 2007 Birkhäuser Verlag Basel/Switzerland

Diagnosis and treatment of community acquired


pneumonia – the German perspective

Tobias Welte

Department of Pulmonary Medicine, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1,


30625 Hannover, Germany

Abstract
Current concepts of diagnosis and treatment of CAP are risk stratified and adapted to
the national resistances of important pathogens. Thorough surveillance systems have to
be implemented in all countries.
The risk of patients can be assessed reliably with a limited number of clinical data
(CRB-65 score). Extended microbiological and laboratory diagnosis is recommended for
hospitaliszed patients only. Outpatient treatment can be performed with classic antibiot-
ics like amoxicillin or doxycyclin. Macrolides are only an alternative in these patients. In
the hospital, treatment has to be adapted to the severity of the disease. Further studies
concerning the duration of treatment and advantageous combinations are necessary.
Recommendations for treatment of CAP have to be adapted to the quickly changing
epidemiology and have to be updated every 2 to 3 years.

Introduction

Pneumonia is a worldwide, serious threat to health, and an enormous


socio-economic burden for healthcare systems. According to recent WHO
data, each year three to four million patients die from pneumonia, a large
proportion of whom are children or elderly people. Pneumonia is the third
most common cause of death among infectious diseases in the world [1].
Detailed epidemiological data is available from the USA, where two
to three million cases of CAP occur each year, leading to around 10 mil-
lion doctor-patient contacts [2]. If an estimated proportion of 20 % (half a
million) of these patients were hospitalised, the incidence is 258 hospital
admissions per 100,000 inhabitants. The requirement for hospitalization
depends on age, with the highest rates observed for patients over age 65,
among whom the necessity for hospital admissions rises by a factor of four
to around 1000 per 100,000 inhabitants [3]. In total, it is estimated that the
costs for pneumonia treatment reach 8 billion dollars annually in the USA.
2 Tobias Welte

The largest proportion of this amount is spent on elderly and hospitalized


patients.
Community Acquired Pneumonia (CAP) results in high mortality (mean
about 8%). In the US, CAP is the sixth most frequent reason for dying, and
there is an increase of 0.5 to 1% per year [2]. The increase is caused by the
growing life expectancy, by aging of the population, and by a better treat-
ment of chronic diseases. Elderly people with concomitant diseases are
more susceptible to infectious diseases [4], and have typically a spectrum
of pathogens (gram negative enterobacteriaceae, staphylococci, legionella,
bacteriaemic pneumococci), which is associated with higher mortality [5, 6].
While the mortality of CAP is low in outpatients (1%), it can rise to up to
12% in hospitalized patients [2].

Definition

Pneumonia is an infection of the alveolar space, with accumulation of


inflammatory cells and secretions in the alveoli, resulting in impaired gas
exchange [8]. Each pneumonia acquired outside of a hospital is defined
as community acquired pneumonia [4], while nosocomial pneumonia is
caused during a stay in the hospital and up to one week after discharge.
A subgroup of CAP is the ”healthcare associated pneumonia” of patients
with frequent contact with the healthcare system (haemodialysis patients,
patients in nursing homes) [9]. Although it is community acquired, this form
is treated similar to a nosocomial infection.
The approach of scientific communities to CAP is very different. A
proof of the disease is the pathologic result, combined with a positive
microbiologic specimen of the tissue. In the clinical routine, biopsies of the
lungs cannot be obtained. The typical signs on the chest radiograph can be
delayed, even with the best technical equipment. Initial diagnostics often
shows no pulmonary infiltration [10]. All other signs which are typical for
pneumonia, such as the typical sounds on auscultation, fever, cough and
sputum expectoration, dispnoea, chest pain and serological markers of
inflammation are not pathognomic, and are nearly not always present in
all patients. In the English-speaking countries, the appearance of a new or
progressive infiltration is absolutely necessary for a diagnosis of pneumo-
nia. All other criteria are of minor importance for the diagnosis [11]. The
European Respiratory Society (ERS) defines pneumonia not via a chest
radiograph finding, since many outpatients do not receive this diagnostic
and therefore chest x-ray cannot be the major criterion. The ERS defines
a “lower respiratory tract infection”, according to the clinical presenta-
tion. This includes tracheo-bronchitis, influenza infection, exacerbation of
COPD, and pneumonia [12]. The recommendations of the ERS are much
broader and cannot been compared with the narrowly focused American
recommendations for CAP.
Diagnosis and treatment of community acquired pneumonia – the German perspective 3

Aetiology

The spectrum of pathogens and resistances varies widely between conti-


nents and countries. Universal guidelines for diagnosing and treatment are
for rough orientation only; the treatment must be adapted to the specific
local situation.
Even under optimal diagnostic conditions, sufficient sputum specimen
can be obtained in only 50% of patients [13]. In the early phase of the
infection, sputum production may still be normal. In about one-third of
all cases, the specimen do not meet international quality standards, which
require a high proportion of leukocytes and a low proportion of squamous
cells (Bartlett-criteria [4]). Depending on the patient group (all patients,
all patients with positive results in the specimen, all patients who were
able to expectorate sputum, all patients who produced purulent sputum),
very different distributions of pathogens has been reported. According
to results from the German competence network for CAP (CAPNETZ
[14]), a reliable microbiologic diagnosis can be established in only 20% of
all cases [15]. Worldwide, the most important pathogen is Streptococcus
pneumoniae, followed by Haemophilus influenzae and Mycoplasma pneu-
moniae. Legionella is rare with a frequency of 4%, but associated with an
excessive mortality. This underlines the importance of the very sensitive
urinary-antigen testing in cases with clinically suspected infection with
legionella (Tab. 1). Infections with enterobacteriaceae are most common
in patients from nursing homes, elderly patients and multi-morbid patients
(cardiac and kidney diseases, neurologic disorders and chronic obstruc-
tive pulmonary disease, CODP). The mortality of these patients is much
higher than in patients who are living in the normal community [16]. In the
USA, Pseudomonas aeruginosa is also a typical pathogen in CAP [6], but
Pseudomonas is not important in middle and northern Europe.
Some studies from Italy and Spain [17] report a high prevalence of
Chlamydia pneumoniae (> 10%). These results come from serologic test-

Table 1. Clinical findings in patients with legionella infection

Fever 100%
Chills 73%
Cough 83%
Purulent sputum 50%
Chest pain 30%
Abdominal symptoms 30%
Polymyositis 78%
Neurological symptoms 23%
4 Tobias Welte

Figure 1. Association of the consumption of penicillins and the prevalence of penicillin resist-
ant pneumococci in Europe (modified according to [22]).
AT, Austria; BE, Belgium; HR, Croatia; CZ, Czech Republic; DK, Denmark; FI, Finland;
FR, France; DE, Germany; HU, Hungary; IE, Ireland; IT, Italy; LU, Luxembourg; NL, The
Netherlands; PL, Poland; PT, Portugal; SI, Slovenia; ES, Spain; UK, England only.

ing. Studies using polymerase chain reaction (PCR) show in less than 3% a
positive finding [15]. The titres of IgA and IgM may remain elevated, even
after previous or oligosymptomatic infections. The use of serologic testing
seem not to be sensible in an acute infection. Chlamydia pneumoniae might
be very prevalent is special outbreaks, but presently, the importance of C.
pneumoniae for CAP seems to be low.
Viruses had been found in a number of studies (with or without PCR)
in 10 to 1 % of all CAP cases [5, 15, 18]. The question of whether viruses
are the responsible pathogen for CAP, or if the virus induced damage of
the bronchial epithelia is the precursor of bacterial infection, is still open.
In winter, influenza viruses are most important (70% of all viruses), under-
lining the importance of influenza vaccination in the elderly because of the
prevalence and severity of pneumonia. Large trials revealed that vaccina-
tion results in lower rates of pneumonia [19]. Similar results could not be
obtained for the vaccination against S. pneumoniae, since the vaccinate did
not cover all serotypes of this pathogen. Bacteriaemic infections could be
prevented, but there is no local protection [14]. American studies revealed
Diagnosis and treatment of community acquired pneumonia – the German perspective 5

that vaccination of children reduced the incidence of severe pneumonia in


adults, albeit different serotypes play the dominant role [20].

Resistances

Problems with resistances of the most important pathogens, especially


pneumococci, vary widely between different countries [21]. The main
reasons are differences in the consumption of antibiotics. There is a direct
correlation between the use of antibiotics and resistances in many countries
(Fig. 1) [22].
The significance of pathogen resistances for the outcome of a patient
is controversially discussed. Increasing resistances of pneumococci against
penicillin did not affect the mortality, even when treatment with penicillin
was continued. On the other hand, resistances against Cefuroxim had been
found to worsen mortality of pneumonia patients [23].
Pneumococci resistant against makrolides seem to be associated with
higher rates of bacteriaemia [24], but the impact on mortality in unclear.
If peumococci are resistant against fluorochinolones (there has been one
epidemic in Hong Kong), treatment with fluorochinolons had worsened the
prognosis of some individual patients [25].
An improvement of resistances can be achieved – as documented in
Scandinavia – by temporary reduction of the use of the respective classes
of antibiotics [26].

Risk stratification

The risk factors for an increased CAP mortality are age, the number of
concomitant diseases, and the place of residence before admission to the
hospital (patients from nursing homes had an eight-fold higher risk for
dying than patients coming from “regular” homes) [7].
Different scores for the estimation of the prognosis of patients with
CAP had been evaluated to substantiate the decision for hospital admission
and the decision of where to treat the patient (regular ward, intermediate
care unit, intensive care unit). Many scores designed for hospital patients
(Pneumonia Severity Index, PSI [27], CURB Score [28]) have the disad-
vantage that they need laboratory testing, what is not available in the out-
patient setting. Recent data revealed that a simple clinical score (CRB-65;
C: Confusion, R: respiratory rate > 30/min, B: blood pressure < 90 mmHg,
65: age > 65 years) allows to stratify patients into a low, moderate, or high
mortality risk group [29, 30].
The risk within the hospital can be best assessed with the modified
ATS Score [31]. If one major criterion (septic shock requiring vasopressor
therapy or mechanical ventilation) or two minor criteria (acute respiratory
6 Tobias Welte

insufficiency (PaO2/FiO2 < 250), multilobular infiltrates in the chest radio-


graph, systolic blood pressure < 90 mmHg) are present, then intensive care
treatment is necessary.

Diagnostic procedures

A chest radiograph (pa (posterior/anterior) and lateral view) is highly rec-


ommended to establish the diagnosis. However, the chest radiograph does
not have 100% sensitivity. High-resolution computed tomography (hrCT)
might show infiltrates which had not been detected in a previous chest x-ray.
If there is a clinical suspicion of pneumonia, the chest radiograph is normal,
and there is no clinical improvement of the patient, a new chest radiograph
should be performed 24 to 48 h later, or a chest computed tomography
should be performed.
Not all infiltrates in the lungs are caused by pneumonia. Congestive heart
failure, tumour, pulmonary infarction, and infiltrates due to interstitial pul-
monary diseases or systemic diseases (Wegener’s granulomatosis, collageno-
ses, and others) are important differential diagnoses for acute pneumonia.
Elevated levels of C-reactive protein (CrP) or procalcitonin III (very
expensive and not currently a routine procedure) are important parameters
[32], even if they do not prove infection. In bacterial pneumonia, leucocy-
tosis and an overrepresentation of young granulocytes can be expected.
Leucopenia might be a sign of sepsis and might therefore be a bad prog-
nostic factor.
In patients with a CRB-65 score of 0, no further laboratory diagnostic
is necessary. In hospitalized patients, the diagnostics should be focused on
inflammation, and further laboratory diagnostics should be performed,
depending on the severity of the disease and on concomitant problems.
Microbiologic testing is recommended, depending on the severity of
the pneumonia and on other risk factors. Patients with a low risk (CRB-
65 = 0) receive no benefit. However, this may change, if in addition to the
urine legionella antigen testing other quick reacting tests become available.
Preliminary results from the pneumococci antigen test do not justify a rou-
tine use of this expensive test.
Microbiologic specimen from the lower airways (sputum, bronchoalveo-
lar lavage and biopsies), pleural effusions, and blood cultures can be better
and faster collected in the hospital than in the outpatient setting. Such diag-
nostics can be recommended for all hospitalized patients in severe cases it
is mandatory.
Blood cultures show positive results in 10 to 20% only and that is a
proof. In severe infections, blood cultures must be obtained.
If legionella infection is suspected (for clinical signs see Table 1), an
antigen testing from urine specimen on Legionella pneumophilia antigen
is recommended.
Diagnosis and treatment of community acquired pneumonia – the German perspective 7

Serological testing (to screen for Mycoplasma, Chlamydia and viruses)


is not generally recommended since results are difficult to interpret (see
above).
Patients who do not improve within 72 h, or who worsen in clinical state,
must be seen as non-responders [34]. In this situation, invasive diagnostic
procedures (bronchoalveolar lavage, transbronchial lung biopsy), and
– depending on the clinical course – further radiologic diagnostic (pleural
ultrasound, CT) might become necessary.

Treatment

All guidelines distinguish patients with low risk of dying (CRB-65 ) 1,


CURB ) 1, PSI I und II), who might be treated as outpatients, and patients
with high risk, who need hospital treatment. Further prognostic factors are
concomitant diseases and the type of pathogen.
Important demographic, epidemiologic, and clinical risk factors for a
specific spectrum of pathogens are:

– Previous antibiotic treatment increases the rate of resistant pathogens.


The previous antibiotic should not be continued [22].
– Recent visits to countries with high prevalence of legionellosis (e.g.
Spain, Italy) and/or contact to contaminated water has to be explored.
– Patients aged 65 and above have a higher prevalence of gram-negative
pathogens, especially if they have co-morbidities (heart, lung, kidney, liver),
previous treatment with antibiotics, or previous hospitalisation [35].
– Patients from nursing homes or with previous hospitalization are at
increased risk for infections with enterobacteriaceae and Staphylococcus
aureus, and have a higher risk for aspiration pneumonia [16].
– Chronic lung diseases are associated with Haemophilus influenzae [36].
In advanced stages of COPD, in patients with cystic fibrosis or bron-
chiectasis, there is a frequent finding of S. aureus and Pseudomonas
aeruginosa [6].
– Contact to animals: Birds: C. psittaci, sheep: C. burnetii.
– Previous treatment with steroids of at least 10 mg/day prednisolone-
aequivalent over at least 4 weeks, or structural lung diseases (COPD,
bronchiectasis, cystic fibrosis) and a hospitalization within the last 30
days (longer than a two-day stay) are associated with P. aeruginosa and
Legionella spp. [9].

The recommendations for treatment distinguish whether there are risk fac-
tors for an infection with Pseudomoas aeruginosa [6]:

– Pulmonary concomitant diseases (structural defects in COPD stadium


GOLD IV, bronchiectasis, cystic fibrosis).
8 Tobias Welte

Table 2. Treatment recommendations for patients with low risk in the outpatient setting.

Substance Dosage (per day) Duration of therapy


First choice
Aminopenicillines
– Amoxicillin * 70 kg: 1 g t.i.d p.o. 7–10 days
< 70 kg: 0.75 g t.i.d. p.o.
Or
Tetracycline
– Doxycycline 200 mg once initially, 7–10 days
* 70 kg: 200 mg once daily
< 70 kg: 100 mg once daily
Alternative
Macrolidea
– Azithromycin 500 mg once daily p.o. 3 days
– Clarithromycin 2 × 500 mg p.o. 3 days, 7–10 days
followed by 250 mg b.i.d
– Roxithromycin 1 × 300 mg oral 7–10 days
aonly in countries with low macrolide resistance

– Hospitalization within the last 30 days (longer than two days stay).
– Risk for aspiration (e.g. neurological diseases).
– Treatment with a broad spectrum antibiotic over more than 7 days
within the last month.

Depending on the number of risk factors, the probability of Pseudomonas


aeruginosa increases exponentially (to more than 50 % in three or more
risk factors).
The guidelines show wide difference in the recommended antibiotics.
The reasons are different local resistances. The recently published recom-
mendations of the ERS are presented here [12]. Updates of the recommen-
dations of the ATS and ISDA are under preparation, but not finalized yet.
Table 2 shows the recommended treatment of CAP patients with a low
risk and treatment in the outpatient setting. A follow-up to assess the suc-
cess of treatment should be performed after 72 h. Patients or persons in
their environment should be advised to contact their doctor again if fever
exceeds 4 days, dyspnoea gets worse, patients stop drinking, or conscious-
ness is decreasing.
Macrolides have been used to treat pathogens such as Chlamydia
and Mycoplasma which did not respond to beta-lactam antibiotics. Some
recently published studies [37, 38] demonstrate that patients with a low
risk of dying have no worse outcome with penicillin derivatives compared
with macrolides. Infections with Chlamydia seem to be rare, and infections
Diagnosis and treatment of community acquired pneumonia – the German perspective 9

Table 3. Treatment recommendations for the calculated initial therapy of hospitalized CAP
patients (no severe pneumonia) without a risk of Pseudomonas infection

Substances Dosage Duration of therapy


Preferred in regions with low pneumococcal
resistance rate
– Penicillin G 1 Mill. IU t.i.d 7–10 days
– Ampicillin 1.5 g t.i.d i.v. 7–10 days
– Amoxicillin/Clavulansäure# 2.2 g t.i.d i.v. 7–10 days
– Ampicillin/Sulbactam 3.0 g t.i.d i.v. 7–10 days
– Cefuroxim 1.5 g t.i.d i.v. 7–10 days
– Ceftriaxon 2.0 g once daily i.v. 7–10 days
– Cefotaxim 2.0 g t.i.d i.v. 7–10 days
± Macrolides (s. Tab. 2)* 7–10 days
Alternatives
(in regions with increased pneumococcal
resistance or intolerance to preferred drugs)
Fluorquinolone§
– Levofloxacin 500 mg once daily i.v. 7–10 days
– Moxifloxacin 400 mg once daily i.v. 7–10 days

# Can be applied as sequential treatment using the same drug


* new macrolides preferred to erythromycin
§ within the fluoroquinolones, moxifloxacin has the highest antipneumococcal activity.
Experience with ketolides is limited but they may offer an alternative when oral treatment is
adequate

with Mycoplasma seem to be self-limiting in low-risk patients. In these sit-


uations, patients might be treated without intracellular active antibiotics.
Doxycyclin had been accepted as first-line treatment despite insufficient
clinical data, since there is no information about treatment failures due to
resistance.
In the outpatient setting, oral treatment is preferred. Oral cephalospo-
rins have a low bioavailability, and there are doubts about their pharmaco-
kinetics and pharmacodynamics in elderly people. Cephalosporins are an
alternative in exceptional cases only.
The recommendations for hospitalized patients are shown in Table 3.
Hospitalized patients should receive parenteral treatment, due to unreli-
able oral pharmacokinetics in elderly and severely affected patients. If the
clinical response is satisfactory, an early switch (after 72 h) to similar oral
substances is possible. Treatment success is assumed if the respiratory rate
is lower than 25/min, and oxygen saturation is above 90%, body tempera-
ture fell by at least 1°C, and the haemodynamic and neurological status are
satisfactory, and if oral feeding is possible [39].
A number of retrospective, and one prospective study [40] show that in
bacteriaemia with S. pneumoniae a combination therapy with a beta-lactam
10 Tobias Welte

Table 4. Criteria for severe community acquired pneumonia (modified according to [46])*

Baseline (minor) criteria assessed at admission


Respiratory rate > 30 min–1
Severe respiratory failure (Pa,O2/Fi,O2 ratio > 250)
Bilateral involvement in chest radiograph
Involvement of more than two lobes in chest radiograph (multilobar involvement)
Systolic blood pressure < 90 mmHg
Diastolic blood pressure < 60 mmHg

Major criteria assessed at admission or during clinical course


Requirement for mechanical ventilation
Increase in the size of infiltrates by * 50% in the presence of clinical nonresponse to
treatment or deterioration (progressive infiltrates)
Requirement of vasopressors > 4 h (septic shock)
Serum-creatinine * 2 mg·dl–1 or increase of * 2 mg·dl–1 in a patient with previous renal
disease or acute renal failure requiring dialysis (acute renal failure)

Pa,O2, arterial oxygen tension; Fi,O2, inspiratory oxygen fraction;


*, the presence of at least two minor criteria or one major criterion defines severe pneumonia,
i.e., pneumonia requiring admission at the ICU.

antibiotic and a macrolide offers the best treatment success. Despite severe
weakness in the design of these studies, combination therapy is recommend-
ed at least for severe infections. Mono therapy with a new fluoroquinolone
(moxifloxacin or levofloxacin) seems to be as effective as combination
therapy, but reliable data is missing [41].
Table 4 shows criteria for severe community acquired pneumonia
(sCAP). In this form of pneumonia, broad initial treatment effective against
all possible pathogens is necessary. The choice of antibiotics is guided by the
probability of Pseudomonas infection (Tab. 5).
In patients with sCAP, the combination therapy between a beta-lactam
and an aminoglycoside or fluorochinolone is also under discussion. Two
Meta analyses [42, 43] could not reveal an additional effect of aminogly-
cosides in infections with gram-negative pathogens. However, the data on
infections with pseudomonas are not sufficient. The recommended combi-
nation of beta-lactams with pseudomonas-active fluoroquinolones seems
to be of advantage, but this combination has not been tested systematically
until today. One abstract, published from the ECCMID meeting 2006 [44]
suggested that mono therapy with moxifloxacin, which penetrates tissue
well, could be sufficient.
Treatment failure is defined as lack of clinical improvement after 72 h of
treatment or worsening of the clinical situation. Besides extended diagnos-
tic procedures, which also allow the recognition of important complications
of CAP (pleural empyema, lung abscess), treatment must be de-escalated
target intracellular pathogens (combinations of macrolides and newer fluo-
Diagnosis and treatment of community acquired pneumonia – the German perspective 11

Table 5. Treatment recommendations for the calculated initial therapy of patients with severe
CAP.

Substances Dosage Duration of therapya


No risk factors for Pseudomonas
– Ceftriaxon 2,0 g once daily i.v. 7–10 days
– + Makrolides (s. Tab. 2)
– or + Fluorquinolone (s. Tab. 3)
Risk factors for Pseudomonasa
– Piperacillin/Inhibitor 4,5 g t.i.d i.v. 7–14 days
– Cefepime 2,0 g t.i.d i.v. 7–14 days
– Carbapenem 1,0 g t.i.d i.v. 7–14 days
– + Ciprofloxacin 400 mg t.i.d i.v. 7–14 days
aminimum treatment is 7 days, maximum 14 days if there is a proof of pseudomonas or L.

pneumophilia.

roquinolones). Gram-negative pathogens and Pseudomonas must be cov-


ered (Tab. 5).
The problem of an optimized duration of treatment is controversially
discussed. First results of short-term treatment (5 days) in patients with
low risk are encouraging [45]. Hospitalized patients who were treated on a
regular ward were nearly completely free of symptoms after five days [41].
The current evidence does not allow a general recommendation of such
shor-t term therapy. Treatment duration of 10 days should only be exceeded
in rare, exceptional cases.

References
1 Lopez AD, Murray CC (1998) The global burden of disease. Nat Med 4: 1241–
1243
2 Center for Disease Control and Prevention (1997) Premature deaths, monthly
mortality and monthly physician contacts-United States. MMWR Morb Mortal
Wkly Rep 46: 556
3 Marston BJ, Plouffe JF, File TM Hackman BA, Salstrom SJ, Lipman HB,
Kolczak MS, Breiman RF (1997) Incidence of community-acquired pneumo-
nia requiring hospitalization. Arch Intern Med 157(15): 1709–1718
4 Bartlett JG, Dowell SF, Mandell LA, File TM Jr, Musher DM, Fine MJ (2000)
Practice guidelines for the management of community-acquired pneumonia in
adults. Infectious Diseases Society of America. Clin Infect Dis 31: 422–425
5 Lim WS, Macfarlane JT, Boswell TC, Harrison TG, Rose D, Leinonen M,
Saikku P (2001) SCAPA: Study of Community Acquired Pneumonia Aetiology
in adults admitted to hospital: implications for management guidelines. Thorax
56: 296–301
12 Tobias Welte

6 Arancibia F, Bauer TT, Ewig S, Mensa J, Gonzalez J, Niederman MS, Torres A


(2002) Community-acquired pneumonia due to gram-negative bacteria and
Pseudomonas aeruginosa: incidence, risk, and prognosis. Arch Intern Med 162:
1849–1858
7 Fine MJ, Smith MA, Carson CA, Mutha SS, Sankey SS, Weissfeld LA, Kapoor
WN (1996) Prognosis and outcome of patients with community acquired pneu-
monia. JAMA 275: 134–141
8 Osler W (1901) Lobar pneumonia. In: J Young (ed) The principles and practice
of medicine. Pentland, Edinburgh & London, 126–129
9 American Thoracic Society; Infectious Diseases Society of America (2005)
Guidelines for the management of adults with hospital-acquired, ventilator-
associated, and healthcare-associated pneumonia. Am J Respir Crit Care Med
171(4): 388–416
10 Syrjala H, Broas M, Suramo I (1998) High-resolution computed tomography for
the diagnosis of community-acquired pneumonia. Clin Infect Dis 27: 358–363
11 Bartlett JG, Breiman RF, Mandell LA, File TM (1998) Community-acquired
pneumonia in adults: Guidelines for management. Clin Infect Dis 26: 811–838
12 Woodhead M, Blasi F, Ewig S, Huchon G, Leven M, Ortqvist A, Schaberg T,
Torres A, van der Heijden G, Verheij TJ (2005) Guidelines for the management
of adult lower respiratory tract infections. Eur Respir J 26(6): 1138–1180
13 Metlay JP, Schulz R, Li YH, Singer DE, Marrie TJ, Coley CM, Hough LJ,
Obrosky DS, Kapoor WN, Fine MJ (1997) Influence of age on symptoms at
presentation in patients with community-acquired pneumonia. Arch Intern
Med 157(13): 1453–1459
14 Welte T, Suttorp N, Marre R (2004) CAPNETZ – community-acquired pneu-
monia competence network. Infection 4: 234–238
15 Welte T, Marre R, Suttorp N; for the CAPNETZ Network (2006) What is new
in the treatment of community-acquired pneumonia? Med Klin (Munich)
101(4): 313–320
16 El-Solh AA, Sikka P, Ramadan F, Davies J (2001) Etiology of severe pneumo-
nia in the very elderly. Am J Respir Crit Care Med 163: 645–651
17 Blasi F (2004) Atypical pathogenes and respiratory tract infections. Eur Respir
J 24: 171–181
18 De Roux A, Marcos MA, Garcia E, Mensa J, Ewig S, Lode H, Torres A (2004)
Viral community-acquired pneumonia in nonimmunocompromised adults.
Chest 125: 1343–1351
19 Nichol KL, Wuorenma J, von Sternberg T (1998) Benefits of influenza vaccina-
tion for low-, intermediate-, and high-risk senior citizens. Arch Intern Med 158:
1769–1776
20 Whitney CG, Farley MM, Hadler J, Harrison LH, Bennett NM, Lynfield R,
Reingold A, Cieslak PR, Pilishvili T, Jackson D et al (2003) Active bacterial
core surveillance of the emerging infections program network. Decline in inva-
sive pneumococcal disease after the introduction of protein-polysaccharide
conjugate vaccine. N Engl J Med 348(18): 1737–1746
21 Felmingham D, Reinert RR, Hirakata Y, Rodloff A (2002) Increasing preva-
lence of antimicrobial resistance among isolates of Streptococcus pneumoniae
from the PROTEKT surveillance study, and compatative in vitro activity of the
ketolide, telithromycin. J Antimicrob Chemother 50 Suppl S1: 25–37
Diagnosis and treatment of community acquired pneumonia – the German perspective 13

22 Goossens H, Ferech M, Vander Stichele R, Elseviers M (2005) Outpatient anti-


biotic use in Europe and association with resistance: a cross national database
study. Lancet 365: 579–587
23 Yu VL, Chiou CC, Feldman C, Ortqvist A, Rello J, Morris AJ, Baddour LM,
Luna CM, Snydman DR, Ip M et al.; International Pneumococcal Study Group
(2003) An international prospective study of pneumococcal bacteremia: cor-
relation with in vitro resistance, antibiotics administered, and clinical outcome.
Clin Infect Dis 37(2): 230–237
24 Lonks JR, Garau J, Gomez L, Xercavins M, Ochoa de Echaguen A, Gareen
IF, Reiss PT, Medeiros AA (2002) Failure of macrolide antibiotic treatment in
patients with bacteremia due to erythromycin–resistant Streptococcus pneu-
moniae. Clin Infect Dis 35 (5): 556–564
25 Davidson R, Cavalcanti R, Brunton JL, Bast DJ, de Azavedo JCS, Kibsey P,
Fleming C, Low DE (2002) Resistance to Levofloxacin and failure of treat-
ment of pneumococcal pneumonia. N Engl J Med 346: 747–750
26 Seppala H, Klaukka T, Vuopio-Varkila J (1997) The effect of changes in the
consumption of macrolide antibiotics on erythromycin resistance in group A
streptococci in Finland. Finnish Study Group for Antimicrobial Resistance. N
Engl J Med 337: 441–446
27 Fine MJ, Auble TE, Yealy DM, Hanusa BH, Weissfeld LA, Singer DE, Coley
CM, Marrie TJ, Kapoor WN (1997) A prediction rule to identify low-risk
patients with community acquired pneumonia. N Engl J Med 336: 243–250
28 Lim WS, van der Eerden MM, Laing R, Boersma WG, Karalus N, Town GI,
Lewis SA, Macfarlane JT (2003) Defining community acquired pneumonia
severity on presentation to hospital: an international derivation and validation
study. Thorax 58: 377–382
29 Capelastegui A, Espana PP, Quintana JM, Areitio I, Gorordo I, Egurrola M,
Bilbao A (2006) Validation of a predictive rule for the management of com-
munity-acquired pneumonia. Eur Respir J 27(1): 151–157
30 Bauer T, Ewig S, Marre R, Suttorp N, Welte T (2006) CURB and CRB–65
scores predict mortality in hospitalised and out-patients with community-
acquired pneumonia (CAP). J Intern Med 260: 93–101
31 Ewig S, de Roux A, Bauer T, Garcia E, Mensa J, Niederman M, Torres A (2004)
Validation of predictive rules and indices of severity for community acquired
pneumonia. Thorax 59 (5): 421–427
32 Christ-Crain M, Jaccard-Stolz D, Bingisser R, Gencay MM, Huber PR, Tamm
M, Muller B (2004) Effect of procalcitonin-guided treatment on antibiotic use
and outcome in lower respiratory tract infections: cluster-randomised, single-
blinded intervention trial. Lancet 363: 600–607
33 Ishida T, Hashimoto T, Arita M, Tojo Y, Tachibana H, Jinnai M (2004) A 3-year
prospective study of a urinary antigen-detection for Streptococcus pneumoniae
in community acquired pneumonia: utility and clinical impact on the reported
etiology. J Infect Chemother 10: 359–363
34 Menendez R, Torres A, Zalacain R, Aspa J, Martin Villasclaras JJ, Borderias
L, Benitez Moya JM, Ruiz-Manzano J, Rodriguez de Castro F, Blanquer J et
al.; Neumofail Group (2004) Risk factors of treatment failure in community
acquired pneumonia: implications for disease outcome. Thorax 59(11): 960–
965
14 Tobias Welte

35 Janssens JP (2005) Pneumonia in the elderly (geriatric) population. Curr Opin


Pulm Med 11(3): 226–230
36 Sethi S, Evans N, Grant BJ, Murphy TF (2002) New strains of bacteria and
exacerbations of chronic obstructive pulmonary disease. N Engl J Med 347(7):
465–471
37 Mills GD, Oehley MR, Arrol B (2005) Effectiveness of beta lactam antibiotics
compared with antibiotics active against atypical pathogens in non-severe com-
munity acquired pneumonia: meta-analysis. BMJ 330: 456
38 Guchev IA, Yu VL, Sinopalnikov A, Klochkov OI, Kozlov RS, Stratchounski
LS (2005) Management of nonsevere pneumonia in military trainees with the
urinary antigen test for Streptococcus pneumoniae: an innovative approach to
targeted therapy. Clin Infect Dis 40: 1608–1616
39 Marrie TJ, Lau CY, Wheeler SL, Wong CJ, Vandervoort MK, Feagan BG (2000)
A controlled trial of a critical pathway for treatment of community-acquired
pneumonia. CAPITAL Study Investigators. Community-Acquired Pneumonia
Intervention Trial Assessing Levofloxacin. JAMA 283: 749–755
40 Baddour LM, Yu VL, Klugman KP, Feldman C, Ortqvist A, Rello J, Morris AJ,
Luna CM, Syndman DR, Ko WC et al (2004) Combination antibiotic therapy
lowers mortality among severely ill patients with pneumococcal bacteremia.
Am J Respir Crit Care Med 170: 440–444
41 Welte T, Petermann W, Schuermann D, Bauer TT, Reimnitz P, Moxirapid Study
Group (2005) Treatment with sequential intravenous/oral moxifloxacin was
associated with faster clinical improvement compared to standard therapy
in hospitalized CAP patients with initial parenteral therapy. Clin Infect Dis
41(12): 1697–1705
42 Paul M, Benuri-Silbiger I, Soares-Weiser K, Leibovici L (2004) `-lactam
monotherapy versus `-lactam-aminoglycoside combination therapy for sepsis
in immunocompetent patients: systematic review and meta-analysis of ran-
domised trials. BMJ 328: 668
43 Safdar N, Handelsman J, Maki DG (2004) Does combination antimicro-
bial therapy reduce mortality in gram-negative bacteraemia? A meta-analysis.
Lancet Infect Dis 4: 519–527
44 Torres A, Read R, Lode H, Carlet J, Winter JH, Garau J, Welte T, Arvis P, Le
Berre MA, Choudhri SH (2006) Once daily sequential intravenous/oral (IV/
PO) moxifloxacin is equivalent to IV ceftriaxone plus twice daily IV/PO levo-
floxacin in the treatment of severe community-acquired pneumonia requiring
hospitalization: the MOTIV study. ECCMID 2006
45 Dunbar LM, Wunderink RG, Habib MP, Smith LG, Tennenberg AM, Khashab
MM, Wiesinger BA, Xiang JX, Zadeikis N, Kahn JB (2003) High-dose, short-
course levofloxacin for community-acquired pneumonia: a new treatment
paradigm. Clin Infect Dis 37: 752–760
46 Niederman MS, Mandell LA, Anzueto A, Bass JB, Broughton WA, Campbell
GD, Dean N, File T, Fine MJ, Gross PA et al (2001) Guidelines for the manage-
ment of adults with community-acquired pneumonia. Diagnosis, assessment of
severity, antimicrobial therapy, and prevention. Am J Respir Crit Care Med 163:
1730–1754
Community-Acquired Pneumonia 15
ed. by N. Suttorp, T. Welte and R. Marre
© 2007 Birkhäuser Verlag Basel/Switzerland

Detection of respiratory bacterial pathogens

Reinhard Marre

University of Ulm, Institute of Medical Microbiology & Hygiene, Robert-Koch-Str. 8,


89081 Ulm, Germany

Abstract
Microbiology of community-acquired pneumonia often is based on indirect or precari-
ous evidence. Time and effort to detect a respiratory pathogen often is not sufficiently
related to its usefulness in guiding therapy. If sputa are accepted by the laboratory for
microbiological studies (Gram stain, culture), they should fulfill quality criteria such as
high number of leukocytes and low number of squamous epithelial cells. Complementary
tests such as antigen detection assays are useful adjuncts for diagnosing pneumococcal
and Legionella pneumonia. Nucleic amplification tests help to overcome the problems in
detecting Chlamydia pneumoniae, Legionella and Mycoplasma pneumoniae.

Introduction

Community acquired pneumonia is caused by a wide variety of differ-


ent bacterial species. Streptococcus pneumoniae is the leading pathogen,
followed by Haemophilus influenzae, Staphylococcus aureus, Chlamydia
pneumoniae, Mycoplasma pneumoniae and a broad spectrum of enterobac-
terial species. While it is generally agreed upon that S. pneumoniae is the
predominant pathogen in CAP, reported incidences of other species vary
considerably depending on patient population, specific epidemiological
situations and microbiological aspects such as type of specimens studied,
detection methods used and pathogen definition. Even in clinical trial con-
ditions detection of a respiratory pathogen often is based on precarious evi-
dence, namely microscopical or cultural detection of a facultative pathogen
in respiratory secretions which may be heavily contaminated by bacteria
of the oropharyngeal tract. Unequivocal, definitive evidence, i.e. growth in
blood or other usually sterile specimens is rare and selects for severe bacte-
remic pneumonia and may thus not be representative. Nucleic acid ampli-
fication techniques (NAT) such as PCR to detect microbial pathogens are
– in the absence of a solid reliable gold standard – not a reliable indicator
16 Reinhard Marre

of a causative agent. In addition, clinical patterns often do not give a clear


clue about the causative micro-organism [1].
Beyond clinical trials the situation is even worse and quality of micro-
biology of respiratory secretions often is regarded as unsatisfactory. This
resulted in the statement of Bartlett that the culture of lower respiratory
specimens may result in more unnecessary microbiologic efforts than any
other type of specimen [2]. Clinical relevance of microscopy of respira-
tory specimens too is regarded as very controversial [3] and guidelines of
the ATS and the IDSA differ with respect to the evaluation of the clinical
impact of a gram stain result of a sputum sample [4].
Because of these limitations and critical appraisal, microbiology of the
CAP may not be useful unless performed by expert microbiologists and
well trained technicians using standardized approaches for specimen work-
up, external and internal quality assurance with an active communication
network between microbiologists and clinicians. The effects of quality on
reported incidence has been illustrated by Bartlett [5], who described an
apparent decrease in the yield of S. pneumoniae from Gram staining and
culture from 80 to < 18% over the last 30 years. In evaluating 15 pub-
lished reports from North America, it was found that S. pneumoniae inci-
dence ranged from 20 to 60% [6], which is in agreement with the German
Multicenter Network CAPNETZ in which the incidence of S. pneumoniae
ranged from 28 to 48% depending of the patients population studied.

Microscopy of respiratory secretions

Microscopy of respiratory sample may help in guiding initial therapy in


CAP if a properly collected specimen is tested. Non-invasive samples such
as sputa and upper respiratory swabs give less conclusive evidence than
invasive samples (Bronchoalveolar lavages (BAL), protected specimen
brushings), but are more easily available. An essential problem is that
sputum may be contaminated by saliva. Several approaches have been
developed to assess the quality of a respiratory sample. These have been
recently described and evaluated by Sharp et al [3]. The bottom line is
that the number of leukocytes related or not to the number of squamous
epithelial cells are a criteria of quality (Fig. 1). Based on this criterion, the
number of CAP patients with a good-quality sputum has been shown by
Rosón et al to be only 40% of all patients and 60% of the patients with a
sputum sample [7]. In 35% of the patients no sputum sample was available.
In a study performed by Kalin et al 76% of the sputa were deemed puru-
lent [8]. In CAPNETZ, a Germany-wide network on CAP with more than
3,000 patients sputum was available in 45 to 70% of patients. In about half
of the patients with sputum, the sample was purulent. Non-purulent sputum
should not be used for culture and therefore rejected. If non-purulent spu-
tum is used for culture non or little pathogenic microorganisms (Candida
Detection of respiratory bacterial pathogens 17

Figure 1: Gram stain of a high-quality (purulent) (below) and a non purulent sputum (above).
18

Figure 2: Frequency of isolates in dependence of the quality of sputum. Filled columns: < 10 leukocytes per 10x field, empty columns 10–25 leukocytes per
Reinhard Marre

10x field, shaddowed columns: > 25 leukocytes per 10x field.


Detection of respiratory bacterial pathogens 19

albicans, Escherichia coli and other enterobacteria) can be detected in high


numbers and in a high proportion of patients, thus perhaps leading to flawed
therapy decisions (Fig. 2). Although these species cannot be regarded as
pathogens in CAP, they may indicate altered colonization. Limiting identifi-
cation and work-up of the sample may lead to improved outcome as shown
convincingly by Barenfanger in the case of detection of yeasts in respiratory
specimens [9]. However, Chlamydia pneumoniae, Mycoplasma pneumoniae,
and Legionella can be detected in non-purulent sputum samples by NAT
techniques.
Many studies state that sputum can also safely be used for microscopic
detection of bacterial pathogens [3]. Positive and negative predictive value
were 90% and 62%, respectively, if patients with a definitive diagnosis of
pneumococcal pneumonia (detection of S. pneumoniae in sterile body flu-
ids or detection of the pneumococcal antigen) were evaluated, and 75 and
98% if patients with a Haemophilus influenzae pneumonia were evaluated
[7]. The corresponding sensitivity values were 35% (S. pneumoniae) and
42% (H. influenzae). Including only patients with a bacteremic pneumo-
coccal pneumonia, Musher reported a sensitivity of 57% [10]. It should,
however, be kept in mind that these studies select for patients with gen-
eralized bacterial infection and that therefore microscopy may be of less
value if patients with a less severe infection are included. In a meta-analy-
sis, Reed et al. reported a range of sensitivity values from 15 to 100%, and
of specificity from 11% to 100% in pneumococcal pneumonia [11], which
illustrates that a Gram stain in the hands of inexperienced may yield mis-
leading results.

Detection of pathogens by culture

Cultural detection of respiratory pathogens in sterile body fluids is usually


specific but limited to CAP with generalized infection. If secretions are
obtained from deeper sites of infections such as bronchoalveolar lavages or
fiber optic brush technique (FB) and if a cut-off level of 103 to 104 cfu/ml is
defined, cultural detection has a sensitivity between 42 and 95% and a speci-
ficity between 45 and 100% (reviewed by Sharp [3]). However, BAL and
FB and sterile body fluids are not available on a routine basis and therefore
microbiology has also to rely on culture of sputum. On the other hand, the
usefulness of sputum cultures is a matter of controversial debate. Heineman
et al stated that cultural detection of pathogens in sputum is meaningless
if not guided by microscopy [12]. Recent publications report either limited
value or nonvalue of microbiological studies to detect a CAP pathogen [13,
14]. Other reports indicate however that sputum cultures may reveal thera-
peutically relevant information in individual cases and that the relevance
can be increased if the sputum sample is purulent, obtained before the start
of antibiotic chemotherapy and if cultured without delay.
20 Reinhard Marre

Streptococcus pneumoniae

S. pneumoniae is the most predominant species in CAP. The incidence


ranges from 20 to 60%. About 20% of these pneumonias are bacteremic.
Pneumococci can be detected out of respiratory samples by microscopy and
culture, in addition by blood cultures and by detection of urinary antigen.
As described above, Gram stain of a sputum sample has a sensitivity of 60%
and specificity of near 100% to detect pneumococci in bacteremic patients
[7, 10]. The examination of a sputum Gram stain, however, is not as trivial as
generally believed and requires expertise of the lab personnel and an active
quality management programs.
The diagnostic value of a sputum culture in bacteremic patients has
been shown to be high if only bacteremic patients with purulent sputum
are included. In these patients it may help to guide therapy and to narrow
the spectrum of the antibiotic. The cultural isolates also allow recognizing
trends in resistance development that have implications on the improve-
ment of guidelines.
Urinary antigen assays for the detection of pneumococci (Binax NOW)
may overcome the limitations of sputum microscopy and culture, since it
is a rapid (15 min) bedside diagnostic, not limited by the availability of
purulent sputum, and does not require experienced lab personnel. The
assay detects the c polysaccharide antigen that is not capsular serotype spe-
cific. Sensitivity is reported to be 70 to 75%, specificity 90 to 95% [15-17].
Patients with a positive sputum culture may be antigen positive in about
only 50% [17], indicating either the low specificity of the sputum culture
or the low sensitivity of the antigen assay in non-bacteremic patients. In
contrast to adults, the test does not discriminate between children with and
without pneumococcal pneumonia [18].
Nucleic acid amplification assays are not useful in diagnosing a pneu-
mococcal pneumonia. When using plasma, buffy coat or urine as specimens,
Murdoch et al. studied the performance of a nested PCR targeting the
pneumolysin gene [19]. Plasma and buffy coat were only positive in 30%
of patients with a positive blood culture, urine was positive in 2% while
the antigen assay was positive in 29% of the patients. Throat swabs were
positive in 55 to 56 % by PCR, irrespective of the status as a control or as
patient.

Haemophilus influenzae

Although H. influenzae is the second or third most common species


in CAP, little attention has being paid to this species as a respiratory
pathogen. Patients with H. influenzae pneumonia often are elderly with
a history of chronic obstructive lung diseases, tumor pathology or other
causes of impaired immunity. Non-encapsulated strains of H. influenzae
Detection of respiratory bacterial pathogens 21

predominate. Lethality of an invasive H. influenzae infection of this patient


population is high [20]. As reported by Roson et al [7] sputum microscopy
has a high sensitivity and specificity for the diagnosis of a H. influenzae
pneumonia of hospitalized patients. Detection of H. influenzae by sputum
culture may overestimate the incidence of this species in CAP since it is a
normal inhabitant of the oropharyngeal tract. Assays to diagnose H. influ-
enzae infection by antigen detection are to the best of my knowledge not
available.

Legionella

Legionella comprises a genus with about 50 different species most of which


are never or rarely encountered in the human host. The overwhelming
majority of Legionella infections are attributed to Legionella pneumophila
serotype 1. Australia, however, may be an exemption where Legionella long-
beachae is more predominant. Legionella occur in aquatic systems where
it can thrive within amoebae. When inhaled by a susceptible person (more
than 60 years of age, smoker, male, immunocompromised), it can cause a
potentially lethal pneumonia. Legionella infections occur with an incidence
of 5 to 10% in CAP in an endemic situation. Morbidity is about 20 cases of
Legionella infection per one million inhabitants.
At the time being there is no one-for-all easy and rapid test to detect
Legionella. If a Legionella infection is suspected by the clinician, differ-
ent assays should be combined. Although Legionella can be cultured from
respiratory secretion when using special media containing cysteine and iron
as essential growth factors, the detection rates are rather low and sensitivity
may not exceed 60%. The advantage of a culture is that a Legionella isolate
for further investigations (source identification) is available. Sputum Gram
stain is not helpful since the Gram negative rod Legionella is stained only
faintly by fuchsin, unless the stain is allowed to act for a prolonged time.
Direct immunofluorescence test using polyclonal or monoclonal antibodies
directed against the lipopolysaccharide can be useful in principal. Cross-
reaction of the polyclonal antiserum with Pseudomonas species [21], the low
sensitivity, the long hands-on time and the need for experienced personnel
have limited the use of this test.
The introduction of the antigen detection assay (Binax or Biotest) has
significantly contributed to a rapid and specific diagnosis of Legionella sero-
type 1 infections. Legionella infections other than Legionella pneumophila
serotype 1, however, are not covered reliably. The antigen detection assay
may also select for severe infections as shown by Yzerman [22]. For patients
with mild Legionella infection, the test sensitivities ranged only from 40
to 53%, whereas for patients with severe Legionnaires Disease (LD) who
needed immediate special medical care, the sensitivities reached 88 to
100%. Guerrero et al. reported that the Bartels EIA and the Biotest EIA
22 Reinhard Marre

had significantly higher sensitivity levels than the Binax NOW immuno-
chromatographic test (65 to 70% vs. 37%) [23].
Nucleic acid amplification assays using respiratory secretions can com-
plement the Legionella test assay system. Real-time PCR is a rapid as well
as reliable method with a sensitivity of 100% when culture proven cases
of Legionella are studied [24, 25]. The 16S rDNA, 5S rDNA or the macro-
phage infectivity potentiator (mip) gene of Legionella commonly are used
as target genes.
Antibody assay is available for the diagnosis of Legionnaire’s Disease
(LD), but its value is limited since some LD patients do not react with a
significant rise of antibodies and others have a persistant IgM antibody titer.
With the advent of molecular tests the indication of serological testing in
LD in acute care is reduced to near null.

Chlamydia pneumoniae

Chlamydia pneumoniae was identified as pulmonary pathogen only 15


years ago. Because of its debated association with coronary heart disease C.
pneumoniae has attracted considerable attention. The reported incidence
ranges from 2 to 40% depending on the epidemiological situation, site and
time of the investigation, detection methods used and patients included.
In recent years reports with low incidences (less than 2%) seem to have
increased [25–27]. The CAPNETZ trial confirmed the low incidences of C.
pneumoniae infections [28].
Most assays for the detection of C. pneumoniae are not applicable for
routine microbiology. Cell culture techniques to grow C. pneumoniae are
restricted to specialized laboratories. Even under optimal conditions (no
delay until culture, high quality specimens) cultural detection is much more
futile than, for example, cultural detection of C. trachomatis. If grown in a
first passage, many isolates do not outlive further passages. The C. pneu-
moniae-specific microimmuno-fluorescence (MIF) test has been introduced
as matter of choice to prove a C. pneumoniae infection and many clinical
CAP studies rely on this assay. In clinical practice, however, serodiagnosis
must often rely on a single serum sample and, therefore, seroconversion and/
or rise in antibody titers are missed in many patients. In addition, about 60%
of adults have antibodies against C. pneumoniae which makes it difficult
to differentiate between an acute or past infection. During the last decade,
molecular methods including conventional and real-time PCR assays have
been developed for sensitive and specific detection of C. pneumoniae DNA
in respiratory tract specimens [29]. However, there is no consensus on the
most suitable specimen, the optimal DNA isolation method and target gene.
Results of multicenter PCR comparison trials have been contradictory and
difficult to interpret [30]. Therefore, further efforts to optimize and stan-
dardize diagnostics in C. pneumoniae are urgently needed.
Detection of respiratory bacterial pathogens 23

Mycoplasma pneumoniae

Detection of Mycoplasma pneumoniae in CAP is dealt with in the chapter


by Waites et al.

References
1 Farr BM, Kaiser DL, Harrison BD, Connolly CK (1989) Prediction of microbial
aetiology at admission to hospital for pneumonia from the presenting clinical
features. British Thoracic Society Pneumonia Research Subcommittee. Thorax
44: 1031–1035
2 Bartlett RC (1974) Establishing clinical relevance. In: RC Bartlett (ed): Medical
microbiology: cost and relevance. John Wiley & Sons, New York, NY
3 Sharp SE, Robinson A, Saubolle M, Santa Cruz M, Carrol K, Baselski V (2004)
Lower respiratory tract infections. In: LA Weissfeld (ed): Cumulative technique
and procedures in clinical microbiology. American Society for Microbiology,
Washington, DC
4 Niederman MS, Mandell LA, Anzueto A, Bass JB, Broughton WA, Campbell
GD, Dean N, File T, Fine MJ, Gross PA et al (2001) Guidelines for the manage-
ment of adults with community-acquired pneumonia. Diagnosis, assessment of
severity, antimicrobial therapy, and prevention. Am J Respir Crit Care Med 163:
1730–1754
5 Bartlett JG (2004) Decline in microbial studies for patients with pulmonary
infections. Clin Infect Dis 39: 170–172
6 Bartlett JG, Mundy LM (1995) Community-cquired pneumonia. New Engl J
Med 333: 1618–1624
7 Roson B, Carratala J, Verdaguer R, Dorca J, Manresa F, Gudiol F (2000)
Prospective study of the usefulness of sputum Gram stain in the initial
approach to community-acquired pneumonia requiring hospitalization. Clin
Infect Dis 31: 869–874
8 Kalin M, Lindberg AA, Tunevall G (1983) Etiological diagnosis of bacterial
pneumonia by Gram stain and quantitative culture of expectorates. Leukocytes
or alveolar macrophages as indicators of sample representativity. Scand J Infect
Dis 15: 153–160
9 Barenfanger J, Arakere P, Cruz RD, Imran A, Drake C, Lawhorn J, Verhulst SJ,
Khardori N (2003) Improved outcomes associated with limiting identification
of Candida spp. in respiratory secretions. J Clin Microbiol 41: 5645–5649
10 Musher DM, Montoya R, Wanahita A (2004) Diagnostic value of microscopic
examination of Gram-stained sputum and sputum cultures in patients with
bacteremic pneumococcal pneumonia. Clin Infect Dis 39: 165–169
11 Reed WW, Byrd GS, Gates RH, Howard RS, Weaver MJ (1996) Sputum Gram’s
stain in community-acquired pneumococcal pneumonia. A meta-analysis. West
J Med 165: 197–204
12 Heineman JS, Chawla JK, Lofton WM (1977) Misinformation from sputum
cultures without microscopic examination. J Clin Microbiol 6: 518–527
13 Lidman C, Burman LG, Lagergren A, Ortqvist A (2002) Limited value of
24 Reinhard Marre

routine microbiological diagnostics in patients hospitalized for community-


acquired pneumonia. Scand J Infect Dis 34: 873–879
14 Theerthakarai R, El Halees W, Ismail M, Solis RA, Khan MA (2001) Nonvalue
of the initial microbiological studies in the management of nonsevere commu-
nity-acquired pneumonia. Chest 119: 181–184
15 Gutierrez F, Masia M, Rodriguez JC, Ayelo A, Soldan B, Cebrian L, Mirete C,
Royo G, Hidalgo AM (2003) Evaluation of the immunochromatographic Binax
NOW assay for detection of Streptococcus pneumoniae urinary antigen in a
prospective study of community-acquired pneumonia in Spain. Clin Infect Dis
36: 286–292
16 Ishida T, Hashimoto T, Arita M, Tojo Y, Tachibana H, Jinnai M (2004) A 3-year
prospective study of a urinary antigen-detection test for Streptococcus pneu-
moniae in community-acquired pneumonia: utility and clinical impact on the
reported etiology. J Infect Chemother 10: 359–363
17 Murdoch DR, Laing RTR, Mills GD, Karalus NC, Town GI, Mirrett S, Reller
LB (2001) Evaluation of a rapid immunochromatographic test for detection of
Streptococcus pneumoniae antigen in urine samples from adults with commu-
nity-acquired pneumonia. J Clin Microbiol 39: 3495–3498
18 Navarro D, Garcia-Maet L, Gimeno C, Escribano A, Garcia-de-Lomas J, and
and the Spanish pneumococcal study network (2004) Performance of the binax
NOW Streptococcus pneumoniae urinary antigen assay for diagnosis of pneu-
monia in children with underlying pulmonary diseases in the absence of acute
pneumococcal infection. J Clin Microbiol 42: 4853–4855
19 Murdoch DR, Anderson TP, Beynon KA, Chua A, Fleming AM, Laing RTR,
Town GI, Mills GD, Chambers ST, Jennings LC (2003) Evaluation of a PCR
assay for detection of Streptococcus pneumoniae in respiratory and nonres-
piratory samples from adults with community-acquired pneumonia. J Clin
Microbiol 41: 63–66
20 Campos J, Hernando M, Roman F, Perez-Vazquez M, Aracil B, Oteo J, Lazaro
E, de Abajo F, and the Group of Invasive Haemophilus Infections of the
Autonomous Community of Madrid (2004) Analysis of invasive Haemophilus
influenzae infections after extensive vaccination against H. influenzae Type b. J
Clin Microbiol 42: 524–529
21 Tenover FC, Edelstein PH, Goldstein LC, Sturge JC, Plorde JJ (1986)
Comparison of cross-staining reactions by Pseudomonas spp. and fluores-
cein-labeled polyclonal and monoclonal antibodies directed against Legionella
pneumophila. J Clin Microbiol 23: 647–649
22 Yzerman EPF, Boer JW, Lettinga KD, Schellekens J, Dankert J, Peeters M
(2002) Sensitivity of three urinary antigen tests associated with clinical sever-
ity in a large outbreak of Legionnaires’ disease in the Netherlands. J Clin
Microbiol 40: 3232–3236
23 Guerrero C, Toldos CM, Yague G, Ramirez C, Rodriguez T, Segovia M (2004)
Comparison of diagnostic sensitivities of three assays (Bartels Enzyme
Immunoassay [EIA], Biotest EIA, and Binax NOW Immunochromatographic
Test) for detection of Legionella pneumophila serogroup 1 antigen in urine. J
Clin Microbiol 42: 467–468
24 Reischl U, Linder H, Lehn N, Landt O, Barratt K, Wellinghausen N (2002)
Detection of respiratory bacterial pathogens 25

Direct detection and differentiation of Legionella spp. and Legionella pneu-


mophila in clinical specimens by dual-color real-time PCR and melting curve
analysis. J Clin Microbiol 40: 3814–3817
25 Templeton KE, Scheltinga SA, van den Eeden WC, Graffelman AW, van den
Broek PJ, Claas EC (2005) Improved diagnosis of the etiology of community-
acquired pneumonia with real-time polymerase chain reaction. Clin Infect Dis
41: 345–351
26 Tsolia MN, Psarras S, Bossios A, Audi H, Paldanius M, Gourgiotis D, Kallergi K,
Kafetzis DA, Constantopoulos A, Papadopoulos NG (2004) Etiology of com-
munity-acquired pneumonia in hospitalized school-age children: evidence for
high prevalence of viral infections. Clin Infect Dis 39: 681–686
27 Weigl JA, Puppe W, Grondahl B, Schmitt HJ (2000) Epidemiological inves-
tigation of nine respiratory pathogens in hospitalized children in Germany
using multiplex reverse-transcriptase polymerase chain reaction. Eur J Clin
Microbiol Infect Dis 19: 336–343
28 Wellinghausen N, Straube E, Freidank H, von Baum H, Marre R, Essig A
(2006) Low prevalence of Chlamydia pneumoniae in community-acquired
pneumonia. Int J Med Microbiol; in press
29 Dowell SF, Peeling R, Boman J, Carlone GM, Fields BS, Guarner J,
Hammerschlag MR, Jackson LA, Chou-Chou K, Maass M et al (2001)
Standardizing Chlamydia pneumoniae assays: recommendations from the
Centers for Disease Control and Prevention (USA) and the Laboratory Centre
for Disease Control (Canada). Clin Infect Dis 33: 492–502
30 Apfalter P, Blasi F, Boman J, Gaydos CA, Kundi M, Maass M, Makristathis A,
Meijer A, Nadrchal R, Persson K et al (2001) Multicenter comparison trial of
DNA extraction methods and PCR assays for detection of Chlamydia pneu-
moniae in endarterectomy specimens. J Clin Microbiol 39: 519–524
Community-Acquired Pneumonia 27
ed. by N. Suttorp, T. Welte and R. Marre
© 2007 Birkhäuser Verlag Basel/Switzerland

Viral pathogens and epidemiology, detection, therapy


and resistance

Walter Hampl and Thomas Mertens

Institute for Virology, University Clinic of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany

Abstract
Worldwide community-acquired pneumonia (CAP) is one of the most frequent infectious
diseases and a leading cause of death. Several studies have shown that a pathogen could
be identified only in 50 to 60% of all patients, although in children < 6 month infectious
agents can be detected in about 90%. Viral infections are most frequent in children < 2
years (80%), whereas bacterial infections increase with age.
RSV, influenzaviruses, rhinoviruses, parainfluenzaviruses and adenoviruses are the
most common viruses associated with CAP in children. Among adenoviruses a pre-
dominance of adenovirus 7 has been reported in several countries with emergence of
highly pathogenic variants with significant lethality in young children. Many childhood
respiratory infections are caused by more than one pathogen and up to 30% mixed viral
/ bacterial infections can be observed. CAP in immunocompetent adults is rare, whereas
persons with underlaying diseases have an increased incidence of CAP. In the elderly,
RSV, influenzaviruses, parainfluenzaviruses and less frequent adenoviruses are predomi-
nant viruses causing pneumonia. Less frequently associated with CAP are the newly dis-
covered human metapneumovirus and the coronaviruses NL63 and HKU1. Hantaviruses,
involved in the hantavirus pulmonary syndrome, belong to the emerging pathogens to
date in North, Middle and South America.
For optimum diagnosis the whole spectrum of potential respiratory viral agents
should be included and multiple diagnostic techniques have to be used.
In view of the high relevance of influenzavirus for CAP influenza vaccination is highly
advisable for prevention of CAP, especially in high-risk groups.

Introduction

Viral infections are involved in 10–25% of CAP. Frequently mixed infec-


tions, viral/viral or viral/bacterial, are detected. CAP in infants and young
children is most commonly due to viral infections, with the predominance
of respiratory syncytial virus (RSV). In developing countries the incidence
of viral pneumonia is higher and is a relevant reason for death of young
children [1, 2].
28 Walter Hampl and Thomas Mertens

Viral pathogens and epidemiology

The major viral pathogens are summarized in Table 1. Usually they cause
mild self-limited illness, mainly restricted to the upper respiratory tract. The
leading viral pathogens for severe disease are RSV and influenzaviruses.
In the past two decades some new, mostly zoonotic viral pathogens have
emerged like SARS-coronavirus, avian influenzaviruses and hantaviruses
associated with hantavirus pulmonary syndrome (HPS), which has crossed
species barriers. Numerous other viruses occasionally can cause severe
respiratory disease in the lower respiratory tract, but they are better known
for other clinical manifestations (Tab. 2).
Respiratory virus infections are more common in winter and early
spring, but some cause respiratory illness without a clear seasonal pattern.
The frequency of detection of respiratory viruses varies in different stud-
ies due to methodological problems. Many studies did not include all known
respiratory viruses. Additionally, the prevalence of viruses may vary over
time and between different geographical areas. In immunocompromised
patients exogenous respiratory viruses may persist with prolonged shed-
ding.
Individuals with a subclinical infection may be an undetectable source
of transmission. Only a few studies have investigated respiratory viruses in
asymptomatic humans with very variable results. An age-dependent occur-
rence of asymptomatic respiratory infections has been reported with the
highest frequency of 68% in young children newborn to 4 years.

Factors influencing occurrence of community-acquired pneumonia


(CAP)

Virus-associated CAP occurs mainly in infants and young children, in


elderly and frail adults, in persons with underlying diseases and in immu-
nocompromised patients. Even “harmless” agents like rhino- or coronavi-
ruses frequently induce acute disorders or exacerbations in people with
chronic cardiopulmonary diseases or asthma. The main host and environ-
ment related risk factors have been investigated, but the influence of vari-
ous other factors, which can predispose CAP is controversial.
In general, the viral-associated CAP decreases with age. In several
recent studies in developed countries a pathogen was detected in 79% to
85% in immunocompetent children with CAP; 25% to 62% of the patients
had evidence of a single viral infection, 8% to 11% for viral/viral infection
and 23% to 43% a viral/bacterial infection. Inflammation and disease were
more severe in viral/bacterial infection [3–6]. In nonimmunocompromised
adults the demonstrated viral etiology is less common. Roux et al. [7] iden-
tified a pathogen only in 38% of the cases with a proportion of viral infec-
tions of approximately 20% (single viral infection 9%) with influenzavirus
Viral pathogens and epidemiology, detection, therapy and resistance 29

Table 1. Major respiratory viruses

Virus family / Genus Virus type


Subfamily
Adenoviridae Mastadenovirus Human adenovirus (1 – 7, 14 and 21)
Coronaviridae Human coronavirus (HCoV-OC43, -229E,
-NH, -NL63, -HKU1)
SARS-coronavirus
Orthomyxoviridae Influenzavirus A, B, C
Avian influenzavirus A (e.g. H5N1)
Paramyxoviridae
Paramyxovirinae Respirovirus Parainfluenzavirus 1, 3
Rubulavirus Parainfluenzavirus 2, 4a/b
Pneumovirinae Pneumovirus Respiratory syncytial virus (A, B)
Metapneumovirus Human metapneumovirus (A, B)
Picornaviridae Rhinovirus Human rhinovirus (HRV 1-102)
Enterovirus Human enterovirus (Cox A10, 16, 21, Cox B1-
6, ECHO 4, 9, 11, 25, 32, HEV 68, 69, HRV 87)

Table 2. Other respiratory viruses

Virus family / Genus Virus type


Subfamily
Bunyaviridae Hantavirus e.g. Sin-Nombre virus, Andes virus, Choclo
virus
Paramyxoviridae Morbillivirus Measles virus
Herpesviridae
Alphaherpesvirinae Herpes simplex virus (HSV) (rare)
Varicella-zoster virus (VZV)
Betaherpesvirinae Cytomegalovirus (CMV)
Gammaherpesvirinae Human herpes virus 6 (HHV6)
Epstein-Barr virus (EBV) (very rare)

as the most frequently identified viral agent. In this study chronic heart
failure (CHF) was a risk factor for virus infections.

Viruses causing CAP

Adenovirus (Adv)

The virus is composed of nonenveloped, icosahedral particles containing a


double-stranded linear DNA genome. They are 70–90 nm in diameter and
consist of 252 capsomers (hexons and pentons) with filamentous glycopro-
teins (fibers) on the penton bases, which display characteristic different
lengths. Human adenoviruses have been classified into six species (subgen-
30 Walter Hampl and Thomas Mertens

era) A-F on the basis of antigenicity and other biological properties (hem-
agglutination, tumorogenicity in animals). This is in quite good agreement
with classification based on genomic differences, like DNA homology of
less than 20% between viruses of the different subgenera. Serotype-spe-
cific epitopes are predominantly found on the hexon capsomere and the
terminal part of the fiber, and are defined by the quantitative neutraliza-
tion test. At present, 51 serotypes have been identified by neutralizing
antibodies and nine of these are documented as respiratory pathogens.
Adenoviruses are very resistant, also against proteolytic enzymes in the
intestinal tract.
The species C adenoviruses (1, 2, 5 and 6) are endemic and are respon-
sible for approximately 60% of all human adenovirus infections (Adv 6 only
for 4%) and for more than 80% of the adenovirus infections (most com-
monly Adv 1 and 2) early in life, whereas they cause 15% of symptomatic
lower respiratory tract infections. After primary infection, C viruses may be
shed in feces for months or even years. Following the initial infection the
C viruses establish a lifelong, asymptomatic, persistent infection, with cur-
rently unknown state of viral persistence. Probable places of persistence are
tonsils and adenoids. New data suggest that human mucosal T-lymphocytes
may harbor C adenoviruses in a latent form [8].
Premature infants are at high risk to develop disseminated neonatal
adenovirus infection with pneumonia and high lethality [9]. In infants and
children adenovirus infections primarily occur between 6 month and 5
years and are responsible for 4–10% of childhood pneumonias. Outbreaks
in predominantly healthy children are most frequently associated with type
7, followed by types 3 and 21 [10–12]. For Adv 7-infected children at high
risk the mortality rate is up to 40% and 12% in healthy children [13]. Adv
differ in their ability to induce inflammatory response in lung tissue, but
particularly Adv 7 is involved in severe lung inflammation and neutrophil
infiltration [14].
The incidence of Adv infection may vary and depends on the detection
assays and the study population. In general it is higher in children (21%)
than in adults (9%) [15, 16]. In 50% of the described cases a viral coinfec-
tion was recognized [8, 17]. It is likely that Adv infections were caused also
by reactivation of endogenous virus. Further, these infections are not linked
to seasonal pattern.
Adenovirus epidemics do occur in human populations living in crowded
conditions with poor hygiene. A large outbreak, predominantly caused by
types 4 and 7, was observed between 1950 and 1960 in 10% of the recruits
in the US military. Ninety percent of the Adv-infected persons developed
pneumonia. Vaccination against adv 4 and 7 since 1971 has effectively
reduced illness from these serotypes [18]. Recent epidemiological studies
have shown that latent subgroup C viruses are involved in some chronic dis-
eases in immmunocompetent patients which are at high risk for pneumonia
induced by other viral pathogens [19, 20].
Viral pathogens and epidemiology, detection, therapy and resistance 31

Table 3. Examples of epidemiological data of adenovirus pneumonia

Patients Age / Risk factor Viral Pneumonia Adv sero- Study


no manifestation infections % % type
7588 < 5 years / ARD mostly none ~ 40 0,4 ? [17]

1640 < 5 years / LRTI mostly yes 43–50 4–10 7, 3, 2 [13]


immuno-
deficient
75 5–14 years / None 65 12 ? [21]
CAP

Therapy and resistance

Disseminated adenovirus infections are difficult to treat and the agents


used, ribavirin and cidofovir, yielded variable results. The benefit of intrave-
nous ribavirin (a synthetic guanosine analog) treatment in life-threatening
disease especially in immunocompromised patients, seemed to be better if
antiviral therapy was started early. Other reports could not show a clear
beneficial effect. In vivo as well as in vitro data suggest that susceptibility
to ribavirin is highly dependent on the virus species [22, 23]. Viruses of sub-
group C were shown to be sensitive to ribavirin, whereas serotypes of the
subgroups A, B, D, E and F were resistant.
Despite a high level and long treatment in some patients no changes in
sensitivity of the virus isolates against ribavirin to date have been report-
ed.
Cidofovir is a nucleotide analogue of cytosine with an effective in vitro
activity against different DNA viruses, adenoviruses included. Treatment is
indicated for severe, disseminated Adv infections, but results are varying
and treatment is limited by severe nephrotoxicity. Emergence of resistant
Adv has been observed only in experimental systems.

Human coronavirus (HCoV)

Coronaviruses are spherical, pleomorphic enveloped viruses with a diam-


eter of 80–200 nm. They possess the largest genomes of all RNA viruses.
The single-stranded positive RNA is associated with the nucleoprotein N
forming the helical nucleoprotein complex. It is surrounded by the enve-
lope, which contains three characteristic surface structures, the S protein,
the membrane protein M and the envelope protein E. Oligomeres of the
S protein are formed to spikes on the virion surface and resemble a solar
corona. The S protein determines cell tropism, is responsible for pathoge-
nicity and is the strongest inducer of neutralizing antibodies. Some coro-
32 Walter Hampl and Thomas Mertens

Table 4. Examples of epidemiologial data of coronavirus pneumonia

Patients Age / Risk factor Viral Pneumonia HCoV Study


no manifestation infections % %
501 < 2 to > 65 years mostly none 37 0,4 -OC43 [25]
ARTI
316 < 65 years CHF, COPD ~ 30 0,6 -OC43, [24]
ARTI -229E
418 > 65 years cardio- ? 2,4 -HKU1 [29]
CAP pulmonary

naviruses contain a fourth envelope protein with a hemagglutinating and


esterase activity. Four serogroups have been distinguished containing three
human pathogens: group 1 with the prototype HCoV-229E, group 2 with
the known HCoV-OC43. In 2003 the severe acute respiratory syndrome
(SARS) led to the detection of a novel coronavirus. The consecutive exten-
sive research in this field led to the discovery of further coronaviruses (see
the following).
After rhinoviruses, coronaviruses are most frequently associated with
the “common cold” (15–30%) in young adults, and they seem not to pose a
risk for healthy elderly. Non SARS coronaviruses rarely cause pneumonia.
However, they may cause diseases of the lower respiratory tract (LRT) in
infants, immunocompromised patients, patients with underlying diseases
and in frail older adults. During endemic outbreaks of HCoV-OC43 and
-229E only elderly patients at risk rarely develop pneumonia [24, 25].
HCoV-NL63 (first described in 2004 in the Netherlands), but although
widespread within the human population it is seldom responsible for pneu-
monia in children [26–28]. Recently, a novel coronavirus HCoV-HKU1
from an 71-year-old patient with COPD and pneumonia was described in
Hong Kong [29]. HKU1-associated pneumonias so far described, occur from
winter to spring, predominantly in the elderly (80% were > 65 years) with
comorbidity [30].
In November 2002, a new emerging disease, SARS was described in
China as a contagious, potentially lethal atypical pneumonia. The SARS-
CoV originated from animal viruses, which could be the result of recom-
bination between mammalian and avian coronaviruses. By June 2003,
worldwide 8447 cases of this illness from > 30 countries were registered with
more than 800 deaths (lethality 9.5%). SARS-CoV seems to have a distinct
cell entry pathway. The initial infection is possible with an extreme low
infectious dose resulting in the generation of proteases in the lung, which
are responsible for a 100- to 1000-fold more efficient rate of infection [31].
In younger children SARS often induces a relatively mild and nonspecific
respiratory illness [32, 33]. In a Chinese study the overall mortality rate was
19.7%, but increased to 78.6% in the patient group with serious underlying
diseases [34].
Viral pathogens and epidemiology, detection, therapy and resistance 33

Therapy and resistance

There is no information about antiviral treatment of HcoV infections and


the rare cases of HCoV associated pneumonia. At present also no standard
antiviral therapy can be recommended for SARS. Ribavirin and cortico-
steroids used in severely ill patients seemed to be effective and a better
outcome was reported after combination therapy with lopinavir/ritonavir,
ribavirin plus steroid [35]. Nothing is known about emergence of resistant
virus variants.

Human influenzavirus / avian influenzavirus

Human and avian influenzaviruses are described in separate chapters within


this book. Only the epidemiological association with CAP and the virologi-
cal diagnosis are discussed in this chapter.
Children (1 to 5 years) and adults with comorbidity, predominantly
chronic heart disease and broncho-pulmonary dysplasia, pregnant women
in the second or third trimester and persons > 65 years are at high risk
for influenza virus-associated pneumonia. Using sensitive methods it has
been shown that the prevalence of influenza infections in older children
with CAP may be higher, and mixed infections, mostly viral/bacterial infec-
tions were documented in up to 35% [3, 21]. Influenza A virus-associated
pneumonia in pregnancy is accompanied by higher morbidity and lethality
and infants may be born preterm with low weight. Although pneumonia in
elderly patients may present only with few respiratory signs, recovery is pro-
longed, especially in the frail elderly, where the incidence of pneumonia is
highest [7, 36]. Mixed infections are more common in the older age groups,
but single virus infections are seen increasingly in the patients over 65 years
[37]. Influenza B virus-associated pneumonia is rare and has been reported
in single cases in children [38, 39].

Table 5. Examples of epidemiologial data of influenzavirus pneumonia

Patients no Age / Risk factor Viral Pneumonia Study


manifestation infections % %
514 CAP 10% premature [40]
23% comorbidity
126 0–1 year 42 0,8
241 1–< 5 years 21 5,8
147 5–16 years 6,8 1,4
75 5–14 years none 65 6,7 [21]
CAP
338 > 65 years CHF 18 11,5 [7]
CAP
34 Walter Hampl and Thomas Mertens

Parainfluenza virus (HPIV)

HPIV are pleomorphic enveloped viruses of between 150 and 300 nm.
They contain a helical nucleocapsid with single-stranded negative RNA.
It encodes at least six structural proteins and two nonstructural proteins.
Important are the two envelope glycoproteins HN (hemagglutinin-neur-
aminidase) and F (fusion), responsible for neutralizing antibodies. Within
the family of the paramyxoviruses only HPIV express neuraminidase activ-
ity. Four major HPIV serotypes exist which are divided into subtypes and
genotypes with steadily occurring antigenic variations. Types 1, 2 and 3 are
distributed worldwide, while type 4 is predominantly spread in America.
The four serotypes are distinct in their epidemiological and clinical
behavior. HPIV 3 is endemic throughout the year, but there are yearly epi-
demic outbreaks from winter to spring. HPIV 1 and 2 are associated with a
biennial epidemic pattern with the peak from fall to winter. Parainfluenza-
viruses 1 to 3, particularly type 3, seem to have the highest virulence and the
capacity to persist.
The majority of type 1 infections occur in children between the second
and third year of life, whereas 60% of type 2 are in children younger than
5 years with a peak incidence between the first and second year. They are
most commonly associated with croup or laryngitis, but may also cause
pneumonia. Type 1 can be found in hospitalized, previously healthy adults
and may be involved in bacterial pneumonias.
HPIV-3 infections occur in 40% of young infants in the first year of
life and in most children in the first two years. Pneumonia with type 3 is
seen primarily in the first 6 months of life, similar to RSV, but with lower
frequency [41]. Like RSV HPIV can reinfect both children and adults with
predominantly mild respiratory tract symptoms accompanied by low and
short virus shedding [42, 43].
In childhood CAP parainfluenzavirus 1–3 infections beside RSV and
influenzaviruses are the most common viral pathogens, which are involved
in up to 10% of the diseases [3, 4, 40, 44]. Parainfluenza virus infections
have been reported in the elderly with pneumonia in up to 12% of cases
[43]. Viral CAP in adults with comorbidity like COPD or CHF is character-
ized by a more severe clinical outcome [7, 45]. Viral/bacterial coinfections
(approximately 20%) are observed frequently.

Therapy and resistance

Ribavirin has in vitro activity against HPIV and both aerosolized and intra-
venous ribavirin have been used. There are anecdotal reports about reduc-
tion of clinical signs and viral load in immunocompromised patients, when
Viral pathogens and epidemiology, detection, therapy and resistance 35

treatment was started early after onset symptoms. Ribavirin resistant virus
variants have not been described.

Respiratory syncytial virus (RSV)

RSV is a negative-strand RNA virus. It is pleomorphic and has a size of


150–300 nm in diameter. The genome encodes eight structural and two
nonstructural proteins. The helical capsid is surrounded by an envelope with
three surface glycoproteins, the fusion (F) protein which mediates mem-
brane fusion with the host cell resulting in viral penetration, the G protein
which is responsible for attachment to the host cell, and an SH protein with
unknown function. In contrast to other paramyxoviruses a hemagglutinin
and neuraminidase function do not exist. The G protein has the highest
degree of antigenic diversity in RSV. It accounts for the strain-specific
epitopes and allows the classification into two antigenic groups RSV-A
and RSV-B with multiple genotypes [46]. Immunologically important are
the F and the G glycoproteins inducing protective neutralizing antibodies.
However, immune-protection is not complete and early reinfections occur.
The G glycoprotein is produced as a membrane bound and a secretioned
form. The secretioned form is able to modulate the innate immune response
to RSV and priming with this protein increases the severity of illness after
RSV reinfection.
In children with severe disease caused by RSV the type 2 Th-cell
response seems to be dominant. The G protein induces an unbalanced Th-
cell response, which in the lung could result in airway hyperresponsiveness,
mucus hypersecretionion, and inflammation, and may contribute to periph-
eral blood and pulmonary eosinophilia [47].
RSV infections occur worldwide peaking in the winter months in tem-
perate climates and in the rainy season in tropical climates. A and B viruses
and their multiple variants generally circulate simultaneously within epi-
demical outbreaks. Yearly outbreaks are possible, because the pattern of
the circulating RSV strains changes, depending on the local strain-specific
immunity in the human population [48].
RSV is the most important cause of acute respiratory tract viral infec-
tion in infants. Primary infections are symptomatic with a spectrum of
clinical manifestations from mild upper tract illness to life-threatening
pneumonia. RSV accounts for 50% of all cases of pneumonia during the
first 2 years of life. The peak incidence of RSV lower respiratory tract
infection (LRTI) is between 1 and 6 months of age. Maternal RSV-spe-
cific antibodies rapidly decrease after birth to approximately 6% at 3
months. This may be the reason for the high frequency of RSV infections
before 3 months of age [49]. Premature infants (28 to 32 weeks) are at
risk for 12 to 6 months after birth. All children get infected once until the
36 Walter Hampl and Thomas Mertens

age of 2 years, but 50% of them already had experienced re-infections [4,
21, 40, 50–52].
There are numerous independent risk factors for severe RSV infection
including genetic factors that are under discussion. In immunocompromised
children, RSV-related mortality (with very high RSV load) was 15%, and
for children with primary immunodeficiencies 40% [53].
In immunocompetent adults younger than 60 years RSV reinfections are
generally mild and may contribute to 2–4% of the lower respiratory tract
infections. In elderly people reinfections can induce life-threatening pneu-
monitis. Similar to children there may exist specific risk factors for severe
RSV disease. In different studies in adult and elderly patients 3–10% devel-
oped RSV diseases depending on risk factors. The infections accounted for
11% of hospitalizations for pneumonia [54-56].
The development of a prophylactic vaccine against RSV was pursued
with a high priority. The formalin-inactivated RSV vaccine developed in the
1960s unfortunately led to a more severe lung disease in vaccinated children
after a subsequent RSV challenge [57].

Therapy and resistance

Ribavirin is licensed to treat severe RSV-associated diseases in children.


Immune globulin for intravenous administration and a humanized mono-
clonal antibody preparation (palivizumab (Synagis)) are designed to pre-
vent or reduce the severity of RSV infection. It has been shown that in
RSV-infected infants with lower respiratory tract disease ribavirin aerosol
therapy improves clinical outcome, but may be effective in adults as well.
Despite high dose and prolonged treatment in some patients no ribavirin-
resistant RSV variants have been isolated [58].

Human metapneumovirus (HMPV)

This recently identified paramyxovirus is most closely related to the pneumo-


virus RSV, but HMPV differs from RSV in two aspects: it is lacking the non-
structural proteins (NS1 and NS2) and it has a different gene constellation.
HPMV is classified into two main lineages, A and B, based on sequence analy-
ses of the F gen. Further sequence analysis of different HMPV genes includ-
ing the G gene are required for refined characterization of the virus isolates.
The two major groups with numerous genotypes cocirculate throughout the
year. Sometimes even genetically distinct strains of HMPV are circulating
during the same year. Most infections have been detected during late winter
and early spring following the peak activity of both RSV and influenzavirus.
The virus was first detected in young children in 2001 in the Netherlands
[59], but serological studies showed that the virus has been circulating in
Viral pathogens and epidemiology, detection, therapy and resistance 37

Table 6. Epidemiologial data of metapneumovirus pneumonia

Patients no Age / Risk factor Viral Pneumonia Study


manifestation infections % %
90 0–1 years none 47 10 [61]
ARD
208 < 3 years 10% 78 1 [62]
ARD
145 adults COPD / CHF / 19 2,8 [64]
CAP+EA asthma

humans for at least 50 years. HMPV has a worldwide distribution and asso-
ciation with respiratory illness in all age groups. It causes upper respiratory
tract infections, but is also associated with lower respiratory tract infections.
It has been suggested that most severe HMPV infections occur in children
< 2 years of age and seem to peak in the third and fifth months of life, some-
what later than RSV. It is found less frequently in hospitalized children
than RSV and the clinical course may be milder. Based on the presence of
HMPV antibodies approximately 55% of children at the age of two and
100% at the age of 5–10 years had a HMPV history. HMPV like RSV may
cause clinical important reinfections in late childhood and adult life, but the
highest infection rate was found in young adults.
HMPV also can be responsible for pneumonia in premature born babies
[60] and other persons at risk. Although in the study of Maggi et al. [61]
the number of infants with age less than 2 years is small, the majority of
the HMPV-infected children developed pneumonia. The incidence in this
study was higher (33%) than in other reports, but the difference is related
to the difference in the population of children studied [62, 63]. The rate of
bronchopneumonias was higher in children with isolated HMPV infection
than in children with mixed infections. Surprising in this study [61] was the
detection of HMPV RNA in plasma of 41% of HMPV-infected children,
like was shown also for RSV infections.
HMPV pneumonia clinically cannot be distinguished from RSV and
influenza, but the disease seems to be somewhat less severe and there is a
greater percentage of cases with underlying diseases (25%) compared to
the influenza- or RSV-associated pneumonia (< 10%). Small studies demon-
strated that HMPV is a relatively important viral pathogen, which can also
lead to pneumonia (14%), especially in elderly and adults with underlying
disease like COPD, CHF, or asthma [64].
Additional studies will be needed, especially year-long active surveil-
lance over consecutive years with analysis of more data from future respi-
ratory seasons to fully define the clinical and epidemic impact of HMPV
infections.
38 Walter Hampl and Thomas Mertens

Therapy and resistance

No antiviral agents or antibody preparations are currently available for the


treatment of HMPV infection. It was shown that ribavirin and a polyclonal
intravenous immunoglobulin had equivalent in vitro activity against both
HMPV and RSV. Treatment may be considered for severe HMPV infection
in immunocompromised patients [65]. No reports about resistant HMPV
exist.

Picornaviruses (human rhinovirus (HRV) / human enterovirus


(HEV))

Picornaviruses are nonenveloped particles and with a diameter of 30 nm


they are very small. They consist of a single positive-strand RNA genome
surrounded by an icosahedral capsid composed of 60 protomers. Each pro-
tomer consists of three nonglycosylated surface proteins (VP1 to VP3) and
internal proteins (VP4). In the center of each protomeric unit is a canyon
where antigenic sites and structures can be found binding to receptors of
the target cells. There are over 100 immunologically distinct rhinovirus
serotypes and two species A and B can be distinguished. Rhinoviruses are
acidlabile and differ in membrane receptor recognition. Further, more than
60 enterovirus serotypes exist.
Rhinoviruses are ubiquitous and infections by relatively low infectious
doses occur throughout the year. They are the most important pathogens of
the “common cold,”, in 80–90% of humans during the peak season in late
autumn. Rhinoviruses are able to infect the lower respiratory tract. It has
been shown in experimental infections that a lower respiratory infection
with rhinoviruses (detected in bronchial biopsy samples) during “common
cold” is not unusual.
Picornaviruses are the most frequently detected virus in respiratory tract
infections in the first year of life. In hospitalized infants with acute expira-
tory wheezing illness respiratory picornaviruses (rhino- and enteroviruses)
are found in 42% of the cases. In older children picornaviruses are predomi-
nant with 65% at the age of 1–2 years and with 82% in children older than
3 years [66, 67].
Earlier studies have not included rhinoviruses, but current data sug-
gest that they are rarely involved in pneumonia in infants, young children
and in older adults. Rhinoviruses can cause pneumonia in children of the
age group 0–6 months, but the highest isolation rate was found in children
between 6 and 12 months with the same frequency as RSV [4, 24, 68, 69].
Coinfections of rhinoviruses and bacteria were found in pediatric patients
with CAP in about 10%. The inflammatory response to rhinovirus infec-
tion is strong and several cytokines are related to pneumonia like IL-6,
which may be an important factor in rhinovirus pathogenesis [68, 70]. In
Viral pathogens and epidemiology, detection, therapy and resistance 39

Table 7. Examples of epidemiologial data of picornavirus pneumonia

Patients no Age / Risk factor Viral Pneumonia Study


manifestation infections % %
502 56% 43 3 [69]
ARTI passive smoking
178 0–6 months 46 2,8
184 6–12 months 40 4.9
96 > 12 months 38 1
254 0,1–17 years ? 62 23 [4]
CAP
316 > 65 years CHF, COPD 30 0,5 [24]
ARTI

a recent review about enterovirus-associated respiratory tract diseases


Rotbart et al. [71] reported, that 13% of infected patients presented with
pneumonia. In contrast, Kellner et al. [69] in a prospective study recovered
enteroviruses only sporadically from children with upper respiratory tract
disease. However, case reports exist about fatal pneumonias in congenital
and neonatal echovirus-infected infants. In these rare and sporadic cases
a maternal disease has been reported in 59 to 68% [71–75]. Epidemic out-
breaks of hand, foot, and mouth disease with enterovirus 70/71 in children
have been observed, where after CNS involvement pulmonary edema
appeared [76].
Using modern diagnostic methods it is increasingly recognized, that
rhinovirus and enterovirus infections are the most common reasons for
unnecessary antibiotic therapy.

Therapy and resistance

The most promising antiviral of the so called WIN compounds is pleconaril


with a broad potent anti-EV and anti-RV activity. It binds to hydrophobic
sites in the base of the capsid canyons and inhibits uncoating of the capsid in
all enteroviruses. Rhinoviruses of the species B, have a significant reduced
susceptibility to pleconaril. For therapeutic application it will be important
to differentiate between natural occurring resistance to pleconaril in B rhi-
noviruses and the emergence of RV resistance under pleconaril treatment
[77].
Pleconaril has been shown to be effective in experimental studies and
to significantly reduce clinical symptoms in treated adult volunteers. It is
available for life threatening enterovirus infections in immunocompromised
patients. Ten percent of the enterovirus isolates have been shown to be
resistant to pleconaril and some patients did not respond (resistant virus
has been identified). Ruprintrivir is an inhibitor of the virally encoded 3C
40 Walter Hampl and Thomas Mertens

protease, which is still under investigation for treatment of rhinovirus infec-


tion in immunocompetent patients [78].

Hantavirus (HV)

Hantaviruses are enveloped, predominantly negative-strand RNA viruses


(ambisense). The genome of these viruses consists of three different single-
stranded RNA segments. The segments L, M and S encode the viral poly-
merase, a glycoprotein processed into G1 and G2 glycoproteins located in the
envelope, and a nucleocapsidprotein. Virus particles are of spherical shape
with a diameter of 80–120 nm, but also elongated forms are seen (170 nm).
Hantaviruses are important zoonotic pathogens, primarily of rodents.
The infections are per-sistent and most of them seem to be asymptomatic
in the natural rodent hosts. Antibodies against hantaviruses are also present
in nonnatural hosts, other wild and domestic animals. Beside the transmis-
sion to humans through human/rodent contacts, infections are acquired by
inhalation of virus-contaminated aerosols of rodent excreta (saliva, urine or
feces) [79]. An occasional transmission may occur from person to person as
was documented for Andes virus in Argentina.
Some hantaviruses belong to the so called emerging pathogens. They
cause an influenza-like acute pulmonary disease in North, Middle and
South America, the hantavirus pulmonary syndrome (HPS). The first HPS
was recognized in 1993 in the USA and was caused by a number of hanta-
virus variants, e.g. Sin Nombre virus (SNV). Symptoms of HPS may vary,
depending on the virus genotype. In 1995, cases of HPS were reported in
South America and a new strain – the Andes hantavirus – was identified. In
late 1999 and early 2000 first outbreaks through an again novel hantavirus,
the choclo virus, were documented in Central America. The overall mortal-
ity was about 44%, but is declining as a consequence of better recogniz-
ing less severe forms of the infections and a better medical management.
Confirmed cases of HPS so far include children, but in the majority of cases
adults (19 to 58 years). SNV accounts for a small number of pediatric cases
in the US, whereas Andes virus was found in pediatric patients (Chile, South
Argentina) in a higher proportion (16%) [80–82].
The incubation period for Andes virus in Chile was 5 to 25 days. Febrile
prodromi last for approximately 4 days followed by a rapid progression to
moderate to severe respiratory distress.

Therapy and resistance

For hantavirus diseases no established specific therapy is currently available.


Ribavirin shows activity against HV, but this has not been well documented
in studies. Emergence of resistant HV is unknown.
Viral pathogens and epidemiology, detection, therapy and resistance 41

Nonconventional respiratory viruses

Measles virus

In developing countries the frequent complications of measles virus infec-


tion are responsible for the mortality rate. Measles virus infection causes
a transient and strong immunosuppression, which is the reason for the
increased susceptibility to other viral or bacterial infections. In 3-4% of the
infected patients measles virus can cause pneumonia, which can be pres-
ent either as primary measles virus pneumonia or as atypical measles virus
pneumonia, but most patients develop secondary bacterial pneumonia.
Measles pneumonia rarely occurs in young adults as shown in 1976 to 1979,
when a measles outbreak in US Air Force recruits was responsible for 106
cases with pneumonia (3.3%), of which two-thirds had a secondary bacterial
infection [83]. During pregnancy measles virus infections induce a higher
number of pneumonias in the mother [84, 85]. There are a few reports about
preterm and newborn infants in which measles pneumonia was observed
[86].

Herpes viruses

After primary infection herpes viruses are able to persist lifelong in their
human host, typically as latent infection, but they also reactivate under
immunosuppression and then some of them, like CMV, HSV, VZV, EBV
and HHV6, may induce pneumonia.
HSV-1 pneumonia may occur occasionally in high-risk persons. Rarely
disseminated HSV infections during pregnancy have been reported, as in
the case of a previously healthy womn with a fatal progressive HSV-2 pneu-
monia in the third trimester of pregnancy [87]. Ramsey et al. reported 20
patients with pneumonia, in which they could differentiate between cases
with focal HSV pneumonia as a result of HSV spreading to the lung paren-
chyma and cases with interstitial pneumonia as a result of a hematogenous
dissemination of HSV [88].
In immunocompetent adults primary VZV infection is uncommon, but
the incidence is increasing (5-10%) and VZV pneumonia is the main com-
plication (incidence 5.5%–16.5%) with high mortality. Some patients may
develop secondary bacterial pneumonia. Most patients (76.7%) do have
at least one known risk factor like pregnancy, smoking or chronic obstruc-
tive pulmonary disease [84, 89–91]. VZV beside influenzavirus is the most
common pathogen, causing pneumonia during pregnancy, predominantly in
the second and third trimester. The risk of primary VZV infection for VZV
pneumonia during pregnancy (0.1–18.3%) is higher if patients are smokers
or manifest multiple (> 100) skin lesions. The mortality rate before a possi-
ble antiviral intervention was significantly higher in pregnancy (41% versus
42 Walter Hampl and Thomas Mertens

1.5–12.1%) [89, 92, 93]. VZV pneumonias as consequence of a primary VZV


infection in childhood are rare [94].
CMV-infected preterm infants can develop a chronic lung disease, which
may be associated with CMV pneumonia with high mortality. Ganciclovir
treatment has been shown to rapidly improve symptoms, a fact that sup-
ports CMV causality of pneumonia [95–97]. Extremely rare are reports
about CMV pneumonia in previously healthy adults [98]. In patients with
lymphoma CMV pneumonia is less common and the incidence after chemo-
therapy and corticosteroid application is approximately 1% with a mortality
rate of 30% [99].
Two genetically distinct variants HHV-6A and -6B do exist, but in
primary infection the variant -6B is dominant. During primary infection
a severe respiratory disease is an extremely rare complication. In a case
report from Knox et al. [100] a fatal pneumonitis due to HHV-6 infection
is documented in an infant with severe T lymphocytopenia. Whereas for
immunocompromised patients (BMT) HHV-6 pneumonia is documented,
in immunocompetent patients an etiologic role of the reactivated HHV-6
infection in pneumonia is not clearly defined [101, 102]. Another report of
an extremely rare case with HHV-6-associated pneumonia is documented
by Merk et al. [103], where an apparently immunocompetent young women
developed a fatal pneumonia.
A mild asymptomatic pneumonitis is described in 5–10% of the cases
with infectious mononucleosis, but a severe pneumonitis as result of pri-
mary EBV infection is rare in immunocompetent patients [84, 104, 105].
In patients with lymphocytic interstitial pneumonia EBV DNA has been
found in lung tissues of infants, children and of adults as well [106–108].
In autopsy cases with diffuse interstitial pneumonia EBV DNA could be
detected in leukocytes and pneumocytes and frequently in airway epithe-
lial cells [109]. The EBV-associated lymphocytic interstitial pneumonia
without HIV infection has been reported predominantly in adults. This
is also true for a rare chronic interstitial lung disease due to EBV, mainly
seen in adults, but also shown in two infants in early months of life [106,
110].

Diagnosis of viral pulmonary infection

Especially in adult patients respiratory virus infections are extremely under-


diagnosed since adequate and possible virological diagnosis is not routinely
performed.
For diagnosis of acute respiratory infections the detection of infec-
tious viruses or viral components (proteins, nucleic acid) is the method
of choice, whereas antibody detection is not relevant and should only be
used in selected patients at later timepoints after infection using paired
sera.
Viral pathogens and epidemiology, detection, therapy and resistance 43

Specimen

Nasopharyngeal aspirate (NPA) is the gold standard for the detection of all
major respiratory viruses, predominantly adopted in infants and children.
NPA is taken by suction of cell-containing mucosal secretion from the naso-
pharyngeal area. Approximately 0.5 ml fluid should be collected into 2 ml
of viral transport medium.
Nasal lavage can be obtained with less discomfort for the patient by gently
instilling 2 ml PBS at room temperature into each nostril and by simultane-
ously suctioning into a sterile trap; the sensitivity for virus detection is compa-
rable to that of NPA, but is controversially reported for RSV [111, 112].
Nasopharyngeal swab, taken usually from adults, is obtained by deep
bilateral nasal and posterior pharynx swabs with sterile cotton swabs, which
should then be placed in 3 ml viral transport medium (VTM) [113]. Calcium
alginate swabs or swabs with wooden sticks may contain substances which
inactivate some viruses and inhibit PCR testing and should not be used!
Tracheal aspirate can be collected from intubated patients after instil-
lation of 4–10 ml sterile normal saline into the endotracheal tube and by
suctioning into a sterile trap; this is the most easily obtained specimen from
lower respiratory tract in these patients [114].
Induced sputum should be collected in the morning after rinsing mouth
and throat with sterile hypertonic saline. Thereafter sputum has to be col-
lected in a sterile container. Induced sputum represents a more complete
sampling of the respiratory tract and is comparable with nasopharyngeal
aspirate specimens. While induced sputum might be used for virus detection
from the lower respiratory tract, the obligate admixture of saliva with the
accompanied flora is an obvious disadvantage. Nevertheless, with minimal
saliva contamination it is a valuable alternative material to BAL [115, 116].
Bronchioalveolare lavage (BAL) has a high diagnostic yield for infec-
tious pathogens predominantly in immunocompromised patients with lung
infiltrates.
Transbronchial biopsy (TBB) / lung biopsy. This invasive technique can
in addition to BAL significantly improve the diagnostic yield [117].
EDTA blood in immunosuppressed patients may facilitate diagnosis of
some viruses (e.g. adenovirus, CMV) and disseminated virus infections and
determination of the virus load may be predictive for disease and outcome;
furthermore, virus load is important for monitoring of therapy.

Methods for detection of viruses

Direct virus detection (rapid antigen detection / nucleic acid assay)

Enzyme/immunoassays (EIA) for rapid viral antigen detection in airway


secretions exist for almost all major respiratory viruses, but have been often
44 Walter Hampl and Thomas Mertens

evaluated only for children and may vary in sensitivity and specificity (range
between 60 and 95%). Results from EIAs for detection of single respira-
tory viruses are usually available between 10 min to 3 h. Alternatively a
screening for respiratory viruses in an airway secretion can be achieved by
indirect immunofluorescent antigen assay (IFA) (sensitivity 85–95%; speci-
ficity 95–99%) using a pool of monoclonal antibodies, whose specificity is
directed against RSV, influenzaviruses A, B, parainfluenzaviruses 1, 2, 3 and
adenovirus. The examination of several cell spots of a cytospin preparation
on slides with pooled and single antisera allows viral antigen detection and
typing within 2 h. The results are strongly dependent on the quality of the
clinical material. For elderly hospitalized and immunocompromised patients
with virus associated LRTI the rapid virusantigen detection is insensitive.
When rapid conventional methods are not available, like for coronavi-
ruses, metapneumoviruses, rhinoviruses or enteroviruses, the PCR has to be
established.

PCR

The PCR may be used alternatively to the mentioned antigen detection


assays. It offers a great sensitivity and can be used for a wider range of viral
pathogens. In principle PCR can detect and differentiate several viruses
simultaneously in a single reaction mixture as multiple RT-PCR (multiplex
PCR), but it provides a lower sensitivity than PCR for single pathogens and
is more difficult to establish. For early detection and monitoring of viral
infections the quantitative PCR represents a sensitive technique that deliv-
ers results within 3 h.
For diagnosis of disseminated virus infections with pulmonary manifes-
tations frequently caused by adenoviruses, RSV in infants or nonconven-
tional respiratory viruses, like CMV additional analysis of plasma or EDTA
blood is helpful.

Detection of infectivity (short term culture / isolation)

Clinical specimen namely secretions of the respiratory tract can be used for
an infectivity assay in centrifuge enhanced shell vial culture (SVC). After
inoculation of airway specimens on different cell systems, which are sus-
ceptible for many of the respiratory viruses, cell cultures are evaluated for
early virus infection after 24–72 h with a pool of virus-specific monoclonal
antibodies and afterwards for typing with the respective individual mono-
clonal antibodies by IFA.
Once isolated, the virus must be typed again preferentially with IFA
using virus-specific monoclonal antibodies or otherwise increasingly by
PCR.
Viral pathogens and epidemiology, detection, therapy and resistance 45

Conventional cell culture with virus isolation has no impact on clinical


decision and management of patients during hospitalization. Virus isola-
tion is useful for a characterization of virus strains involved in epidemic
outbreaks.

Antibody detection

Serology is not useful during the acute phase of infection. Due to the
delayed onset of the antibody response, approximately at the end of the
first week of disease serological methods can detect antibodies. To use
only serological methods for diagnosis of viral infections in CAP is not at
all sufficient. Paired serum samples may be used to detect seroconversion
or a fourfold increase in antibody titer comparing the first and the second
serum.
Serology in addition is of limited value in newborns with maternal anti-
bodies, in immunocompromised patients, in the elderly and patients receiv-
ing blood products. Antibody detection can be done with the complement
fixation test (CFT, increasingly obsolete), ELISA-IgG, -IgA, -IgM, immuno-
fluorescent assays, immunoblot and neutralization test.

Diagnostic comments on specific viruses

Adenovirus

Viruses can be detected in 25 to 72% of patients with disseminated infection


in peripheral blood for more than 3 weeks. Detection of virus from multiple
sites correlates with more severe disease and higher mortality. It is essential
to monitor viral load during antiviral therapy. Phenotyping of isolates can
be done by neutralization test, by hemagglutination test or by serotype-spe-
cific monoclonal antibodies and finally by PCR.
Group-specific antigens are used in CFT for antibody detection in the
second week of disease. CFT is insufficient to detect antibodies in infants.
IgG antibodies are detectable by ELISA at the end of the second week,
whereas IgM antibodies are not detectable in all cases with primary infec-
tion [10].

Coronavirus

HCoVs antigen can be detected by IFT using rabbit antisera (no monoclo-
nals available). Coronaviruses RNA can be detected with RT-PCR also in
stool. SARS-CoV can be detected in plasma, but in urine and stool longer
than 4 weeks. Virus load is unusually low in the early phase of SARS. A
46 Walter Hampl and Thomas Mertens

chip-based test detecting 10 respiratory viral pathogens including coronavi-


ruses has been developed. Non-SARS human coronaviruses are difficult to
isolate. With SARS-HCoV no IgG and IgM antibodies can be found within
the first 7 days of disease, seroconversion may be delayed up to 8 weeks and
IgG does not persist [34, 118, 119].

Influenzavirus

Rapid antigen detection tests are available (15 min) which differentiate
between A and B viruses and some now include avian influenza virus H5N1.
Sensitivity and specificity range from 60–95% and 52–99% respectively. RT-
PCR is much more sensitive and allows virus typing. Virus isolation is still
important for characterization of circulating influenza strains.
Antibody detection with ELISA-IgG, -IgA and IFA-IgG, -IgA is pos-
sible, but shows low levels of both IgG and IgA, a delayed peak antibody
titer and shorter persistence of antibodies in elderly patients.

Parainfluenzavirus and respiratory syncytial virus

In adults and elderly people virus shedding is lower and shorter. RT-PCR
is more sensitive in patients with parainfluenzavirus and RSV infections,
particularly reinfections, and it might be useful especially for rapid diagnosis
in elderly.
For rapid RSV antigen detection in pediatric patients different antigen
detection assays (20 min) with sensitivities between 61 to 92% and specifici-
ties between 93 to 98% are available [120]. Alternatively the rapid immuno-
fluorescent antigen assay can be used.
Since RSV is thermolabile, it is important to use a qualified transport
medium, and to have a cooled and short transport until processing (30
min). The shell vial culture assay shows the highest sensitivity (94.3%) and
specificity (96.9%) for detecting RSV from NPA in children younger than
1 year.
Only 41% of RSV-infected children could be identified by serology and
antibodies titers that develop are usually low, although both serum and
secretory antibodies are produced [121].

Human metapneumovirus

Commercial monoclonal antibodies are available for IFA. The sensitivity


and specificity of this test is 73.3 and 97% respectively and results agree
with RT-PCR in 89.6% [122].
Viral pathogens and epidemiology, detection, therapy and resistance 47

Isolation of HMPV is difficult and regardless of CPE the use of RT-PCR


to enhance the HMPV identification in cell culture is indicated.
No commercial tests are available for antibody detection.

Picornaviruses (rhinovirus / enterovirus)

Due to more than 100 distinct serotypes a rapid detection of rhinovirus


capsid antigen cannot be developed. The application of molecular assays has
markedly increased the detection rate of picornaviruses in acute respira-
tory infections. Rhinovirus culture is the “gold standard,” but it takes 3 to
7 days. The sensitivity of PCR is superior to the infectivity detection in the
cell culture.
No reliable typespecific serological test for rhinovirus infections exists.
Entervoirus serology by neutralization tests is possible but difficult and not
completely reliable.

Hantavirus

Currently no antigen detection assay is available. RT-PCR for hantavirus


RNA detection is useful to detect infection and to identify the viral geno-
type. RT-PCR for viral RNA detection can be done from whole blood or
serum during the acute phase in the first 10 days of illness.
Virus isolation is inefficient and there is no routine assay.
Serum specimens were tested for IgM and IgG antibodies by ELISA
using Sin Nombre virus antigen, according to the guidelines of the US
Centers for Disease Control (CDC). Positive results indicate infections with
new world hantaviruses. IgM antibodies can be detected in all acute cases,
maximal IgG level occurs during the first week of illness and are detectable
after disease for a relatively long period.

HSV, VZV and CMV

Since HSV, VZV and CMV pneumonia can be effectively treated it is


important to rapidly investigate specimens from the lower respiratory
tract, because pneumonia can only be established on the basis of BAL or
lung tissue examined by PCR (HSV, VZV) and/or shell vial culture (HSV).
Serologic tests are not relevant in case of HSV reactivation.
Primary VZV infection can be confirmed by IgM detection and by IgG
seroconversion.
In premature infants with risk for CMV pneumonia CMV DNA can be
found directly by PCR in urine and in the pharynx. Virus detection in air-
48 Walter Hampl and Thomas Mertens

way specimens (virus load in BAL) and lung tissue will be positive before
seronconversion.

Summary

In various studies and case reports it has been shown that respiratory viral
pathogens are frequently involved in community-acquired pneumonia. Age
groups at risk are the very young and elderly, as well as persons with co-
morbidity, where immune response is restricted.
The frequency of detection of respiratory viruses varies in different stud-
ies due to methodological problems, patient selection and the fact that only
detection of specific viruses was performed. With modern diagnostic tools
it has been increasingly realized that rhinovirus and enterovirus infections
are the most common reasons for unnecessary antibiotic therapy, often also
adenovirus infections. Future studies have to consider the whole range of
viral agents that can be involved in community-aquired pneumonia.
In recent decades several new respiratory viruses have been described
and it is reasonable to assume that new viruses or virus types from differ-
ent virus families will emerge in the future, often after having crossed a
host species barrier with high pathogenic potential for severe pulmonary
diseases.
Most of the relevant infections can be diagnosed by virus detection.
Several studies in children have documented a significant proportion
(25 to 30%) of viral/bacterial infections beside isolated virus infections.
Children with isolated virus infection tended to be younger than coinfected
children. The implications of coinfecting agents in epidemiology, pathoge-
nicity and clinical outcome have to be elucidated.

References
1 Welte T, Marre R, Suttorp N (2004) Competence network “community acquired
pneumonia” (CAPNETZ). A first interim report. Internist (Berl) 393–401
2 Shah PB, Giudice JC, Griesback R Jr, Morley TF, Vasoya A (2004) The newer
guide-lines for the management of community-acquired pneumonia. J Am
Osteopath Assoc 104: 521–526
3 Michelow IC, Olsen K, Lozano J, Rollins NK, Duffy LB, Ziegler T, Kauppila J,
Leinonen M, McCracken GH, Jr (2004) Epidemiology and clinical characteris-
tics of community-acquired pneumonia in hospitalized children. Pediatrics 113:
701–707
4 Juven T, Mertsola J, Waris M, Leinonen M, Meurman O, Roivainen M, Eskola
J, Saikku P, Ruuskanen O (2000) Etiology of community-acquired pneumonia
in 254 hospitalized children. Pediatr Infect Dis J 19: 293–298
5 Juven T, Mertsola J, Waris M, Leinonen M, Ruuskanen O (2004) Clinical
Viral pathogens and epidemiology, detection, therapy and resistance 49

response to antibiotic therapy for community-acquired pneumonia. Eur J


Pediatr 163: 140–144
6 Virkki R, Juven T, Mertsola J, Ruuskanen O (2005) Radiographic follow-up of
pneumonia in children. Pediatr Pulmonol 40: 223–227
7 de Roux A., Marcos MA, Garcia E, Mensa J, Ewig S, Lode H, Torres A (2004)
Viral community-acquired pneumonia in nonimmunocompromised adults.
Chest 125: 1343–1351
8 Garnett CT, Erdman D, Xu W, Gooding LR (2002) Prevalence and quantita-
tion of species C adenovirus DNA in human mucosal lymphocytes. J Virol 76:
10608–10616
9 Rieger-Fackeldey E, Aumeier S, Genzel-Boroviczeny O (2000) Disseminated
adenovirus infection in two premature infants. Infection 28: 237–239
10 Aberle SW, Aberle JH, Steininger C, Matthes-Martin S, Pracher E, Popow-
Kraupp T (2003) Adenovirus DNA in serum of children hospitalized due to an
acute respiratory adenovirus infection. J Infect Dis 187: 311–314
11 Carballal G, Videla C, Misirlian A, Requeijo PV, Aguilar MC (2002) Adenovirus
type 7 associated with severe and fatal acute lower respiratory infections in
Argentine children. BMC Pediatr 2: 6
12 Chuang YY, Chiu CH, Wong KS, Huang JG, Huang YC, Chang LY, Lin TY
(2003) Severe adenovirus infection in children. J Microbiol Immunol Infect 36:
37–40
13 Hong JY, Lee HJ, Piedra PA, Choi EH, Park KH, Koh YY, Kim WS (2001)
Lower respiratory tract infections due to adenovirus in hospitalized Korean
children: epidemiology, clinical features, and prognosis. Clin Infect Dis 32:
1423–1429
14 Booth JL, Coggeshall KM, Gordon BE, Metcalf JP (2004) Adenovirus type 7
induces interleukin–8 in a lung slice model and requires activation of Erk. J
Virol 78: 4156–4164
15 Leruez-Ville M, Minard V, Lacaille F, Buzyn A, Abachin E, Blanche S, Freymuth
F, Rouzioux C (2004) Real-time blood plasma polymerase chain reaction for
management of disseminated adenovirus infection. Clin Infect Dis 38: 45–52
16 Howard DS, Phillips II GL, Reece DE, Munn RK, Henslee-Downey J, Pittard
M, Barker M, Pomeroy C (1999) Adenovirus infections in hematopoietic stem
cell transplant recipients. Clin Infect Dis 29: 1494–1501
17 Rocholl C, Gerber K, Daly J, Pavia AT, Byington CL (2004) Adenoviral infec-
tions in children: the impact of rapid diagnosis. Pediatrics 113: e51-e56
18 Gray GC, Goswami PR, Malasig MD, Hawksworth AW, Trump DH, Ryan MA,
Schnurr DP (2000) Adult adenovirus infections: loss of orphaned vaccines pre-
cipitates military respiratory disease epidemics. Clin Infect Dis 31: 663–670
19 Hayashi S (2002) Latent adenovirus infection in COPD. Chest 121: 183S–187S
20 Fujii T, Hogg JC, Keicho N, Vincent R, Van Eeden SF, Hayashi S (2003)
Adenoviral E1A modulates inflammatory mediator expression by lung epi-
thelial cells exposed to PM10. Am J Physiol Lung Cell Mol Physiol 284: L290-
L297
21 Tsolia MN, Psarras S, Bossios A, Audi H, Paldanius M, Gourgiotis D, Kallergi K,
Kafetzis DA, Constantopoulos A, Papadopoulos NG (2004) Etiology of com-
50 Walter Hampl and Thomas Mertens

munity-acquired pneumonia in hospitalized school-age children: evidence for


high prevalence of viral infections. Clin Infect Dis 39: 681–686
22 Morfin F, Dupuis-Girod S, Mundweiler S, Falcon D, Carrington D, Sedlacek P,
Bierings M, Cetkovsky P, Kroes AC, van Tol MJ, Thouvenot D (2005) In vitro
susceptibility of adenovirus to antiviral drugs is species-dependent. Antivir
Ther 10: 225–229
23 Lankester AC, Heemskerk B, Claas EC, Schilham MW, Beersma MF, Bredius
RG, van Tol MJ, Kroes AC (2004) Effect of ribavirin on the plasma viral DNA
load in patients with disseminating adenovirus infection. Clin Infect Dis 38:
1521–1525
24 Falsey AR, Walsh EE, Hayden FG (2002) Rhinovirus and coronavirus infection-
associated hospitalizations among older adults. J Infect Dis 185: 1338–1341
25 Vabret A, Mourez T, Gouarin S, Petitjean J, Freymuth F (2003) An outbreak of
coronavirus OC43 respiratory infection in Normandy, France. Clin Infect Dis
36: 985–989
26 Vabret A, Mourez T, Dina J, van der HL, Gouarin S, Petitjean J, Brouard J,
Freymuth F (2005) Human coronavirus NL63, France. Emerg Infect Dis 11:
1225–1229
27 Moes E, Vijgen L, Keyaerts E, Zlateva K, Li S, Maes P, Pyrc K, Berkhout B,
van der HL, Van RM (2005) A novel pancoronavirus RT-PCR assay: frequent
detection of human coronavirus NL63 in children hospitalized with respiratory
tract infections in Belgium. BMC Infect Dis 5: 6
28 van der HL, Pyrc K, Jebbink MF, Vermeulen-Oost W, Berkhout RJ, Wolthers
KC, Wertheim-van Dillen PM, Kaandorp J, Spaargaren J, Berkhout B (2004)
Identification of a new human coronavirus. Nat Med 10: 368–373
29 Woo PC, Lau SK, Chu CM, Chan KH, Tsoi HW, Huang Y, Wong BH, Poon
RW, Cai JJ, Luk WK, Poon LL, Wong SS, Guan Y, Peiris JS, Yuen KY (2005)
Characterization and complete genome sequence of a novel coronavirus, coro-
navirus HKU1, from patients with pneumonia. J Virol 79: 884–895
30 Woo PC, Lau SK, Tsoi HW, Huang Y, Poon RW, Chu CM, Lee RA, Luk WK,
Wong GK, Wong BH, Cheng VC, Tang BS, Wu AK, Yung RW, Chen H, Guan
Y, Chan KH, Yuen KY (2005) Clinical and molecular epidemiological features
of coronavirus HKU1-associated community-acquired pneumonia. J Infect Dis
192: 1898–1907
31 Matsuyama S, Ujike M, Morikawa S, Tashiro M, Taguchi F (2005) Protease-
mediated enhancement of severe acute respiratory syndrome coronavirus
infection. Proc Natl Acad Sci USA 102: 12543–12547
32 Bitnun A, Allen U, Heurter H, King SM, Opavsky MA, Ford-Jones EL, Matlow
A, Kitai I, Tellier R, Richardson S, Manson D, Babyn P, Read S (2003) Children
hospitalized with severe acute respiratory syndrome-related illness in Toronto.
Pediatrics 112: e261
33 Cheng FW, Ng PC, Chiu WK, Chu WC, Li AM, Lo KL, Hon EK, Nelson EA,
Leung TF, Ng WH, Wong E, Ip P, Fok TF (2005) A case-control study of SARS
versus community-acquired pneumonia. Arch Dis Child 90: 747–749
34 Mo HY, Xu J, Ren XL, Zeng GQ, Tan YX, Chen RC, Chan-Yeung M, Zhong
NS (2005) Evaluation by indirect immunofluorescent assay and enzyme linked
immunosorbent assay of the dynamic changes of serum antibody responses
Viral pathogens and epidemiology, detection, therapy and resistance 51

against severe acute respiratory syndrome coronavirus. Chin Med J (Engl) 118:
446–450
35 Cheng VC, Tang BS, Wu AK, Chu CM, Yuen KY (2004) Medical treatment
of viral pneumonia including SARS in immunocompetent adult. J Infect 49:
262–273
36 Marrie TJ (2000) Community-acquired pneumonia in the elderly. Clin Infect
Dis 31: 1066–1078
37 van Gageldonk-Lafeber AB, Heijnen ML, Bartelds AI, Peters MF, van der Plas
SM, Wilbrink B (2005) A case-control study of acute respiratory tract infection
in general practice patients in The Netherlands. Clin Infect Dis 41: 490–497
38 Lu KC, Chen PY, Huang FL, Yu HW, Kao CH, Lau YJ (2004) Influenza B virus
associated pneumonia: report of one case. Acta Paediatr Taiwan 45: 242–245
39 van den Dungen FA, van Furth AM, Fetter WP, Zaaijer HL, van Elburg RM
(2001) Fatal case of influenza B virus pneumonia in a preterm neonate. Pediatr
Infect Dis J 20: 82–84
40 Weigl JA, Puppe W, Belke O, Neususs J, Bagci F, Schmitt HJ (2005) Population-
based incidence of severe pneumonia in children in Kiel, Germany. Klin Padiatr
217: 211–219
41 Hall CB (2001) Respiratory syncytial virus and parainfluenzavirus. N Engl J
Med 344: 1917–1928
42 Henrickson KJ (2003) Parainfluenzaviruses. Clin Microbiol Rev 16: 242–264
43 Treanor J, Falsey A (1999) Respiratory viral infections in the elderly. Antivir
Res 44: 79–102
44 Farha T, Thomson AH (2005) The burden of pneumonia in children in the
developed world. Paediatr Respir Rev 6: 76–82
45 Lieberman D, Lieberman D, Gelfer Y, Varshavsky R, Dvoskin B, Leinonen M,
Friedman MG (2002) Pneumonic vs nonpneumonic acute exacerbations of
COPD. Chest 122: 1264–1270
46 Sato M, Saito R, Sakai T, Sano Y, Nishikawa M, Sasaki A, Shobugawa Y, Gejyo
F, Suzuki H (2005) Molecular epidemiology of respiratory syncytial virus infec-
tions among children with acute respiratory symptoms in a community over
three seasons. J Clin Microbiol 43: 36–40
47 Elliott MB, Pryharski KS, Yu Q, Boutilier LA, Campeol N, Melville K, Laughlin
TS, Gupta CK, Lerch RA, Randolph VB, LaPierre NA, Dack KM, Hancock
GE (2004) Characterization of recombinant respiratory syncytial viruses with
the region responsible for type 2 T-cell responses and pulmonary eosinophilia
deleted from the attachment (G) protein. J Virol 78: 8446–8454
48 Peret TC, Hall CB, Schnabel KC, Golub JA, Anderson LJ (1998) Circulation
patterns of genetically distinct group A and B strains of human respiratory
syncytial virus in a community. J Gen Virol 79 ( Pt 9): 2221–2229
49 Hacimustafaoglu M, Celebi S, Aynaci E, Sinirtas M, Koksal N, Kucukerdogan
A, Ercan I, Goral G, Ildirim I (2004) The progression of maternal RSV antibod-
ies in the offspring. Arch Dis Child 89: 52–53
50 Kaneko M, Watanabe J, Ueno E, Hida M, Sone T (2001) Risk factors for severe
respiratory syncytial virus-associated lower respiratory tract infection in chil-
dren. Pediatr Int 43: 489–492
51 Virkki R, Juven T, Rikalainen H, Svedstrom E, Mertsola J, Ruuskanen O
52 Walter Hampl and Thomas Mertens

(2002) Differentiation of bacterial and viral pneumonia in children. Thorax 57:


438–441
52 Openshaw PJ, Tregoning JS (2005) Immune responses and disease enhance-
ment during respiratory syncytial virus infection. Clin Microbiol Rev 18:
541–555
53 Schmidt AC, Couch RB, Galasso GJ, Hayden FG, Mills J, Murphy BR, Chanock
RM (2001) Current research on respiratory viral infections: third international
symposium. Antivir Res 50: 157–196
54 Falsey AR, Hennessey PA, Formica MA, Cox C, Walsh EE (2005) Respiratory
syncytial virus infection in elderly and high-risk adults. N Engl J Med 352:
1749–1759
55 Falsey AR, Walsh EE (2005) Respiratory syncytial virus infection in elderly
adults. Drugs Aging 22: 577–587
56 Falsey AR, Walsh EE (2000) Respiratory syncytial virus infection in adults. Clin
Microbiol Rev 13: 371–384
57 Kapikian AZ, Mitchell RH, Chanock RM, Shvedoff RA, Stewart CE (1969) An
epidemiologic study of altered clinical reactivity to respiratory syncytial (RS)
virus infection in children previously vaccinated with an inactivated RS virus
vaccine. Am J Epidemiol 89: 405–421
58 Hall CB, McBride JT, Gala CL, Hildreth SW, Schnabel KC (1985) Ribavirin
treatment of respiratory syncytial viral infection in infants with underlying
cardiopulmonary disease. JAMA 254: 3047–3051
59 van den Hoogen BG, de Jong JC, Groen J, Kuiken T, de GR, Fouchier RA,
Osterhaus AD (2001) A newly discovered human pneumovirus isolated from
young children with respiratory tract disease. Nat Med 7: 719–724
60 Ulloa-Gutierrez R, Skippen P, Synnes A, Seear M, Bastien N, Li Y, Forbes JC
(2004) Life-threatening human metapneumovirus pneumonia requiring extra-
corporeal membrane oxygenation in a preterm infant. Pediatrics 114: e517-
e519
61 Maggi F, Pifferi M, Vatteroni M, Fornai C, Tempestini E, Anzilotti S, Lanini
L, Andreoli E, Ragazzo V, Pistello M, Specter S, Bendinelli M (2003) Human
metapneumovirus-associated with respiratory tract infections in a 3-year study
of nasal swabs from infants in Italy. J Clin Microbiol 41: 2987–2991
62 Boivin G, De SG, Cote S, Gilca R, Abed Y, Rochette L, Bergeron MG, Dery
P (2003) Human metapneumovirus infections in hospitalized children. Emerg
Infect Dis 9: 634–640
63 Williams JV, Harris PA, Tollefson SJ, Halburnt-Rush LL, Pingsterhaus JM,
Edwards KM, Wright PF, Crowe JE, Jr (2004) Human metapneumovirus and
lower respiratory tract disease in otherwise healthy infants and children. N
Engl J Med 350: 443–450
64 Hamelin ME, Cote S, Laforge J, Lampron N, Bourbeau J, Weiss K, Gilca R,
DeSerres G, Boivin G (2005) Human metapneumovirus infection in adults
with community-acquired pneumonia and exacerbation of chronic obstructive
pulmonary disease. Clin Infect Dis 41: 498–502
65 Wyde PR, Chetty SN, Jewell AM, Boivin G, Piedra PA (2003) Comparison of
the inhibition of human metapneumovirus and respiratory syncytial virus by
ribavirin and immune serum globulin in vitro. Antiviral Res 60: 51–59
Viral pathogens and epidemiology, detection, therapy and resistance 53

66 Jartti T, Lehtinen P, Vuorinen T, Osterback R, van den HB, Osterhaus AD,


Ruuskanen O (2004) Respiratory picornaviruses and respiratory syncytial virus
as causative agents of acute expiratory wheezing in children. Emerg Infect Dis
10: 1095–1101
67 Legg JP, Warner JA, Johnston SL, Warner JO (2005) Frequency of detection of
picornaviruses and seven other respiratory pathogens in infants. Pediatr Infect
Dis J 24: 611–616
68 Papadopoulos NG (2004) Do rhinoviruses cause pneumonia in children?
Paediatr Respir Rev 5 (Suppl A): S191-S195
69 Kellner G, Popow-Kraupp T, Kundi M, Binder C, Wallner H, Kunz C (1988)
Contribution of rhinoviruses to respiratory viral infections in childhood: a
prospective study in a mainly hospitalized infant population. J Med Virol 25:
455–469
70 Seemungal TA, Harper-Owen R, Bhowmik A, Jeffries DJ, Wedzicha JA (2000)
Detection of rhinovirus in induced sputum at exacerbation of chronic obstruc-
tive pulmonary disease. Eur Respir J 16: 677–683
71 Rotbart HA, Hayden FG (2000) Picornavirus infections: a primer for the prac-
titioner. Arch Fam Med 9: 913–920
72 Boyd MT, Jordan SW, Davis LE (1987) Fatal pneumonitis from congenital
echovirus type 6 infection. Pediatr Infect Dis J 6: 1138–1139
73 Toce SS, Keenan WJ (1988) Congenital echovirus 11 pneumonia in association
with pulmonary hypertension. Pediatr Infect Dis J 7: 360–362
74 Cheeseman SH, Hirsch MS, Keller EW, Keim DE (1977) Fatal neonatal pneu-
monia caused by Echovirus type 9 Am J Dis Child 131: 1169
75 Ventura KC, Hawkins H, Smith MB, Walker DH (2001) Fatal neonatal echovi-
rus 6 infection: autopsy case report and review of the literature. Mod Pathol 14:
85–90
76 Chang LY, Lin TY, Hsu KH, Huang YC, Lin KL, Hsueh C, Shih SR, Ning HC,
Hwang MS, Wang HS, Lee CY (1999) Clinical features and risk factors of pul-
monary oedema after enterovirus–71-related hand, foot, and mouth disease.
Lancet 354: 1682–1686
77 Ledford RM, Collett MS, Pevear DC (2005) Insights into the genetic basis for
natural phenotypic resistance of human rhinoviruses to pleconaril. Antiviral
Res 68: 135–138
78 Rotbart HA (2002) Treatment of picornavirus infections. Antiviral Res 53:
83–98
79 Lednicky JA (2003) Hantaviruses. a short review. Arch Pathol Lab Med 127:
30–35
80 Zeier M, Handermann M, Bahr U, Rensch B, Muller S, Kehm R, Muranyi W,
Darai G (2005) New ecological aspects of hantavirus infection: a change of a
paradigm and a challenge of prevention – a review. Virus Genes 30: 157–180
81 Miedzinski L (2005) Community-acquired pneumonia: new facets of an old
disease – Hantavirus pulmonary syndrome. Respir Care Clin N Am 11: 45–58
82 Ferres M, Vial P (2004) Hantavirus infection in children. Curr Opin Pediatr 16:
70–75
83 Gremillion DH, Crawford GE (1981) Measles pneumonia in young adults. An
analysis of 106 cases. Am J Med 71: 539–542
54 Walter Hampl and Thomas Mertens

84 Kim EA, Lee KS, Primack SL, Yoon HK, Byun HS, Kim TS, Suh GY, Kwon OJ,
Han J (2002) Viral pneumonias in adults: radiologic and pathologic findings.
Radiographics 22 (Spec No): S137–S149
85 Chiba ME, Saito M, Suzuki N, Honda Y, Yaegashi N (2003) Measles infection
in pregnancy. J Infect 47: 40–44
86 Drut R, Drut RM (1988) Measles pneumonia in a newborn. Pediatr Pathol 8:
553–557
87 Frederick DM, Bland D, Gollin Y (2002) Fatal disseminated herpes simplex
virus infection in a previously healthy pregnant woman. A case report. J
Reprod Med 47: 591–596
88 Ramsey PG, Fife KH, Hackman RC, Meyers JD, Corey L (1982) Herpes sim-
plex virus pneumonia: clinical, virologic, and pathologic features in 20 patients.
Ann Intern Med 97: 813–820
89 Jones AM, Thomas N, Wilkins EG (2001) Outcome of varicella pneumonitis
in immunocompetent adults requiring treatment in a high dependency unit. J
Infect 43: 135–139
90 Gasparetto EL, Warszawiak D, Tazoniero P, Escuissato DL, Marchiori E (2005)
Varicella pneumonia in immunocompetent adults: report of two cases, with
emphasis on high-resolution computed tomography findings. Braz J Infect Dis
9: 262–265
91 Chou DW, Lee CH, Chen CW, Chang HY, Hsiue TR (1999) Varicella pneumo-
nia complicated by acute respiratory distress syndrome in an adult. J Formos
Med Assoc 98: 778–782
92 Goodnight WH, Soper DE (2005) Pneumonia in pregnancy. Crit Care Med 33:
S390-S397
93 Harger JH, Ernest JM, Thurnau GR, Moawad A, Momirova V, Landon MB,
Paul R, Miodovnik M, Dombrowski M, Sibai B, Van DP (2002) Risk factors and
outcome of varicella-zoster virus pneumonia in pregnant women. J Infect Dis
185: 422–427
94 Pfeiffer H, Varchmin-Schultheiss K, Brinkmann B (2006) Sudden death in
childhood due to varicella pneumonia: a forensic case report with clinical impli-
cations. Int J Legal Med 120: 33–35
95 Brayer C, Bony C, Salles M, Samperiz S, Pilorget H, Attali T, Alessandri JL
(2004) [Bronchopulmonary dysplasia and cytomegalovirus pneumonia]. Arch
Pediatr 11: 223–225
96 Sawyer MH, Edwards DK, Spector SA (1987) Cytomegalovirus infection
and bron-chopulmonary dysplasia in premature infants. Am J Dis Child 141:
303–305
97 Suzumura H, Sakurai K, Kano K, Ichimura T (1996) Chronic respiratory failure
after acquired cytomegalovirus infection in a very low birthweight infant. Acta
Paediatr Jpn 38: 677–680
98 Pedrazzini AG, Petralli C, Pedrazzini A, Luscieti P, Pedrinis E, Kauzlaric D
(1981) [Cytomegalovirus pneumonia in primarily healthy adults]. Schweiz Med
Wochenschr 111: 943–947
99 Chemaly RF, Torres HA, Hachem RY, Nogueras GM, Aguilera EA, Younes A,
Luna MA, Rodriguez G, Tarrand JJ, Raad II (2005) Cytomegalovirus pneumo-
nia in patients with lymphoma. Cancer 104: 1213–1220
Viral pathogens and epidemiology, detection, therapy and resistance 55

100 Knox KK, Pietryga D, Harrington DJ, Franciosi R, Carrigan DR (1995)


Progressive immunodeficiency and fatal pneumonitis associated with human
herpesvirus 6 infection in an infant. Clin Infect Dis 20: 406–413
101 Cone RW, Huang ML, Hackman RC (1994) Human herpesvirus 6 and pneumo-
nia. Leuk Lymphoma 15: 235–241
102 Plummer G, Benyesh-Melnick M (1964) A plaque reduction neutralization test
for human cytomegalovirus. Proc Soc Exp Biol Med 117: 145–150
103 Merk J, Schmid FX, Fleck M, Schwarz S, Lehane C, Boehm S, Salzberger B,
Birnbaum DE (2005) Fatal pulmonary failure attributable to viral pneumonia
with human herpes virus 6 (HHV6) in a young immunocompetent woman. J
Intensive Care Med 20: 302–306
104 Gautschi O, Berger C, Gubler J, Laube I (2003) Acute respiratory failure and
cerebral hemorrhage due to primary Epstein-Barr virus infection. Respiration
70: 419–422
105 Ankermann T, Claviez A, Wagner HJ, Krams M, Riedel F (2003) Chronic inter-
stitial lung disease with lung fibrosis in a girl: uncommon sequelae of Epstein-
Barr virus infection. Pediatr Pulmonol 35: 234–238
106 Mueller GA, Pickoff AS (2003) Pediatric lymphocytic interstitial pneumonitis
in an HIV-negative child with pulmonary Epstein-Barr virus infection. Pediatr
Pulmonol 36: 447–449
107 Kaan PM, Hegele RG, Hayashi S, Hogg JC (1997) Expression of bcl-2 and
Epstein-Barr virus LMP1 in lymphocytic interstitial pneumonia. Thorax 52:
12–16
108 Marzouk K, Corate L, Saleh S, Sharma OP (2005) Epstein-Barr-virus-induced
interstitial lung disease. Curr Opin Pulm Med 11: 456–460
109 Oda Y, Okada Y, Katsuda S, Nakanishi I (1994) Immunohistochemical study on
the infection of herpes simplex virus, human cytomegalovirus, and Epstein-Barr
virus in secondary diffuse interstitial pneumonia. Hum Pathol 25: 1057–1062
110 Pfleger A, Eber E, Popper H, Zach MS (2000) Chronic interstitial lung disease
due to Epstein-Barr virus infection in two infants. Eur Respir J 15: 803–806
111 Balfour-Lynn IM, Girdhar DR, Aitken C (1995) Diagnosing respiratory syncy-
tial virus by nasal lavage. Arch Dis Child 72: 58–59
112 Heikkinen T, Marttila J, Salmi AA, Ruuskanen O (2002) Nasal swab versus
nasopharyngeal aspirate for isolation of respiratory viruses. J Clin Microbiol
40: 4337–4339
113 Falsey AR, Formica MA, Walsh EE (2002) Diagnosis of respiratory syncytial
virus infection: comparison of reverse transcription-PCR to viral culture and
serology in adults with respiratory illness. J Clin Microbiol 40: 817–820
114 Akhtar N, Ni J, Stromberg D, Rosenthal GL, Bowles NE, Towbin JA (1999)
Tracheal aspirate as a substrate for polymerase chain reaction detection of viral
genome in childhood pneumonia and myocarditis. Circulation 99: 2011–2018
115 Simpson JL, Moric I, Wark PA, Johnston SL, Gibson PG (2003) Use of induced
sputum for the diagnosis of influenza and infections in asthma: a comparison of
diagnostic techniques. J Clin Virol 26: 339–346
116 Xiang X, Qiu D, Chan KP, Chan SH, Hegele RG, Tan WC (2002) Comparison
of three methods for respiratory virus detection between induced sputum
56 Walter Hampl and Thomas Mertens

and nasopharyngeal aspirate specimens in acute asthma. J Virol Methods 101:


127–133
117 Jain P, Sandur S, Meli Y, Arroliga AC, Stoller JK, Mehta AC (2004) Role of flex-
ible bronchoscopy in immunocompromised patients with lung infiltrates. Chest
125: 712–722
118 Woo PC, Lau SK, Wong BH, Tsoi HW, Fung AM, Kao RY, Chan KH, Peiris JS,
Yuen KY (2005) Differential sensitivities of severe acute respiratory syndrome
(SARS) coronavirus spike polypeptide enzyme linked immunosorbent assay
(ELISA) and SARS coronavirus nucleocapsid protein ELISA for serodiagno-
sis of SARS coronavirus pneumonia. J Clin Microbiol 43: 3054–3058
119 Niedrig M, Leitmeyer K, Lim W, Peiris M, Mackenzie JS, Zambon M (2005)
First ex-ternal quality assurance of antibody diagnostic for SARS-new corona-
virus. J Clin Virol 34: 22–25
120 Gregson D, Lloyd T, Buchan S, Church D (2005) Comparison of the RSV
respi-strip with direct fluorescent-antigen detection for diagnosis of respiratory
syncytial virus infection in pediatric patients. J Clin Microbiol 43: 5782–5783
121 Brandenburg AH, Groen J, van Steensel-Moll HA, Claas EC, Rothbarth PH,
Neijens HJ, Osterhaus AD (1997) Respiratory syncytial virus specific serum
antibodies in infants under six months of age: limited serological response upon
infection. J Med Virol 52: 97–104
122 Ebihara T, Endo R, Ma X, Ishiguro N, Kikuta H (2005) Detection of human
metapneumovirus antigens in nasopharyngeal secretions by an immunofluo-
rescent-antibody test. J Clin Microbiol 43: 1138–1141
Community-Acquired Pneumonia 57
ed. by N. Suttorp, T. Welte and R. Marre
© 2007 Birkhäuser Verlag Basel/Switzerland

Resistance in Streptococcus pneumoniae

Mathias W. R. Pletz1, Lesley McGee2 and Tobias Welte1


1Department of Respiratory Medicine, Hannover Medical School, Carl- Neuberg-Str.1,
Hannover, 30625, Germany; 2Hubert Department of Global Health, Emory University, 1518
Clifton Road, Atlanta, GA, 30322, USA

Abstract
Streptococcus pneumoniae is a leading cause of community-acquired lower respiratory
tract infections, sinusitis, meningitis, and bloodstream infections. Pneumococci are Gram
positive, encapsulated bacteria and exhibit more than 90 different capsular serotypes.
Resistance to penicillin in clinical isolates was reported anecdotally as early as 1965,
but was not considered a major concern until the mid-1990s. In the 1990s, there was a
tremendous global increase in resistance to penicillins and this led to the increased use
of macrolides and tetracyclines to treat infections. After several years, the resistance rates
to these antibiotics began to increase as well. Currently, fluoroquinolones are used most
frequently to treat community-acquired respiratory infections in adults and resistance
rates globally are still low.
Pneumococci are naturally competent bacteria and frequently acquire resistance by
intraspecies or interspecies gene transfer. Resistance to `-lactams is due to the acquisi-
tion of different mutations within the pencillin-binding proteins that have been demon-
strated to originate from the less pathogenic viridans streptococci. Other mechanisms of
antibiotic resistance include enzymes and efflux pumps on mobile genetic elements (e.g.
erm and mef), or resistance arising through spontaneous mutations. Clinical studies show
that resistance, particularly to penicillins, is not always related to clinical failure.
The global increase in resistance rates in pneumococci is in part due to the spread of
a limited number of highly sucessful multiresistant pneumococcal clones. Isolates belong-
ing to a specific clone, defined by sequence types according to multilocus sequencing,
often exhibit the same serotype. However, capsular switching due to genetic rearrange-
ments within the same clone has been observed. The recently introduced seven-valent
conjugated pneumococcal vaccine has been shown to decrease disease and carrier rates
of the included serotypes. Since some of the multiresistant clones exhibit vaccine sero-
types, resistance rates to penicillin, macrolides and fluoroquinolones have been decreas-
ing since the introduction of this vaccine.

INTRODUCTION

S. pneumoniae is the most frequent pathogen causing community-acquired


pneumonia and for many years penicillin or ampicillin were the drugs of
choice for all pneumococcal infections.
58 Mathias W. R. Pletz et al.

In contrast to other bacterial species, such as staphylococci or Gram-


negative bacteria, pneumococci had remained susceptible to almost all
antibiotics for decades. Penicillin-resistant strains were selected in the labo-
ratory already in the 1940s, but it took more than 30 years before the first
clinical isolates with penicillin-resistance were detected. The subsequent
global emergence of penicillin-resistance in the 1980s and 1990s led to the
increased use of macrolides, other non-beta lactam antibiotics and the fluo-
roquinolones. Due to these new selective pressures, resistance to macrolides
and tetracyclines emerged rapidly. For both classes it could be demonstrated
that the increase in resistance rates paralleled the use of these individual
classes of antibiotics [1]. It had also been shown that this trend could be
reversed when the use of the concerned antibiotic was restricted [2]. This
data explains, in part, the considerable differences in rates of antibiotic
resistance published between different countries and reflects the antibiotic
prescription behavior. In general, antibiotic resistance in countries where
antibiotics can be purchased without a prescription (over the counter) as in
Spain and Mexico, are higher than in countries where antibiotics can only
be prescribed by physicians. Recently, resistance to fluoroquinolones has
emerged, but there are global differences reported with individual cases
described in Austria and rates as high as 13.3% in Hong Kong [3, 4]. Due to
the tremendous cost for the development of antibiotics and the challenges
associated with the safety of newly developed drugs, many pharmaceutical
companies have discontinued their development of new antibiotics. During
the last few years only a limited number of new antibiotics were licensed,
such as telithromycin, linezolid and quinupristin-dalfopristin.
Clinical pneumococcal isolates with resistance to quinupristin-dalfopris-
tin, telithromycin and linezolid have already been reported [5–7]. In this
context it is also of concern that pneumococci with tolerance to vancomycin,
an antibiotic used as a reserve against multi-resistant Gram-positive bacte-
ria, have already been documented [8].
Resistance rates parallel antibiotic usage and exhibit therefore tre-
mendous regional differences and trends [3, 9]. Besides numerous local
and regional surveillance studies, several large longitudinal surveillance
projects monitor globally the trends of antibiotic resistance in pneumococci
(i.e. the Alexander project, Prospective Resistant Organism Tracking and
Epidemiology for the Ketolide Telithromycin – PROTEKT). Table 1 shows
resistance rates in eight European countries according to results of the
PneumoWorld study from 2001–2003 [3].

Genetic background

Bacterial resistance may be “intrinsic” or an inherent – naturally occurring


property of an organism, i.e., these organisms may lack the appropriate
drug-susceptible target or possess natural barriers that prevent the agent
Resistance in Streptococcus pneumoniae 59

Table 1. MIC90 and antibiotic resistance of 2,279 isolates of S. pneumoniae in eight European
countries according to Reinert et al. [(3] (Abbreviations: I, intermediate; R, resistant)

Country (no. of strains) Antibiotic MIC90 % Resistance (I+R)


Austria (n = 160) Penicillin G 0.03 4.4
Cefuroxime 0.06 0.6
Clarithromycin 0.25 10.0
Levofloxacin 1 0
Tetracycline 4 10.6
Belgium (n = 148) Penicillin G 0.125 11.5
Cefuroxime 0.25 8.8
Clarithromycin 32 23.7
Levofloxacin 1 0.7
Tetracycline 32 23.7
France (n = 443) Penicillin G 2 47.6
Cefuroxime 8 39.1
Clarithromycin 32 46.1
Levofloxacin 1 0.9
Tetracycline 32 40.4
Germany (n = 530) Penicillin G 0.03 6.0
Cefuroxime 0.06 1.5
Clarithromycin 1 10.6
Levofloxacin 1 0.4
Tetracycline 4 11.3
Italy (n = 462) Penicillin 0.25 13.0
Cefuroxime 0.5 7.4
Clarithromycin 32 35.5
Levofloxacin 1 1.3
Tetracycline 32 23.8
Portugal (n = 174) Penicillin G 2 19.0
Cefuroxime 8 14.4
Clarithromycin 1 10.3
Levofloxacin 1 1.2
Tetracycline 8 12.1
Spain (n = 310) Penicillin G 2 61.9
Cefuroxime 8 43.9
Clarithromycin 32 43.6
Levofloxacin 1 1
Tetracycline 32 47.1
Switzerland (n = 52) Penicillin G 0.5 17.3
Cefuroxime 2 11.5
Clarithromycin 0.25 17.3
Levofloxacin 1 0
Tetracycline 4 13.5
60 Mathias W. R. Pletz et al.

from reaching its target. Acquired resistance results either from spontane-
ous mutations in existing DNA or the acquisition of foreign genetic material
by genetic elements such as plasmids and bacteriophages (horizontal gene
transfer).
To date, no plasmids have been detected in any resistant S. pneumoniae
strain and genetic changes conferring resistance are therefore all chromo-
somal. Pneumococci are one of the few bacterial species that are naturally
competent and are able to acquire resistance genes through horizontal gene
transfer, usually via the process of transformation.
Pneumococci are able to bind, internalize and integrate free DNA by
recombination without antecedent stimulation as it is, for example, neces-
sary for E. coli. The origin of the genetic material for these transformations
may be from the same (intraspecies recombination) or a different – but
closely related – species (interspecies recombination). The results of inter-
species recombination result in mosaic genes, which are genes consisting
of both pneumococcal sequences and sequences from other species. The
higher the degree of homology between the donor and recipient DNA the
higher is the likelihood of successful homologous recombination. Therefore
the amino acid sequence of the donor and recipient DNA is often identical.
In contrast, due to the degeneration of the genetic code the 3rd nucleotide
of a codon coding for the same amino acid can be different. The resulting
“codon usage bias” is species specific. A characteristic feature of integrated
DNA derived from another species is a repeated mismatch of the 3rd
nucleotide leading to a decreased similarity of the recombined sequence
when compared to a reference sequence (Fig. 1). Viridans streptococci have
been frequently observed as a donor species for the transfer of genetic
resistance determinants to S. pneumoniae. Viridans streptococci belong to
the commensal flora and are exposed to the antibiotics administered to the
host and resistant strains should therefore be easily selected. Since viridans
streptococci are to a high degree homologous with S. pneumoniae, recom-
bination between the two species exhibits a high success rate. In addition,
both species colonize the human nasopharynx and so contact between the
two species is guaranteed, which is necessary for the exchange of genetic
material. Since commensal bacteria rarely cause disease, they are not rou-
tinely tested for resistance and surveillance studies addressing resistance in
commensal bacteria are rare. In pneumococci acquisition of foreign genetic
material and the consequent spread of resistant strains seems to be the pri-
mary cause for the increase in resistance to different antibiotics.

Spread of resistance in pneumococci – the clone concept

The global spread of penicillin-resistant pneumococci has been documented


by numerous surveillance studies. Genetic fingerprinting using techniques
like pulsed-field gel electrophoresis of pneumococcal DNA digested by
Resistance in Streptococcus pneumoniae 61

Figure 1. Alignment of PBP 2b of a penicillin-resistant S. pneumoniae strain (#402) with the sus-
ceptible pneumococcal laboratory reference strain R6. The red part of the sequence is derived
from S. oralis. (Courtesy of Dr B. Beall, CDC)

the restriction enzyme SmaI, has revealed that the majority of the resistant
clinical isolates belong to a small number of highly successful clones, some
of which have spread globally [10]. More recently, another tool to investi-
gate the genetic relatedness of resistant isolates became available, namely
multi-locus sequence typing (MLST) [11]. MLST is based on the sequencing
of seven highly conserved “housekeeping genes.” The sequences at each of
the seven loci are compared with all of the known alleles at that locus which
is available at the pneumococcal MLST web site (http: //spneumoniae.mlst.
net/). The alleles at each of the seven loci define the allelic profile of each
isolate and their sequence type (ST). If isolates have identical sequence
types, or differ at less than three loci, they are considered to be related.
Many studies have used PFGE for the rapid clustering of genetically
related isolates and MLST to assign nonsubjective and electronically por-
table clone identifiers to clusters of related isolates.
In order to create a nomenclature to monitor the international spread of
resistant pneumococci, in 1997 the Pneumococcal Molecular Epidemiology
Network (PMEN) was established under the auspices of the International
Union of Microbiological Societies with the aim of characterizing, stan-
dardizing, naming, and classifying antibiotic-resistant pneumococcal clones
(http: //www.sph.emory.edu/PMEN/) [12]. Currently, the PMEN describes 26
international clones, some of which have a global distribution and have been
reported in many countries with varying antibiograms. The global spread of
the PMEN-clone 1, Spain 23F-1, is illustrated in Figure 2. Pneumococci have
the ability to switch their capsular serotypes which can be due to mutations
or the exchange of capsular genes. For surveillance it is therefore not appro-
priate to equal serotype and sequence type (clone) since capsular variants
of clones exist, e.g., 19F capsular variants of PMEN-clone 1 is designated
Spain 23F-1-19F.
62 Mathias W. R. Pletz et al.

Figure 2. Global distribution of the PMEN strain Spain23F-1.

Serotype switching serves also as a mechanism to avoid the selective


pressure due to vaccination with vaccines containing only a limited number
of serotypes.
PMEN clones are spreading more successfully than other pneumo-
cocci. The reason for that fitness advantage is not yet clear, but it has been
observed that resistance to some classes of antibiotics has emerged rapidly
once it has been introduced into these clones [13].

Beta-lactams

Beta-lactams bind and inactivate the so-called penicillin-binding proteins


(PBPs). These proteins are enzymes that catalyze the terminal stages of
murein synthesis which is the main component of the bacterial cell wall [14].
The action of `-lactams leads to cell death, i.e. the action is bactericidal. Most
of the bacterial species exhibiting `-lactam resistance secrete or contain `-
lactamase enzymes that cleave the `-lactams. In contrast to other bacterial
species, `-lactam resistant pneumococci do not express any `-lactamases.
The pneumococcal mechanism of resistance to `-lactams consists of mul-
tiple mutations within several penicillin-binding proteins (PBPs)[15]. These
mutations mediate changes in the structure of the penicillin-binding domain
of the PBP leading to a decreased affinity to `-lactams. Six types of PBPs
are found in pneumococci: PBP 1a (92–100 kDa), PBP 1b (89–95 kDa), PBP
2x (85 kDa), PBP2a (80–81 kDa), PBP 2b (77–78 kDa), and PBP 3 (43–52
kDa) (K 64, 117). High-level resistance to third-generation cephalosporins
has occurred primarily by the development of altered forms of PBP1a and
2x, whereas high-level penicillin resistance additionally requires alterations
of PBP2b [16]. It has been found that the genes of low-affinity forms of
Resistance in Streptococcus pneumoniae 63

PBPs 1a, 2b and 2x from clinical isolates contain sequence blocks with up
to 20% divergence in their nucleotide sequence. Such sequence blocks have
not evolved by spontaneous mutations and the majority of these altera-
tions do not confer amino acid substitutions (silent mutation). The mosaic
blocks are shared between resistant S. pneumoniae and the closely related
viridans streptococci (S. oralis, S. mitis). Based on these observations, it has
been assumed that `-lactam resistance developed originally in viridans
streptococci and was transferred to pneumococci by interspecies recom-
bination [17]. This hypothesis is supported by the fact that genes adjacent
to the pbps (e.g., ddl) are frequently part of the recombination and appear
in resistant strains. This was recently observed in very high level penicillin
resistant (MIC * 8 mg/l), invasive pneumococcal isolates from the CDC’s
ABC Surveillance study. The ddl locus in two of these strains was from S.
oralis, suggesting that the origin of very-high-level resistance in some strains
may result from the transformation and incorporation of resistance deter-
minants from viridans group streptococci [18].
There is evidence that altered PBPs from resistant pneumococci have a
preference for branched peptides in regard to the cell wall synthesis [19].
This is in contrast to PBPs from susceptible strains which use primarily
linear stem peptides. A newly identified protein MurM which, together
with MurN, is involved in the synthesis of short peptide branches in the
pneumococcal cell wall. Cells in which MurM was inactivated produced cell
walls without branches and also completely lost penicillin resistance [20].
Currently, the role of MurM in `-lactam resistant strains is the subject of
further studies.

Macrolides

Macrolides consist of saccharides that are attached to 14-membered (eryth-


romycin, clarithromycin), 15-membered (azithromycin) or 16-membered
rings. The use of the latter is usually restricted to veterinary purposes.
Macrolides act bacteriostatically by binding to the 23S ribosomal RNA
of the 50S subunit of the bacterial ribosome and consequently blocking
the elongation step of protein synthesis. There are two main mechanisms
of macrolide resistance: active efflux and target site modification. Active
efflux is performed by an energy dependent cell membrane transporter
protein that is encoded by the mef gene, which is located on a conjugative
transposon. The mef gene provides lower levels of resistance and confers
the so-called M-phenotype, i.e. resistance primarily to macrolides. Two vari-
ants of the mef gene, mef(A) (primarily found in S. pyogenes) and mef(E)
have been identified in pneumococci. However, due to confusion in the
nomenclature many authors do not differentiate between the two and call
it mef(A) regardless of the variant. Target site modification is performed by
a methylase enzyme which adds a CH3-group to an adenine residue on the
64 Mathias W. R. Pletz et al.

23S rRNA. The methylation blocks the binding site of not only macrolides,
but also lincosamides and streptogramins and the resulting phenotype is
called MLSB. The methylase enzyme is encoded by the erm gene which
is also located on a transposon. Among bacterial species there exists sev-
eral variants of the erm gene, in pneumococci primarily the erm(B) gene is
found. Some macrolide resistant pneumococci have been documented to
exhibit both the erm and mef genes. These strains appear to be highly clonal
and most belong to the PMEN clone Taiwan19F-14 [21].
A third, currently emerging mechanism of resistance to macrolides
consists of mutations conferring amino acid substitutions in the ribosomal
proteins and nucleotide mutations in the 23S rRNA itself. Frequently, the
ribosomal proteins L4 and L22 are involved and mutations in the rRNA and
ribosomal proteins can confer new resistance phenotypes with combined
resistance to macrolides and lincosamides (ML) or macrolides, ketolides
and streptogramins (MKSB). Usually, the decrease in susceptibility to
ketolides (for example telithromycin) is less than the decrease in suscepti-
bility for other MLKS(B) agents [22].

Tetracyclines

Tetracyclines are antibiotics derived from Streptomyces species and inhibit


bacterial protein synthesis by blocking attachment of the aminoacyl-tRNA
to the ribosome. Resistance to tetracyclines in pneumococci is conferred
by ribosomal protection proteins Tet(M) and Tet(O). These proteins are
homologous to the elongation factors EF-Tu and EF-G. It is assumed that
Tet(M) and Tet(O) induce the detachment of tetracyclines from the bacte-
rial ribosome, the detailed mechanism of action is not known. Tet(M) and
Tet(O) are encoded by the tet(M) and tet(O) genes that are located on a
transposon. In species other than pneumococci, resistance to tetracyclines is
frequently mediated by plasmids.

Fluoroquinolones

Resistance to fluoroquinolones in pneumococci is caused by efflux and/or


by mutations in the quinolone resistance-determining regions (QRDR)
of the genes coding for type II topoisomerase enzymes: DNA gyrase and
topoisomerase IV.
Mutations conferring resistance occur in a stepwise fashion, with
mutations observed in either parC or gyrA (depending on the selecting
fluoroquinolone) or both, leading to decreased fluoroquinolone suscepti-
bility [23]. Strains usually become fully fluoroquinolone resistant with the
addition of a mutation in the other target gene (either gyrA or parC) and
mutations in parE and gyrB may contribute to resistance in some isolates.
Resistance in Streptococcus pneumoniae 65

QRDR mutations can arise spontaneously or be transferred by the inte-


gration of foreign genetic material. In comparison to `-lactam resistance,
horizontal gene transfer seems to play a minor role in fluoroquinolone
resistance. Studies addressing that question found evidence for horizontal
gene transfer in 0–11% of fluoroquinolone resistant isolates and interest-
ingly, this ratio seems to be higher in respiratory isolates than in invasive
isolates [24–26]. This discrepancy might be explained by the hypothesis
that interspecies recombination takes place in the nasopharynx, which is
colonized by both pneumococci and viridans streptococci. One could spec-
ulate that the antecedent nasopharyngeal colonization period for invasive
strains is shorter than that of respiratory isolates. Therefore, there may be
less time for interspecies recombination in invasive isolates.
The impact of efflux on fluoroquinolone resistance seems to be more
limited and selective. To date, no highly resistant isolate has been found
with efflux as the only mechanism of resistance. Fluoroquinolones with
a small molecule size, e.g. ciprofloxacin, seem to be affected to a higher
extent than larger molecules such as moxifloxacin. A putative efflux pump,
PmrA was described by Gill et al., that exhibits homology to the multidrug
efflux pumps NorA and Bmr [27]. PmrA consists of 12 transmembrane
segments as efflux proteins of the proton-dependent pumps. In addition,
a recent article describes the presence of a non-PmrA pump in S. pneu-
moniae.
Many studies have addressed the epidemiology of PmrA by comparing
ciprofloxacin MICs in the absence and the presence of the pump inhibitor
reserpine. In contrast to Mef(A), PmrA seems not to be encoded by a resis-
tance gene but rather over expression. However, little is know about the
mechanism of the expression regulation of PmrA.

Resistance to new antibiotics

Since 1962, only a few classes of novel antibiotics have been introduced,
and all since 1999, including the streptogramins (quinupristin/dalfopristin),
the oxazolidinones (linezolid), and the lipopeptides (daptomycin). All other
recently introduced drugs, such as tigecycline and ertapenem are derived
from antibiotic classes that are already in use. The ketolides (e.g., telithro-
mycin) were developed from the macrolides to overcome antibiotic resis-
tance in pneumococci and are characterized by the lack of the L-cladinose
sugar at position 3 of the erythronolide A moiety, which is replaced by a
keto group.
Farrell and Felmingham recently reported only 10 isolates to be telithro-
mycin resistant among a worldwide collection of 13,874 S. pneumoniae iso-
lates isolated between 1999 and 2003. The strains isolated in France, Italy,
Spain, Hungary, and Japan had telithromycin MICs of 4 to 8 +g/ml and
showed an erm(B) genotype [28]. Reinert et al. recently described a telithro-
66 Mathias W. R. Pletz et al.

mycin resistant isolate in Germany (MIC 8 +g/ml) that exhibited in addition


to the erm(B) genotype, mutations in the L4 (S20N) protein [6]. The S20N
L4 alteration has been shown to contribute to the increase in macrolide
MICs in pneumococci. It may be assumed that a combination of erm(B) and
L4 mutations confer ketolide resistance. Also a single base deletion within
the 23S-rRNA has been detected to confer resistance to macrolides and
telithromycin [29]. Another clinical isolate resistant to telithromycin and
fluoroquinolones with a still unidentified mechanism was recently found in
Argentina [30].
Streptogramin (quinupristin-dalfopristin, Q/D) resistance among Gram-
positive cocci has been very uncommon [31]. Two clinical isolates among
8,837 (0.02%) S. pneumoniae isolates were discovered in 2001 to 2002
with Q/D MICs of 4 +g/ml. Each had a 5-amino-acid tandem duplication
(RTAHI) in the L22 ribosomal protein gene (rplV) preventing synergistic
ribosomal binding of the streptogramin combination [7].
Recently, two clinical S. pneumoniae isolates, identified as nonsuscep-
tible to linezolid and as resistant to macrolides and chloramphenicol, were
found to contain 6-bp deletions in the gene encoding riboprotein L4 [5].

Clinical relevance of resistance

MIC based breakpoints have been suggested by different organizations


to distinguish between susceptible, intermediate and resistant strains. The
breakpoints published by the CLSI (formerly the NCCLS) are widely used
in routine clinical laboratories and in surveillance studies. However, in some
cases these breakpoints are unable to predict clinical failure in patients
who were discordantly treated. Pharmacodynamic parameters are superior
to MIC in predicting bacteriological eradication that is related to clinical
outcome. To calculate these parameters, ideally antimicrobial susceptibil-
ity (measured by MIC) is compared to the achievable unbound fraction
of antibiotic at the site of infection. Since the latter is difficult to measure,
usually serum/plasma concentrations obtained from clinical studies with
healthy volunteers are used instead. This may lead to some limitations since
recent pharmacokinetic studies have clearly demonstrated the there can
be tremendous differences in the concentration of an antibiotic between
healthy subjects and severely ill patients, in regard to the patient’s age and
the body site.

Clinical relevance of `-lactam resistance

In pneumococcal pneumonia caused by penicillin-resistant strains, no


association between treatment failure and discordant therapy could be
observed in several studies, as long as the administration was intravenous
Resistance in Streptococcus pneumoniae 67

and drugs were penicillin, ampicillin, amoxicillin, cefotaxime or ceftri-


axone. The only studies that have found such an association were not
stratified for the severity of disease or did not document the susceptibility
of the pneumococci to the drug that failed. The basis of that seeming con-
tradiction is high serum concentrations of these `-lactams and relatively
low breakpoints defining resistance [32]. The main PK/PD parameter
of `-lactams which are time-dependent killing antibiotics is the propor-
tion of time of the dose interval during which the plasma concentration
exceeds the MIC (T > MIC) [33]. Pharmacodynamic calculations predict
that high doses of intravenous penicillin remain useful for the treatment
of pneumococcal pneumonia up to the MICs of 4 mg/l (non-susceptibil-
ity according to CLSI is defined by MIC, > 0.12 mg/l). Pharmacodynamic
also predicts that `-lactams with less anti-pneumococcal activity may lead
to failure. Examples of such failures have been documented for cefazolin,
cefuroxime and ticarcillin.
Only a few data are available on the clinical failure of oral `-lactam
therapy. However, due to the decreased bioavailability of ampicillin and
amoxicillin such failure could be expected and the failure of low-dose amox-
icillin prophylaxis to prevent penicillin-resistant pneumonia in patients
with sickle cell disease has been documented. In a recent cohort study
presented at the 45th Interscience Conference on Antimicrobial Agents
and Chemotherapy (45th, ICAAC, Washington, D.C., 2005) Iannini et al.
investigated treatment failure in outpatients with pneumococcal pneumonia
(abstract # 896). In patients treated orally with cephalosporines they found
cephalosporine resistance associated treatment failure, however, this was
not found in outpatients treated orally with penicillins. This might be also
caused by the unfavourable bioavailability of cephalosporins. In another
abstract presented at the same conference File et al. described the eradiac-
tion of penicillin resistant pneumococci (amoxicillin MIC, ) 4mg/l) in 43
out of 44 patients with pneumonie by amoxicillin/clavulanic acid (abstract
# 707). Since pneumococci do not express `-lactamases this effect has to be
contributed primarily to amoxicillin.

Macrolides

The clinical relevance of macrolide resistance has been a matter of debate.


After the first break through bacteraemias during macrolide treatment,
it was thought that only resistant strains harboring the erm(B) mecha-
nism could cause treatment failure [34]. It has since become clear that
also strains with the efflux mechanism can exhibit high MIC’s resulting
in treatment failure. Of particular concern is the recent observation that
mutations in the ribosomal proteins or the 23S rRNA itself can develop
during macrolide treatment and confer MIC’s high enough to cause treat-
ment failure [35].
68 Mathias W. R. Pletz et al.

Clinical relevance of fluoroquinolone-resistance and first step mutants

Whereas most studies have shown no increase in mortality in patients with


invasive disease infected with penicillin-resistant pneumococci, FQ resis-
tance has been associated with clinical failure [36, 37].
In 2002, Davidson et al. published a paper on a topic that was receiv-
ing broad attention [38]. They described four patients treated with levo-
floxacin with pneumococcal pneumonia in whom treatment failure due to
fluoroquinolone resistance was observed. Two of the patients died. Prior
to this publication several reports had documented treatment failure due
to fluoroquinolones resistance, however, most treatment failures occurred
with ciprofloxacin, a fluoroquinolone with weak antipneumococcal activity.
Meanwhile, several other reports of fluoroquinolone resistance associated
treatment failure have been published and were reviewed in a recent paper
by Fuller and Low [39].
During the stepwise acquisition of mutations leading to full resistance,
so called “first-step mutants” are evolving. These first-step mutants are
frequently phenotypically susceptible and therefore cannot be detected
by routine resistance testing [40]. Their detection therefore requires the
sequencing of the topoisomerase genes, a method outside the scope of
most routine susceptibility testing in clinical laboratories, but an important
method for surveillance laboratories. Once a mutation in one of the target
enzymes is present there is a significant increased likelihood for the acquisi-
tion of mutations in the second target enzyme leading to complete resistance
[41]. Croisier et al. exposed susceptible first-step mutants with mutations
within parC to levofloxacin and moxifloxacin in a rabbit model, matching
human pharmacokinetics and found that double mutants (i.e. containing
mutations in parC and gyrA) resistant to these agents were selected under
both treatment regimens [42]. This is particularly likely to occur when large
pneumococcal populations, as observed in COPD for example, are exposed
to fluoroquinolones [42].Therefore, FQ treatment of infections caused by
first-step mutants can lead to the selection of resistant isolates resulting in
treatment failure and a general increase in FQ resistance. Some authors
consider first-step mutants to be a key parameter for the spread of FQ resis-
tance and estimate that their prevalence may be several times higher than
the prevalence of phenotypically resistant pneumococci [40, 43].

Limiting the spread of resistance – the impact of vaccines

The main mechanism driving the increase of antibiotic resistance in pneu-


mococci is clonal spread. Even if “clone” does not equal “serotype”, the
most frequent isolates of the same clone exhibit also the same serotype.
The majority of the serotypes exhibited by the main antibiotic resistant
pneumococcal clones are covered by the 23-valent non-conjugated pneu-
Resistance in Streptococcus pneumoniae 69

mococcal polysaccharide vaccine. The vaccine was demonstrated to reduce


the severity of pneumococcal vaccine types but, because the vaccine does
only induce a B-cell response, it cannot prevent the colonization of the
nasopharynx by strains exhibiting the vaccine serotypes. In contrast, the
seven-valent conjugated vaccine does induce also a T-cell response. Studies
have shown that the overall prevalence of strains of vaccine serotypes
(both in the vaccine target group and all age groups) has been reduced
since the introduction of the conjugate vaccine in 2000 in the United States
[44]. Obviously the immunological response introduced by the vaccine
enables carrier to clear strains of vaccine serotypes. Since most of the vac-
cine serotypes are exhibited by antibiotic resistant clones, antibiotic resis-
tance should decrease. This has already been demonstrated for penicillin
and macrolide resistance [44–46]. Surveillance data indicate that the rise
of fluoroquinolone resistance seems also to plateau in the United States
after the introduction of the vaccine [47]. However, little is known about
the long-term impact of the vaccine, and the extent to which vaccine sero-
types will be replaced by nonvaccine types is still not clear. Pneumococci
are naturally transformable, and capsular switching is a well-documented
mechanism for the potential evasion of the host immune response. Reports
of the emergence of penicillin-nonsusceptible clones of NVT sharing
genetic relatedness with internationally established clones targeted by the
vaccine [48, 49] have indicated the need to track phenotypic and genotypic
changes within invasive pneumococci.

References
1 Hyde TB, Gay K, Stephens DS, Vugia DJ, Pass M, Johnson S et al (2001)
Macrolide resistance among invasive Streptococcus pneumoniae isolates.
JAMA 286(15): 1857–1862
2 Seppala H, Klaukka T, Vuopio-Varkila J, Muotiala A, Helenius H, Lager K et
al (1997) The effect of changes in the consumption of macrolide antibiotics
on erythromycin resistance in group A streptococci in Finland. Finnish Study
Group for Antimicrobial Resistance. N Engl J Med 337(7): 441–446
3 Reinert RR, Reinert S, van der Linden M, Cil MY, Al-Lahham A, Appelbaum
P (2005) Antimicrobial susceptibility of Streptococcus pneumoniae in eight
European countries from 2001 to 2003. Antimicrob Agents Chemother 49(7):
2903–2913
4 Ho PL, Yung RW, Tsang DN, Que TL, Ho M, Seto WH et al (2001) Increasing
resistance of Streptococcus pneumoniae to fluoroquinolones: results of a Hong
Kong multicentre study in 2000 J Antimicrob Chemother 48(5): 659–665
5 Wolter N, Smith AM, Farrell DJ, Schaffner W, Moore M, Whitney CG et al
(2005) Novel mechanism of resistance to oxazolidinones, macrolides, and chlor-
amphenicol in ribosomal protein L4 of the pneumococcus. Antimicrob Agents
Chemother 49(8): 3554–3557
6 Reinert RR, van der Linden M, Al-Lahham A (2005) Molecular characteriza-
70 Mathias W. R. Pletz et al.

tion of the first telithromycin-resistant Streptococcus pneumoniae isolate in


Germany. Antimicrob Agents Chemother 49(8): 3520–3522
7 Jones RN, Farrell DJ, Morrissey I (2003) Quinupristin-dalfopristin resistance in
Streptococcus pneumoniae: novel L22 ribosomal protein mutation in two clini-
cal isolates from the SENTRY antimicrobial surveillance program. Antimicrob
Agents Chemother 47(8): 2696–2698
8 Gillis LM, White HD, Whitehurst A, Sullivan DC (2005) Vancomycin-toler-
ance among clinical isolates of Streptococcus pneumoniae in Mississippi during
1999–2001 Am J Med Sci 330(2): 65–68
9 Castanheira M, Gales AC, Mendes RE, Jones RN, Sader HS (2004) Antimicrobial
susceptibility of Streptococcus pneumoniae in Latin America: results from five
years of the SENTRY Antimicrobial Surveillance Program. Clin Microbiol
Infect 10(7): 645–651
10 Lefevre JC, Faucon G, Sicard AM, Gasc AM (1993) DNA fingerprinting of
Streptococcus pneumoniae strains by pulsed-field gel electrophoresis. J Clin
Microbiol 31(10): 2724–2728
11 Enright MC, Spratt BG (1998) A multilocus sequence typing scheme for
Streptococcus pneumoniae: identification of clones associated with serious
invasive disease. Microbiology 144 (Pt 11): 3049–3060
12 McGee L, McDougal L, Zhou J, Spratt BG, Tenover FC, George R et al (2001)
Nomenclature of major antimicrobial-resistant clones of Streptococcus pneu-
moniae defined by the pneumococcal molecular epidemiology network. J Clin
Microbiol 39(7): 2565–2571
13 Klugman KP (2002) The successful clone: the vector of dissemination of resis-
tance in Streptococcus pneumoniae. J Antimicrob Chemother 50 (Suppl S2):
1–5
14 Tomasz A (1979) From penicillin-binding proteins to the lysis and death of
bacteria: a 1979 view. Rev Infect Dis 1(3): 434–467
15 Zighelboim S, Tomasz A (1980) Penicillin-binding proteins of multiply antibi-
otic-resistant South African strains of Streptococcus pneumoniae. Antimicrob
Agents Chemother 17(3): 434–442
16 Coffey TJ, Dowson CG, Daniels M, Spratt BG (1995) Genetics and molecular
biology of beta-lactam-resistant pneumococci. Microb Drug Resist 1(1): 29–34
17 Hakenbeck R (1998) Mosaic genes and their role in penicillin-resistant
Streptococcus pneumoniae. Electrophoresis 19(4): 597–601
18 Schrag SJ, McGee L, Whitney CG, Beall B, Craig AS, Choate ME et al (2004)
Emergence of Streptococcus pneumoniae with very-high-level resistance to
penicillin. Antimicrob Agents Chemother 48(8): 3016–3023
19 Dessen A, Mouz N, Gordon E, Hopkins J, Dideberg O (2001) Crystal structure
of PBP2x from a highly penicillin-resistant Streptococcus pneumoniae clinical
isolate: a mosaic framework containing 83 mutations. J Biol Chem 276(48):
45106–45112
20 Filipe SR, Tomasz A (2000) Inhibition of the expression of penicillin resistance
in Streptococcus pneumoniae by inactivation of cell wall muropeptide branch-
ing genes. Proc Natl Acad Sci USA 97(9): 4891–4896
21 McGee L, Klugman KP, Wasas A, Capper T, Brink A (2001) Serotype 19f mul-
tiresistant pneumococcal clone harboring two erythromycin resistance deter-
Resistance in Streptococcus pneumoniae 71

minants (erm(B) and mef(A)) in South Africa. Antimicrob Agents Chemother


45(5): 1595–1598
22 Edelstein PH (2004) Pneumococcal resistance to macrolides, lincosamides,
ketolides, and streptogramin B agents: molecular mechanisms and resistance
phenotypes. Clin Infect Dis 38 (Suppl 4): S322–327
23 Pan XS, Ambler J, Mehtar S, Fisher LM (1996) Involvement of topoisomerase
IV and DNA gyrase as ciprofloxacin targets in Streptococcus pneumoniae.
Antimicrob Agents Chemother 40(10): 2321–2326
24 Balsalobre L, Ferrandiz MJ, Linares J, Tubau F, de la Campa AG (2003) Viridans
group streptococci are donors in horizontal transfer of topoisomerase IV genes
to Streptococcus pneumoniae. Antimicrob Agents Chemother 47(7): 2072–2081
25 Bast DJ, de Azavedo JC, Tam TY, Kilburn L, Duncan C, Mandell LA et al
(2001) Interspecies recombination contributes minimally to fluoroquinolone
resistance in Streptococcus pneumoniae. Antimicrob Agents Chemother 45(9):
2631–2634
26 Pletz MW, McGee L, Beall B, Whitney CG, Klugman KP (2005) Interspecies
recombination in type II topoisomerase genes is not a major cause for fluo-
roquinolone resistance in invasive Streptococcus pneumoniae isolates in the
United States. Antimicrob Agents Chemother 49: 779–780
27 Gill M, Brenwald NP, Wise R (1999) Identification of an efflux pump gene,
pmrA, associated with fluoroquinolone resistance in Streptococcus pneumoni-
ae. Antimicrob Agents Chemother 43: 187–189
28 Farrell DJ, Felmingham D (2005) The PROTEKT global study (year 4) dem-
onstrates a continued lack of resistance development to telithromycin in
Streptococcus pneumoniae. J Antimicrob Chemother 56(4): 795–797
29 Canu A, Malbruny B, Coquemont M, Davies TA, Appelbaum PC, Leclercq R
(2002) Diversity of ribosomal mutations conferring resistance to macrolides,
clindamycin, streptogramin, and telithromycin in Streptococcus pneumoniae.
Antimicrob Agents Chemother 46(1): 125–131
30 Faccone D, Andres P, Galas M, Tokumoto M, Rosato A, Corso A (2005)
Emergence of a Streptococcus pneumoniae clinical isolate highly resistant to
telithromycin and fluoroquinolones. J Clin Microbiol 43(11): 5800–5803
31 Hancock RE (1997) Peptide antibiotics. Lancet 349(9049): 418–422
32 Feldman C (2004) Clinical relevance of antimicrobial resistance in the man-
agement of pneumococcal community-acquired pneumonia. J Lab Clin Med
143(5): 269–283
33 Craig WA (1998) Pharmacokinetic/pharmacodynamic parameters: rationale for
antibacterial dosing of mice and men. Clin Infect Dis 26(1): 1–10; quiz 11–12
34 Nuermberger E, Bishai WR (2004) The clinical significance of macrolide-resis-
tant Streptococcus pneumoniae: it’s all relative. Clin Infect Dis 38(1): 99–103
35 Perez-Trallero E, Marimon JM, Iglesias L, Larruskain J (2003) Fluoroquinolone
and macrolide treatment failure in pneumococcal pneumonia and selection of
multidrug-resistant isolates. Emerg Infect Dis 9(9): 1159–1162
36 Ewig S, Ruiz M, Torres A, Marco F, Martinez JA, Sanchez M et al (1999)
Pneumonia acquired in the community through drug-resistant Streptococcus
pneumoniae. Am J Respir Crit Care Med 159(6): 1835–1842
37 Goldstein EJ, Garabedian-Ruffalo SM (2002) Widespread use of fluoroqui-
72 Mathias W. R. Pletz et al.

nolones versus emerging resistance in pneumococci. Clin Infect Dis 35(12):


1505–1511
38 Davidson R, Cavalcanti R, Brunton JL, Bast DJ, de Azavedo JC, Kibsey P et
al (2002) Resistance to levofloxacin and failure of treatment of pneumococcal
pneumonia. N Engl J Med 346(10): 747–750
39 Fuller JD, Low DE (2005) A review of Streptococcus pneumoniae infection
treatment failures associated with fluoroquinolone resistance. Clin Infect Dis
41(1): 118–121
40 Lim S, Bast D, McGeer A, de Azavedo J, Low DE (2003) Antimicrobial suscep-
tibility breakpoints and first-step parC mutations in Streptococcus pneumoniae:
redefining fluoroquinolone resistance. Emerg Infect Dis 9(7): 833–837
41 Gillespie SH, Voelker LL, Ambler JE, Traini C, Dickens A (2003)
Fluoroquinolone resistance in Streptococcus pneumoniae: evidence that gyrA
mutations arise at a lower rate and that mutation in gyrA or parC predisposes
to further mutation. Microb Drug Resist 9(1): 17–24
42 Croisier D, Etienne M, Bergoin E, Charles PE, Lequeu C, Piroth L et al (2004)
Mutant selection window in levofloxacin and moxifloxacin treatments of
experimental pneumococcal pneumonia in a rabbit model of human therapy.
Antimicrob Agents Chemother 48(5): 1699–1707
43 Pletz MW, Shergill AP, McGee L, Beall B, Whitney C, Klugman KP (2006)
Prevalence of first-step mutants among levofloxacin-susceptible invasive
Streptococcus pneumoniae in the United States. Antimicrob Agents Chemother
50(4): 1561–1563
44 Whitney CG, Farley MM, Hadler J, Harrison LH, Bennett NM, Lynfield R et
al (2003) Decline in invasive pneumococcal disease after the introduction of
protein-polysaccharide conjugate vaccine. N Engl J Med 348(18): 1737–1746
45 McEllistrem MC, Adams JM, Patel K, Mendelsohn AB, Kaplan SL, Bradley JS
et al (2005) Acute otitis media due to penicillin-nonsusceptible Streptococcus
pneumoniae before and after the introduction of the pneumococcal conjugate
vaccine. Clin Infect Dis 40(12): 1738–1744
46 Stephens DS, Zughaier SM, Whitney CG, Baughman WS, Barker L, Gay K et
al (2005) Incidence of macrolide resistance in Streptococcus pneumoniae after
introduction of the pneumococcal conjugate vaccine: population-based assess-
ment. Lancet 365(9462): 855–863
47 Pletz MW, McGee L, Jorgensen J, Beall B, Facklam RR, Whitney CG et al
(2004) Levofloxacin-resistant invasive Streptococcus pneumoniae in the United
States: evidence for clonal spread and the impact of conjugate pneumococcal
vaccine. Antimicrob Agents Chemother 48(9): 3491–3497
48 Porat N, Arguedas A, Spratt BG, Treffler R, Brilla E, Loaiza C (2004)
Emergence of penicillin-nonsusceptible Streptococcus pneumoniae clones
expressing serotypes not present in the antipneumococcal conjugate vaccine. J
Infect Dis190: 2154–2161
49 Pai R, Moore MR, Pilishvili T, Gertz RE, Whitney CG, Beall B and the Active
Bacterial Core Surveillance Team (2005) Postvaccine genetic structure of
Streptococcus pneumoniae serotype 19A from children in the United States. J
Infect Dis 192: 1988–1995
Community-Acquired Pneumonia 73
ed. by N. Suttorp, T. Welte and R. Marre
© 2007 Birkhäuser Verlag Basel/Switzerland

Influenza

Hans-Dieter Klenk

Institut für Virologie, Philipps-Universität Marburg, Hans-Meerwein-Str. 3, 35043 Marburg,


Germany

Abstract
Influenza-A-viruses have a wide host range and occur with a wide spectrum of variants
defined by 16 HA and 9 NA subtypes. All of these subtypes occur in birds, whereas only
some of them have so far been observed in man, pig, horse, and a number of other mam-
mals. In contrast, influenza-B and C-viruses occur only in man, and there are no subtypes
of these viruses. Influenza-A-viruses occasionally can be transmitted from aquatic birds,
their natural reservoir, to terrestrial birds and mammals. On rare occasions, they adapt
to the new species and establish thus new virus lineages. Adaptation requires multiple
mutations and it may involve gene reassortment after co-infection with another virus.
By these mechanisms, viruses with new surface glycoproteins and therefore a distinct
change in antigenicity are generated. If a new virus with such an antigenic shift occurs in
man, it causes a pandemic. Antigenic drift, unlike antigenic shift, is characterized by slight
changes in antigenicity resulting from successive mutations in HA and NA. Antigenic drift
is responsible for the annual human epidemics. It occurs not only with influenza-A-viruses,
but also with influenza-B viruses.
Influenza is a highly contagious disease that is transmitted by aerosols. Virus replica-
tion occurs in airway epithelia and reaches its peak 2–3 days after infection. Symptoms
typically include high fever, chills, headache, sore throat, dry cough, myalgias, anorexia,
and malaise. Complications include primary viral pneumonia, secondary bacterial pneu-
monia, or combined bacterial and viral pneumonia. Serious complications of influenza
most often occur in people 65 years of age and older, in the very young, and in those of
any age with underlying chronic cardiac, pulmonary, or metabolic disease. Vaccination is
the most potent instrument for influenza control. Prime candidates for vaccination are
persons at risk for complications and individuals who might transmit influenza to such
persons. Inactivated vaccines obtained from infected chicken embryos are most commonly
used. Neuraminidase inhibitors are the influenza antivirals of choice. Application is lim-
ited to a relatively small time window shortly before or after infection.

Introduction

Influenza viruses are segmented negative stranded RNA viruses. They


have a high genetic variability and occur with a large number of different
74 Hans-Dieter Klenk

variants in man and animals. Human influenza viruses are unique in their
ability to cause recurrent seasonal epidemics of different severity as well
as global pandemics during which acute febrile respiratory disease occurs
explosively in large parts of the population. Influenza outbreaks can be
traced back to the Middle Ages and antiquity. Particularly disastrous was
the Spanish influenza which cost more than 40 million lives in 1918 and
1919. In the 1930s, influenza viruses have been identified in man and pigs.
Twenty years later it was discovered that influenza viruses occur also in
birds. It was then possible to systematically investigate these pathogens.
Despite detailed knowledge on structure and replication of influenza
viruses, many aspects of epidemiology and pathogenesis are still poorly
understood.

Classification and structure of influenza viruses

Influenzaviruses belong to the family orthomyxoviridae. There are three


genera or types: influenza-A-viruses, influenza-B-viruses, and influenza-C-
viruses. Influenza-C-viruses do not play a major role as human pathogens.
The genome of influenza-A and B-viruses consists of eight single-stranded
RNA molecules of negative polarity each of which encodes one or two
viral proteins. The entire genome of influenza-A and B-viruses has a length
of 13,600 and 14,600 nucleotides, respectively. The individual RNA seg-
ments have a circular structure resulting from base pairing at the 3’ and
5’ ends. The segmentation of the genome is of particular biological sig-
nificance, since it is a precondition for the high genetic variability of these
viruses. The RNA genome forms together with the polymerase proteins
PB1, PB2, and PA the helical nucleocapsid core of virus particles. It is sur-
rounded by the lipid-containing viral envelope that is lined on its inner side
by the matrix protein M1 and has glycoprotein spikes on its outer surface.
Influenza-A and B-viruses have hemagglutinin (HA) spikes with receptor
binding and fusion functions and neuraminidase (NA) spikes with recep-
tor destroying activity. The fusion activity of HA depends on proteolytic
cleavage by cellular proteases and a pH dependent conformational change.
This activation process results in exposure of the fusion domain of HA and
its penetration into the target membrane and thus in membrane fusion.
There are a number of other proteins. With influenza-A-viruses, these are
the M2 protein that forms an ion channel in the viral envelope, the NS2
protein present in small amounts in virions, and the non-structural protein
NS1 (Fig. 1).
The surface glycoproteins of influenza viruses are the subtype and
strain specific antigenic determinants. They are the major viral components
responsible for eliciting protective immunity. Matrix and nucleocapsid pro-
teins show type-specific antigenicity and allow the serological differentia-
tion of types A, B, and C [1].
Influenza 75

Figure 1. Structure of influenza-virus. Upper panel: Electronmicrograph of a virus particle


(diameter 100 nm). The spikes at the surface (length 10 nm) can be seen. Lower panel: Virus
model. The envelope contains hemagglutinin (HA) and neuraminidase (NA) spikes as well
the M2 protein. The interior of the particle is formed by ribonucleoproteins, consisting of the
genomic RNA-segments, the nucleocapsid protein NP, and the polymerase proteins PB1, PB2,
and PA. The inner side of the envelope is lined by the matrix protein M1. Virions contain also
small amounts of the NS2 protein.

Replication cycle

HA initiates infection by mediating binding to neuraminic acid-containing


receptors and membrane fusion following endocytosis. The M2 ion chan-
nel plays an important role in uncoating by lowering the pH within the
virus particle and, thus, allowing dissociation of the internal components.
The nucleocapsid complexes are then transported into the nucleus, where
transcription and replication take place. The genomic RNA (vRNA) serves
as template for two different RNA species: complementary RNA (cRNA)
which is a complete copy of vRNA, and mRNA with a cap structure at the
5’ end and with the 3’ terminal nucleotides of cRNA being replaced by a
poly-A-tail. Non-coding sequences at the 3’ end of vRNA serve as primers
for mRNA synthesis. The cRNA is the template for new vRNA molecules.
The cap structures of the viral mRNAs are derived from cellular mRNA
molecules. Viral mRNA utilizes the cellular translation machinery for the
76 Hans-Dieter Klenk

synthesis of viral proteins. The virus enhances the coding capacity of its
genome by several mechanisms. These include splicing, expression of two
cistrons in tandem position, and start of translation at two different ini-
tiation codons. Translation of nucleocapsid protein, polymerase proteins,
matrix protein, and two non-structural proteins NS1 and NS2 occurs on
free polysomes. Ribonucleoproteins are assembled in the nucleus and sub-
sequently exported into the cytoplasm. The envelope proteins are translated
at the rough endoplasmic reticulum and then transported by the exocytotic
apparatus to the plasma membrane where virus particles are assembled in
a budding process. The neuraminidase mediates virus release by removing
receptors from the infected cell [2].

Epidemiology

The epidemiology of influenza is largely determined by its high genetic


variability. This is particularly obvious with influenza-A-viruses. They have
a wide host range and occur with a wide spectrum of variants defined by 16
HA and 9 NA subtypes. All of these subtypes occur in birds, whereas only
some of them have so far been observed in man, pig, horse, and a number of
other mammals. In fact, aquatic birds appear to be the natural reservoir of
influenza-A-viruses. In contrast, influenza-B and C-viruses occur only with
man, and there are no subtypes of these viruses.
The host barrier is not an insurmountable obstacle for influenza-A
viruses. Thus, they occasionally can be transmitted from aquatic birds, their
natural reservoir, to terrestrial birds and mammals with transient outbreaks
of disease. On rare occasions, they adapt to the new species and thus estab-
lish new virus lineages. Adaptation requires multiple mutations and it may
involve gene reassortment after co-infection with another virus. By these
mechanisms, viruses with new surface glycoproteins and therefore a distinct
change in antigenicity are generated. If a new virus with such an antigenic
shift occurs in man, it causes a pandemic. There were three pandemics in
the last century. After the Spanish influenza (H1N1) in 1918, an antigenic
shift in HA and NA gave rise to the Asian influenza (H2N2) in 1957, and
another shift in HA to the Hong Kong influenza (H3N2) in 1968. In 1977
the H1N1 subtype re-emerged and co-circulates now in man with H3N2
and influenza-B virus. More recently avian viruses of subtypes H5, H7, and
H9 have been transmitted, but not yet adapted to man. This includes H5N1
viruses that emerged in 1997 in South-East Asia and caused large outbreaks
in domestic and wild birds, and are now spreading into many other parts of
the world (Fig. 2).
Antigenic drift, unlike antigenic shift, is characterized by slight changes
in antigenicity resulting from successive mutations in HA and NA. Antigenic
drift is responsible for the annual human epidemics. It occurs not only with
influenza-A-viruses, but also with influenza-B viruses [1].
Influenza 77

Figure 2. Influenza-A periods in man. Circulation periods of H1N1, H2N2, and H3N2 viruses
are shown. In recent years, transmission of avian strains of subtypes H5, H7, and H9 to man
have also been observed. These viruses have not adapted yet to man. Of particular concern,
however, are the H5N1 viruses (“bird flu viruses”) that are now endemic in birds in Asia from
where they rapidly spread to other parts of the world.

Pathogenesis

Influenza viruses exhibit large differences in organ tropism and serverity


of disease. In man and other mammals, influenza usually causes respiratory
disease. In contrast, most avian viruses cause asymptomatic enteric infec-
tion, whereas a few H5 and H7 viruses lead to fowl plague or bird flu, a
highly lethal systemic disease.
Experimental studies at the molecular level have shown that many bio-
logical properties of influenza viruses contribute to their pathogenicity, such
as replication efficiency, organ tropism, spread of infection, and sensitivity
to host defense mechanisms. Thus, the immune status of the human popula-
tion and the extent of the antigenic differences between viruses are largely
the reason that new pandemic viruses induce more severe disease than
interpandemic ones. In each phase of the viral life cycle, there are specific
interactions between viral proteins and host factors, such as cell receptors,
nuclear proteins, and proteases. The role of these interactions in pathogen-
esis is particularly evident with proteolytic activation of HA. The highly
pathogenic avian viruses are activated by the protease furin that is ubiq-
78 Hans-Dieter Klenk

Figure 3. Cells infected with influenza virus in human airway epithelium. Ciliae are stained
black. Cells infected with influenza virus A/Memphis/14/96 (H1N1) are stained brown. Virus
infection targets specifically non-ciliated cells [5].

uitous and therefore promotes rapid virus spread within the organism. In
contrast, the viruses causing local infection including the human influenza
viruses are activated by proteases confined to specific tissues. Staphylococcus
aureus, Streptococcus pneumoniae, and Haemophilus influenzae as well as
several other bacteria secrete also HA activating proteases. After co-infec-
tion with these microorganisms, influenzavirus infections show therefore a
particularly severe course of disease [3]. Other mechanisms that contribute
to pathogenicity are the interferon antagonism of the NS1 protein and an
activity optimum of the viral polymerase probably also depending on host
factors.
Examination of tissues obtained from patients revealed that viral repli-
cation can occur throughout the respiratory tract. These studies also indi-
cated that ciliated epithelial cells are a primary site of infection, whereas
recent studies carried out on cultured airway epithelia revealed that human
influenza viruses target specifically non-ciliated cells (Fig. 3). Inflammation
of the larynx, trachea, and bronchi, and desquamation of ciliated columnar
epithelium into the lumen of the bronchus was observed in individuals with
uncomplicated acute influenza infections.
Influenza 79

Regeneration of the respiratory epithelial cells takes ca. 3–4 weeks,


during which time pulmonary function abnormalities may persist. In these
typical cases of influenza in which infection is confined to the respiratory
tract, prostration, fever, and myalgia often seem to be disproportionate to
objective clinical signs or observed pathological changes.
Lungs from fatal cases of primary viral pneumonia most notably show
hyaline membrane coverage of alveolar walls together with extensive
intra-alveolar edema and hemorrhage. Tracheitis and bronchitis are also
observed. Patients with secondary bacterial pneumonia have changes char-
acteristic of bacterial pneumonia in addition to the tracheobronchial find-
ings of influenza.

Immune response

Innate immunity is an important defense mechanism early in infection.


There are profound changes in the expression of a large number of cel-
lular genes involved in interferon production and signaling, apoptosis,
and oxidative stress. Influenza-infected patients trigger the activation
of antiviral genes and production of cytokines, such as interleukin (IL)-
6, IFN-alpha, and IL-8. IL-8 is correlated with lower respiratory tract
involvement. Influenza viruses have evolved mechanisms to counter the
innate antiviral responses of the host. The viral NS1 protein antagonizes
interferon production and the activation of PKR in influenza-infected
cells [1].
The adaptive immune response to infection with influenza viruses leads
to induction of both virus-specific B- and T-cell immune responses that
clear infection and generate long-lasting specific immunologic memory.
Antibodies are made against the viral external glycoproteins HA and NA
as well as the internal type-specific proteins NP and M1. Neutralizing anti-
body directed against the HA is the primary immune mediator of protec-
tion from infection and clinical illness due to influenza viruses. Antibodies
and T-cells play complementary roles in clearing the infection and promot-
ing recovery. CD4 + (Th1 and Th2) and CD8 + T-cell responses to influenza
are type-specific and are largely cross-reactive among influenza A viruses
of different subtypes. In naturally infected humans the CD4 + cell response
recognizes epitopes on the internal proteins NP and M1 as well as the sur-
face proteins NA and HA. The CD8 + cytotoxic T lymphocyte response is
directed to multiple surface and internal influenza proteins, in which many
epitopes are potentially recognized, as determined by their binding to class
I MHC molecules. Recent animal model studies have shown that both
CD4 + and CD8 + T-cells can contribute to immunity to influenza viruses.
The increased severity of influenza infections in geriatric patients has been
correlated with a combination of functional impairments in their T-cell
responses to influenza [4].
80 Hans-Dieter Klenk

Clinical manifestations

Influenza is a highly contagious disease that is transmitted by aerosols.


Virus replication occurs in airway epithelia and reaches its peak 2–3 days
after infection. Patients shed virus for about 7 days, with primary infections
for up to 2 weeks. Early symptoms in adults typically include fever, chills,
headache, sore throat, dry cough, myalgias, anorexia, and malaise. A fever
of 38–40°C that peaks within 24 h of onset is common, but peaks as high as
41°C can also occur. Pyrexia typically lasts 3 days, but may last from 1 to 5
days or longer. Other symptoms that occur less frequently include subster-
nal soreness, photophobia and other ocular symptoms, nausea, abdominal
pain, and diarrhea. Although most symptoms typically resolve within a
week, cough and malaise may persist for 1 or more weeks after fever has
subsided. In children, symptoms are similar to those in adults, but gastroin-
testinal symptoms such as vomiting, abdominal pain, and diarrhea are seen
more frequently. Maximum temperatures also tend to be higher in children
than in adults, and febrile convulsions can occur. In addition, myositis, croup,
and otitis media occur more frequently in children. Influenza infection of
neonates can be life-threatening and may be manifest only as an unex-
plained febrile illness.
Complications of the upper respiratory tract after influenza infection
include bacterial sinusitis and otitis media. Lower respiratory tract compli-
cations include exacerbation of chronic obstructive pulmonary disease and
chronic congestive heart failure, croup, bronchitis, bronchiolitis, wheezing
attacks in asthmatics, and pneumonia (primary viral pneumonia, secondary
bacterial pneumonia, or combined bacterial and viral pneumonia). Primary
influenza pneumonia develops abruptly and progresses rapidly. It has been a
frequent cause of death in the 1918 pandemic and is also responsible for the
high case fatality rate of the human infections with the ongoing H5N1 out-
break. However, with ordinary influenza epidemics, this type of pneumonia
is uncommon and occurs mainly among those at increased risk for complica-
tions of influenza. Rapid respiration rate, tachycardia, cyanosis, high fever,
and hypotension are frequent symptoms. Diffuse pulmonary infiltrates and
acute respiratory failure with a high mortality rate are also features of this
disease. Combined viral and bacterial pneumonia is more common than
primary viral pneumonia and may be clinically indistinguishable from it.
Secondary bacterial infections typically occur 5–10 days after initial onset
of influenza symptoms and are responsible for most pneumonias during
influenza epidemics. Productive cough, pleuritic chest pain, and chills are
common symptoms of this type of pneumonia. Streptococcus pneumoniae,
Staphylococcus aureus, and Haemophilus influenzae are the organisms most
commonly involved. These illnesses respond to appropriate antimicrobial
agents and have a lower case fatality rate than primary viral pneumonia.
Other reported but less frequent complications of influenza include
myositis, myocarditis and pericarditis, acute renal failure, encephalopathy,
Influenza 81

encephalitis, transverse myelitis, Guillain-Barré syndrome and a range of


other neurological complications, Reye’s syndrome, and toxic shock syn-
drome. Higher rates of spontaneous abortion, stillbirths, and premature
births were reported among pregnant women during the major pandemics
of 1918/19 and 1957/58.
Serious complications of influenza most often occur in people 65 years
of age and older, in the very young, and in those of any age with underlying
chronic cardiac, pulmonary, or metabolic disease. Complications in elderly
people, particularly among those with pulmonary, cardiovascular, or other
chronic diseases, account for most of the mortality in influenza epidemics
[4].

Laboratory diagnosis

Virus is isolated from nose or throat secretions by inoculation of cell cul-


tures (MDCK cells) or embryonated eggs. Isolation is usually only success-
ful within the first days of infection. Rapid diagnosis is possible by antigen
detection in cells obtained from throat swabs. Viral RNA can be detected by
PCR in clinical specimens. This approach is now replacing virus culture as
“gold standard” for virus detection, because it is more sensitive, more rapid,
and allows more precise characterization.
Serological diagnostics involves complement fixation, hemagglutina-
tion inhibition, neutralization, and enzyme immunoassays. Antibody titres
increase rapidly in the course of infection and decrease within a couple of
weeks to low levels. It is often impossible to detect a fourfold increase in
titre in blood samples taken with an interval of 2 weeks, since high antibod-
ies reach high titres already in the acute disease phase. Therefore a single
high antibody titre matched by the typical clinical symptoms is usually con-
sidered to be sufficient for the diagnosis of the infection.

Prophylaxis and therapy

Vaccination is the most potent instrument for influenza control. Prime can-
didates for vaccination are persons at risk for complications and individuals
who might transmit influenza to such persons. Inactivated vaccines obtained
from infected chicken embryos are most commonly used. They contain HA
and NA antigens as protective components and are each year formulated
according to the circulating influenza-A and B strains. Because of antigenic
drift, vaccination protects only against known viruses. Vaccinations should
therefore be done every year. Current vaccines protect 70–90 percent of
healthy adults. With older people, i.e. the most important target group, effec-
tiveness is lower. Live vaccines are only available in some countries, such as
the USA. Development of pandemic vaccines is a particular challenge, since
82 Hans-Dieter Klenk

they will have to be manufactured within a very short time period in very
large quantities.
Antiviral compounds were previously available only in the form of
amantadine and rimantadine. Both compounds block the M2 ion channel
of influenza-A-viruses and are therefore not effective against influenza-B-
virus infections. Prophylactic application provides 70–90 percent protec-
tion. Concerns about side effects involving the central nervous system have
limited the use of amantadine. Development of amantadine resistant virus
strains, which have emerged recently with increasing frequency, is another
problem.
The neuraminidase inhibitors zanamivir and oseltamivir that interfere
with virus spread compare favorably with amantadine by having less side
effects and a wider therapeutic spectrum that includes also influenza-B
viruses. The neuraminidase inhibitors are therefore now the influenza anti-
virals of choice. However, as is the case with amantadine, application is lim-
ited to a relatively small time window shortly before or after infection, and
there is increasing evidence for the development of virus resistance to these
drugs, too. Nevertheless, neuraminidase inhibitors are expected to play an
important role in the control of a future pandemic.

References
1 Cox NJ, Neumann G, Donis RO, Kawaoka Y (2005) Orthomyxoviruses: influ-
enza. In: BWJ Mahy, V ter Meulen (eds): Topley and Wilson’s microbiology and
microbial infections, Virology, Vol. 1. Hodder Arnold, London, 634–698
2 Noah DL, Krug RM (2005) Influenza virus virulence and its molecular deter-
minants. Adv Virus Res 65: 121–145
3 Klenk H-D, Garten W (1994) Host cell proteases controlling virus pathogenic-
ity. Trends Microbiol 2: 39–43
4 Nicholson KG (1998) Human influenza. In: KG Nicholson, RG Webster, AJ
Hay (eds): Textbook of influenza. Blackwell Science, Oxford, 219–264
5 Matrosovich MN, Matrosovich TY, Gray T, Roberts NA, Klenk H-D (2004)
Human and avian influenza viruses target different cell types in cultures of
human airway epithelium. Proc Natl Acad Sci USA 101: 4620–4624
Community-Acquired Pneumonia 83
ed. by N. Suttorp, T. Welte and R. Marre
© 2007 Birkhäuser Verlag Basel/Switzerland

Pathogenesis of Chlamydophila pneumoniae infections –


epidemiology, immunity, cell biology, virulence factors

Matthias Krüll and Norbert Suttorp

Dept. Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité


Universitätsmedizin Berlin, Augustenburgerplatz 1, 13353 Berlin

Abstract
Chlamydophila (Chlamydia) pneumoniae, a Gram-negative obligate intracellular bacte-
rium, is a widespread respiratory pathogen causing sinusitis, pharyngitis, bronchitis and
pneumonia. Repetitive or chronic persistent infections have been associated with an
increased risk for asthma, chronic obstructive pulmonary disease (COPD) or vascular
lesions. Although the genome of C. pneumoniae has been sequenced completely this
information has not led yet to an understanding of the mechanisms of infection and target
cell activation nor to the identification of potential chlamydial virulence factors. In this
review we will give an overview on the pathogenesis of C. pneumoniae-induced acute and
chronic infections.

Epidemiology, diagnosis and clinical manifestation

Chlamydophila pneumoniae, a Gram-negative obligate intracellular bac-


terium, is a widespread respiratory pathogen causing sinusitis, pharyngitis,
bronchitis and pneumonia [1–3]. The majority of C. pneumoniae infections
are subclinical, but severe pulmonary infection and profound lymphocytic
alveolitis are observed [4]. In addition, chronic-persistent or recurrent infec-
tions may be an important risk factor for adult-onset asthma [5], chronic
obstructive lung disease (COLD, [6]) and development of vascular lesions
[7–10]. Regarding vascular diseases, the field, however, is troubled by the
“hen vs. egg” problem and causative proof is difficult because besides dif-
ferent anti-chlamydial isotypes of antibodies there are no good markers to
differentiate among new vs. old (IgM vs. IgG) as well as acute vs. chronic
persistent (IgM vs. IgA) C. pneumoniae infection.

Epidemiology

C. pneumoniae was first isolated 1965 from the conjunctiva of a Taiwanese


child (strain TW183, [11]). The first respiratory strain (AR 39) could be
84 Matthias Krüll and Norbert Suttorp

isolated 1983. Both strains had a DNA-homology of > 99.5% and were
established in 1986 as a new (third) human-pathogenic chlamydia species
“Chlamydia pneumoniae,” synonym TWAR [1]. Until now about 50 strains
have been isolated worldwide, DNA-homology between these strains is 94-
100%.
Chlamydiae have been placed in their own order, Chlamydiales, with one
family, Chlamydiaceae. Molecular evaluation of rRNA sequences confirms
that chlamydiae are bacteria, but with only very distant relationship to other
bacterial divisions [12, 13]. Although it has been proposed that Chlamydia
trachomatis and Chlamydia pneumoniae represent different genera [14],
their gene content and genome organization are extremely similar [15] as
are their structure and biology [16]. The newly proposed nomenclature – as
used throughout this chapter – “Chlamydophila pneumoniae”, however, has
not been generally accepted [17].
Chlamydophila pneumoniae is found worldwide. Seroepidemiologic
surveys have demonstrated that more than 70% of all adults have been
exposed to this organism during their lifetimes [18]. Age-specific prevalence
rates start to rise early in childhood, although there is only little disease
associated with these early timepoints of infection. The most rapid rise (in
the Western world) in age-specific prevalence occurs during the ages of 5- to
20-years.
In spite of a high percentage of C. pneumoniae-seropositive adults
reinfections are common. According to these seroepidemiologic studies,
C. pneumoniae infection seems to be both endemic and epidemic with fre-
quent reinfection during a lifetime. Currently available data suggest that
C. pneumoniae is primarily transmitted from human to human without any
animal reservoir. Transmission seems to be inefficient, although household
outbreaks with high transmission rates are reported [19].

Diagnosis

For differential diagnosis of an acute, repetitive or chronic/persistent C.


pneumoniae infection the kinetics of the antibody expression has to be
taken into consideration. In patients with primary infection IgM antibod-
ies appear about 2–3 weeks post infection and remain elevated/detectable
up to 2–6 months. IgG antibodies may not reach high titers until 6-8 weeks
after onset of clinical symptoms. C. pneumoniae does not induce a persistent
protective immunity and reinfections are common. In case of reinfection,
the level of IgG antibodies increase rapidly within 1–2 weeks while IgM
antibodies may not appear again [19]. Until now there is no reference test
for validating a chronic/persistent infection by means of serological testing.
Persistently elevated IgG or the presence of IgA antibodies has been sug-
gested as potential serological marker. Several studies have proposed that
high IgA titers might be a better marker for a chronic infection than IgG
Pathogenesis of Chlamydophila pneumoniae infections… 85

titers because serum IgA has a half- life of about 5–7 days, whereas IgG
has a half-life of weeks to months [9, 20]. Further evaluation, however, and
confirmation by other tests have to be awaited.
A lack of validated and standardized diagnostic techniques in diagnosis
of acute, recurrent or chronic persistent infection with C. pneumoniae has
made interpretation of published data difficult. Recommendations for stan-
dardized approaches were recently published by the Centers for Disease
Control and Prevention (CDC, Atlanta) and the Laboratory Centre for
Disease Control (Ottawa, Canada) in cooperation with members of the C.
pneumoniae-study group [21]. Diagnostic approaches include serological
testing, isolation/culture, nucleic acid-based amplification techniques such
as PCR, and tissue diagnostics like immunofluorescence, immunohisto-
chemistry (IHC) or in situ hybridization.

Serological testing

The microimmunofluorescence (MIF) test, developed by Wang et al. in the


1970s [22] is currently the serological testing method of choice for diagnosis
of acute C. pneumoniae infection. This test allows the quantitative deter-
mination of antibody reactivity to formalinized elementary bodies from C.
pneumoniae, C. trachomatis and C. psittaci fixed as dots on a single glass
slide. Dilution of sera are placed over the antigen dots and incubated. Use
of the MIF test allows definition of criteria for serologic evidence of acute
infection (defined by a four-fold rise in IgG between acute and convalescent
samples or an acute IgM titer * 1: 16), or past exposure (indicated by an
IgG titer * 1: 16). Kits based on the MIF format are commercially available,
however, it should be noted that the quality of commercially available MIF
kits varies and interpretation of the results is subjective [23]. Due to the
lack of species specificity or sensitivity, other serological tests like comple-
ment fixation (CF), whole fluorescence, ELISA and EIA cannot currently
be endorsed [24, 25].

Culture

Chlamydophila pneumoniae is an obligate intracellular pathogen and


must be cultivated on specific eukaryotic host cells. All currently estab-
lished culture techniques involve inoculation of specimens onto a human
cell line via centrifugation, incubation for up to 72 h and subsequent
staining with fluorescent-labeled species-specific antibodies to visualize
intracellular inclusion bodies [8, 26, 27] (see Fig. 1). Sensitivity is low due
to the complexicity of the assay. Specificity is dependent on the ability
of the lab staff to distinguish between true chlamydia inclusions and
artifacts.
86 Matthias Krüll and Norbert Suttorp

Figure 1. Chlamydia pneumoniae strain TW183 inclusion bodies in freshly isolated human
airway epithelial cells. Inclusion bodies were visualized, using a fluorescein isothiocy-
anate (FITC)–conjugated, genus-specific monoclonal antibody (DakoCytomation, Hamburg,
Germany).

PCR

Although many in-house PCR techniques for detection of C. pneumoniae


have been published [28-31], sensitivity and specificity remain almost
unknown. Due to a broad variety of methods for specimen-collection and
specimen-processing, primer design, nucleic acid extraction, amplification
product detection and identification of false-positive and false-negative
results, no commercially standardized tests have been approved by the
FDA.

Tissue diagnostics/immunohistochemistry (IHC)

Tissue diagnostic methods offers the advantage of preserving tissue mor-


phology and permitting localization of the infectious agent to specific areas
and cells. IHC has been the most frequently used in published studies [32,
33]. Detection rates are higher than those of PCR. This is attributed to a
faster degradation of DNA, difficulties of extracting DNA from tissues and
the presence of PCR inhibitors. The most widely used IHC-technique is
the avidin-biotinylated immune-complex method. However, interpretation
Pathogenesis of Chlamydophila pneumoniae infections… 87

of IHC-results is a critical challenge due to difficulties in distinguishing


between true- and false-positive results of the staining.
Specific recommendations for standardized culture/isolation or detec-
tion tests (e.g., specimen-collection, -transport and -processing conditions)
have been suggested elsewhere (s.a., [21]).
Taken together, diagnosis of C. pneumoniae infection is difficult because
cell culture techniques are not available for routine clinical use and non-
culture techniques using antigen detection methods (IHC) or DNA probes
have not been developed for commercial use. The MIF test is currently the
serological testing method of choice for diagnosis of acute C. pneumoniae
infection (“gold standard”), although the test is technically complex, requir-
ing experience in fluorescence microscopy, interpretation is therefore sub-
jective.

Clinical manifestation

Chlamydophila pneumoniae causes acute respiratory diseases and is


responsible for approximately 5% of bronchitis and sinusitis cases and 5-
10% of community acquired pneumonia (CAP) cases in adults worldwide
[34–37] (see Fig. 2). However, a broad geographic diversity and periodicity
with higher incidence rates of C. pneumoniae-mediated CAP-cases for 2
or 3 years followed by 4 or 5 years with low incidence has been suggested
[18]. A recently submitted study from the German “community acquired
pneumonia-network, CAPNETZ”-study group, including more than 4000
CAP-patients in Germany demonstrated, that between 2001 and 2004 using
recommended standardized diagnosis protocols (MIF test, PCR from BAL-
fluid) C. pneumoniae could be identified as causative agent in < 1 % of all
CAP-cases (personal communication N. Suttorp, speaker of CAPNETZ).
C. pneumoniae infection has also been implicated in the pathogenesis of
asthma in both adults and children. This hypothesis is based on clinical stud-
ies and on the evidence of specific IgE production, direct epithelial dam-
age, induction of T-cell immunopathologic diseases, and vascular smooth
cell infection. In addition, asthma patients, especially those with moderate
asthma, had significantly higher serum IgA antibody levels to chlamydial
heat shock protein 60 (cHsp60) than healthy controls [5, 38–41]. Moreover,
recurrent or chronic persistent C. pneumoniae infection seems to be com-
mon in patients with chronic bronchitis whether exacerbated or not, and is
characterized by a strong humoral immune response to this intracellular
pathogen, which is present in the majority of patients with severe chronic
bronchitis. Increased antichlamydial IgG and IgA indicated acute exacerba-
tions with C. pneumoniae in COPD patients [5, 42-44]. Some authors suggest
a possible role of this organism in the etiology of lung cancer; future studies,
using measures of chronic C. pneumoniae status, however are necessary, to
substantiate these data [45, 46].
88 Matthias Krüll and Norbert Suttorp

Figure 2. Chest x-ray of a 28-year-old man reveals basal infiltration in both lungs as well as a
generally pronounced interstitial pattern. Chlamydophila pneumonia infection could be con-
firmed by MIF test and PCR from BAL-fluid.

Chronic-persistent or recurrent Chlamydophila pneumoniae infections


may be a trigger and promoter of inflammation which may cause vascular
lesions and atherosclerosis [8, 9, 31, 47, 48]. The theory is supported by a
serological association between C. pneumoniae infection and coronary heart
disease as well as other vascular diseases (arterial occlusive disease, carotid
artery stenosis and stroke [9, 20, 49, 50]), the demonstration of C. pneumoni-
ae in atherosclerotic plaques by electronmicroscopy, immunocytochemistry,
PCR, and isolation of viable chlamydia (indicating a productive chlamydial
infection [8, 10, 29, 31, 51, 52]), and different animal models, demonstrating
that intranasal infection of mice and rabbit with C. pneumoniae leads to
pneumonia, perimyocarditis, septic circulatory dysregulation and – more
delayed – systemic spread of chlamydia into spleen, lymphnodes, peritone-
um and atherosclerotic plaques of arterial blood vessels [7, 53–56]. Current
antibiotic treatment options for acute chlamydial infection, however, were
Pathogenesis of Chlamydophila pneumoniae infections… 89

proven to be ineffective with respect to clinical outcome in different groups


of atherosclerotic patients (WIZARD, AZACS, ACES or PROVE-IT study
[55, 57–62]). Interpretation of these clinical trials is challenging as definite
markers of chronic vascular C. pneumoniae infection are still missing and
serologic testing seems to be inaccurate in patient populations with high
chlamydial IgG seroprevalence. Moreover and probably more important, C.
pneumoniae in the persistent state, as observed in blood monocytes, shows
high resistance against macrolides, tetracyclines and rifampicin [63–65]. This
observation may explain the ineffectiveness of all interventional studies in
atherosclerotic patients.
Several recent studies have suggested a chronic/persistent C. pneumoni-
ae infection as a possible risk factor for central nervous system diseases like
Alzheimer’s disease or multiple sclerosis, data, however, are poor and clear
evidence is still missing [66, 67].

Innate and adaptive immunity

Little is known about a humoral or cell-mediated immunity (“CMI”)


induced during acute or chronic/persistent infection with Chlamydophila
pneumoniae. Most of the data have been elaborated using other Chlamydiae
species (C. trachomatis, C. psittaci). As intracellular bacteria, Chlamydiae
pose an extra challenge for the defense mechanisms of the host. While
importance of anti-chlamydial antibodies still is controversially discussed
[68], cell-mediated immune responses are decisive, at least in mice. CMI
against C. pneumoniae, however, is weak since recurrent infections as well
as persistency of viable pathogens in different target cells are common
phenomena.

Innate immunity

C. pneumoniae is able to infect and to replicate in a multitude of target cells


[69–71]. Subsequently, different intracellular signal transduction pathways
are activated to induce a proinflammatory phenotype (for review see [72]).
However, there is still limited knowledge of the mechanisms of Chlamydiae
entry into host cells. Wuppermann et al. showed that heparan sulfate-like
glycosaminoglycans (GAG) might act as possible chlamydial receptors on
the surface of epithelial cells (HEp-2 cells), the importance of the estrogen
receptor complex is controversially discussed [73, 74]. In a recent paper,
Puolakkainen et al. demonstrated, that C. pneumoniae uses the mannose 6-
phosphate/insulin-like growth factor 2 receptor for infection of endothelial
cells, additional studies, however are necessary to confirm these results [75].
Several recent studies demonstrated the involvement of extracellular Toll-
like receptor-2 (TLR2) and -4 (TLR4) in initiation of innate immune cell
90 Matthias Krüll and Norbert Suttorp

activation by C. pneumoniae or chlamydial components [76–80]. Moreover,


we were able to demonstrate that the recently identified nucleotide-binding
oligomerization domain (Nod) proteins might act as intracellular receptors
during C. pneumoniae-infection of target cells [81].
C. pneumoniae is internalized by macrophages as well as by nonprofes-
sional phagocytes (innate cells), where it survives and replicates [64]. In these
cells IFN-a synergizes with bacterial products to activate various bactericidal
or bacteriostatic mechanisms [82]. IFN-a is a strong activator of indoleamine
2,3-dioxygenase (IDO), limiting the availability of L-tryptophan to intra-
cellular microorganisms [83]. Induction of IDO has been demonstrated to
inhibit chlamydial growth in vitro [84, 85]. IFN-a can also activate inducible
nitric oxide synthase (iNOS) which catalyzes production of NO from L-argi-
nine. Inhibition of chlamydial growth through induction of iNOS has also
been reported [86]. Moreover, stimulation of neutrophils or monocytes with
IFN-a induced transcription of the gp91 component of NADPH oxidase (ox)
mRNA with subsequent enhanced respiratory burst of phagocytic cells [82].
In addition, Rottenberg et al. could demonstrate that IFN-a-receptor-double
knock-out mice (IFN-a-R–/–) showed a dramatically increased susceptibility
to C. pneumoniae, mediated via a reduced iNOS-mRNA accumulation, inde-
pendent of diminished levels of specific antibodies. An increased susceptibil-
ity of iNOS–/– mice substantiated the protective role of this enzyme-activity
during infection with C. pneumoniae [87]. These data suggested a relevant
protective role of IFN-a-dependent innate mechanisms of protection. For
a more extended view of the signal transduction pathways involved in
Chlamydia-mediated innate immune cell activation see Figure 3, in addition
we would like to refer to our recent review [72].

Adaptive immunity

In C. trachomatis infection models, both CD4+ and CD8+ cells have been
shown to confer protection, although the former are considered of major
importance [88, 89]. In C. psittaci infection, CD8+ rather than CD4+ cells
have been reported to confer protection in mice [90].
Since the overall DNA homology between C. pneumoniae and C. tracho-
matis or C. psittaci is less than 5 or 10%, respectively [15] the parameters of
infection identified with the later two cannot be directly extrapolated to C.
pneumoniae.
Most data have been acquired using mouse models for C. pneumoniae-
mediated pneumonia [91, 92]. The situation, however, is complicated due to
mouse strain-specific differences studying mechanisms of adaptive immu-
nity. In addition, importance for a protective role of T-cell subspecies during
primary infection and reinfection still is controversially discussed.
Using C57BL/6J mice genomically lacking T-cell coreceptors or cytokine
receptors, Rottenberg et al. demonstrated that CD4 + T-cells played a dual
Pathogenesis of Chlamydophila pneumoniae infections… 91

Figure 3. Scheme of the supposed signaling cascades induced by C. pneumoniae infection in


human cells (TyrKi, tyrosin kinase; ERK, extracellular regulated kinase; PKC, protein kinase C;
reproduction from [75] courtesy of the authors and of Schattauer Verlag, Germany).

role, one deleterious, promoting bacterial growth and disease early after
infection, but also participating in the control of bacterial growth at later
timepoints as well as in protection against reinfection. The early damag-
ing effect of CD4 + cells in the absence of CD8 + cells was associated with
enhanced IL-4 and interleukin10 (IL-10) mRNA levels and delayed IFN-a
mRNA accumulation in the lungs of mice [87]. CD8 + T cells inhibited this
CD4 + activity. The CD8 + T-cell mediated protective immunity during early
stages of primary infection was perforin independent and associated with
an altered cytokine balance as indicated by increased IL-4 and IL-10 and
delayed accumulation of IFN-a mRNA in CD8–/– mice. The early higher
susceptibility of CD8-deficient mice correlated with an immune deviation
from a normal Th1 response to a Th2 cytokine pattern [87]. These results
were confirmed by Wizel et al. demonstrating the importance of peptide-
specific CD8 + T-cell lines in local and systemic compartments after primary
(intranasal) infection with C. pneumoniae. These CTL lines suppressed
chlamydial growth in vitro by direct lysis of infected target cells and by
secretion of IFN-a. In addition, they were able to identify 18 H-2(b) binding
peptides representing sequences from 12 C. pneumoniae antigens [93]. The
importance of CD8 + CTL during reinfection could be confirmed by Penttilä
et al. They demonstrated in a BALB/c nude mice model with absence of
92 Matthias Krüll and Norbert Suttorp

all T-cells, that the overall clearance kinetic after primary C. pneumoniae
infection was not dependent on either CD4 + or CD8 + cells alone [94], but,
after reinfection, acquired immunity was strongly CD8 dependent with an
enhanced IFN-a-production and an increased local lymphoid reaction in the
lungs [94, 95].
Analyzing lymphocytes from male patients with respiratory tract infec-
tion Halme et al. demonstrated a C. pneumoniae-specific CMI response
during acute, primary infection early after onset of disease symptoms
and simultaneously with a humoral response (increase of C. pneumoniae-
specific IgM-antibodies). C. pneumoniae-induced lymphocyte activation
involved CD8 + T-cells in the early phase of infection and CD4 + cells in the
later stage [96].
The mechanism underlying the protection mediated by the CD8 + cells
in C. pneumoniae infection is unclear. CTL specific for C. trachomatis have
been demonstrated in C. trachomatis-infected mice [97, 98]. CD8 + cells may
function by secreting cytokines such as IFN-a. Rottenberg et al. showed
that C57BL/6 mice produce IFN-a in response to C. pneumoniae primary
infection and suggested a IFN-a-mediated protection mechanisms [82].
These results were supported by Vuola et al. demonstrating an altered bac-
terial load in anti-IFN-a-treated C57BL/6 mice with markedly exacerbated
signs of pulmonary inflammation [99]. Results are different in BALB/c
mice. Pentillä et al. and Vuola et al. demonstrated an IFN-a-independent
cellular response in BALB/c mice [95, 99]. Interestingly although BALB/c
mice appear not to be dependent on IFN-a during primary infection, they
do not develop a typical Th2 type response either [99]. During reinfection,
neutralization of IFN-a exacerbated the infection in both strains [82, 95].
CD8 + cells may therefore at least partially function through IFN-a produc-
tion, however actively IFN-a-producing cells have also been demonstrated
in CD8-depleted mice, in which the acquired immunity seen during reinfec-
tion is abolished [94]. Thus, IFN-a is an important, but not the only effector
mechanism in acquired immunity.
Overall, a different mouse model of C. pneumoniae-infection demon-
strated that immunity is critically dependent on CD8 + CTL. In a recent
study the group of Wizel et al. was able for the first time to successfully
immunize C57BL/6 mice with a CD8 + T-cell heptaepitope based DNA
vaccine to induce a protective immunity against C. pneumoniae [100].
These results were confirmed by Pentillä et al., demonstrating that DNA
immunization is a promising possibility for developing much wanted vac-
cines against important chlamydial pathogens [101]. Further studies are
now required to elaborate the optimal design of (multicomponent) anti-C.
pneumoniae vaccines for humans.
Infection with C. pneumoniae induces a strong serum response. Little
is known about the immunogenic antigens of C. pneumoniae. The 40 kDa
major outer membrane protein (MOMP) is the most important immuno-
dominant structure during C. trachomatis-infection; during infection with C.
Pathogenesis of Chlamydophila pneumoniae infections… 93

pneumoniae it is a relatively immunorecessive antigen [102, 103]. This lack


of antigenicity has not been resolved yet. Among the different antigens of
C. pneumoniae the 60 kDa heat-shock protein 60 (cHsp 60, GroEL-1) has
been suggested to be a key player during C. pneumoniae-induced humoral
immunity, further studies, however, are required to identify the importance
of this antigen during acute and chronic infection (see also the chapter on
“virulence factors”) [102, 104]. Antibodies to different structures on the C.
pneumoniae elementary body can neutralize the organism in cell culture,
however, the epitope specificity of these neutralizing monoclonal antibodies
to specific C. pneumoniae proteins remains undefined at a molecular level
[105]. Moreover, in vivo, protective effects of antibodies seem to be weak
since reinfections or chronic chlamydial infections are common despite high
antibody titers [18, 19].
Under some circumstances, antibody response during C. pneumoniae-
infection might be immunopathological; for details please see chapter
“virulence factors”.

Cell biology

Monocytes, macrophages, smooth muscle cells, endothelial cells, human air-


way epithelial cells (HAEC), BEAS-2B (bronchoepithelial cell line), HEp-2
and Hela-229 cells have all been shown to be susceptible for C. pneumoniae
infection [69-71]. Following inhalation, bronchial epithelial cells, however,
are the first line of defense in getting in contact with C. pneumoniae and
respiratory epithelium has been identified as the primary target of infec-
tion [70, 71]. Little is known about Chlamydiae-induced epithelial cell
alterations and C. pneumoniae-mediated interactions among all cell types
involved in orchestrating airway inflammation (e.g. lymphocytes, macro-
phages, granulocytes).
Chlamydiae have a unique development cycle with two functionally
and morphologically distinct forms, the condensed, “spore-like” infectious
but metabolically inactive elementary body (EB, 0.3 +m) and the labile,
noninfectious metabolically active reticulate body (RB, 0.9 +m, see Fig. 4)
Infection or invasion is an active process requiring the existence of viable
chlamydia, heat- or UV-inactivated bacteria are not able to invade tar-
get cells. However, there is still limited knowledge of the mechanisms of
Chlamydiae entry into host cells. The chlamydial growth cycle is initiated
when an infectious elementary body (EB) attaches to a susceptible target
cell, promoting entry into a host cell-derived phagocytic vesicle. EB are
internalized, dissociated from the endocytotic pathway by actively modify-
ing the vacuole to become fusogenic with exocytic vesicles. Interaction with
this secretory pathway appears to provide a pathogenic mechanism that
allows chlamydia to establish themselves in a site that is not destined to
fuse with lysosomes [63, 106]. Coombes and Mahony suggested a receptor-
94 Matthias Krüll and Norbert Suttorp

Figure 4. Transmission electron micrographs of Chlamydophila pneumoniae-infected HL cells


72 h postinfection. Large inclusions filled with C. pneumoniae particles are found in the HL
cells. Mature EBs are small with an electrodense core (thick arrow), while RBs are large with
a coarse inner structure (thin arrow, reproduction from [157] courtesy of the authors and the
American Society for Microbiology).

mediated induction of specific cell signaling by Chlamydiae as an essential


step in C. pneumoniae invasion of epithelial cells [107]. After internaliza-
tion EB then develop into reticular bodies (RB), a process which could
be detected metabolically within 15 min and microscopically 12-15 h after
addition of Chlamydiae to HEp-2 and Hela-229 cells [70, 71]. RBs are first
observed dividing by binary fission after about 12 h. As the RBs multiply,
the inclusion membrane expands to accommodate the increasing numbers
of bacteria. After about 18-24 hpostinfection, the first RBs begin differen-
tiating back into EB which accumulate in the lumen of the inclusion as the
remainder of the RBs continue to multiply. Forty-eight to 72 h after com-
pleting the development cycle, depending primarily on the infecting species,
infectious EB are released either by lysis of the host cell or by fusion of the
inclusion membrane with the plasma membrane to release the content of
the inclusion into the environment (for review see [108, 109]).
Under some conditions, however, such as in the presence of IFN-a, or
penicillin or depletion of essential nutrients (iron, tryptophan) Chlamydiae
Pathogenesis of Chlamydophila pneumoniae infections… 95

can achieve a state of intracellular chronic, persistent infection in which they


remain viable, but metabolically quiescent and do not replicate [63, 110, 111].
Because of the reduced or negative ribosomal cell activities, these bacteria
have no adequate targets for the known chlamydia-targeting antibiotics [64,
112]. Persistent Chlamydiae fail to complete development from RBs into
EBs are enlarged and morphologically aberrant, and form small inclusions.
Moreover, they exhibit characteristic gene and protein expression profiles
showing reduced levels of outer membrane proteins like the major outer
membrane protein OmpA or OmcB and substantially increased levels of
Heat shock protein 60 (Hsp 60) [113]. Chlamydiae may be reactivated from
persistence by removal of the inducing stimulus.
A second strategy of Chlamydiae to prolong intracellular survival is
to inhibit pro-apoptotic pathways of the infected host cells. It is thought
that C. pneumoniae can protect infected cells by inhibiting the release of
cytochrome c from mitochondria and upregulate the expression of the anti-
apoptotic mediators IAP and MCL-1. Thus, protection against apoptosis
may be another strategy that the Chlamydiae use to maintain a persistent,
chronic infection. For a more extensive view of the mechanisms involved in
Chlamydia interference with apoptosis signaling in host cells please refer to
the review of Byrne et al. [114].

Virulence factors

Although the genome of C. pneumoniae was sequenced completely in 1999


(1.23 × 106 nt encoding for approx. 1052 proteins, Gen-Bank No.: AE001363,
[15]), until now little is known about structures on the chlamydial surface
(proteins, glycolipids) initiating and mediating bacterial contact to target
cells and inducing subsequent target cell activation. A multitude of the
identified sequences from the chlamydial genome encode for proteins of
bacterial metabolism. Other sequences demonstrated homologies to known
proteins and virulence factors of other bacterial pathogens. Most of these
factors of C. pneumoniae and C. trachomatis have now been identified by
proteom-analysis of both species [115, 116]. Proteins of the “outer mem-
brane complex” (OmpA/B, Omp3, OmcB, POMP), chlamydial lipopolysac-
charide (cLPS), chlamydial heat-shock-proteins (e.g. chsp60/GroEL-1), a
type III secretion apparatus (TTS), the “chlamydial protease- or protea-
some-like activity factor” (CPAF) or peptidoglycans and peptidoglycan-like
structures are likely candidates as possible virulence factors.

Chlamydial envelope/outer membrane complex

The chlamydial outer membrane complex is composed primarily of three


proteins specifically the outer membrane protein A, OmpA, formerly
96 Matthias Krüll and Norbert Suttorp

referred to as major outer membrane protein (MOMP) and two cysteine-


rich proteins, the outer membrane complex B protein (OmcB) and the outer
membrane complex A protein (OmcA) [16]. The gene that encodes MOMP,
ompA [117] exhibits extensive DNA sequence variations that is confined
mainly to four variable segments or domains (VS or VD 1 to VD 4) that
contain subspecies- and serovar-specific antigenic determinants [118]. Until
recently, OmpA was the only protein unequivocally shown to be expressed
on the surface of all chlamydia. The protein is the most important immu-
nodominant structure in all chlamydial strains except C. pneumoniae [102,
103]. Until now, this lack of antigenicity has not been resolved. It might be
possible that either the exposed variable regions of C. pneumoniae OmpA
are not recognized by human or animal immune systems, or that surface
exposure of the OmpA is somehow masked, perhaps by one of the poly-
morphic outer membrane proteins (POMP) as suggested by Christiansen et
al. [103]. OmpA of the chlamydial elementary body is the main component
for chlamydial protection against the environment outside the host, defense
against the host immune response, and attachment to host cells [108].
OmcB, encoded by omcB, does not appear to be surface exposed but is
thought to form a supramolecular lattice in the periplasm. Another impor-
tant difference is that OmcB is extremely highly conserved (for review see
[119]). Stephens et al. could demonstrate that the 60 kDa cystein-rich outer
chlamydial membrane complex protein OmcB is also able to bind heparin
[120]. This protein therefore might be the link mediating chlamydial attach-
ment and initial steps of invasion into target cells.
Although recent studies indicated a functional peptidoglycan (PG)
pathway in chlamydia [121, 122], a clear cut biochemical evidence for the
synthesis of peptidoglycans in chlamydia is missing [123, 124]. Chlamydia,
however, are sensitive to antibiotics that inhibit peptidoglycan synthesis
[125]. This phenomenon has been referred to the “chlamydial anomaly,”
Recent studies, using genomics or proteomics suggested the expression of
peptidoglycan-like structures, not on the surface of elementary bodies, but
– after invasion of the target cells – during cell division on the subsequently
developed reticular bodies [121, 126]. Elucidating the existence of PG in
Chlamydia is of significance for the development of novel antibiotics target-
ing the chlamydial cell wall.

Chlamydial heat shock protein 60 (chsp, GroEL-1)

Heat shock proteins (Hsps) belong to a family of evolutionarily highly con-


served proteins, which are produced by eucaryotic and procaryotic cells dur-
ing a variety of conditions such as heat shock, nutrient deprivation, infec-
tions, and inflammatory reactions, functioning to stabilize cellular proteins.
Several studies using neutralizing monoclonal antibodies or puri-
fied recombinant chlamydial heat-shock protein 60 (cHsp60, GroEL-1),
Pathogenesis of Chlamydophila pneumoniae infections… 97

however, suggested that this protein can act as an extracellular agonist


and might be a key player in activation of different intracellular signal
transduction pathways with a subsequent expression of a profound and
prolonged proinflammatory phenotype in treated cells [76, 79, 127].
Moreover, it has recently been demonstrated, that GroEL-1 is highly
expressed in IFN-a-induced persistent infections of tissue culture cells [63,
128]. Sasu et al. demonstrated that GroEL-1 is a potent inducer of human
vascular smooth muscle cell proliferation and that this effect is mediated
by rapid TLR4-mediated activation of ERK1/2 [80]. We were able to show
that purified recombinant GroEL-1 induced a rapid phosphorylation of
ERK1/2 and p38-MAPK with subsequent enhanced release of IL-8 from
human umbilical vein endothelial cells (HUVEC) [72]. Moreover, GroEL-
1 protein has been shown to stimulate a hyperinflammatory response in
animal models [129–131]. The responses were mediated via a TLR2- and
TLR4-dependent fashion similar to the whole microorganism and differed
markedly from responses induced by endotoxin or CpG oligonucleotides
[77, 78].
Several groups have demonstrated that elevated IgA antibody-titers
of chlamydial heat shock proteins are predictors of chronic chlamydial
infections like bronchial asthma, COPD, arteriosclerosis or pelvic inflam-
matory disease, PID [41, 104, 127, 131]. The molecular mechanisms, how-
ever, by which C. pneumoniae might contribute to development of chronic
diseases remain unclear. Due to the close structural homology between
human and bacterial Hsps and their highly immunogenic nature, Hsps
– especially Hsp60 – have been proposed as key antigens in the devel-
opment of autoimmune diseases [132]. Bachmaier et al. showed that a
peptide from the murine heart muscle-specific _-myosin heavy chain that
has sequence homology to the Hsp60 of C. pneumoniae, C. psittaci, and C.
trachomatis, was shown to induce autoimmune inflammatory heart dis-
ease in mice, suggesting that Chlamydia-mediated heart disease is induced
by antigenic mimicry of a heart muscle-specific protein [133]. In addition,
Kol et al. demonstrated that cHsp60, produced in large amounts during
chronic chlamydial infections, colocalizes within plaque macrophages
with human Hsp60 [127]. Human Hsp60, when expressed by heat-shocked
endothelial cells, can provoke an autoimmune reaction mediating endo-
thelial cytotoxicity [134]. Chlamydial Hsp60 might therefore augment ath-
erosclerosis and/or stimulate humoral and cellular immunity in atheroma
[127, 134].

Chlamydial LPS

Although chlamydia are Gram-negative bacteria, the common LPS group


antigen of all chlamydial species (cLPS) differs significantly from LPS of
other Gram-negative pathogens. In 1998, a first monoclonal antibody was
98 Matthias Krüll and Norbert Suttorp

isolated which recognizes LPS and neutralizes the infectivity of C. pneu-


moniae strain TW183 [135]. This antibody, however, does not neutralize
other strains of C. pneumoniae suggesting the presence of more than a
genus-specific epitope on cLPS. Until now, at least two important differenc-
es between cLPS and LPS from of , for example, enteric bacteria have been
demonstrated: 1) the core trisaccharide 3-deoxy-D-manno-octulosonic acid
(KDO) structure of chlamydial LPS contains a 1-8 linkage, a genus specific
epitope as well as a 1–4 linkage similar to that of other bacteria, encoded
by a single multifunctional KDO transferase [136–138], and 2) the chla-
mydial LPS has low endotoxic activity, although inducing some cytokines
[139, 140]. Immunogold studies suggested that surface exposure of LPS
is greater on RBs than on EBs [141, 142]. Moreover, using different anti-
bodies which recognized either RB or EB Birkelund et al. suggested that
epitope exposure, or the chemical structure of LPS might differ during the
development cycle [143]. cLPS can be released from intracellular, intrain-
clusion Chlamydiae to the inclusion membrane, to the host cell cytoplasm
and surface, and to surrounding infected cells [144–146]. Although this
release might have an impact on the pathogenesis of chlamydial infections
and the host’s immune disposition of infected cells, several studies have
demonstrated that cLPS plays only a minor role for target cell activation
[79, 138].

Chlamydial protease- or proteasome-like activity factor (CPAF)

In 2001, Zhong et al. demonstrated for the first time that a chlamydial
species, C. pneumoniae, secreted a protease into the cytoplasm of infected
target cells. This protease, “chlamydial protease- or proteasome-like
activity factor” (CPAF) split host cell transcription factors necessary for
MHC-I (RFX5) and -II (“upstream stimulatory factor 1”, USF-1) antigen
presentation. Shaw et al. and Dong et al. showed that CPAF is secreted by
different chlamydial species [147, 148]. Heuer et al. suggested that CPAF
could be located in the inclusion lumen or associated with bacteria during
the first 48 h of an acute infection. Seventy-two hours and later, CPAF was
present predominantly in the cytoplasm of the infected cells. Translocation
of CPAF into cytoplasm correlated in time with degradation of the tran-
scription factor RFX5 [149]. CPAF does not preexist in chlamydial organ-
isms and synthesis requires organism replication in cells. Moreover, mice
inoculated with viable chlamydial organisms produced a strong antibody
response to CPAF. In addition, sera from women diagnosed with C. tra-
chomatis cervicitis displayed higher levels of antibodies to CPAF than to
either chlamydial major outer membrane protein or heat shock protein 60.
This sera neutralized the proteolytic activity of CPAF in vitro, suggesting
that CPAF is both produced and immunogenic during human chlamydial
infection [150, 151].
Pathogenesis of Chlamydophila pneumoniae infections… 99

Type III secretion system (TTS)

The first genetic evidence that Chlamydia might have a type III secretion
(TTS) system was presented by Hsia et al. in 1997, when they described
four genes homologous to structural and regulatory components of a con-
tact-dependent or TTS apparatus share high homology with TTS systems
of other bacterial pathogens [152]. Subsequently, these results could be
confirmed by genome and proteom analysis as well as by microscopic obser-
vations [15, 115, 153, 154]. Since TTS systems have been shown to play a
major role in the pathogen-host interaction in several other pathogens like,
for example, Shigella or Salmonella, the TTS may also act as a key virulence
mechanism of Chlamydia [155].
One can speculate about a role for TTS, both in the initial stages of infec-
tion where Chlamydia first comes into contact with the host cell as well as
in the intracellular phase of chlamydial development using the structure of
the TTS system to translocate different effectors into the host cell, depend-
ing on the phase of the developmental cycle. TTS genes expressed in the
mid- to late stage of the developmental cycle appear to be down-regulated
by IFN-a treatment [156]. This suppression may play a role in maintaining
C. pneumoniae in a persistent or altered state within the host cell. It will
therefore be important to determine what structures are present in C. pneu-
moniae and what role each of them plays in the development and possible
persistence of Chlamydia.

Future research directions

Overall, the data presented suggest that Chlamydophila pneumoniae are able
to infect a multitude of target cells and subsequently to activate and trig-
ger a cascade of early and prolonged signal transduction events. Additional
studies are required to determine the relationship between distinct steps
of initial attachment, the chlamydial development cycle, importance of
different chlamydial virulence factors and initiation of host cell signaling
pathways that could lead to target cell damage and inflammation which in
turn may result in acute diseases like bronchitis or (community-acquired)
pneumonia or may promote chronic diseases like COPD, bronchial asthma
or atherosclerosis. New techniques of biochemical and genetical analysis
(genomics, proteomics) are now available to improve our understanding
about pathomechanisms of infection and inflammation and offer unprec-
edented opportunities to address many fundamental questions regarding
chlamydial interactions with the host cells. These results subsequently will
direct new research avenues in terms of diagnostics, therapeutics, as well as
vaccination strategies. Especially chronic persistent infections with meta-
bolically aberrant chlamydiae that are refractory to current antimicrobial
treatment schemes are a challenge for the development of new therapeutic
100 Matthias Krüll and Norbert Suttorp

strategies. In addition, vaccination against Chlamydophila pneumoniae (e.g.,


via DNA immunization as recently demonstrated [101]) could be a benefi-
cial approach for either preventing or controlling infection by this human
respiratory pathogen.

Acknowledgements

The authors apologize for not citing more original manuscripts due to
space limitations and hope that the cited reviews will provide more detail.
This work was in part supported by the Deutsche Forschungsgemeinschaft
to M.K. and N.S. (Kr 2197/1-2), as well as by the Bundesministerium für
Bildung und Forschung (BMBF) to N.S. (CAPNETZ).

References
1 Grayston JT, Kuo CC, Wang SP, Altman J (1986) A new Chlamydia psittaci
strain, TWAR, isolated in acute respiratory tract infections. N Engl J Med 315:
161–168
2 Kuo CC, Jackson LA, Campbell LA, Grayston JT (1995) Chlamydia pneu-
moniae (TWAR). Clin Microbiol Rev 8: 451–461
3 Grayston JT, Aldous MB, Easton A, Wang SP, Kuo CC, Campbell LA, Altman
J (1993) Evidence that Chlamydia pneumoniae causes pneumonia and bronchi-
tis. J Infec Dis 168: 1231–1235
4 Dalhoff K, Maass M (1996) Chlamydia pneumoniae pneumonia in hospitalized
patients. Clinical characteristics and diagnostic value of polymerase chain reac-
tion detection in BAL. Chest 110: 351–356
5 Hahn DL (1999) Chlamydia pneumoniae, asthma, and COPD: what is the evi-
dence? Ann Allergy Asthma Immunol 83: 271–288, 291
6 Sethi S, Murphy TF (2001) Bacterial infection in chronic obstructive pulmonary
disease in 2000: a state-of-the-art review. Clin Microbiol Rev 14: 336–363
7 Fong IW, Chiu B, Viira E, Jang D, Mahony JB (1999) De novo induction of
atherosclerosis by Chlamydia pneumoniae in a rabbit model. Infect Immunol
67: 6048–6055
8 Maass M, Bartels C, Engel PM, Mamat U, Sievers HH (1998) Endovascular
presence of viable Chlamydia pneumoniae is a common phenomenon in coro-
nary artery disease. J Am Coll Cardiol 31: 827–832
9 Saikku P, Leinonen M, Tenkanen L, Linnanmaki E, Ekman MR, Manninen V,
Manttari M, Frick MH, Huttunen JK (1992) Chronic Chlamydia pneumoniae
infection as a risk factor for coronary heart disease in the Helsinki Heart Study.
Ann Int Med 116: 273–278
10 Shor A, Kuo CC, Patton DL (1992) Detection of Chlamydia pneumoniae in
coronary arterial fatty streaks and atheromatous plaques. S Afr Med J 82:
158–161
Pathogenesis of Chlamydophila pneumoniae infections… 101

11 Kuo CC, Chen HH, Wang SP, Grayston JT (1986) Identification of a new group
of Chlamydia psittaci strains called TWAR. J Clin Microbiol 24: 1034–1037
12 Pace NR (1997) A molecular view of microbial diversity and the biosphere.
Science 276: 734–740
13 Weisburg WG, Hatch TP, Woese CR (1986) Eubacterial origin of chlamydiae. J
Bacteriol 167: 570–574
14 Everett KD, Bush RM, Andersen AA (1999) Emended description of the order
Chlamydiales, proposal of Parachlamydiaceae fam. nov. and Simkaniaceae fam.
nov., each containing one monotypic genus, revised taxonomy of the family
Chlamydiaceae, including a new genus and five new species, and standards for
the identification of organisms. Int J Syst Bacteriol 49 Pt 2: 415–440
15 Kalman S, Mitchell W, Marathe R, Lammel C, Fan J, Hyman RW, Olinger
L, Grimwood J, Davis RW, Stephens RS (1999) Comparative genomes of
Chlamydia pneumoniae and C. trachomatis. Nat Gen 21: 385–389
16 Hatch TP (1999) Development Biology. In: RS Stephens (ed): Chlamydia:
Intracellular biology, pathogenesis, and immunity. ASM Press, Washington, DC,
29–67
17 Schachter J, Stephens RS, Timms P, Kuo C, Bavoil PM, Birkelund S, Boman
J, Caldwell H, Campbell LA, Chernesky M et al (2001) Radical changes to
chlamydial taxonomy are not necessary just yet. Int J Syst Evol Microbiol 51:
249–253
18 Schachter J (1999) Infection and disease epidemiology. In: RS Stephens (ed):
Chlamydia: Intracellular biology, pathogenesis, and immunity. ASM Press,
Washington, DC, 139–170
19 Blasi F, Tarsia P, Arosio C, Fagetti L, Allegra L (1998) Epidemiology of
Chlamydia pneumoniae. Clin Microbiol Infect 4 (Suppl 4): S1–S6
20 Saikku P, Leinonen M, Mattila K, Ekman MR, Nieminen MS, Makela PH,
Huttunen JK, Valtonen V (1988) Serological evidence of an association of a
novel Chlamydia, TWAR, with chronic coronary heart disease and acute myo-
cardial infarction. Lancet 2: 983–986
21 Dowell SF, Peeling RW, Boman J, Carlone GM, Fields BS, Guarner J,
Hammerschlag MR, Jackson LA, Kuo CC, Maass M et al (2001) Standardizing
Chlamydia pneumoniae assays: recommendations from the Centers for Disease
Control and Prevention (USA) and the Laboratory Centre for Disease Control
(Canada). Clin Infect Dis 33: 492–503
22 Wang SP, Grayston JT (1970) Immunologic relationship between genital TRIC,
lymphogranuloma venereum, and related organisms in a new microtiter indi-
rect immunofluorescence test. Am J Ophthalmol 70: 367–374
23 Peeling RW, Wang SP, Grayston JT, Blasi F, Boman J, Clad A, Freidank H,
Gaydos CA, Gnarpe J, Hagiwara T et al (2000) Chlamydia pneumoniae serol-
ogy: interlaboratory variation in microimmunofluorescence assay results. J
Infect Dis 181 (Suppl 3): S426–S429
24 Peeling RW (1999) Serology for Chlamydia pneumoniae (TWAR). In: L
Allegra, F Blasi (eds): Chlamydia pneumoniae: the lung and the heart. Springer-
Verlag Italia, Milano, 33–42
25 Kutlin A, Tsumura N, Emre U, Roblin PM, Hammerschlag MR (1997)
Evaluation of Chlamydia immunoglobulin M (IgM), IgG, and IgA rELISAs
102 Matthias Krüll and Norbert Suttorp

Medac for diagnosis of Chlamydia pneumoniae infection. Clin Diagn Lab


Immunol 4: 213–216
26 Maass M, Essig A, Marre R, Henkel W (1993) Growth in serum-free medium
improves isolation of Chlamydia pneumoniae. J Clin Microbiol 31: 3050–3052
27 Tjhie JH, Roosendaal R, MacLaren DM, Vandenbroucke-Grauls CM (1997)
Improvement of growth of Chlamydia pneumoniae on HEp-2 cells by pretreat-
ment with polyethylene glycol in combination with additional centrifugation
and extension of culture time. J Clin Microbiol 35: 1883–1884
28 Boman J, Allard A, Persson K, Lundborg M, Juto P, Wadell G 1997 Rapid diag-
nosis of respiratory Chlamydia pneumoniae infection by nested touchdown
polymerase chain reaction compared with culture and antigen detection by
EIA. J Infect Dis 175: 1523–1526
29 Campbell LA, O’Brien ER, Cappuccio AL, Kuo CC, Wang SP, Stewart D,
Patton DL, Cummings PK, Grayston JT (1995) Detection of Chlamydia
pneumoniae TWAR in human coronary atherectomy tissues. J Infect Dis 172:
585–588
30 Gaydos CA, Roblin PM, Hammerschlag MR, Hyman CL, Eiden JJ, Schachter J,
Quinn TC (1994) Diagnostic utility of PCR-enzyme immunoassay, culture, and
serology for detection of Chlamydia pneumoniae in symptomatic and asymp-
tomatic patients. J Clin Microbiol 32: 903–905
31 Kuo CC, Shor A, Campbell LA, Fukushi H, Patton DL, Grayston JT (1993)
Demonstration of Chlamydia pneumoniae in atherosclerotic lesions of coro-
nary arteries. J Infect Dis 167: 841–849
32 Wong YK, Gallagher PJ, Ward ME (1999) Chlamydia pneumoniae and athero-
sclerosis. Heart 81: 232–238
33 Kuo C, Campbell LA (2000) Detection of Chlamydia pneumoniae in arterial
tissues. J Infect Dis 181 (Suppl 3): S432-S436
34 Lim WS, Macfarlane JT, Boswell TC, Harrison TG, Rose D, Leinonen M,
Saikku P (2001) Study of community acquired pneumonia aetiology (SCAPA)
in adults admitted to hospital: implications for management guidelines. Thorax
56: 296–301
35 Luna CM, Famiglietti A, Absi R, Videla AJ, Nogueira FJ, Fuenzalida AD, Gene
RJ (2000) Community-acquired pneumonia: etiology, epidemiology, and out-
come at a teaching hospital in Argentina. Chest 118: 1344–1354
36 Ruiz M, Ewig S, Marcos MA, Martinez JA, Arancibia F, Mensa J, Torres A
(1999) Etiology of community-acquired pneumonia: impact of age, comorbid-
ity, and severity. Am J Respir Crit Care Med 160: 397–405
37 Wattanathum A, Chaoprasong C, Nunthapisud P, Chantaratchada S, Limpairojn
N, Jatakanon A, Chanthadisai N (2003) Community-acquired pneumonia in
southeast Asia: the microbial differences between ambulatory and hospitalized
patients. Chest 123: 1512–1519
38 Johnston SL, Martin RJ (2005) Chlamydophila pneumoniae and Mycoplasma
pneumoniae: a role in asthma pathogenesis? Am J Respir Crit Care Med 172:
1078–1089:
39 Webley WC, Salva PS, Andrzejewski C, Cirino F, West CA, Tilahun Y, Stuart ES
(2005) The bronchial lavage of pediatric patients with asthma contains infec-
tious Chlamydia. Am J Respir Crit Care Med 171: 1083–1088
Pathogenesis of Chlamydophila pneumoniae infections… 103

40 Biscione GL, Corne J, Chauhan AJ, Johnston SL (2004) Increased frequency of


detection of Chlamydophila pneumoniae in asthma. Eur Respir J 24: 745–749
41 Savykoski T, Harju T, Paldanius M, Kuitunen H, Bloigu A, Wahlstrom E, Rytila
P, Kinnula V, Saikku P, Leinonen M (2004) Chlamydia pneumoniae infection
and inflammation in adults with asthma. Respiration 71: 120–125
42 Branden E, Koyi H, Gnarpe J, Gnarpe H, Tornling G (2005) Chronic Chlamydia
pneumoniae infection is a risk factor for the development of COPD. Respir
Med 99: 20–26
43 Blasi F, Damato S, Cosentini R, Tarsia P, Raccanelli R, Centanni S, Allegra L
(2002) Chlamydia pneumoniae and chronic bronchitis: association with severity
and bacterial clearance following treatment. Thorax 57: 672–676
44 Wu L, Skinner SJ, Lambie N, Vuletic JC, Blasi F, Black PN (2000.)Immunohist
ochemical staining for Chlamydia pneumoniae is increased in lung tissue from
subjects with chronic obstructive pulmonary disease. Am J Respir Crit Care
Med 162: 1148–1151
45 Littman AJ, White E, Jackson LA, Thornquist MD, Gaydos CA, Goodman GE,
Vaughan TL (2004) Chlamydia pneumoniae infection and risk of lung cancer.
Cancer Epidemiol Biomarkers Prev 13: 1624–1630
46 Kocazeybek B (2003) Chronic Chlamydophila pneumoniae infection in lung
cancer, a risk factor: a case-control study. J Med Microbiol 52: 721–726
47 Thom DH, Grayston JT, Siscovick DS, Wang SP, Weiss NS, Daling JR (1992)
Association of prior infection with Chlamydia pneumoniae and angiographi-
cally demonstrated coronary artery disease. JAMA 268: 68–72
48 Ngeh J, Anand V, Gupta S (2002) Chlamydia pneumoniae and atherosclerosis
– what we know and what we don’t. Clin Microbiol Infect 8: 2–13
49 Elkind MS, Lin IF, Grayston JT, Sacco RL( 2000) Chlamydia pneumoniae and
the risk of first ischemic stroke: The Northern Manhattan Stroke Study. Stroke
31: 1521–1525
50 Sander D, Winbeck K, Klingelhofer J, Etgen T, Conrad B (2001) Enhanced pro-
gression of early carotid atherosclerosis is related to Chlamydia pneumoniae
(Taiwan acute respiratory) seropositivity. Circulation 103: 1390–1395
51 Ramirez JA (1996) Isolation of Chlamydia pneumoniae from the coronary
artery of a patient with coronary atherosclerosis. The Chlamydia pneumoniae/
Atherosclerosis Study Group. Ann Intern Med 125: 979–982
52 Taylor-Robinson D, Thomas BJ (2000) Chlamydia pneumoniae in atheroscle-
rotic tissue. J Infect Dis 181 (Suppl 3): S437–S440
53 Moazed TC, Kuo C, Grayston JT, Campbell LA (1997) Murine models of
Chlamydia pneumoniae infection and atherosclerosis. J Infect Dis 175: 883–
890
54 Blessing E, Campbell LA, Rosenfeld ME, Chough N, Kuo CC 2001 Chlamydia
pneumoniae infection accelerates hyperlipidemia induced atherosclerotic
lesion development in C57BL/6J mice. Atherosclerosis 158: 13–17
55 Muhlestein JB. 2003 Antibiotic treatment of atherosclerosis. Curr Opin Lipidol
14: 605–614
56 Gieffers J, van Zandbergen G, Rupp J, Sayk F, Kruger S, Ehlers S, Solbach W,
Maass M (2004) Phagocytes transmit Chlamydia pneumoniae from the lungs to
the vasculature. Eur Respir J 23: 506–510
104 Matthias Krüll and Norbert Suttorp

57 Cercek B, Shah PK, Noc M, Zahger D, Zeymer U, Matetzky S, Maurer G,


Mahrer P (2003) Effect of short-term treatment with azithromycin on recurrent
ischaemic events in patients with acute coronary syndrome in the Azithromycin
in Acute Coronary Syndrome (AZACS) trial: a randomised controlled trial.
Lancet 361: 809–813
58 Gelfand EV, Cannon CP (2004) Antibiotics for secondary prevention of coro-
nary artery disease: an ACES hypothesis but we need to PROVE IT. Am Heart
J 147: 202–209
59 Grayston JT (2003) Antibiotic treatment of atherosclerotic cardiovascular dis-
ease. Circulation 107: 1228–1230
60 Gurfinkel E, Bozovich G, Daroca A, Beck E, Mautner B (1997) Randomised
trial of roxithromycin in non-Q-wave coronary syndromes: ROXIS Pilot Study.
ROXIS Study Group. Lancet 350: 404–407
61 Gurfinkel E (2000) Inflammation, infection, or both in atherosclerosis: the
ROXIS trial in perspective. J Infect Dis 181 (Suppl 3): S566–S568
62 O‘Connor CM, Dunne MW, Pfeffer MA, Muhlestein JB, Yao L, Gupta S,
Benner RJ, Fisher MR, Cook TD (2003) Azithromycin for the secondary pre-
vention of coronary heart disease events: the WIZARD study: a randomized
controlled trial. JAMA 290: 1459–1466
63 Beatty WL, Morrison RP, Byrne GI (1994) Persistent chlamydiae: from cell
culture to a paradigm for chlamydial pathogenesis. Microbiol Rev 58: 686–699
64 Gieffers J, Fullgraf H, Jahn J, Klinger M, Dalhoff K, Katus HA, Solbach W,
Maass M (2001) Chlamydia pneumoniae infection in circulating human mono-
cytes is refractory to antibiotic treatment. Circulation 103: 351–356
65 Malinverni R, Kuo CC, Campbell LA, Lee A, Grayston JT (1995) Effects of
two antibiotic regimens on course and persistence of experimental Chlamydia
pneumoniae TWAR pneumonitis. Antimicrob Agents Chemother 39: 45–49
66 Stratton CW, Sriram S (2003) Association of Chlamydia pneumoniae with cen-
tral nervous system disease. Microbes Infect 5: 1249–1253
67 Yucesan C, Sriram S (2001) Chlamydia pneumoniae infection of the central
nervous system. Curr Opin Neurol 14: 355–359
68 Peterson EM, Cheng X, Qu Z, de la Maza LM (1996) Characterization of the
murine antibody response to peptides representing the variable domains of the
major outer membrane protein of Chlamydia pneumoniae. Infect Immun 64:
3354–3359
69 Gaydos CA, Summersgill JT, Sahney NN, Ramirez JA, Quinn TC (1996)
Replication of Chlamydia pneumoniae in vitro in human macrophages, endo-
thelial cells, and aortic artery smooth muscle cells. Infec Immu 64: 1614–1620
70 Roblin PM, Dumornay W, Hammerschlag MR (1992) Use of HEp–2 cells for
improved isolation and passage of Chlamydia pneumoniae. J Clin Microbiol 30:
1968–1971
71 Wong KH, Skelton SK, Chan YK (1992) Efficient culture of Chlamydia
pneumoniae with cell lines derived from the human respiratory tract. J Clin
Microbiol 30: 1625–1630
72 Krull M, Maass M, Suttorp N, Rupp J (2005) Chlamydophila pneumoniae.
Mechanisms of target cell infection and activation. Thromb Haemost 94: 319–
326
Pathogenesis of Chlamydophila pneumoniae infections… 105

73 Wuppermann FN, Hegemann JH, Jantos CA (2001) Heparan sulfate-like gly-


cosaminoglycan is a cellular receptor for Chlamydia pneumoniae. J Infect Dis
184: 181–187
74 Davis CH, Raulston JE, Wyrick PB (2002) Protein disulfide isomerase, a com-
ponent of the estrogen receptor complex, is associated with Chlamydia tracho-
matis serovar E attached to human endometrial epithelial cells. Infect Immun
70: 3413–3418
75 Puolakkainen M, Kuo CC, Campbell LA (2005) Chlamydia pneumoniae uses
the mannose 6-phosphate/insulin-like growth factor 2 receptor for infection of
endothelial cells. Infect Immun 73: 4620–4625
76 Bulut Y, Faure E, Thomas L, Karahashi H, Michelsen KS, Equils O, Morrison
SG, Morrison RP, Arditi M (2002) Chlamydial heat shock protein 60 activates
macrophages and endothelial cells through Toll-like receptor 4 and MD2 in a
MyD88-dependent pathway. J Immunol 168: 1435–1440
77 Costa CP, Kirschning CJ, Busch D, Durr S, Jennen L, Heinzmann U, Prebeck S,
Wagner H, Miethke T (2002) Role of chlamydial heat shock protein 60 in the
stimulation of innate immune cells by Chlamydia pneumoniae. Eur J Immunol
32: 2460–2470
78 Da Costa CU, Wantia N, Kirschning CJ, Busch DH, Rodriguez N, Wagner H,
Miethke T (2004) Heat shock protein 60 from Chlamydia pneumoniae elicits
an unusual set of inflammatory responses via Toll-like receptor 2 and 4 in vivo.
Eur J Immunol 34: 2874–2884
79 Netea MG, Kullberg BJ, Galama JM, Stalenhoef AF, Dinarello CA, Van der
Meer JW (2002) Non-LPS components of Chlamydia pneumoniae stimulate
cytokine production through Toll-like receptor 2-dependent pathways. Eur J
Immunol 32: 1188–1195
80 Sasu S, LaVerda D, Qureshi N, Golenbock DT, Beasley D (2001) Chlamydia
pneumoniae and chlamydial heat shock protein 60 stimulate proliferation of
human vascular smooth muscle cells via toll-like receptor 4 and p44/p42 mito-
gen-activated protein kinase activation. Circ Res 89: 244–250
81 Opitz B, Forster S, Hocke AC, Maass M, Schmeck B, Hippenstiel S, Suttorp N,
Krull M (2005) Nod1-mediated endothelial cell activation by Chlamydophila
pneumoniae. Circ Res 96: 319–326
82 Rottenberg ME, Gigliotti RA, Gigliotti D, Ceausu M, Une C, Levitsky V,
Wigzell H (2000) Regulation and role of IFN-gamma in the innate resistance
to infection with Chlamydia pneumoniae. J Immunol 164: 4812–4818
83 Hassanain HH, Chon SY, Gupta SL (1993) Differential regulation of human
indoleamine 2,3-dioxygenase gene expression by interferons-gamma and -
alpha. Analysis of the regulatory region of the gene and identification of an
interferon-gamma-inducible DNA-binding factor. J Biol Chem. 268: 5077–
5084
84 Thomas SM, Garrity LF, Brandt CR, Schobert CS, Feng GS, Taylor MW,
Carlin JM, Byrne GI (1993) IFN-gamma-mediated antimicrobial response.
Indoleamine 2,3-dioxygenase-deficient mutant host cells no longer inhibit
intracellular Chlamydia spp. or Toxoplasma growth. J Immunol 150: 5529–
5534
85 Summersgill JT, Sahney NN, Gaydos CA, Quinn TC, Ramirez JA (1995)
106 Matthias Krüll and Norbert Suttorp

Inhibition of Chlamydia pneumoniae growth in HEp-2 cells pretreated with


gamma interferon and tumor necrosis factor alpha. Infec Immu 63: 2801–2803
86 Igietseme JU, Perry LL, Ananaba GA, Uriri IM, Ojior OO, Kumar SN, Caldwell
HD (1998) Chlamydial infection in inducible nitric oxide synthase knockout
mice. Infect Immun 66: 1282–1286
87 Rottenberg ME, Gigliotti Rothfuchs AC, Gigliotti D, Svanholm C, Bandholtz
L, Wigzell H (1999) Role of innate and adaptive immunity in the outcome
of primary infection with Chlamydia pneumoniae, as analyzed in genetically
modified mice. J Immunol 162: 2829–2836
88 Magee DM, Williams DM, Smith JG, Bleicker CA, Grubbs BG, Schachter J,
Rank RG (1995) Role of CD8 T cells in primary Chlamydia infection. Infect
Immun 63: 516–521
89 Su H, Caldwell HD (1995) CD4 + T cells play a significant role in adoptive
immunity to Chlamydia trachomatis infection of the mouse genital tract. Infect
Immun 63: 3302–3308
90 Buzoni-Gatel D, Guilloteau L, Bernard F, Bernard S, Chardes T, Rocca A
(1992) Protection against Chlamydia psittaci in mice conferred by Lyt-2 + T
cells. Immunology 77: 284–288
91 Yang ZP, Kuo CC, Grayston JT (1993) A mouse model of Chlamydia pneu-
moniae strain TWAR pneumonitis. Infect Immun 61: 2037–2040
92 Kaukoranta-Tolvanen SE, Laurila AL, Saikku P, Leinonen M, Laitinen K
(1995) Experimental Chlamydia pneumoniae infection in mice: effect of rein-
fection and passive immunization. Microb Pathog 18: 279–288
93 Wizel B, Starcher BC, Samten B, Chroneos Z, Barnes PF, Dzuris J, Higashimoto
Y, Appella E, Sette A (2002) Multiple Chlamydia pneumoniae antigens prime
CD8 + Tc1 responses that inhibit intracellular growth of this vacuolar pathogen.
J Immunol 169: 2524–2535
94 Penttila JM, Anttila M, Varkila K, Puolakkainen M, Sarvas M, Makela PH,
Rautonen N (1999) Depletion of CD8 + cells abolishes memory in acquired
immunity against Chlamydia pneumoniae in BALB/c mice. Immunology 97:
490–496
95 Penttila JM, Anttila M, Puolakkainen M, Laurila A, Varkila K, Sarvas M,
Makela PH, Rautonen N. 1998 Local immune responses to Chlamydia pneu-
moniae in the lungs of BALB/c mice during primary infection and reinfection.
Infect Immun 66: 5113–5118
96 Halme S, Latvala J, Karttunen R, Palatsi I, Saikku P, Surcel HM (2000) Cell-
mediated immune response during primary Chlamydia pneumoniae infection.
Infect Immun 68: 7156–7158
97 Beatty PR, Stephens RS (1994) CD8 + T lymphocyte-mediated lysis of
Chlamydia-infected L cells using an endogenous antigen pathway. J Immunol
153: 4588–4595
98 Starnbach MN, Bevan MJ, Lampe MF (1994) Protective cytotoxic T lym-
phocytes are induced during murine infection with Chlamydia trachomatis. J
Immunol 153: 5183–5189
99 Vuola JM, Puurula V, Anttila M, Makela PH, Rautonen N (2000) Acquired
immunity to Chlamydia pneumoniae is dependent on gamma interferon in two
Pathogenesis of Chlamydophila pneumoniae infections… 107

mouse strains that initially differ in this respect after primary challenge. Infect
Immun 68: 960–964
100 Pinchuk I, Starcher BC, Livingston B, Tvninnereim A, Wu S, Appella E, Sidney
J, Sette A, Wizel B (2005) A CD8 + T cell heptaepitope minigene vaccine
induces protective immunity against Chlamydia pneumoniae. J Immunol 174:
5729–5739
101 Penttila T, Tammiruusu A, Liljestrom P, Sarvas M, Makela PH, Vuola JM,
Puolakkainen M (2004) DNA immunization followed by a viral vector booster
in a Chlamydia pneumoniae mouse model. Vaccine 22: 3386–3394
102 Campbell LA, Kuo CC, Wang SP, Grayston JT (1990) Serological response to
Chlamydia pneumoniae infection. J Clin Microbiol 28: 1261–1264
103 Christiansen G, Ostergaard L, Birkelund S (1997) Molecular biology of the
Chlamydia pneumoniae surface. Scan J Infec Dis Supp 104: 5–10
104 Huittinen T, Hahn D, Anttila T, Wahlstrom E, Saikku P, Leinonen M (2001)
Host immune response to Chlamydia pneumoniae heat shock protein 60 is
associated with asthma. Eur Respir J 17: 1078–1082
105 Kuo CC, Jackson LA, Campbell LA, Grayston JT (1995) Chlamydia pneu-
moniae (TWAR). Clin Microbiol Rev 8: 451–461
106 Hackstadt T, Fischer ER, Scidmore MA, Rockey DD, Heinzen RA (1997)
Origins and functions of the chlamydial inclusion. Trends Microbiol 5: 288–
293
107 Coombes BK, Mahony JB (2002) Identification of MEK- and phosphoinositide
3-kinase-dependent signalling as essential events during Chlamydia pneumoni-
ae invasion of HEp2 cells. Cell Microbiol 4: 447–460
108 Hackstadt T (1999) Cell biology. In: RS Stephens (ed): Chlamydia:
Intracellularbiology, pathogenesis and immunity, pp. 101–138, ASM Press,
Washington, D.C.
109 Moulder JW (1991) Interaction of chlamydiae and host cells in vitro. Microbiol
Rev 55: 143–190
110 Gieffers J, Rupp J, Gebert A, Solbach W, Klinger M (2004) First-choice anti-
biotics at subinhibitory concentrations induce persistence of Chlamydia pneu-
moniae. Antimicrob Agents Chemother 48: 1402–1405
111 Hogan RJ, Mathews SA, Mukhopadhyay S, Summersgill JT, Timms P (2004)
Chlamydial persistence: beyond the biphasic paradigm. Infect Immun 72:
1843–1855
112 Kutlin A, Roblin PM, Hammerschlag MR (1999) In vitro activities of azithro-
mycin and ofloxacin against Chlamydia pneumoniae in a continuous-infection
model. Antimicrob Agents Chemother 43: 2268–2272
113 Beatty WL, Byrne GI, Morrison RP (1993) Morphologic and antigenic char-
acterization of interferon gamma-mediated persistent Chlamydia trachomatis
infection in vitro. Proc Natl Acad Sci USA 90: 3998–4002
114 Byrne GI, Ojcius DM (2004) Chlamydia and apoptosis: life and death decisions
of an intracellular pathogen. Nat Rev Microbiol 2: 802–808
115 Vandahl BB, Birkelund S, Demol H, Hoorelbeke B, Christiansen G,
Vandekerckhove J, Gevaert K (2001) Proteome analysis of the Chlamydia
pneumoniae elementary body. Electrophoresis 22: 1204–1223
116 Shaw AC, Gevaert K, Demol H, Hoorelbeke B, Vandekerckhove J, Larsen
108 Matthias Krüll and Norbert Suttorp

MR, Roepstorff P, Holm A, Christiansen G, Birkelund S (2002) Comparative


proteome analysis of Chlamydia trachomatis serovar A, D and L2. Proteomics
2: 164–186
117 Stephens RS, Mullenbach G, Sanchez-Pescador R, Agabian N (1986) Sequence
analysis of the major outer membrane protein gene from Chlamydia trachoma-
tis serovar L2. J Bacteriol 168: 1277–1282
118 Millman KL, Tavare S, Dean D (2001) Recombination in the ompA gene
but not the omcB gene of Chlamydia contributes to serovar-specific differ-
ences in tissue tropism, immune surveillance, and persistence of the organism.
J Bacteriol 183: 5997–6008
119 Hatch TP (1999) Development Biology. In: RS Stephens (ed): Chlamydia:
Intracellular biology, pathogenesis, and immunity. ASM Press, Washington, DC,
29–67
120 Stephens RS, Koshiyama K, Lewis E, Kubo A (2001) Heparin-binding outer
membrane protein of chlamydiae. Mol Microbiol 40: 691–699
121 McCoy AJ, Sandlin RC, Maurelli AT (2003) In vitro and in vivo functional activ-
ity of Chlamydia MurA, a UDP-N-acetylglucosamine enolpyruvyl transferase
involved in peptidoglycan synthesis and fosfomycin resistance. J Bacteriol 185:
1218–1228
122 Hesse L, Bostock J, Dementin S, Blanot D, Mengin-Lecreulx D, Chopra I
(2003) Functional and biochemical analysis of Chlamydia trachomatis MurC,
an enzyme displaying UDP-N-acetylmuramate: amino acid ligase activity. J
Bacteriol 185: 6507–6512
123 Chopra I, Storey C, Falla TJ, Pearce JH (1998) Antibiotics, peptidoglycan
synthesis and genomics: the chlamydial anomaly revisited. Microbiology 144:
2673–2678
124 Fox A, Rogers JC, Gilbart J, Morgan S, Davis CH, Knight S, Wyrick PB (1990)
Muramic acid is not detectable in Chlamydia psittaci or Chlamydia trachomatis
by gas chromatography-mass spectrometry. Infect Immun 58: 835–837
125 Moulder JW (1993) Why is Chlamydia sensitive to penicillin in the absence of
peptidoglycan? Infect Agents Dis 2: 87–99
126 Bavoil PM, Hsia R, Ojcius DM (2000) Closing in on Chlamydia and its intracel-
lular bag of tricks. Microbiology 146: 2723–2731
127 Kol A, Sukhova GK, Lichtman AH, Libby P (1998) Chlamydial heat shock pro-
tein 60 localizes in human atheroma and regulates macrophage tumor necrosis
factor-alpha and matrix metalloproteinase expression. Circulation 98: 300–307
128 Beatty WL, Morrison RP, Byrne GI (1995) Reactivation of persistent Chlamydia
trachomatis infection in cell culture. Infect Immun 63: 199–205
129 Morrison RP, Belland RJ, Lyng K, Caldwell HD (1989) Chlamydial disease
pathogenesis. The 57-kD chlamydial hypersensitivity antigen is a stress
response protein. J Exp Med 170: 1271–1283
130 Patton DL, Sweeney YT, Kuo CC (1994) Demonstration of delayed hypersen-
sitivity in Chlamydia trachomatis salpingitis in monkeys: a pathogenic mecha-
nism of tubal damage. J Infect Dis 169: 680–683
131 Wagar EA, Schachter J, Bavoil P, Stephens RS (1990) Differential human sero-
logic response to two 60,000 molecular weight Chlamydia trachomatis antigens.
J Infect Dis 162: 922–927
Pathogenesis of Chlamydophila pneumoniae infections… 109

132 Zugel U, Kaufmann SH (1999) Role of heat shock proteins in protection from
and pathogenesis of infectious diseases. Clin Microbiol Rev 12: 19–39
133 Bachmaier K, Neu N, de la Maza LM, Pal S, Hessel A, Penninger JM (1999)
Chlamydia infections and heart disease linked through antigenic mimicry.
Science 283: 1335–1339
134 Schett G, Xu Q, Amberger A, van der ZR, Recheis H, Willeit J, Wick G (1995)
Autoantibodies against heat shock protein 60 mediate endothelial cytotoxicity.
J Clin Invest 96: 2569–2577
135 Peterson EM, de la Maza LM, Brade L, Brade H (1998) Characterization
of a neutralizing monoclonal antibody directed at the lipopolysaccharide of
Chlamydia pneumoniae. Infect Immun 66: 3848–3855
136 Belunis CJ, Mdluli KE, Raetz CR, Nano FE (1992) A novel 3-deoxy-D-manno-
octulosonic acid transferase from Chlamydia trachomatis required for expres-
sion of the genus-specific epitope. J Biol Chem 267: 18702–18707
137 Brade L, Nano FE, Schlecht S, Schramek S, Brade H (1987) Antigenic and
immunogenic properties of recombinants from Salmonella typhimurium and
Salmonella minnesota rough mutants expressing in their lipopolysaccharide a
genus-specific chlamydial epitope. Infect Immun 55: 482–486
138 Lobau S, Mamat U, Brabetz W, Brade H (1995) Molecular cloning, sequence
analysis, and functional characterization of the lipopolysaccharide biosynthetic
gene kdtA encoding 3-deoxy-alpha-D-manno-octulosonic acid transferase of
Chlamydia pneumoniae strain TW-183. Mol Microbiol 18: 391–399
139 Ingalls RR, Rice PA, Qureshi N, Takayama K, Lin JS, Golenbock DT (1995)
The inflammatory cytokine response to Chlamydia trachomatis infection is
endotoxin mediated. Infect Immun 63: 3125–3130
140 Ivins BE, Wyrick PB (1978) Response of C3H/HeJ and C3H/HeN mice and
their peritoneal macrophages to the toxicity of Chlamydia psittaci elementary
bodies. Infect Immun 22: 620–622
141 Collett BA, Newhall WJ, Jersild RA Jr, Jones RB (1989) Detection of surface-
exposed epitopes on Chlamydia trachomatis by immune electron microscopy. J
Gen Microbiol 135: 85–94
142 Kuo CC, Chi EY (1987) Ultrastructural study of Chlamydia trachomatis surface
antigens by immunogold staining with monoclonal antibodies. Infect Immun 55:
1324–1328
143 Birkelund S, Lundemose AG, Christiansen G (1989) Immunoelectron micros-
copy of lipopolysaccharide in Chlamydia trachomatis. Infect Immun 57: 3250–
3253
144 Richmond SJ, Stirling P (1981) Localization of chlamydial group Antigen in
McCoy cell monolayers infected with Chlamydia trachomatis or Chlamydia
psittaci. Infect Immun 34: 561–570
145 Karimi ST, Schloemer RH, Wilde CE, III (1989) Accumulation of chlamydial
lipopolysaccharide antigen in the plasma membranes of infected cells. Infect
Immun 57: 1780–1785
146 Campbell S, Richmond SJ, Yates PS, Storey CC (1994) Lipopolysaccharide in
cells infected by Chlamydia trachomatis. Microbiology 140: 1995–2002
147 Dong F, Zhong Y, Arulanandam B, Zhong G (2005) Production of a proteolyti-
110 Matthias Krüll and Norbert Suttorp

cally active protein, chlamydial protease/proteasome-like activity factor, by five


different Chlamydia species. Infect Immun 73: 1868–72
148 Shaw AC, Vandahl BB, Larsen MR, Roepstorff P, Gevaert K, Vandekerckhove
J, Christiansen G, Birkelund S (2002) Characterization of a secreted Chlamydia
protease. Cell Microbiol 4: 411–424
149 Heuer D, Brinkmann V, Meyer TF, Szczepek AJ (2003) Expression and trans-
location of chlamydial protease during acute and persistent infection of the
epithelial HEp–2 cells with Chlamydophila (Chlamydia) pneumoniae. Cell
Microbiol 5: 315–322
150 Sharma J, Bosnic AM, Piper JM, Zhong G (2004) Human antibody responses
to a Chlamydia-secreted protease factor. Infect Immun 72: 7164–7171
151 Sharma J, Dong F, Pirbhai M, Zhong G (2005) Inhibition of proteolytic activity
of a chlamydial proteasome/protease-like activity factor by antibodies from
humans infected with Chlamydia trachomatis. Infect Immun 73: 4414–4419
152 Hsia RC, Pannekoek Y, Ingerowski E, Bavoil PM (1997) Type III secretion
genes identify a putative virulence locus of Chlamydia. Mol Microbiol 25:
351–359
153 Matsumoto A (1981) Electron microscopic observations of surface projections
and related intracellular structures of Chlamydia organisms. J Electron Microsc
(Tokyo) 30: 315–320
154 Nichols BA, Setzer PY, Pang F, Dawson CR (1985) New view of the surface
projections of Chlamydia trachomatis. J Bacteriol 164: 344–349
155 Hueck CJ (1998) Type III protein secretion systems in bacterial pathogens of
animals and plants. Microbiol Mol Biol Rev 62: 379–433
156 Slepenkin A, Motin V, de la Maza LM, Peterson EM (2003) Temporal expres-
sion of type III secretion genes of Chlamydia pneumoniae. Infect Immun 71:
2555–2562
157 Airenne S, Surcel HM, Alakarppa H, Laitinen K, Paavonen J, Saikku P, Laurila
A (1999) Chlamydia pneumoniae infection in human monocytes. Infect Immun
67: 1445–1449
Community-Acquired Pneumonia 111
ed. by N. Suttorp, T. Welte and R. Marre
© 2007 Birkhäuser Verlag Basel/Switzerland

Legionnaires’ disease and its agent


Legionella pneumophila

Dina M. Bitar1, Marina Santic2, Yousef Abu Kwaik3 and Maëlle Molmeret3
1Department of Microbiology and Department of Medical Microbiology and Immunology,
Faculty of Medicine, Al-Quds University, Jerusalem, 19356, Israel; 2Department of Microbiology
and Parasitology, Medical Faculty, University of Rijeka, Croatia; 3Department of Microbiology
and Immunology, Room MS-410, University of Louisville, Louisville, KY 40209, USA

Abstract
Legionella pneumophila, the agent responsible for Legionnaire’s disease, is a facultative
intracellular pathogen that can replicate within protozoa and macrophages. Protozoa are
considered to play a central role in the pathogenesis and ecology of L. pneumophila. In
humans, L. pneumophila reaches the lungs, where it is ingested by alveolar macrophages.
Unlike phagosomes containing inert particles or avirulent bacteria, the L. pneumophila-
containing vacuoles avoid fusion with lysosomes, recruiting rough endoplasmic reticulum
and mitochondria. The formation of this specialized vacuole is directed by the type IV
secretion system encoded by the dot/icm genes in mammalian and protozoan cells. Killing
of mammalian cells by L. pneumophila has been proposed to occur through induction
of apoptosis during the early stages of the infection. A rapid induction of necrosis by L.
pneumophila also occurs upon entry into the post-exponential phase of growth within
both macrophages and protozoa, when the bacteria become cytotoxic. Before the lysis of
the mammalian or protozoan plasma membrane, the bacteria egress into the cytoplasm.
In vivo, clearance of Legionella from the lungs depends on the host production of IFN-a
in A/J mice, while in BALB/c mice IFN-a is not produced. Intracellular replication of L.
pneumophila is inhibited in IFN-a-activated mouse and human primary macrophages.
Both antigen-specific humoral and cell-mediated immune responses are induced dur-
ing Legionella infection. Although Legionella-specific antibodies are produced during
human or murine infection, acquired cell-mediated immune response is believed to play
a stronger role in Legionella clearance. Both macrophages and DCs are able to pres-
ent microbial antigens on major histocompatibility class I and class II molecules, which
stimulate antigen-specific T-cell response. Identification of antigens and determination of
vesicular trafficking mechanisms involved in processing and presentation remain to be
understood in greater detail.

Introduction

The first recognized outbreak of pneumonia due to Legionella pneumophila


occurred in Philadelphia, during the summer of 1976 among 180 persons
attending the 56th annual American Legion Convention. Twenty-nine
patients died and the disease became known as Legionnaires’ disease [1].
112 Dina M. Bitar et al.

Guinea pigs were infected with postmortem lung tissue from the patients
with fatal Legionnaires’ disease, and embryonated yolk sacs were inoculated
with spleen homogenates from the infected guinea pigs. In January of 1977,
a Gram-negative bacterium was isolated and designated L. pneumophila [2].
The source of the infection during the Legionnaires’ convention was later
found to be the air conditioning system in the hotel.
It has been documented that the hallmark of Legionnaires’ disease is
the intracellular replication of L. pneumophila in the alveolar spaces. At
least 48 species of legionellae have been identified, some of which are asso-
ciated with disease while others are environmental isolates and whether
they can cause disease is not known [3]. L. pneumophila is responsible
for more than 80% of cases of Legionnaires’ disease, and among the 13
serogroups of L. pneumophila, serogroup 1 is responsible for more than
95% of Legionnaires’ disease cases. It is estimated that L. pneumophila is
responsible for at least 25,000 cases of pneumonia/year in the US, which is
most probably an underestimate due to the difficulty in bacterial isolation
from clinical samples. Since L. pneumophila is the most frequent cause of
Legionnaires’ disease, most pathogenic and environmental studies have
focused on L. pneumophila.
Free-living amoebae are important predators controlling microbial com-
munities. They are ubiquitous and have been isolated from various natural
sources such as soil, freshwater, salt water, dust, and air. Although their
presence in soil is limited, they have been implicated in the stimulation
of phosphorus and nitrogen turnover and thus play an important role in
soil ecosystems [4]. Free-living amoebae are also frequently isolated from
anthropogenic ecosystems, such as tap water, air conditioning units and cool-
ing towers, feeding on the microbial biofilms present in those systems [5, 6].
However, several bacteria have developed mechanisms to survive phagocy-
tosis by free-living amoebae and are able to exploit them as hosts [7, 8]; see
review in [9]. Transient association with amoebae have been reported for a
number of different bacteria including Legionella pneumophila, mycobacte-
rium sp., Francisella tularensis, or Escherichia coli O157, among others [8, 10–
12]. As most of these bacteria are human pathogens, amoebae have been sug-
gested to represent their environmental reservoirs, acting as “Trojan horses”
of the microbial world [9, 13]. To date only the interaction of L. pneumophila,
a facultative intracellular pathogen of humans causing Legionnaire’s disease,
with free-living amoebae has been studied in greater detail.
L. pneumophila has a very similar intracellular fate within both mam-
malian and protozoan cells. Intracellular multiplication of Legionella with
protozoa such as Acanthamoeba polyphaga and macrophages requires the
Dot/Icm secretion system for biogenesis of the phagosome and intracellular
replication [14, 15]. The dot/icm genes are located in two different regions.
The region I includes seven genes (dotA–D, icmV,W,X) and the larger
region II contains the remaining 17 members (icmT,S,R,Q,P,O,N,M,L,K,E,
G,C,D,J,B,F) [16–19]. The dot/icm genes of the Type IV secretion system are
Legionnaires’ disease and its agent Legionella pneumophila 113

thought to encode proteins involved in the translocation of effector mol-


ecules into the host cell that will prevent the bacteria from being killed [16,
20–23]. The dot/icm loci are highly similar to the transfer region of plasmid
R64 and other IncI1 plasmids, which suggests that dot/icm virulence genes
share a common ancestor with plasmid conjugation system [15, 24–28]. It is
not known whether the dot/icm genes derive from a single plasmid, which
has been separated into the two gene clusters, or there were the result of
multiple gene transfer events. However, only Coxiella burnettii has homo-
logs of the full icm/dot genes, which are contained in a single locus [29-33].
Besides the dot/icm Type IVB secretion system, L. pneumophila possess a
second Type IV secretion system, the lvh/lvr genes that surprisingly are not
involved in its virulence [26]. The Type IV secretion system has been shown
to be the main virulence system of L. pneumophila.

Legionellae facultative intracellular pathogen of free-living amoebae

In 1980, Rowbotham described the ability of L. pneumophila to multiply


intracellularly within protozoa [7, 8]. Since then, L. pneumophila has been
described to multiply in many species of protozoa, and this host-parasite
interaction is central to the pathogenesis and ecology of L. pneumophila.
At least 14 species of amoebae and 2 species of ciliated protozoa have
been shown to support intracellular replication of L. pneumophila [34, 35].
Among the most predominant amoebae in water sources are Hartmannellae
and Acanthamoebae, which have also been isolated from water sources
associated with Legionnaires’ disease outbreaks [34]. Interaction between
L. pneumophila and protozoa is considered to be central to the pathogen-
esis and ecology of L. pneumophila [8, 9, 36]. In humans, L. pneumophila
reaches the lungs after inhalation of contaminated aerosol droplets [34, 37].
The main sources of contaminated water droplets are hot water and air con-
ditioning systems, but the bacteria have been isolated from fountains, spas,
pools, dental and hospital units and other man-made water systems [37]. No
person to person transmission has ever been described. Once in the lungs,
L. pneumophila are ingested by alveolar macrophages, which are thought to
be the major site of bacterial replication. This results in an acute and severe
pneumonia. Approximatively one-half of the 48 species of Legionella have
been associated with human disease. L. pneumophila is responsible for 95 %
of cases of Legionnaires’ disease. However, all the Legionella species under
appropriate conditions may be capable of intracellular growth and infliction
of human disease. Infections due to less common species of legionellae are
not frequently diagnosed and reported, and are less studied than L. pneu-
mophila (review in [38, 39]).
In addition to recognized Legionella species, a number of Legionella-
related bacteria designated Legionella-like amoebal pathogens (LLAPs) [8,
40] have been described [41]. Interestingly, many LLAPs have been asso-
114 Dina M. Bitar et al.

ciated with Legionnaires’ disease [42, 43]. In contrast to other Legionella


species, however, most of LLAPs cannot be cultured in vitro on artificial
media, but are isolated by co-culture with protozoa [34]. Considering that
approximately 50% of the 0.5 million annual cases of pneumonia in the US
are of unknown etiology, the LLAPs may be responsible for at least some
of these cases. The recent developments in using the polymerase chain reac-
tion for bacterial identification in environmental samples will facilitate bet-
ter identification of legionellae and LLAPs. Further cellular and molecular
biology studies are needed to better understand the intracellular life of
these endosymbionts.

The role of amoebae in persistence of Legionella in the environment

It is most likely that the association of legionellae with protozoa is a major


factor in continuous presence of the bacteria in the environment. Many
strategies have been used to eradicate legionellae from sources of infec-
tion in water and plumbing systems that have been associated with disease
outbreaks. These strategies include chemical biocides such as chlorine,
overheating of the water, and UV irradiation [44–46]. Such interventions
have been successful for short periods of time after which the bacteria reap-
pear in these sources [46, 47]. Thus, eradication of L. pneumophila from the
environmental sources of infection requires continuous treatment of the
water with agents such as monochloramine or copper-silver ions in addi-
tion to maintenance of the water temperature above ~55 °C [45, 48–50].
Compared to in vitro-grown L. pneumophila, amoebae-grown bacteria have
been shown to be highly resistant to chemical disinfectants and to treat-
ment with biocides [51]. Amoebae-grown L. pneumophila have been shown
to manifest a dramatic increase in their resistance to harsh environmental
conditions such as fluctuation in temperature, osmolarity, pH, and exposure
to oxidizing agents [52]. Protozoa have been shown to release vesicles con-
taining L. pneumophila that are highly resistant to biocides [53]. The ability
of L. pneumophila to survive within amoebic cysts, further contributes to
resistance of L. pneumophila to physical and biochemical agents used in
bacterial eradication [51, 54]. It is likely that eradication of the bacteria from
the environment should start by preventing protozoan infection, an integral
part of the infectious cycle of L. pneumophila. Extracellular L. pneumophila
is more susceptible to environmental conditions and is not protected from
biocides and disinfectants.

The role of amoebae in pathogenesis of Legionella

There are many lines of evidence to suggest that protozoa play major roles
in transmission of L. pneumophila as infectious particles for Legionnaires’
Legionnaires’ disease and its agent Legionella pneumophila 115

disease ([7]; review in [9]). First, many protozoan hosts have been identi-
fied that allow intracellular bacterial replication, the only documented
means of bacterial amplification in the environment [34, 36, 55]. Second, in
outbreaks of Legionnaires’ disease, amoebae and bacteria have been iso-
lated from the same source of infection and the isolated amoebae support
intracellular replication of the bacteria [56]. Third, following intracellular
replication within protozoa, L. pneumophila exhibit a dramatic increase
in resistance to harsh conditions including high temperature, acidity, and
high osmolarity, which may facilitate bacterial survival in the environment
[57-59]. Fourth, intracellular L. pneumophila within protozoa are more
resistant to chemical disinfection and biocides compared to in vitro-grown
bacteria [51, 54, 60]. Fifth, protozoa have been shown to release vesicles
of respirable size that contain numerous L. pneumophila. The vesicles are
resistant to freeze-thawing and sonication, and the bacteria within the
vesicles are highly resistant to biocides [53]. Sixth, following their release
from the protozoan host, the bacteria exhibit a dramatic increase in their
infectivity for mammalian cells in vitro [61]. In addition, it has been dem-
onstrated that intracellular bacteria within H. vermiformis are dramatically
more infectious and are highly lethal in mice [62]. Seventh, the number of
bacteria isolated from the source of infection of Legionnaires’ disease is
usually very low or undetectable, and thus, enhanced infectivity of intracel-
lular bacteria within protozoa may compensate for the low infectious dose
[63]. Eight, viable but non-culturable L. pneumophila can be resuscitated
by co-culture with protozoa [64]. This observation may suggest that fail-
ure to isolate the bacteria from environmental sources of infection may
be due to this “dormant” phase of the bacteria that cannot be recovered
on artificial media. Ninth, there has been no documented case of bacte-
rial transmission between individuals. The only source of transmission is
environmental droplets generated from man-made devices such as shower
heads, water fountains, whirlpools, and cooling towers of air conditioning
systems [34]. These findings indicate a rather sophisticated host-parasite
interaction and a tremendous adaptation of legionellae to parasitize pro-
tozoa. This host-parasite interaction is central to the pathogenesis and
ecology of these bacteria.

Intracellular infection of L. pneumophila

Entry of L. pneumophila into protozoa

The attachment and entry mechanisms of L. pneumophila into its proto-


zoan hosts and variations in their mechanisms have been reported for both
amoeba Hartmanella vermiformis and Acanthamoeba spp. Attachment of
L. pneumophila to H. vermiformis is mediated by adherence to a protozoan
receptor characterized as a putative galactose/N-acetyl-galactosamine (Gal/
116 Dina M. Bitar et al.

GalNAc) lectin [65–67]. Host protein synthesis by A. polyphaga is not


required for invasion by L. pneumophila whereas it is required for inva-
sion of H. vermiformis [66]. Integrins are heterodimeric protein tyrosine
kinase receptors that undergo tyrosine phosphorylation upon engagement
to ligands, which subsequently results in recruitment and rearrangements
of the cytoskeleton. Interestingly, attachment of L. pneumophila to the
Gal/GalNAc of H. vermiformis triggers signal transduction events in H.
vermiformis that are manifested in dramatic tyrosine dephosphorylation
of the lectin receptor and other proteins [65, 68]. Similar observations have
been obtained upon infection of H. vermiformis by another species. of
legionellae, L. micdadei [69]. Among the L. pneumophila-induced tyrosine
dephosphorylated proteins in H. vermiformis are the cytoskeletal proteins
paxillin, vinculin, and focal adhesion kinase [65, 68]. Tyrosine phospha-
tases have been shown to disrupt the cytoskeleton in mammalian cells.
Thus, the induced tyrosine phosphatase activity in H. vermiformis is prob-
ably manifested in disruption of the protozoan cytoskeleton to facilitate
entry through a cytoskeleton-independent receptor-mediated endocytosis.
Interestingly, in addition to these manipulations of the signal transduction
of H. vermiformis by L. pneumophila, bacterial invasion is also associ-
ated with specific induction of gene expression in protozoa, and inhibi-
tion of this gene expression blocks entry of the bacteria [70]. Following
this initial host-parasite interaction, uptake of L. pneumophila by A.
castellanii occurs by coiling phagocytosis [11, 12]. However, the uptake
of L. pneumophila by H. vermiformis occurs mainly through cup-shaped
invaginations (or zipper phagocytosis) on the surface of the amoeba, in
addition to occasional coiling phagocytosis [11, 12]. Human macrophages
are able to phagocytose heat- or formalin-killed L. pneumophila by coiling
phagocytosis [71], which indicates that coiling phagocytosis does not play
a role in the intracellular fate of L. pneumophila. However, the infectiv-
ity of A. castellanii and macrophages by L. pneumophila has been shown
to be similar [72]. In addition, the adherence receptors of macrophages
used by L. pneumophila does not seem to affect its intracellular survival
profoundly as the bacterium multiplies within phagocytes after entry
under different opsonizing or non-opsonizing conditions [73–76]. Uptake
of L. pneumophila by another protozoan host, A. polyphaga, is not com-
pletely blocked by Gal or GalNAc and is associated with partial tyrosine
dephosphorylation of a 170 kDa protein, which may be related to the Gal/
GalNAc lectin of H. vermiformis [66]. Thus, entry of the bacteria into A.
polyphaga is partially mediated by the Gal/GalNAc lectin and additional
receptors may be involved for bacterial attachment and entry. The hetero-
geneity in the uptake mechanisms of L. pneumophila into H. vermiformis
and A. polyphaga has been confirmed using invasion defective mutants of
L. pneumophila. Several mutants that were severely defective in attach-
ment to A. polyphaga exhibited minor reductions in attachment to H.
vermiformis [66].
Legionnaires’ disease and its agent Legionella pneumophila 117

The mode of entry into mammalian and protozoan host cells have been
shown to occur by both dot/icm dependent and independent mechanisms
[72, 77, 78]. Studies have shown that phagocytosis of wild-type L. pneu-
mophila is more efficient than uptake of dot/icm mutants within macro-
phages and A. castallanii, indicating that this mechanism is independent of
adherence receptors [72, 77]. However, when the hosts are infected with sta-
tionary-phase cultures that have been incubated overnight in pH 6.4 buffer,
a treatment which enhances the resistance to acid, hydrogen peroxise and
antibiotics stress, entry into A. castellanii and macrophages do not require
functional dot/icm genes [78]. In addition, a “repeats in structural toxin”
(RTX) gene, rtxA has also been shown to play a role in adherence and entry
and replication within human macrophages, A. castellanii and in vivo [79,
80]. These data indicate the remarkable adaptation of L. pneumophila for
attachment and invasion into different host cells.

Bacterial invasion of macrophages cells and genetic susceptibility

Invasion and intracellular replication of L. pneumophila within pulmo-


nary cells in the alveoli is the hallmark of Legionnaires’ disease [81].
Uptake of L. pneumophila by monocytes and macrophages has been
shown to occur through conventional and coiling phagocytosis [71, 82-85].
It has been recently shown that the enhanced phagocytosis of L. pneu-
mophila by mammalian cells is dot/icm-dependent [72]. Interestingly, the
dot/icm genes delay uptake and induce macropinocytosis in A/J mice mac-
rophages [77]. With the exception of A/J mice, most of the inbred mouse
strains are not permissive to L. pneumophila infection, neither are macro-
phages isolated from these mice [86, 87]. Macropinosomes containing L.
pneumophila in A/J mice macrophages are induced transiently and shrink
rapidly (5-15 min) [77], and this mode of uptake is linked to the lgn1 locus
on chromosome 13 of mice [77, 88, 89]. In macrophages of non permissive
strains of mice, the macropinocytic uptake of L. pneumophila is reduced
[77]. The lgn1 allele causes the bacteria to behave as if they are lacking
the dot/icm system [77]. Thus, the lgn1 allele is required for dot/icm-
dependent macropinocytosis and delayed uptake by mice macrophages
[77]. Whether this mode of uptake plays a role in subsequent trafficking
of L. pneumophila is not known. The intracellular growth of others spe-
cies than pneumophila are not under lgn1’s control [90]. The mouse lgn1
region includes six copies of the neuronal apoptosis inhibitory protein
(naip) gene. These Naip proteins have been shown to be direct inhibitors
of caspase 3 and 7 [91]. Recently, naip5, also known as birc1e (baculoviral
inhibitory apoptosis protein repeat-containing 1), has been shown to be
the allele within lgn1 responsible for susceptibility to Legionella [92, 93].
The mechanism by which naip5 regulates susceptibility to L. pneumophila
is not yet known.
118 Dina M. Bitar et al.

Manipulation of vesicle traffic by the Dot/Icm system and biogenesis


of a replicative ER-derived vacuole

L. pneumophila utilize the Dot/Icm secretion system to transfer macromol-


ecules into the host cell to evade endocytic fusion [25, 27]. The dot/icm loci
may be involved in the insertion of a pore in the host cellular membrane
through which the effector proteins are exported into the host cell [94, 95].
The effector molecules involved in intracellular trafficking and evasion of
the lysosomal fusion within mammalian cells are cis-acting on the phago-
some, but do not alter endocytic fusion in the rest of the cell [96]. With few
exceptions, the function of individual Dot/Icm proteins is unknown.
Several studies demonstrated that upon internalization of L. pneumophila
by the host cell, the Legionella-containing vacuole recruits organelles such as
vesicles, mitochondria and ER [11, 12, 97, 98] (Fig. 1). Within 5 min following
entry of the bacteria into host cells, the L. pneumophila phagosome is sur-
rounded by host cell vesicles, and RER [98-100]. It has been shown that the
Legionella-containing phagosome within mammalian macrophages and pro-
tozoa does not fuse to lysosomes [12, 71, 101, 102]. Interestingly, examination
of the intracellular infection of macrophages, alveolar epithelial cells, and
protozoa by another legionellae spp., L. micdadei, showed that within all of
these host cells, the bacteria were localized to RER-free phagosomes [103].
Whether other legionellae species replicate within RER-free phagosomes is
still to be determined. Recent studies suggest that fusion [104], or exchange
of lipid bilayer with ER vesicles on the L. pneumophila-containing phago-
some [99] allows the phagosomal membrane to become as thin as the ER
membrane with similar characteristics [99, 104]. Within 5 min of uptake, host
vesicles come into contact with wild-type Legionella-containing phagosomes
and flatten along the surface of the phagosome, and this process is completed
within 15 min [99], and is dot/icm-dependent [99]. This is consistent with
earlier studies that have shown that after 4 h of infection, there are only few
vesicles associated with the phagosomal membrane, but there are ribosomes
studding the phagosomal membrane [98]. It is likely that the recruitment of
the ER may be involved in the biogenesis of the phagosome that is depen-
dent on the type IV secretion system, since the dot/icm mutants are unable to
recruit RER and their phagosomes fuse to the lysosomes [105]. In addition,
it has been shown that the L. pneumophila-containing phagosome is a transi-
tional ER (tER)-derived organelle [106]. Its biogenesis involves intercepting
early secretory vesicles exiting from tER [106].
Similar to its intracellular fate within macrophages, L. pneumophila is
enclosed, after entry into amoebae, in a phagosome surrounded by host
cell organelles such as mitochondria, vesicles, and a multilayer membrane
derived from the rough endoplasmic reticulum (RER) of amoeba [11,
98–100]. A few hours after internalization and formation of the ER-derived
replicative organelle, bacterial replication is initiated. Both, evasion of
endocytic fusion and recruitment of early secretory vesicles as they exit
Legionnaires’ disease and its agent Legionella pneumophila 119

Figure 1. L. pneumophila are contained within intact phagosome at 8 h post-infection.


Representative electron micrographs of L. pneumophila-infected macrophages 8 h post-infec-
tion are shown in (a) and (b). The membrane of the LCP is intact as indicated by the thin
arrows. Lpn for L. pneumophila, N for nucleus. Adapted from [140].

the ER is controlled by the Dot/Icm type IV secretion system [11, 98, 99,
106–108]. Most or all of the Dot/Icm structural proteins have been shown
to be essential for biogenesis of the LCP and for intracellular multiplication
within both protozoa and mammalian cells [25, 27]. The role of the RER
in the intracellular infection is unknown, but the RER is not required as a
source of proteins for the bacteria [109].
Recently, the soil amoeba, Dictyostelium discoideum has been studied as
a new host model for Legionella, in particular to understand how Legionella
establish its replicative niche within phagocytic host cells. The advantages of
using this model for the infection of Legionella is that D. discoideum can be
genetically manipulated, that cellular markers are commercially available,
and more importantly, the growth of L. pneumophila is similar to that in
macrophages and fresh-water amoebae including the requirement for a func-
tional dot/icm Type IV secretion system [110, 111]. D. discoideum is found in
soil as a unicellular free-living amoeba that feeds on bacteria. Under starva-
tion conditions, the organism undergoes a complex developmental cycle dur-
ing which it aggregates to form a multicellular motile phototactic slug. This
slug can develop into a fruiting body forming viable spores supported by a
column of stalk. In a rtoA mutant of D. discoideum where vesicle trafficking
event is lowered, the intracellular growth of L. pneumophila is depressed
[112]. In addition, Cytoskeleton-associated proteins and calcium-binding
120 Dina M. Bitar et al.

proteins of the ER, calnexin and calreticulin, specifically influence the


uptake and the intracellular growth of L. pneumophila within D. discoideum
[113]. Therefore, ER recruitment plays an important role in the intracellular
multiplication of L. pneumophila within this host. One of the hypotheses
regarding the role of the recruitment of ER to the LCP is that autophagy
mechanism involved in cellular homeostasis was highjacked by Legionella in
order to establish its replicative niche. As macroautophagy genes have been
identified in D. discoideum, they have been used to show that macroautoph-
agy is dispensable for the intracellular multiplication of L. pneumophila in
D. discoideum [114]. Therefore, it is rather clear that ER-derived vesicles and
proteins are part of a system that leads to the establishment of the replicative
vacuole of L. pneumophila but autophagy does not appear play a role in this
mechanism within Legionella protozoan hosts.

Role of the dot/icm genes in evasion of the endocytic pathway

The main virulence system of L. pneumophila is the dot/icm Type IV secre-


tion system. Because the Dot/Icm secretion system is ancestrally related to
Type IV secretion systems that mediate conjugal DNA transfer between
bacteria [16], L. pneumophila may utilize this transporter to transfer macro-
molecules into the host cell to evade endocytic fusion [104]. The Dot/Icm-
mediated transfer is thought to occur through the insertion of a pore in the
host cellular membrane through which the effector proteins are transported
[94, 95]. With few exceptions, the function of individual Dot/Icm proteins is
unknown. However, the dot/icm genes are present in all tested Legionella
species [115].
Most of the dot/icm genes required for intracellular growth within
human cells, are also required for intracellular growth in the protozoan host
Acanthamoeba castellanii [15]. Although some loci have been shown to be
only essential for the intracellular growth of L. pneumophila in macrophages
[116], numerous loci have been identified as essential for survival and intra-
cellular replication of L. pneumophila in A. polyphaga or H. vermiformis
and in macrophages [14, 117, 118]. D. discoideum has been shown to support
intracellular multiplication of L. pneumophila [110, 111, 119]. As stated ear-
lier, the intracellular fate of L. pneumophila is very similar in infected D. dis-
coideum to that in macrophages, including the recruitment of RER, evasion
of lysosomal fusion [119], and dependence of intracellular growth on dot/icm
gene functions [119]. The similarity between the infection by L. pneumophila
of different protozoa, supports the idea that the ability of L. pneumophila to
parasitize macrophages and hence to cause human disease is a consequence
of its prior adaptation to intracellular growth within protozoa.
The Type IV secretion system of L. pneumophila is though to be an effec-
tive apparatus to translocate effector molecules into the host cell and modu-
late the host cell physiology in order for the bacteria to establish a replicative
Legionnaires’ disease and its agent Legionella pneumophila 121

niche by recruiting ER-derived vesicles and evading the endocytic pathway.


Genetic screens that enable the identification of the set of 26 dot/icm genes
have failed to identify the genes encoding the Type IV secretion system
substrates secreted into host cells [17, 19, 27]. Different strategies have been
used for this purpose. First, strategies, which are not based on intracellular
growth defect of the bacteria, but based on homology to eukaryotic protein
have allowed the identification of Type IV secretion system effectors such as
RalF, LepA and LepB [20, 21]. Most of these substrates do not express an
intracellular replication deficient phenotype (or a minor one) within both
mammalian and protozoan host cells, which explain why they had not been
isolated previously in genetic screens for intracellular growth mutants. RalF
protein, containing a Sec7-homology domain is produced by L. pneumophila
and is injected into the host cells by the Dot/Icm transporter, functioning as
an exchange factor that activates members of the ARF protein family [21,
120]. LepA and LepB, which have a weak homology to SNAREs have been
shown to be delivered to host cells by a Type IV secretion system-depen-
dent mechanism. The double mutant of the paralogues lepA lepB exhibited
a defect in release of L. pneumophila-containing “fecal” or “respirable”
vesicles from A. castellanii and D. discoideum [20]. Second, a strategy based
on that fact that some translocated proteins also function to maintain the
integrity of the Dot/Icm translocator and mutations that destroy this func-
tion are predicted to result in a Dot/Icm complex that poisons the bacterium,
resulting in reduced viability, has allowed identifying an effector called LidA.
This substrate LidA, identified by a complex genetic screen has been shown
to be associated with the cytoplasmic face of the L. pneumophila-containing
phagosome [23]. Third, an interbacterial protein transfer assay is another
strategy used to identify the substrate SidC (substrate of icm/dot complex)
identified among other Sid substrates also secreted into macrophages [22].
Finally, some substrates have been identified using chaperone proteins
belonging to the dot/icm genes secretion system, as bait. IcmS has been
hypothesized to serve as a chaperone for secreted substrates since it is pre-
dicted to be located in the bacterial cytoplasm, has no homology to bacterial
conjugal transfer proteins and has similar biochemical properties of secre-
tion chaperones from the type II secretion system [121, 122]. IcmS has been
used as a bait to identify potential secreted substrates such as SidE family
proteins (SdeA, SdeB, SdeC, SidE) [123] family proteins have been shown to
be secreted by L. pneumophila at early stages of infection of macrophages
across the phagosomal membrane and have been shown to be required for
full virulence in Acanthamoeba castellanii although individual deletions of a
number of substrates had a modest or no effect in intracellular replication
[22] probably resulting from the export of functionally redundant proteins by
the Type IV secretion system. In addition, as the complex IcmS-IcmW form a
stable protein complex and play an important role in subtrate translocation,
IcmW has also been used as a bait in a yeast two-hybrid system to identify
substrate proteins translocated into host cells by the type IV secretion sys-
122 Dina M. Bitar et al.

tem [124]. IcmW-interacting proteins (Wips) have been recovered including


SidG and SidH identified previously with the interbacterial protein transfer
screen [22] as well as WipA, which is translocated into mammalian host cells.
A paralogue called wipB, which is also translocated into macrophages by the
Type IV secretion system, was found in the Legionella genome data base and
the double mutant wipA wipB had no growth defect in A. castallanii [124].
The functions of the identified substrates are under investigation. There are
probably hundred of effectors that need to be identified in order to under-
stand how Legionella establish a successful replicative niche within its host
cells. Some of these substrates may also be host-dependent and be functional
only in one of the various host cells of Legionella.

Growth phase-regulated virulence

L. pneumophila obtained from post-exponential cultures, expresses traits


that are correlated with virulence, in contrast to exponentially-growing
bacteria, [7, 8, 57, 58]. During the replication phase, L. pneumophila are
sodium resistant and aflagellated [7, 8, 57, 58]. When L. pneumophila egress
from host cells, the bacteria are flagellated and sodium-sensitive [7, 8, 57,
58]. It has been hypothesized that amino acid limitation in vitro induces the
virulent phenotype [57, 58]. When L. pneumophila enters into post expo-
nential growth phase or is subjected to amino acid limitation, the bacteria
accumulate the stringent response signal, guanosine 3’,5’-bispyrophosphate
(ppGpp) through the ppGpp synthetase, RelA [125]. The accumulation of
ppGpp increases the amount of the stationary-phase sigma factor RpoS,
which triggers the expression of the stationary-phase genes [125]. A rpoS
mutant of L. pneumophila replicates within HL60 and THP-1 monocytic
cell lines, but is attenuated in A. castellanii [126]. In bone marrow-derived
macrophages from A/J mice, L. pneumophila rpoS mutants replicate poorly
because they traffic rapidly to a late endosome-like compartment [125].
These phenotypic differences in different host cells may be due to differ-
ent parental strains used to construct the rpoS mutants. It has been shown
that some genotypic and phenotypic differences exist between the AA100,
JR32 and LP01 stains, which are the most commonly used in virulence
studies [127] and AA100 is clearly the most virulent. Therefore, the differ-
ences between L. pneumophila strains and/or between the host cells may
explain the different intracellular growth observed for the rpoS mutants.
Sodium sensitivity and maximal expression of flagellin also requires RpoS
[125]. Therefore, some RpoS-regulated traits could be critical for efficient
transmission or infection [126]. L. pneumophila in post-exponential phase
becomes cytotoxic by an RpoS-independent pathway [125]. It is proposed
that when nutrient levels and other conditions are favorable, L. pneumoph-
ila replicates within host cells, and when amino acids become rare, intracel-
lular bacteria express several traits that permit escape from the host cell,
Legionnaires’ disease and its agent Legionella pneumophila 123

survival in the environment and the transmission to a new host. However,


the intracellular signals that trigger phenotypic transition at the post-expo-
nential phase are still to be determined.
Although a previous study has shown that RpoS, which accumulates
when RelA is activated, is required for intracellular growth in A. castellanii
[126], Zusman et al. have shown that relA gene product is dispensable for
intracellular growth in HL-60-derived human macrophages and in A. cas-
tellanii [128]. Moreover, it has also been shown that RelA and RpoS have
minor effects on expression of some of the dot/icm genes [128]. Thus, the
role of RpoS in the intracellular infection seems to be host cell-specific, but
that may be due to differences between the parental strain used to construct
the rpoS mutants.
Interestingly, it has been shown that the conserved RNA binding protein
CsrA is a global repressor of phenotypic transition of L. pneumophila at the
post-exponential phase [129]. Overexpression of CsrA in L. pneumophila
blocks many phenotypic traits that are expressed at the post-exponential
phase such as flagellation and reduction of cell size [129]. This is also asso-
ciated with reduction in transcription of fliA and flaA [129]. In addition,
the global response two-component regulator of L. pneumophila, LetA/S,
is involved in regulation of phenotypic transition, since letA mutants of L.
pneumophila exhibit reduced infectivity and are more resistant to oxidative
and acid stress in addition to a severe defect in intracellular replication in
Acanthamoeba [130, 131]. However, the letA mutant is not defective for
intracellular replication in human-derived macrophages, similar to the rpoS
mutant and mutants in the type II secretion system [131]. Thus, RpoS and
LetA/S regulate phenotypic transition of L. pneumophila at the post-expo-
nential phase and both are essential for expression of L. pneumophila genes
required for replication in the protozoan host but not in human-derived
macrophages. Since both regulatory systems are triggered by ppGpp syn-
thesized by RelA, and RelA is not required for the intracellular infection
of macrophages or amoeba, it is not clear how other regulatory pathways
contribute to the regulation of phenotypic transition.

Cytolysis of the host and bacterial egress

A fundamental step in the pathogenic life cycle of intracellular bacteria is


the ability to lyse the host cell and to egress. Apoptosis and necrosis are
the two commonly observed types of cell death. Necrosis is characterized
by physical damage causing cell death. Apoptosis is a regulated suicide
program of the cell manifesting morphological and biochemical features
distinct from those of necrosis [132]. Killing of mammalian cells by L.
pneumophila has been proposed to occur in two phases [133, 134]. In the
first phase, L. pneumophila induces apoptosis in macrophages, monocytes
and alveolar epithelial cells during the early stages of the infection [133,
124 Dina M. Bitar et al.

Figure 2. L. pneumophila is cytoplasmic at 24 h post-infection. Representative electron micro-


graphs of L. pneumophila-infected macrophages at 24 h post-infection are shown in (c) and (d).
L. pneumophila (Lpn) are cytoplasmic where bacteria are surrounded by numerous vesicles
(V), lysosomal contents (white arrows), mitochondria (M), and amorphous elements (A). No
distinguishable phagosomal membrane surrounds the bacteria. N for nucleus. Adapted from
[107].

135–137], which is mediated through the activation of caspase-3 [134].


Induction of apoptosis is largely independent of the bacterial growth phase
[134]. The second phase is mediated through rapid induction of necrosis by
L. pneumophila upon entry into the post-exponential phase of growth when
the bacteria become cytotoxic [138, 139]. Our working model of bacterial
egress can be presented in three steps. First, upon exiting the exponential
phase of intracellular growth, an „egress pore“ is inserted into the phago-
somal membrane leading to its disruption. Second, the bacteria egress into
cytoplasm. Third, disruption of organelles and the plasma membrane occurs,
culminating in lysis of the host cells and bacterial egress.
A detailed ultrastructural analysis of late stages of intracellular replica-
tion has been performed to examine egress of L. pneumophila from both
macrophages and amoebae by electron microscopy [140]. The membrane
of the L. pneumophila-containing phagosome (LCP) within both macro-
phages and Acanthamoeba polyphaga is intact up to 8 h postinfection [140].
However, at 12 h, the majority of the LCPs are disrupted within both hosts,
while the plasma membrane remains intact [140]. At 18 and 24 h postinfec-
tion, cytoplasmic elements such as mitochondria, lysosomes, vesicles, and
amorphous material are dispersed among the bacteria and these bacteria
are considered cytoplasmic [140] (Fig. 2). Thus, by 18 h–24 h postinfection,
the majority of the remaining host cells harbor cytoplasmic bacteria and
this transient cytoplasmic presence of L. pneumophila precedes lysis of the
plasma membrane. Interestingly, within both macrophages and amoebae,
bacterial replication proceeds in the cytoplasm.
Legionnaires’ disease and its agent Legionella pneumophila 125

Therefore, the phagosomal membrane is disrupted first, rather than by


simultaneous lysis of both the phagosomal and the plasma membranes.
These disruptions of LCP may be the result of multifactorial events linked
to apoptosis, the pore forming activity (PFA) of the Type IV secretion system
and mechanical pressure due to the increase of the phagosomal size [95].

Immunity to intracellular L. pneumophila

The Toll-like receptors

As the interaction between host phagocytes and Legionella results in the


induction of the host cell response that is thought to activate the innate
immune system through the stimulation of TLRs on the surface. It leads
to the production of pro-inflammatory cytokines that recruit lymphocytes
to the infection sites and activate macrophages. Macrophages express both
TLR2 and 4, which can recognize different bacterial products (peptido-
glycan, LPS, etc.). However, surprisingly TLR2 plays a major role in the
response to Legionella, whereas TLR4, which recognizes predominantly
LPS, is less involved in the process [141, 142]. In addition, TLR5 may also be
important in immunity to Legionella as TLR5 recognizes a conserved region
in most bacterial flagellin including Legionella flagellin.

The cytokine production

Neutrophils, macrophages and dendritic cells have been shown to produce


cytokines after Legionella infection, which is comparable to the cytokine
production involved in the Th1 response [143, 144]. IL-12 is produced by
many cells, including macrophages, DC and neutrophils after exposure
to bacteria and has been shown to have an important role in eliminating
Legionella in vivo [145]. IL-18 is also crucial in the induction of IFN-a
production from T-cells, B cells and NK cells in vitro and during in vivo
infection with Legionella [146]. When they are depleted independently or
together, the levels of IFN-a drop significantly, which lead to a decreased
ability of clearing Legionella from the lungs of the mice [146]. Intracellular
replication of L. pneumophila is inhibited in gamma interferon (IFN-a)-
activated bone marrow-derived mouse macrophages and IFN-a-activated
human monocyte-derived macrophages in a dose-dependent manner. This
inhibition of intracellular replication is associated with the maturation of
the LCP into a phagolysosome [147]. Together with IFN-a, TNF-_ plays
also a role in the clearance of Legionella from the lungs via activation of
macrophages. Added independently or together, both IFN-a and TNF-_
can also significantly restrict the intracellular growth of Legionella [148,
149].
126 Dina M. Bitar et al.

The clearance of Legionella from the lungs depends on the host produc-
tion of IFN-a in A/J mice [150]. Additionally, Legionella infection cannot be
cleared efficiently in BALB/c mice, which do not produce IFN-a compared
to infection in mice producing IFN-a [151]. Similarly, when human mono-
cytes and alveolar macrophages are treated with IFN-a, it results in dose-
dependent restricted growth of Legionella [152]. Restriction of Legionella
growth is, in part, due to low availability of intracellular iron, a process
mediated by the transferring receptor which is downregulated in IFN-a
activated monocytes [153]. A recent study also showed that activated mac-
rophages infected by L. pneumophila can downregulate T-cell responses via
production of prostaglandins, which may play a role in limiting unnecessary
immune-mediated damage of host tissues [154].
Activated macrophages can also produce nitric oxide after bacterial
infection, which has a direct lethal effect on many pathogens. In the case of
Legionella infection, the role of NO may not be direct. Inhibitors of NO syn-
thesis had an effect intracellular replication of L. pneumophila in BALB/c
alveolar macrophages, but not in A/J mice macrophages. It had, however, a
effect on the Legionella infection of A/J mice model [149, 151, 155-157].

Acquired immune response

Both antigen-specific humoral and cell-mediated immune responses are


induced during Legionella infection. Although Legionella specific antibod-
ies are produced during human infection, or in the guinea pigs model, their
role in controlling Legionella infection has not been clearly demonstrated
[158]. Antibody opsonization does not inhibit intracellular multiplication of
L. pneumophila [159].
However, when CD4 and CD8 T-cells are depleted in mice, susceptibility
to Legionella infection is enhanced, suggesting that acquired cell-mediated
immune response play a role in Legionella clearance [160]. Both macro-
phages and DCs are able to present microbial antigens on major histo-
compatibility (MHC) class I and MHC class II molecules, which stimulate
antigen specific T-cell response. DCs appear to play an important role in
producing antigen-specific immune responses and in priming T-cells. DCs
are able to restrict the intracellular growth of Legionella without prevent-
ing traffic of Legionella-containing vacuole or bacterial proteins synthesis.
Therefore, resctriction of growth is possible without the Legionella vacuole
fusing to lysosomes, suggesting that the range of proteins presented by DCs
to naïve T-cells is very similar to those presented by macrophages [161, 162].
Taken together, these data showed that Legionella antigens synthesized
in a non-degradative ER-derived vacuole are most likely processed and
loaded onto MHC class II molecules for presentation to CD4 + T-cells [161].
In addition, it has recently been shown that in mice primary macrophages,
trafficking of Legionella-containing phagosome to lysosomes is required for
Legionnaires’ disease and its agent Legionella pneumophila 127

optimal antigen processing and MHC-II presentation to CD4 T-cells [163].


Identification of antigens and determination of vesicular trafficking mecha-
nisms involved in processing and presentation remain to be understood in
greater detail.

Conclusions

L. pneumophila has become a paradigm of an intracellular pathogen that


manipulates several processes involved in endocytosis and vesicle traffic
between host cell organelles, and many of these manipulations are con-
trolled by the Dot/Icm secretion system and the effectors exported through
this system. This organism avoids phagosomes-lysosomes fusion within the
first few minutes of the intracellular infection and the phagosomes are con-
verted into endoplasmic reticulum-derived organelles that support intra-
cellular replication [99]. This process involves intercepting early secretory
vesicles exiting from the tER [106]. After 4 h, L. pneumophila starts to repli-
cate within this replicative organelle. Between 8 h and 18 h, the phagosomal
membrane is gradually disrupted and the bacteria become cytoplasmic and
are dispersed among cytoplasmic organelles such as mitochondria and lyso-
somes prior to lysis of the host cell [140]. Legionella infection triggers both
antigen-specific humoral and cell-mediated immune responses, but the cell-
mediated responses appear to play a greater role in Legionella clearance
from the lungs. Understanding the unique intracellular fate at the molecular
and the cellular level will unravel fascinating aspects of the intricate balance
in the evolution of this host-parasite interaction and the ability to control
Legionella propagation in human and in environment.

Acknowledgements

Our work is supported by Public Health Service grant RO1AI43965 award-


ed to Y.A.K.

References:
1 Fraser DW, Tsai TR, Orenstein W, Parkin WE, Beecham HJ, Sharrar RG, Harris
J, Mallison GF, Martin SM, McDade JE et al (1977) Legionnaires’ disease:
description of an epidemic of pneumonia. N Engl J Med 297: 1189–1197
2 McDade JE, Shepard CC, Fraser DW, Tsai TR, Redus MA, Dowdle WR (1977)
Legionnaires’ disease: isolation of a bacterium and demonstration of its role in
other respiratory disease. N Engl J Med 297: 1197–1203
3 Adeleke AA, Fields BS, Benson RF, Daneshvar MI, Pruckler JM, Ratcliff
RM, Harrison TG, Weyant RS, Birtles RJ, Raoult D et al (2001) Legionella
drozanskii sp nov, Legionella rowbothamii sp nov and Legionella fallonii sp
128 Dina M. Bitar et al.

nov: three unusual new Legionella species. Int J Syst Evol Microbiol 51(Pt 3):
1151–1160
4 Rodriguez-Zaragoza S (1994) Ecology of free-living amoebae. Crit Rev
Microbiol 20(3): 225–241
5 Murga R, Forster TS, Brown E, Pruckler JM, Fields BS, Donlan RM (2001)
Role of biofilms in the survival of Legionella pneumophila in a model potable-
water system. Microbiology 147(Pt 11): 3121–3126
6 Sheehan KB, Henson JM, Ferris MJ (2005) Legionella species diversity in
an acidic biofilm community in Yellowstone National Park. Appl Environ
Microbiol 71(1): 507–11
7 Rowbotham TJ (1980) Preliminary report on the pathogenicity of Legionella
pneumophila for freshwater and soil amoebae. J Clin Pathol 33: 1179–1183
8 Rowbotham TJ (1986) Current views on the relationships between amoebae,
legionellae and man. Isr J Med Sci 22: 678–689
9 Molmeret M, Horn M, Wagner M, Santic M, Abu Kwaik Y (2005) Amoebae as
training grounds for intracellular bacterial pathogens. Appl Environ Microbiol
71(1): 20–8
10 Newsome AL, Baker RL, Miller RD, Arnold RR (1985) Interactions between
Naegleria fowleri and Legionella pneumophila. Infect Immun 50: 449–452
11 Abu Kwaik Y (1996) The phagosome containing Legionella pneumophila
within the protozoan Hartmanella vermiformis is surrounded by the rough
endoplasmic reticulum. Appl Environ Microbiol 62: 2022–2028
12 Bozue JA, Johnson W (1996) Interaction of Legionella pneumophila with
Acanthamoeba catellanii: uptake by coiling phagocytosis and inhibition of
phagosome-lysosome fusion. Infect Immun 64: 668–673
13 Barker J, Brown MR (1994) Trojan horses of the microbial world: protozoa and
the survival of bacterial pathogens in the environment. Microbiology 140 (Pt 6):
1253–1259
14 Gao L-Y, Harb OS, Abu Kwaik Y (1997) Utilization of similar mechanisms by
Legionella pneumophila to parasitize two evolutionarily distant hosts, mam-
malian and protozoan cells. Infect Immun 65: 4738–4746
15 Segal G, Shuman HA (1999) Legionella pneumophila utilizes the same genes
to multiply within Acanthamoeba castellanii and human macrophages. Infect
Immun 67: 2117–2124
16 Christie PJ, Vogel JP (2000) Bacterial type IV secretion: conjugation systems
adapted to deliver effector molecules to host cells. Trends Microbiol 8(8):
354–360
17 Segal G, Shuman HA (1997) Characterization of a new region required for
macrophage killing by Legionella pneumophila. Infect Immun 65(12): 5057–
5066
18 Brand B, Sadosky AB, Shuman HA (1994) The Legionella pneumophila icm
locus: a set of genes required for intracellular multiplication in human macro-
phages. Mol Microbiol 14: 797–808
19 Berger KH, Isberg RR (1993) Two distinct defects in intracellular growth com-
plemented by a single genetic locus in Legionella pneumophila. Mol Microbiol
7: 7–19
20 Chen J, de Felipe KS, Clarke M, Lu H, Anderson OR, Segal G, Shuman HA
Legionnaires’ disease and its agent Legionella pneumophila 129

(2004) Legionella effectors that promote nonlytic release from protozoa.


Science 303(5662): 1358–1361
21 Nagai H, Kagan JC, Zhu X, Kahn RA, Roy CR (2002) A bacterial guanine
nucleotide exchange factor activates ARF on Legionella phagosomes. Science
295(5555): 679–682
22 Luo ZQ, Isberg RR (2004) Multiple substrates of the Legionella pneumophila
Dot/Icm system identified by interbacterial protein transfer. Proc Natl Acad Sci
USA 101(3): 841–846
23 Conover GM, Derre I, Vogel JP, Isberg RR (2003) The Legionella pneumophila
LidA protein: a translocated substrate of the Dot/Icm system associated with
maintenance of bacterial integrity. Mol Microbiol 48(2): 305–321
24 Komano T, Yoshida T, Narahara K, Furuya N (2000) The transfer region of
IncI1 plasmid R64: similarities between R64 tra and legionella icm/dot genes.
Mol Microbiol 35(6): 1348–1359
25 Segal G, Purcell M, Shuman HA (1998) Host cell killing and bacterial conjuga-
tion require overlapping sets of genes within a 22-kb region of the Legionella
pneumophila chromosome. Proc Natl Acad Sci USA 95: 1669–1674
26 Segal G, Russo JJ, Shuman HA (1999) Relationships between a new type IV
secretion system and the icm/dot virulence system of Legionella pneumophila.
Mol Microbiol 34(4): 799–809
27 Vogel JP, Andrews HL, Wong SK, Isberg RR (1998) Conjugative transfer by the
virulence system of Legionella pneumophila. Science 279: 873–876
28 Sexton JA, Vogel JP (2002) Type IVB secretion by intracellular pathogens.
Traffic 3(3): 178–185
29 Seshadri R, Paulsen IT, Eisen JA, Read TD, Nelson KE, Nelson WC, Ward NL,
Tettelin H, Davidsen TM, Beanan MJ et al (2003) Complete genome sequence
of the Q-fever pathogen Coxiella burnetii. Proc Natl Acad Sci USA 100(9):
5455–5460
30 Zamboni DS, McGrath S, Rabinovitch M, Roy CR (2003) Coxiella burnetii
express type IV secretion system proteins that function similarly to compo-
nents of the Legionella pneumophila Dot/Icm system. Mol Microbiol 49(4):
965–976
31 Zusman T, Yerushalmi G, Segal G (2003) Functional similarities between the
icm/dot pathogenesis systems of Coxiella burnetii and Legionella pneumophila.
Infect Immun 71(7): 3714–3723
32 Feldman M, Zusman T, Hagag S, Segal G (2005) Coevolution between non-
homologous but functionally similar proteins and their conserved partners in
the Legionella pathogenesis system. Proc Natl Acad Sci USA 102(34): 12206–
12211
33 Segal G, Feldman M, Zusman T (2005) The Icm/Dot type-IV secretion systems
of Legionella pneumophila and Coxiella burnetii. FEMS Microbiol Rev 29(1):
65–81
34 Fields BS (1996) The molecular ecology of legionellae. Trends Microbiol 4:
286–290
35 Shadrach WS, Rydzewski K, Laube U, Holland G, Ozel M, Kiderlen AF, Flieger
A (2005) Balamuthia mandrillaris, free-living ameba and opportunistic agent
130 Dina M. Bitar et al.

of encephalitis, is a potential host for Legionella pneumophila bacteria. Appl


Environ Microbiol 71(5): 2244–2249
36 Harb OS, Gao L-Y, Abu Kwaik Y (2000) From protozoa to mammalian cells:
A new paradigm in the life cycle of intracellular bacterial pathogens. Environ
Microbiol 2: 251–265
37 Fliermans CB (1996) Ecology of Legionella: From data to knowledge with a
little wisdom. Microb Ecol 32(2): 203–228
38 Fields BS, Benson RF, Besser RE (2002) Legionella and Legionnaires’ disease:
25 years of investigation. Clin Microbiol Rev 15(3): 506–526
39 Molmeret M, Bitar DM, Han L, Kwaik YA (2004) Cell biology of the intracel-
lular infection by Legionella pneumophila. Microbes Infect 6(1): 129–139
40 Rowbotham TJ (1983) Isolation of Legionella pneumophila from clinical
specimens via amoebae, and the interaction of those and other isolates with
amoebae. J Clin Pathol 36: 978–986
41 Adeleke A, Pruckler J, Benson R, Rowbotham T, Halablab M, Fields BS (1996)
Legionella-like amoebal pathogens-phylogenetic status and possible role in
respiratory disease. Emerg Infect Dis 2: 225–229
42 Marrie TJ, Raoult D, La Scola B, Birtles RJ, de Carolis E (2001) Legionella-like
and other amoebal pathogens as agents of community-acquired pneumonia.
Emerg Infect Dis 7(6): 1026–1029
43 Birtles RJ, Rowbotham TJ, Raoult D, Harrison TG (1996) Phylogenetic diver-
sity of intra-amoebal legionellae as revealed by 16S rRNA gene sequence
comparison. Microbiology 142 (Pt 12): 3525–3530
44 Muraca P, Stout JE, Yu VL (1987) Comparative assessment of chlorine, heat,
ozone, and UV light for killing Legionella pneumophila within a model plumb-
ing system. Appl Environ Microbiol 53: 447–453
45 Kool JL, Carpenter JC, Fields BS (1999) Effect of monochloramine disinfec-
tion of municipal drinking water on risk of nosocomial Legionnaires’ disease.
Lancet 353(9149): 272–7
46 Biurrun A, Caballero L, Pelaz C, Leon E, Gago A (1999) Treatment of a
Legionella pneumophila-colonized water distribution system using copper-
silver ionization and continuous chlorination. Infect Control Hosp Epidemiol
20(6): 426–8
47 Yamamoto H, Ezaki T, Ikedo M, Yabuuchi E (1991) Effects of biocidal treat-
ments to inhibit the growth of legionellae and other microorganisms in cooling
towers. Microbiol Immunol 35: 795–802
48 Kusnetsov J, Iivanainen E, Elomaa N, Zacheus O, Martikainen PJ (2001)
Copper and silver ions more effective against legionellae than against myco-
bacteria in a hospital warm water system. Water Res 35(17): 4217–4225
49 Darelid J, Lofgren S, Malmvall BE (2002) Control of nosocomial Legionnaires’
disease by keeping the circulating hot water temperature above 55 degrees C:
experience from a 10-year surveillance programme in a district general hospi-
tal. J Hosp Infect 50(3): 213–219
50 Hoebe CJ, Kool JL (2000) Control of Legionella in drinking-water systems.
Lancet 355(9221): 2093–2094
51 Barker J, Brown MRW, Collier PJ, Farrell I, Gilbert P (1992) Relationships
between Legionella pneumophila and Acanthamoebae polyphaga: physiologi-
Legionnaires’ disease and its agent Legionella pneumophila 131

cal status and susceptibility to chemical inactivation. Appl Environ Microbiol


58: 2420–2425
52 Abu Kwaik Y, Gao L-Y, Harb OS, Stone BJ (1997) Transcriptional regulation
of the macrophage-induced gene (gspA) of Legionella pneumophila and phe-
notypic characterization of a null mutant. Mol Microbiol 24: 629–642
53 Berk SG, Ting RS, Turner GW, Ashburn RJ (1998) Production of respirable
vesicles containing live Legionella pneumophila cells by two Acanthamoeba
spp. Appl Environ Microbiol 64: 279–286
54 Barker J, Scaife H, Brown MRW (1995) Intraphagocytic growth induces an
antibiotic-resistant phenotype of Legionella pneumophila. Antimicrob Agents
Chemother 39: 2684–2688
55 Abu Kwaik Y, Gao L-Y, Stone BJ, Venkataraman C, Harb OS (1998) Invasion
of protozoa by Legionella pneumophila and its role in bacterial ecology and
pathogenesis. Appl Environ Microbiol 64: 3127–3133
56 Fields BS, Nerad TA, Sawyer TK, King CH, Barbaree JM, Martin WT, Morrill
WE, Sanden GN (1990) Characterization of an axenic strain of Hartmannella
vermiformis obtained from an investigation of nosocomial legionellosis. J
Protozool 37: 581–583
57 Byrne B, Swanson MS (1998) Expression of Legionella pneumophila virulence
traits in response to growth conditions. Infect Immun 66: 3029–3034
58 Hammer BK, Swanson MS (1999) Co-ordination of Legionella pneumophila
virulence with entry into stationary phase by ppGpp. Mol Microbiol 33(4):
721–731
59 Hammer BK, Tateda ES, Swanson MS (2002) A two-component regulator
induces the transmission phenotype of stationary-phase Legionella pneumoph-
ila. Mol Microbiol 44(1): 107–118
60 Barker J, Lambert PA, Brown MRW (1993) Influence of intra-amoebic and
other growth conditions on the surface properties of Legionella pneumophila.
Infect Immun 61: 3503–3510
61 Cirillo JD, Tompkins LS, Falkow S (1994) Growth of Legionella pneumophila
in Acanthamoeba castellanii enhances invasion. Infect Immun 62: 3254–3261
62 Brieland JK, Fantone JC, Remick DG, LeGendre M, McClain M, Engleberg
NC (1997) The role of Legionella pneumophila-infected Hartmanella vermifor-
mis as an infectious particle in a murine model of Legionnaires’ disease. Infect
Immun 65: 4892–4896
63 O’Brien SJ, Bhopal RS (1993) Legionnaires’ disease: the infective dose para-
dox. Lancet 342: 5–6
64 Steinert M, Emody L, Amann R, Hacker J (1997) Resuscitation of viable but
nonculturable Legionella pneumophila Philadelphia JR32 by Acanthamoeba
castellanii. Appl Environ Microbiol 63: 2047–2053
65 Venkataraman C, Haack BJ, Bondada S, Abu Kwaik Y (1997) Identification of
a Gal/GalNAc lectin in the protozoan Hartmannella vermiformis as a potential
receptor for attachment and invasion by the Legionnaires’ disease bacterium,
Legionella pneumophila. J Exp Med 186: 537–547
66 Harb OS, Venkataraman C, Haack BJ, Gao L-Y, Abu Kwaik Y (1998)
Heterogeneity in the attachment and uptake mechanisms of the Legionnaires’
132 Dina M. Bitar et al.

disease bacterium, Legionella pneumophila, by protozoan hosts. Appl Environ


Microbiol 64: 126–132
67 Mann BJ, Torian BE, Vedvick TS, Petri WAJ (1991) Sequence of a cysteine-rich
galactose-specific lectin of Entamoeba histolytica. Proc Natl Acad Sci USA 88:
3248–3252
68 Venkataraman C, Gao L-Y, Bondada S, Abu Kwaik Y (1998) Identification of
putative cytoskeletal protein homologues in the protozoan Hartmannella ver-
miformis as substrates for induced tyrosine phosphatase activity upon attach-
ment to the Legionnaires’ disease bacterium, Legionella pneumophila. J Exp
Med 188: 505–514
69 Abu Kwaik Y, Venkataraman C, Gao L-Y, Harb OS (1998) Signal transduction
in the protozoan host Hartmannella vermiformis upon attachment and invasion
by its bacterial parasite, the Legionnaires’ disease agent, Legionella micdadei.
Appl Environ Microbiol 64: 3134–3139
70 Abu Kwaik Y, Fields BS, Engleberg NC (1994) Protein expression by the
protozoan Hartmannella vermiformis upon contact with its bacterial parasite
Legionella pneumophila. Infect Immun 62: 1860–1866
71 Horwitz MA (1984) Phagocytosis of the Legionnaires’ disease bacterium
(Legionella pneumophila) occurs by a novel mechanism: engulfment within a
pseudopod coil. Cell 36: 27–33
72 Hilbi H, Segal G, Shuman HA (2001) Icm/dot-dependent upregulation of
phagocytosis by Legionella pneumophila. Mol Microbiol 42(3): 603–617
73 Horwitz MA, Silverstein SC (1981) Interaction of the legionnaires’ disease
bacterium (Legionella pneumophila) with human phagocytes. II. Antibody
promotes binding of L. pneumophila to monocytes but does not inhibit intra-
cellular multiplication. J Exp Med 153: 398–406
74 Payne NR, Horwitz MA (1987) Phagocytosis of Legionella pneumophila is
mediated by human monocyte complement receptors. J Exp Med 166: 1377–
1389
75 Marra A, Horwitz MA, Shuman HA (1990) The HL-60 model for the inter-
action of human macrophages with the Legionnaires’ disease bacterium. J
Immunol 144: 2738–2744
76 Cirillo JD, Cirillo SL, Yan L, Bermudez LE, Falkow S, Tompkins LS (1999)
Intracellular growth in Acanthamoeba castellanii affects monocyte entry mech-
anisms and enhances virulence of Legionella pneumophila. Infect Immun 67(9):
4427–4434
77 Watarai M, Derre I, Kirby J, Growney JD, Dietrich WF, Isberg RR (2001)
Legionella pneumophila is internalized by a macropinocytotic uptake pathway
controlled by the Dot/Icm system and the mouse Lgn1 locus. J Exp Med 194(8):
1081–1096
78 Bandyopadhyay P, Xiao H, Coleman HA, Price-Whelan A, Steinman HM
(2004) Icm/dot-independent entry of Legionella pneumophila into amoeba and
macrophage hosts. Infect Immun 72(8): 4541–4551
79 Cirillo SL, Lum J, Cirillo JD (2000) Identification of novel loci involved in entry
by Legionella pneumophila. Microbiology 146(Pt 6): 1345–1359
80 Cirillo SL, Bermudez LE, El-Etr SH, Duhamel GE, Cirillo JD (2001)
Legionnaires’ disease and its agent Legionella pneumophila 133

Legionella pneumophila entry gene rtxA is involved in virulence. Infect Immun


69(1): 508–17
81 Abu Kwaik Y (1998) Fatal attraction of mammalian cells to Legionella pneu-
mophila. Mol Microbiol 30: 689–696
82 Elliott JA, Winn WC Jr (1986) Treatment of alveolar macrophages with cyto-
chalasin D inhibits uptake and subsequent growth of Legionella pneumophila.
Infect Immun 51: 31–36
83 Rechnitzer C, Blom J (1989) Engulfment of the Philadelphia strain of Legionella
pneumophila within pseudopod coils in human phagocytes. Comparison with
the other Legionella strains and species. Acta Pathol Microbiol Immunol Scand
[B] 97: 105–114
84 Weinbaum DL, Benner RR, Dowling JN, Alpern A, Pasculle AW, Donowitz
GR (1984) Interaction of Legionella micdadei with human monocytes. Infect
Immun 46: 68–73
85 Dowling JN, Saha AK, Glew RH (1992) Virulence factors of the family
Legionellaceae. Microbiol Rev 56: 32–60
86 Yamamoto Y, Klein TW, Friedman H (1992) Genetic control of macrophage
susceptibility to infection by Legionella pneumophila. FEMS Microbiol
Immunol 89: 137–146
87 Beckers MC, Yoshida S, Morgan K, Skamene E, Gros P (1995) Natural resis-
tance to infection with Legionella pneumophila: chromosomal localization of
the Lgn1 susceptibility gene. Mamm Genome 6(8): 540–545
88 Beckers MC, Ernst E, Diez E, Morissette C, Gervais F, Hunter K, Housman D,
Yoshida S, Skamene E, Gros P (1997) High-resolution linkage map of mouse
chromosome 13 in the vicinity of the host resistance locus Lgn1. Genomics
39(3): 254–263
89 Dietrich WF, Damron DM, Isberg RR, Lander ES, Swanson MS (1995) Lgn1, a
gene that determines susceptibility to Legionella pneumophila, maps to mouse
chromosome 13. Genomics 26(3): 443–50
90 Miyamoto H, Maruta K, Ogawa M, Beckers MC, Gros P, Yoshida S (1996)
Spectrum of Legionella species whose intracellular multiplication in murine
macrophages is genetically controlled by Lgn1. Infect Immun 64(5): 1842–5
91 Maier JK, Lahoua Z, Gendron NH, Fetni R, Johnston A, Davoodi J, Rasper D,
Roy S, Slack RS, Nicholson DW et al (2002) The neuronal apoptosis inhibitory
protein is a direct inhibitor of caspases 3 and 7. J Neurosci 22(6): 2035–2043
92 Wright EK, Goodart SA, Growney JD, Hadinoto V, Endrizzi MG, Long EM,
Sadigh K, Abney AL, Bernstein-Hanley I, Dietrich WF (2003) Naip5 affects
host susceptibility to the intracellular pathogen Legionella pneumophila. Curr
Biol 13(1): 27–36
93 Diez E, Lee SH, Gauthier S, Yaraghi Z, Tremblay M, Vidal S, Gros P (2003)
Birc1e is the gene within the Lgn1 locus associated with resistance to Legionella
pneumophila. Nat Genet 33(1): 55–60
94 Kirby JE, Isberg RR (1998) Legionnaires‘ disease: the pore macrophage and
the legion of terror within. Trends Microbiol 6: 256–258
95 Kirby JE, Vogel JP, Andrews HL, Isberg RR (1998) Evidence for pore-forming
ability by Legionella pneumophila. Mol Microbiol 27: 323–336
96 Coers J, Monahan C, Roy CR (1999) Modulation of phagosome biogenesis
134 Dina M. Bitar et al.

by Legionella pneumophila creates an organelle permissive for intracellular


growth. Nat Cell Biol 1(7): 451–453
97 Katz SM, Hashemi S (1982) Electron microscopic examination of the inflam-
matory response to Legionella pneumophila in guinea pigs. Lab Invest 46:
24–32
98 Horwitz MA (1983) Formation of a novel phagosome by the Legionnaires’
disease bacterium (Legionella pneumophila) in human monocytes. J Exp Med
158: 1319–1331
99 Tilney LG, Harb OS, Connelly PS, Robinson CG, Roy CR (2001) How the para-
sitic bacterium Legionella pneumophila modifies its phagosome and transforms
it into rough ER: implications for conversion of plasma membrane to the ER
membrane. J Cell Sci 114 (Pt 24): 4637–4650
100 Swanson MS, Isberg RR (1995) Formation of the Legionella pneumophila rep-
licative phagosome. Infect Agents Dis 2: 269–271
101 Horwitz MA (1983) The Legionnaires’ disease bacterium (Legionella pneu-
mophila) inhibits phagosome-lysosome fusion in human monocytes. J Exp Med
158: 2108–2126
102 Horwitz MA, Maxfield FR (1984) Legionella pneumophila inhibits acidifica-
tion of its phagosome in human monocytes. J Cell Biol 99: 1936–1943
103 Gao L-Y, Susa M, Ticac B, Abu Kwaik Y (1999) Heterogeneity in intracellular
replication and cytopathogenicity of Legionella pneumophila and Legionella
micdadei in mammalian and protozoan cells. Microb Pathog 27: 273–287
104 Roy CR, Tilney LG (2002) The road less traveled: transport of Legionella to the
endoplasmic reticulum. J Cell Biol 158(3): 415–419
105 Swanson MS, Isberg RR (1995) Association of Legionella pneumophila with
the macrophage endoplasmic reticulum. Infect Immun 63: 3609–3620
106 Kagan JC, Roy CR (2002) Legionella phagosomes intercept vesicular traffic
from endoplasmic reticulum exit sites. Nat Cell Biol 4(12): 945–54
107 Kagan JC, Stein MP, Pypaert M, Roy CR (2004) Legionella subvert the func-
tions of Rab1 and Sec22b to create a replicative organelle. J Exp Med 199(9):
1201–1211
108 Derre I, Isberg RR (2004) Legionella pneumophila replication vacuole forma-
tion involves rapid recruitment of proteins of the early secretory system. Infect
Immun 72(5): 3048–3053
109 Abu Kwaik Y (1998) Induced expression of the Legionella pneumophila gene
encoding a 20-kilodalton protein during intracellular infection. Infect Immun
66: 203–212
110 Hagele S, Kohler R, Merkert H, Schleicher M, Hacker J, Steinert M (2000)
Dictyostelium discoideum: a new host model system for intracellular pathogens
of the genus Legionella. Cell Microbiol 2(2): 165–171
111 Solomon JM, Isberg RR (2000) Growth of Legionella pneumophila in
Dictyostelium discoideum: a novel system for genetic analysis of host-pathogen
interactions. i 8(10): 478–480
112 Li Z, Solomon JM, Isberg RR (2005) Dictyostelium discoideum strains lacking
the RtoA protein are defective for maturation of the Legionella pneumophila
replication vacuole. Cell Microbiol 7(3): 431–442
113 Fajardo M, Schleicher M, Noegel A, Bozzaro S, Killinger S, Heuner K, Hacker
Legionnaires’ disease and its agent Legionella pneumophila 135

J, Steinert M (2004) Calnexin, calreticulin and cytoskeleton-associated proteins


modulate uptake and growth of Legionella pneumophila in Dictyostelium dis-
coideum. Microbiology 150(Pt 9): 2825–2835
114 Otto GP, Wu MY, Clarke M, Lu H, Anderson OR, Hilbi H, Shuman HA,
Kessin RH (2004) Macroautophagy is dispensable for intracellular replication
of Legionella pneumophila in Dictyostelium discoideum. Mol Microbiol 51(1):
63–72
115 Alli OA, Zink S, von Lackum NK, Abu-Kwaik Y (2003) Comparative assess-
ment of virulence traits in Legionella spp. Microbiology 149(Pt 3): 631–641
116 Gao L-Y, Harb OS, Abu Kwaik Y (1998) Identification of macrophage-specific
infectivity loci (mil) of Legionella pneumophila that are not required for infec-
tivity of protozoa. Infect Immun 66: 883–892
117 Cianciotto NP, Fields BS (1992) Legionella pneumophila mip gene potentiates
intracellular infection of protozoa and human macrophages. Proc Natl Acad Sci
USA 89: 5188–5191
118 Stone BJ, Brier A, Kwaik YA (1999) The Legionella pneumophila prp locus;
required during infection of macrophages and amoebae. Microb Pathog 27(6):
369–376
119 Solomon JM, Rupper A, Cardelli JA, Isberg RR (2000) Intracellular growth
of Legionella pneumophila in Dictyostelium discoideum, a system for genetic
analysis of host-pathogen interactions. Infect Immun 68(5): 2939–2947
120 Nagai H, Cambronne ED, Kagan JC, Amor JC, Kahn RA, Roy CR (2005) A
C-terminal translocation signal required for Dot/Icm-dependent delivery of the
Legionella RalF protein to host cells. Proc Natl Acad Sci USA 102(3): 826–831
121 Coers J, Kagan JC, Matthews M, Nagai H, Zuckman DM, Roy CR (2000)
Identification of icm protein complexes that play distinct roles in the biogenesis
of an organelle permissive for Legionella pneumophila intracellular growth.
Mol Microbiol 38(4): 719–736
122 Feldman MF, Cornelis GR (2003) The multitalented type III chaperones: all
you can do with 15 kDa. FEMS Microbiol Lett 219(2): 151–158
123 Bardill JP, Miller JL, Vogel JP (2005) IcmS-dependent translocation of SdeA
into macrophages by the Legionella pneumophila type IV secretion system.
Mol Microbiol 56(1): 90–103
124 Ninio S, Zuckman-Cholon DM, Cambronne ED, Roy CR (2005) The Legionella
IcmS-IcmW protein complex is important for Dot/Icm-mediated protein trans-
location. Mol Microbiol 55(3): 912–926
125 Bachman MA, Swanson MS (2001) RpoS co-operates with other factors
to induce Legionella pneumophila virulence in the stationary phase. Mol
Microbiol 40(5): 1201–1214
126 Hales LM, Shuman HA (1999) The Legionella pneumophila rpoS gene is
required for growth within Acanthamoeba castellanii. J Bacteriol 181(16):
4879–4889
127 Samrakandi MM, Cirillo SL, Ridenour DA, Bermudez LE, Cirillo JD (2002)
Genetic and phenotypic differences between Legionella pneumophila strains. J
Clin Microbiol 40(4): 1352–1362
128 Zusman T, Gal-Mor O, Segal G (2002) Characterization of a Legionella pneu-
136 Dina M. Bitar et al.

mophila relA insertion mutant and toles of RelA and RpoS in virulence gene
expression. J Bacteriol 184(1): 67–75
129 Fettes PS, Forsbach-Birk V, Lynch D, Marre R (2001) Overexpresssion of a
Legionella pneumophila homologue of the E. coli regulator csrA affects cell
size, flagellation, and pigmentation. Int J Med Microbiol 291(5): 353–360
130 Lynch D, Fieser N, Gloggler K, Forsbach-Birk V, Marre R (2003) The response
regulator LetA regulates the stationary-phase stress response in Legionella
pneumophila and is required for efficient infection of Acanthamoeba castel-
lanii. FEMS Microbiol Lett 219(2): 241–248
131 Gal-Mor O, Segal G (2003) The Legionella pneumophila GacA homolog
(LetA) is involved in the regulation of icm virulence genes and is required for
intracellular multiplication in Acanthamoeba castellanii. Microb Pathog 34(4):
187–194
132 Cohen JJ (1993) Overview: Mechanisms of apoptosis. Immunol Today 14:
126–130
133 Gao L-Y, Abu Kwaik Y (1999) Apoptosis in macrophages and alveolar epithe-
lial cells during early stages of infection by Legionella pneumophila and its role
in cytopathogenicity. Infect Immun 67: 862–870
134 Gao L-Y, Abu Kwaik Y (1999) Activation of caspase-3 in Legionella pneu-
mophila-induced apoptosis in macrophages. Infect Immun 67(9): 4886–4894
135 Hagele S, Hacker J, Brand BC (1998) Legionella pneumophila kills human
phagocytes but not protozoan host cells by inducing apoptotic cell death.
FEMS Microbiol Lett 169(1): 51–58
136 Molmeret M, Zink SD, Han L, Abu-Zant A, Asari R, Bitar DM, Abu Kwaik Y
(2004) Activation of caspase-3 by the Dot/Icm virulence system is essential for
arrested biogenesis of the Legionella-containing phagosome. Cell Microbiol
6(1): 33–48
137 Zink SD, Pedersen L, Cianciotto NP, Abu-Kwaik Y (2002) The Dot/Icm type
IV secretion system of Legionella pneumophila is essential for the induction of
apoptosis in human macrophages. Infect Immun 70(3): 1657–1663
138 Alli OAT, Gao L-Y, Pedersen LL, Zink S, Radulic M, Doric M, Abu Kwaik Y
(2000) Temporal pore formation-mediated egress from macrophages and alve-
olar epithelial cells by Legionella pneumophila. Infect Immun 68: 6431–6440
139 Molmeret M, Alli OA, Zink S, Flieger A, Cianciotto NP, Kwaik YA (2002) icmT
is essential for pore formation-mediated egress of Legionella pneumophila
from mammalian and protozoan cells. Infect Immun 70(1): 69–78
140 Molmeret M, Bitar DM, Han L, Kwaik YA (2004) Disruption of the phagosom-
al membrane and egress of Legionella pneumophila into the cytoplasm during
the last stages of intracellular infection of macrophages and Acanthamoeba
polyphaga. Infect Immun 72(7): 4040–4051
141 Arnoult D, Tatischeff I, Estaquier J, Girard M, Sureau F, Tissier JP, Grodet A,
Dellinger M, Traincard F, Kahn A et al (2001) On the evolutionary conserva-
tion of the cell death pathway: mitochondrial release of an apoptosis-induc-
ing factor during Dictyostelium discoideum cell death. Mol Biol Cell 12(10):
3016–3030
142 Akamine M, Higa F, Arakaki N, Kawakami K, Takeda K, Akira S, Saito A
Legionnaires’ disease and its agent Legionella pneumophila 137

(2005) Differential roles of Toll-like receptors 2 and 4 in in vitro responses of


macrophages to Legionella pneumophila. Infect Immun 73(1): 352–361
143 Tateda K, Matsumoto T, Ishii Y, Furuya N, Ohno A, Miyazaki S, Yamaguchi K
(1998) Serum cytokines in patients with Legionella pneumonia: relative pre-
dominance of Th1-type cytokines. Clin Diagn Lab Immunol 5(3): 401–403
144 Tateda K, Moore TA, Deng JC, Newstead MW, Zeng X, Matsukawa A, Swanson
MS, Yamaguchi K, Standiford TJ (2001) Early recruitment of neutrophils deter-
mines subsequent T1/T2 host responses in a murine model of Legionella pneu-
mophila pneumonia. J Immunol 166(5): 3355–3361
145 Gao L-Y, Stone BJ, Brieland JK, Abu Kwaik Y (1998) Different fates of
Legionella pneumophila pmi and mil mutants within human-derived macro-
phages and alveolar epithelial cells. Microb Pathog 25: 291–306
146 Brieland JK, Jackson C, Hurst S, Loebenberg D, Muchamuel T, Debets
R, Kastelein R, Churakova T, Abrams J, Hare R, O’Garra A (2000)
Immunomodulatory role of endogenous interleukin-18 in gamma interferon-
mediated resolution of replicative Legionella pneumophila lung infection.
Infect Immun 68(12): 6567–6573
147 Santic M, Molmeret M, Abu Kwaik Y (2005) Maturation of the Legionella
pneumophila-containing phagosome into a phagolysosome within gamma
interferon-activated macrophages. Infect Immun 73(5): 3166–3171
148 Skerrett SJ, Bagby GJ, Schmidt RA, Nelson S (1997) Antibody-mediated
depletion of tumor necrosis factor-alpha impairs pulmonary host defenses to
Legionella pneumophila. J Infect Dis 176(4): 1019–1028
149 Brieland JK, Remick DG, Freeman PT, Hurley MC, Fantone JC, Engleberg NC
(1995) In vivo regulation of replicative Legionella pneumophila lung infection
by endogenous tumor necrosis factor alpha and nitric oxide. Infect Immun 63:
3253–3258
150 Brieland J, Freeman P, kunkel R, Chrisp C, Hurley M, Fantone J, Engleberg NC
(1994) Replicative Legionella pneumophila lung infection in intratracheally
inoculated A/J mice: A murine model of human Legionnaires’ disease. Am J
Pathol 145: 1537–1546
151 Brieland J, McClain M, Heath L, Chrisp C, Huffnagle G, LeGendre M, Hurley
M, Fantone J, Engleberg C (1996) Coinoculation with Hartmannella vermifor-
mis enhances replicative Legionella pneumophila lung infection in a murine
model of Legionnaires’ disease. Infect Immun 64(7): 2449–2456
152 Nash TW, Libby DM, Horwitz MA (1988) IFN-gamma-activated human alveo-
lar macrophages inhibit the intracellular multiplication of Legionella pneu-
mophila. J Immunol 140: 3978–3981
153 Byrd TF, Horwitz MA (1989) Interferon gamma-activated human monocytes
downregulate transferrin receptors and inhibit the intracellular multiplication
of Legionella pneumophila by limiting the availability of iron. J Clin Invest 83:
1457–1465
154 Neild AL, Shin S, Roy CR (2005) Activated macrophages infected with
Legionella inhibit T cells by means of MyD88-dependent production of prosta-
glandins. J Immunol 175(12): 8181–8190
155 Gebran SJ, Yamamoto Y, Newton C, Klein TW, Friedman H (1994) Inhibition
of Legionella pneumophila growth by gamma interferon in permissive A/J
138 Dina M. Bitar et al.

mouse macrophages: role of reactive oxygen species, nitric oxide, tryptophan,


and iron(III). Infect Immun 62(8): 3197–3205
156 Yamamoto Y, Klein TW, Friedman H (1996) Immunoregulatory role of nitric
oxide in Legionella pneumophila-infected macrophages. Cell Immunol 171(2):
231–9
157 Yamamoto Y, Klein TW, Newton CA, Widen R, Friedman H (1988) Growth of
Legionella pneumophila in thioglycolate-elicited peritoneal macrophages from
A/J mice. Infect Immun 56: 370–375
158 Breiman RF, Horwitz MA (1987) Guinea pigs sublethally infected with aero-
solized Legionella pneumophila develop humoral and cell-mediated immune
responses and are protected against lethal aerosol challenge. A model for
studying host defense against lung infections caused by intracellular pathogens.
J Exp Med 165: 799–811
159 Horwitz MA, Silverstein SC (1981) Interaction of the Legionnaires‘ disease
bacterium (Legionella pneumophila) with human phagocytes. I. L. pneumoph-
ila resists killing by polymorphonuclear leukocytes, antibody, and complement.
J Exp Med 153: 386–397
160 Susa M, Ticac T, Rukavina T, Doric M, Marre R (1998) Legionella pneumoph-
ila infection in intratracheally inoculated T cell depleted or non-depleted A/J
mice. J Immunol 160: 316–321
161 Neild AL, Roy CR (2003) Legionella reveal dendritic cell functions that
facilitate selection of antigens for MHC class II presentation. Immunity 18(6):
813–823
162 Neild AL, Roy CR (2004) Immunity to vacuolar pathogens: what can we learn
from Legionella? Cell Microbiol 6(11): 1011–1018
163 Neild A, Murata T, Roy CR (2005) Processing and major histocompatibility
complex class II presentation of Legionella pneumophila antigens by infected
macrophages. Infect Immun 73(4): 2336–2343
Community-Acquired Pneumonia 139
ed. by N. Suttorp, T. Welte and R. Marre
© 2007 Birkhäuser Verlag Basel/Switzerland

Pathogenesis of Streptococcus pneumoniae infections:


adaptive immunity, innate immunity, cell biology,
virulence factors

Sven Hammerschmidt1, Gavin K. Paterson2, Simone Bergmann1 and


Timothy J. Mitchell2
1Research Center for Infectious Diseases, University of Würzburg, Röntgenring 11, 97070
Würzburg, Germany; 2Division of Infection and Immunity, Institute of Biomedical and Life
Science, Joseph Black Building, University of Glasgow G12–8QQ, UK

Abstract
During the past two decades the intense study of the infection process of Streptococcus
pneumoniae has elucidated multifaceted interactions of the human pathogenic bacterium
with the host. A broad spectrum of pneumococcal virulence factors, which are adapted
successfully to different host niches, is involved either predominantly in nasopharyngeal
colonization or subsequently in dissemination and transmigration of host tissue barriers.
The severe course of infections becomes manifest in invasive diseases like pneumonia,
meningitis and septicaemia.
To escape the risk of increasing antibiotic resistance and to combat the threat of
pneumococcal infections pneumococcal vaccines have been developed. The carrier pro-
tein of the current available heptavalent vaccine is not derived from pneumococci there-
fore it is thought to substitute this carrier by a highly conserved and immunogenic pneu-
mococcal-specific protein. S. pneumoniae is a versatile microorganism and has evolved
numerous successful strategies to colonize its host and to evade host defence mechanisms.
In this report we discuss the bacterial repertoire of virulence factors and provide insights
into the surface protein variability. In addition, we show the impact of these virulence
factors on interactions with host components, including cellular receptors and how the
function of these proteins contributes to colonization and virulence of S. pneumoniae.
The non-invasive and invasive infections are accompanied by immune responses of both
the innate and adaptive immune system. These two systems operate in concert to combat
infections, but pneumococci have developed highly sophisticated mechanisms to subvert
the host immune system. We introduce pattern recognition receptors that recognize spe-
cific structures of pneumococci and stimulate thereby host defence mechanisms.

Introduction

Streptococcus pneumoniae (the pneumococcus) is a serious human patho-


gen causing local infections including otitis media and sinusitis and life-
threatening invasive diseases, such as lobar pneumonia, sepsis and men-
140 Sven Hammerschmidt et al.

ingitis [1]. The burden of disease is highest in the youngest and elderly
population and in patients with immunodeficiencies. The pneumococcus is
the prime cause of community-acquired pneumonia in adults and accounts
for 50–75% cases. The incidence of pneumococcal pneumonia remains high,
between 68–260 cases per 100,000 and year [2]. Despite the use of antibiot-
ics and availability of vaccines the mortality rate remains high. Each year,
1 million children younger than 5 years die from pneumonia and invasive
diseases [3]. Even more, community-acquired pneumococcal meningitis has
a very high case-fatality rate. The survivors often develop long-term clinical
sequelae including hearing loss, neurological deficits, and neurophysiologi-
cal impairment [4]. The infections caused by pneumococci are preceded by
an asymptomatic carrier status and accompanied by the transmigration of
tissue barriers by the pathogen. The clinical outcome of disease has been
shown to be dependent on both the pathogen and host susceptibility for
the individual pathogen. Pneumococci are endowed with a multitude of
factors that contribute to the pathogenic potential of this versatile patho-
gen. Biological activities that have been attributed to these virulence fac-
tors include the subversion of host immunity and adoption of host-protein
functions to facilitate adherence and invasion. Pneumococci disseminate
and gain access to the ear, lungs, blood or meninges. The adaptation of
pneumococci to different host milieus including the nasopharynx has been
correlated with a differential expression of several pneumococcal factors
[5–9]. A comprehensive understanding of critical steps during pneumococ-
cal pathogenesis including colonization, progression to pneumonia, dis-
semination in the bloodstream, and transition of the blood-brain-barrier is
crucial to combat the threat of pneumococcal infections and hence, reduce
the mortality exacted by this pathogen. This review will evaluate our current
understanding of the mechanisms employed by pneumococci to encounter
the human host.

Nasopharyngeal colonization

S. pneumoniae is a complex microorganism divided into over 90 serotypes


depending on the antigenic structures of their capsular polysaccharides [10].
Pneumococci of different serotypes are able to simultaneously colonize the
nasopharyngeal cavity of healthy individuals [11]. The rates of carriage and
acquisition depend on age, genetic background, socioeconomic conditions
and geographical area [12]. Europe and the United States show similar
serotype distributions with minor differences in several serotypes. Disease is
most commonly due to strains representing 20 of the > 90 different pneumo-
coccal serotypes and these 20 serotypes have been covered by the 23-valent
polysaccharide (PS) vaccine [13, 14]. By contrast, the serotype distribution
in Asia is slightly different, which had resulted in only < 70% effectiveness
of the 23-valent conjugate PS vaccine [15]. The current vaccine, a seven-
Pathogenesis of Streptococcus pneumoniae infections… 141

Figure 1. Adherence of Streptococcus pneumoniae to human epithelial cells after 4 h infec-


tion as seen by high-resolution field emission scanning electron microscopy. (Image courtesy
Manfred Rohde, German Research Centre for Biotechnology, Braunschweig, Germany.)

valent conjugate vaccine (Prevnar, Wyeth, USA), covers the most prevalent
serotypes 4, 6B, 9V, 14, 18C, 19F, and 23F. It is noteworthy that invasive
disease originates from nasopharyngeal colonization with the homologous
serotype [16]. Interestingly, certain serotypes including 14 and 18C clones
have a high potential to cause invasive disease, whereas the most commonly
carried serotypes 6B, 19F, and 23F are least invasive. By contrast, infrequent
colonizers and non-vaccine serotypes including serotypes 8, 38, 33F appear
to be more invasive [12, 17].

Transition from colonization to pneumonia and invasive infections

The mechanisms of how the pneumococcus makes the transition from a


commensal with asymptomatic carriage to a virulent pathogen causing dis-
ease have not yet been fully explained. It is, however, known that the local
immune response has an important regulatory role during colonization and
subsequent infections [18]. The successful conversion of the commensal to
an invasive microorganism is accompanied by the transmigration of tissue
barriers and the subsequent adaptation of the pathogen to different host
niches. A prerequisite for transmigration is the ability of the pneumococ-
cus to adhere to mucosal cells of the respiratory tract. It has been shown
that pneumcoccal colonization of the nasopharynx correlates with observed
differences in colony phenotypes. S. pneumoniae undergoes spontaneous,
reversible opacity phase variation with a frequency of 10–3 to 10–6 resulting
in opaque and transparent colonies [19]. Differences in colonial opacity
have previously been shown to correlate with different levels of capsular
polysaccharide (CPS) [19]. The transparent phenotype produces lower
amounts of CPS and has an enhanced ability to reside on mucosal surfaces
when tested in several animal models [20, 21]. By contrast, the opaque
variant is more virulent in systemic infections [22]. The higher amounts of
142 Sven Hammerschmidt et al.

CPS produced by these variants make them more resistant to complement-


mediated opsonophagocytotic killing [22]. Although higher amounts of CPS
have been demonstrated to block pneumococcal adherence, a basal level
of encapsulation is essential for colonization [23, 24]. Strikingly, electron
micrographs illustrated that the intimate contact of pneumococci with host
cells is associated with a reduction of encapsulation [25].
Pneumococci spread presumably to the lungs by aspiration and attach-
ment of pneumococci has been indicated to bronchial epithelial cells [26]
or components of the basement membrane including laminin, collagen and
fibronectin [27, 28]. Binding is promoted by an impaired ciliary beating
frequency due to the damage caused by smoking or the release of pneumo-
lysin, a cytolysin of pneumococci. Pneumolysin inhibits the normal beating
of cilia on epithelial and endothelial cells thereby facilitating penetration
of pneumococci into the bloodstream [29, 30]. The effect of the hydrogen
peroxide released by pneumococci, which is as toxic as pneumolysin to
ependymal ciliary cells, is masked in the presence of pneumolysin [31].
Within the alveoli pneumococci adhere to type II pneumocytes and can
spread rapidly into the blood by crossing the vascular endothelium [32].
Three stages of lesions were distinguished during pneumococcal pneu-
monia: (1) engorgement, (2) red hepatization, and (3) gray hepatization.
Engorgment is associated with the accumulation of a serous exudate in
the alveoli and is followed by a leakage of erythrocytes into the alveoli.
At the stage of gray hepatization bacterial multiplication peaks, fibrin is
formed through the procoagulant activity induced by pneumococci and
polymorphonuclear leukocytes (PMNs) recruited to the infection site start
to control pneumococcal multiplication [32]. Clearance of pneumococci
by opsonophagocytosis is further dependent on complement and facili-
tated by anti-capsular antibodies [15]. One of the remarkable features of
pneumococcal pneumonia is that the lungs of patients who withstand the
inflammatory response of the host and survive almost invariably return to
normal, irrespective of the severity of the systemic or pulmonary condition
when the disease was at its peak [33].

Cell wall structures and virulence factors of S. pneumoniae

Pneumococci are encased by a capsular polysaccharide that has been recog-


nized as a sine qua non of virulence. Survival in the bloodstream depends on
the expression of capsule. The degree of activation of the classical or alter-
native pathways of complement, the deposition and degradation of comple-
ment components, and the degree of protection against complement-medi-
ated opsonophagocytosis is determined by the biochemical structure of the
CPS rather than by the thickness of the CPS [15].
The layer underneath the capsule, the pneumococcal outer cell wall, is
comprised of peptidoglycan, teichoic (TA) and lipoteichoic acids (LTA),
Pathogenesis of Streptococcus pneumoniae infections… 143

Figure 2. Infections caused by pneumococci. Depicted are encapsulated pneumococci (arrow-


heads) in the cerebrospinal fluid (top left panel) and the histopathology of purulent meningitis
caused by pneumococci (top right panel). The photomicrograph of a section of the subarach-
noid space, in hematoxylin-eosin stain, shows the infiltration of leukocytes (arrowheads) and
purulent meningitis (arrows). Image courtesy Roland Nau, University of Göttingen, Germany.
Chest X-ray of a lobar pneumonia (bottom, left panel) and section of lung representing a stage
of hepatization.
144 Sven Hammerschmidt et al.

and phosphorylcholine (PCho). The PCho is covalently linked to the TA


and LTA, which differ only in their attachment to the pneumococcal cell
wall. Although PCho is part of the cell wall of other respiratory pathogens
including Neisseria spp., Haemophilus influenzae, and Pseudomonas, only
pneumococcal PCho anchors a special class of proteins, the choline-bind-
ing proteins (CBP) non-covalently on the surface. CBPs have a modular
organization and consist in general of a leader peptide, a biologically active
N-terminal domain and the conserved choline-binding domain (CBD) that
targets the aminoalcohol PCho. The CBD, generally located at the carboxy
terminus and preceded by a proline-rich sequence, consists of highly homol-
ogous 20-amino acid long repetitive sequences.
Pneumococci can produce 13 to 16 different CBPs and the number of
produced CBPs depends on the pneumococcal strain. To date, the extensive-
ly characterized CBPs include the pneumococcal surface protein A (PspA),
the pneumococcal surface protein C (also referred to as CbpA or SpsA),
and four cell wall hydrolases. In general, the bacterial cell wall hydrolases
(CWHs) are endogenous enzymes that specifically cleave covalent bonds of
the cell wall. The pneumococcal CWHs are: the major autolysin LytA (N-
acetyl-muramoyl-L-alanine amidase), a `-N-acetylglucosamidase (LytB), a
`-N-acetylmuramidase (LytC; lysozyme), and a phosphorylcholine esterase
(Pce or CbpE). LytA has been characterized in detail and recently, the struc-
tural analysis of the CBD of LytA demonstrated that this module adopts a
peculiar solenoid structure [34]. LytB is highly expressed in the early expo-
nential growth phase and has been shown to be important for cell separa-
tion of pneumococci. In contrast to other CBPs, LytB and LytC possess the
CBD as an N-terminal domain [34]. Apart from these, CbpD, CbpG, CbpJ,
CbpI and, PcpA represent further members of this family [35, 36].
Three clusters of surface proteins can be distinguished by genome analy-
sis: the lipoproteins including peptide permeases and ATP binding cassette
(ABC) transporter (42 in R6 and 47 in TIGR4), the above mentioned family
of CBP (10 in R6 and 15 in TIGR4) and proteins with an LPxTG motif (13
in R6 and 19 in TIGR4). The latter represent typical Gram-positive surface
proteins that are covalently anchored in the cell wall after cleavage of the
LPxTG sequence by a transpeptidase, designated sortase. Strikingly, many
of these LPxTG proteins contain enzymatic activity including neuramini-
dases, hyaluronidase, IgA1-protease and zinc metalloproteases (ZmpB,
ZmpC, and ZmpD). In addition to these predicted surface proteins, non-
classical surface proteins lacking a classical leader peptide and membrane
anchoring motifs, have been identified on the pneumococcal surface. These
proteins have received considerable attention for their contribution to viru-
lence of pneumococci and other pathogenic bacteria. To date, the mecha-
nism of secretion and anchoring of these proteins that include the enolase,
the glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and the PavA
(pneumococcal adherence and virulence factor A) remain unknown for
pneumococci and many other Gram-positive pathogens.
Pathogenesis of Streptococcus pneumoniae infections… 145

Another important molecule that is associated with pneumococcal


virulence, but not surface-exposed due to the lack of secretion signal,
is pneumolysin. This hemolysin is intracellularly produced by pneumo-
cocci and released by the action of cell-bound autolysin. Pneumolysin is a
sulfhydryl(thiol)-activated pore-forming cytolysin that binds to cholesterol
in the plasma membrane of host cells [37]. Soluble pneumolysin monomers
form ring-shaped oligomeric pores [38, 39]. The observation of the pore-
forming process by cryo-electron microscopy has indicated that the mem-
brane bound form with an intact bilayer is the prepore and that the confor-
mational transition from the prepore to the pore form is accompanied with
a separation of monomers and a substantial refolding of protein domains
[40]. The multiple biological activities of pneumolysin have been shown to
interfere with eukaryotic host-cell function and the immune system.

Cellular biology of pneumococcal infections

Bacteria attach to eukaryotic host cells via surface-exposed adhesins which


specifically interact with cellular host receptors or adhesive glycoproteins
of the extracellular matrix (ECM) which connect the microorganism with
cellular receptors [41]. It has been shown that pneumococci translocate the
respiratory barrier and gain access to the blood circulation through the
intracellular route [42]. However, the strategies used by pneumococci for
translocation are not yet fully elucidated and a paracellular route of entry
cannot be excluded as an alternative route.

Adherence to host cells

In the early stages of the infectious process pulmonary epithelial cells and
vascular endothelial cells are targeted by pneumococci. Attachment to rest-
ing lung cells and vascular endothelial cells occurs via the recognition of two
classes of glycoconjugates. The disaccharides N-acetyl-D-galactosamin `1-
3/4 galactose (GalNAc (`1-3/4) Gal) and sialylated N-acetyl-D-glucosamine
`1-3 galactose (GlcNAc (`1-3) Gal) are recognized by pneumococcal viru-
lence determinants that have not been identified so far [43, 44]. Binding of
pneumococci to resting cells was completely abolished by a combination of
these two carbohydrates as represented by asialo-GM and globoside [45].
It is has been suggested that the multiple neuraminidases (NanA, NanB
and probably NanC) of pneumococci cleave terminal sialic acid (N-acetyl-
neuraminic acid) from glycolipids, glycoproteins, and oligosaccharides on
host-cell surfaces and body fluids thereby enhancing intimate adherence. In
fact, NanA has been shown to cleave the terminal sialic acids of lipooligo-
saccharides from Haemophilus influenzae and Neisseria meningitidis [46].
Because terminal sialic acids protect these respiratory pathogens against
146 Sven Hammerschmidt et al.

complement mediated phagocytosis, desialylation of competitors may pro-


vide an advantage during colonization of host niches. In addition, NanA has
been shown to be implicated in desialylation of human proteins exhibiting
sialic acid including the secretory component, lactoferrin and IgA2 [47].
These human proteins are pneumococcal host targets and the removal of
sialic acid may facilitate bacterial persistence in the respiratory tract. NanA
sequence diversity is restricted to regions that are not required for enzy-
matic activity and has been suggested to provide an important advantage in
evading the adaptive immune response [48]. In a chinchilla infection model,
loss of NanA has been shown to impair pneumococcal persistence in the
nasopharynx and middle ear [49]. In contrast, the nanA knockout was not
attenuated in an intraperitoneal infection model [50]. A similar role is sug-
gested for the hyaluronidase (Hyl), which hydrolases primarily hyaluronan.
Although hyl knockout strains are attenuated in an intraperitoneal mouse
infection model [51], the precise function of the Hyl during pathogenesis
has yet to be clarified.
S. pneumoniae produce up to four zinc metallo proteases including IgA1-
protease, ZmpB, ZmpC, and ZmpD, which are anchored to the cell wall by
an N-terminal LPxTG motif. The IgA1-protease is produced virtually by all
pneumococci and Weiser and co-workers [52] demonstrated that cleavage
of surface-bound serotype-specific IgA1 by the IgA1-protease markedly
enhanced adherence of pneumococci to host cells. It has been assumed that
bound Fab fragments neutralize the negatively charged capsule and negate
the anti-adhesive effects of the capsule.
A role in colonization has also been suggested for the lipoprotein SlrA,
and the choline-binding proteins CbpD and CbpG, which is thought to be a
serine protease [35]. Recent reports indicated that CbpD is a competence-
stimulating-peptide-inducible protein and a function as a murein hydro-
lase has been proposed. CbpD has been demonstrated to assist LytA in
competence-induced cell lysis [53]. A further study provided experimental
evidence that the biological activity of CbpD is involved in the ability of
competent bacteria to trigger release of virulence factors from non-com-
petent S. pneumoniae [54]. The surface-exposed lipoprotein SlrA is a func-
tional, cyclophilin-type peptidyl-prolyl isomerase. The deficiency in SlrA
caused a less efficient nasopharyngeal colonization of mice, and this has
been attributed to the decreased ability of the knockout mutants to adhere
to non-professional cells [55].
The pneumococcal surface adhesin A (PsaA), which is the substrate-
binding lipoprotein of an ATP binding cassette (ABC)-type manganese
transport system [56] has also been demonstrated to affect adherence
of pneumococci. Mutations in psaA caused pleiotropic effects including
reduced adherence of pneumococci to host cells, attenuation in an intra-
nasal and intraperitoneal mouse infection model and increased sensitivity
to oxidative stress [9, 57] . After detecting the pleiotropic effects it was
assumed that PsaA does not function itself as an adhesin, however, a recent
Pathogenesis of Streptococcus pneumoniae infections… 147

report has demonstrated that antibodies against PsaA reduce the adherence
of pneumococci to nasopharyngeal epithelial cells [58]. This is consistent
with the finding that mucosal immunization of mice with PsaA is highly
protective against pneumococcal carriage [59] and that psaA is upregulated
during atttachment of pneumococci to epithelial cells [7, 9].

The major pneumococcal adhesin

The pneumococcal surface protein C (PspC; also known as SpsA or CbpA)


is a multifunctional CBP of pneumococci; PspC promotes uptake of pneu-
mococci into nasopharyngeal epithelial cells and interferes with components
of the innate immune system. PspC promotes pneumococcal adherence via
a human specific interaction with the secretory component (SC), which is
found on the polymeric immunoglobulin receptor and secretory forms of
IgA and IgM on mucosal surfaces [60–62]. The SC consists of five Ig-like
ectodomains (D1 to D5) and independent reports have indicated that the
human specificity of the PspC-SC interaction is determined by amino acid
differences in the ectodomains D3 and D4 of the SC [63, 64]. Despite such
human specificity, loss of function in PspC has been shown to reduce colo-
nization of infant rats [65] and pIgR knockout mice [62]. The underlying
mechanisms of these in vivo effects are poorly defined. However, PspC has
also been shown to bind to immobilized sialic acid and lacto-N-neotetraose
[65] and to interact with complement components including C3 and factor
H [66, 67].
The amino-terminal part of most of the PspC molecules contains repeat-
ed domains, designated R1 and R2. Structural analysis of R1 (aa175 to 285)
and R2 (aa327 to 442) of PspC derived from TIGR4 (PspC allele PspC3.4
[68]) demonstrated that the R domains adopt an unusual and simple struc-
ture comprised of three _-helices. These R domains of PspC contain the
minimal and conserved SC-binding site Y/RRNYPT [61]. The structure
indicated that bundling of the helices through _-helix/_-helix interactions
results in a flat, raft-like structure in which the critical residues YPT of the
minimal SC-binding motif are located in a loop between helix 1 and helix 2
and form a “tyrosine fork” structure [69].

Interaction with ECM components and recruitment of proteolytic


activity

There is experimental evidence that binding of microorganisms to ECM


components may be of importance for pathogenesis. Pneumococci have
been shown to bind to various ECM components, however, the impact of
these interactions on colonization are not yet clarified. The PavA protein
(pneumococcal adherence and virulence factor A) has been identified as a
148 Sven Hammerschmidt et al.

pneumococcal adhesin for fibronectin and, probably more important, as a


crucial virulence determinant in pneumococcal infections. In a systemic and
experimental mouse meningitis model of infection the pneumococcal pavA
knockout of strain D39 was substantially attenuated [70, 71]. In addition,
PavA is most likely involved indirectly and in a fibronectin-independent
manner in pneumococcal adherence to host cells. However, the precise
function of PavA has yet to be clarified, because deficiency in PavA does
not affect expression and function of known virulence factors [71].
S. pneumoniae acquires host proteolytic activity by binding plasmin(ogen),
and interestingly, the glycolytic enzymes enolase and GAPDH were identi-
fied as plasmin(ogen)-binding proteins displayed on the cell wall [72, 73].
The enolase has been shown to potentiate degradation of extracellular
matrix (ECM), dissolution of fibrin and pneumococcal transmigration [74].
The key pneumococcal binding site in enolase responsible for plasmin-
mediated ECM degradation has been attributed to the nonameric peptide
“FYDKERKVYD” [75]. Crystal structure analysis indicated an octameric
composition of the pneumococcal enolase and depicted the nonameric plas-
minogen-binding site on the surface of the protein [76]. The importance of
enolase in virulence has been demonstrated in intranasal mice infections of
mice. Enolase mutants with functionally inactive plasminogen-binding sites
were significantly attenuated compared to the isogenic D39 parental strain
[75].

Invasion of host cells by pneumococci

The proportion of internalized pneumococci in resting pulmonary cells was


only 0.1 % of the adherent bacteria [77]. In contrast, activation of vascular
endothelial cells with thrombin or tumor necrosis factor-_ (TNF-_) caused
a substantial increase in pneumococcal uptake. The cell activation is associ-
ated with an increase in expression of novel cell adhesion molecules such as
receptor for the platelet-activating factor (PAF). The PAF receptor is rap-
idly internalized after interaction with its ligand PAF and pneumococci have
been shown to engage the upregulated PAF receptor for internalization
[77]. It has been shown that the cell wall structural component phosphor-
ylcholine (PCho), which is also present in PAF, function as an adhesin for
the PAF receptor. Binding and uptake of pneumococci to activated endo-
thelial cells was inhibited by PAF antagonists, purified pneumococcal cell
wall components, or anti-PCho antibodies. The absence of PCho in pneu-
mococci reduced adherence to levels indicated for resting cells, indicating
that PCho directly interacts as an adhesive molecule with the PAF receptor
[77]. Binding of pneumococci via PCho to activated cells was inhibited
by glycoconjugates N-acetyl-glucosmamine or lacto-N-neotetraose. These
sugars showed no effect on adherence of pneumococci to resting cells. This
implicates that pneumococci most likely bind to the PAF receptor at two
Pathogenesis of Streptococcus pneumoniae infections… 149

Figure 3. Surface proteins of pneumococci contributing to colonization and invasion of host


cells.
A. The extracellular matrix (ECM) of the mucosal cavity represents the first mechanical bar-
rier for pneumococcal colonization. Colonization of pneumcocci is facilitated by cleavage of
bound IgA1 by the IgA1-protase and by removing of terminal sialic acids by NanA. The glyco-
lytic enzymes enolase and GAPDH bind human plasminogen (PLG), which is converted to the
protease plasmin (PA) and promotes degradation of various ECM compounds. ZmpC activates
the matrix metallo protease 9 (MMP-9) which has collagenase activity and degrades the ECM.
The non-classical surface associated protein PavA has been shown to bind to immobilized
fibronectin (Fn) and to modulate pneumococcal adherence.
B. The intimate contact of pneumococci with cellular receptors is mediated by adhesins. The
major adhesin of pneumococci identified is PspC, which interacts with the ectodomain of the
human polymeric immunoglobulin receptor (pIgR). PCho mediates pneumococcal adher-
ence to stimulated cells via an interaction with upregulated platelet activating factor receptor
(PAFr). It is thought that the lipoprotein PsaA may also function as an adhesin, but the cellular
receptor is not defined yet. Reprinted from Microbiology SGM [209] with permission of the
publisher.

sites: one shared with the natural ligand PAF and the other at a site of gly-
cosylation of the receptor. Alternatively, the glycosyl determinant can be
located on a putative co-receptor that interacts with the PAF receptor. The
PCho-PAF receptor interaction represents further a specific mechanism for
pneumococcal targeting of the blood-brain barrier (BBB) and transmigra-
tion of pneumococci across the BBB [24].
150 Sven Hammerschmidt et al.

The efficiency of colonization and invasion of brain microvascular


endothelial cells correlates with the higher amounts of PCho on transpar-
ent pneumococci. In addition, the amount of PCho is modulated by the
phosphorylcholine esterase activity of Pce (also referred to as CbpE) which
removes PCho from the cell wall and causes changes in colony phenotype
[78, 79]. The crystal structure reveals that Pce belongs to the metallo-`-
lactamase family and that only PCho residues that are located at the end
of the teichoic acid chains are accessible to the catalytic center of Pce [80,
81]. It is thought that Pce may have a dual function and favor both colo-
nization and invasive infection by modulating the amount of PCho on the
pneumococcal cell wall. The physiological significance of the PCho-PAF
receptor interaction was demonstrated in a rabbit model of pneumonia.
The PAF-receptor antagonist blocked activation-dependent adherence and
transition of pneumococci from the alveolus into the blood. This resulted in
reduced nasal colonisation and attenuated development of pneumococcal
bacteremia [77].

Cell signaling induced by S. pneumoniae

The PAF receptor is a G-protein coupled receptor and binding of PAF acti-
vates phopholipase C [82]. In contrast, internalization of pneumococci via
the PAF receptor was independent of the G-protein pathway and failed to
induce signal transduction [77]. The endocytosis of pneumococci requires
both the PAF receptor as a portal of entry and `-arrestin. It has been dem-
onstrated that pneumococci induced the translocation of `-arrestin at the
plasma membrane, where it colocalized with the PAF receptor. This event
caused a G-protein independent activation of the MAP kinase ERK-1/
ERK-2 and pneumococi moved into clathrin-coated vesicles. At least half
of the pneumococci proceeded through Rab5 to Rab7 marked endosomes
toward lysosomes. Other vacuoles acquire Rab11, which is consistent with
the known recycling of the bacteria to the apical surface [83].
In pneumococcal pneumonia massive leukocyte recruitment to the lung
is observed [84]. Invasion of leukocytes is accompanied by secretion of pro-
inflammatory and chemotactic cytokines by lung epithelium and cells of the
innate immune response including alveolar macrophages. It has been shown
that the pneumococcal cell wall mediated signaling induces the expression
of transcription factor NF-kB and induces the production of TNF-_, IL-8,
IL-6 and IL-8 [85-87]. Schmeck and co-workers demonstrated that in pneu-
mococcal pneumonia the induction of NF-gB and p38 MAPK signaling
pathways contribute to the secretion of IL-8 and GM-CSF. Activation of
NF-gB was IgB-kinase dependent, but activation was independent of p38
MAPK. It has been demonstrated that p38 MAPK did not affect inducible
nuclear translocation of NF-gB/RelA to the IL-8 promotor but regulates
IL-8 transcription on the level of phosphorylated RelA at the promoter.
Pathogenesis of Streptococcus pneumoniae infections… 151

Both, p38 MAPK and NF-gB have been shown to be required to modulate
the host response at the transcriptional level in response to pneumococci
infection [88].

Pulmonary virulence determinants

Large-scale in vivo screens including signature-tagged mutagenesis (STM),


differential fluorescence induction (DIF) and whole-genome microarray
analysis have provided an extended list of putative pulmonary virulence
factors [5–9]. These approaches confirmed already known virulence factors
including CbpD, ComD, Hyl, IgA1-protease, LytA, NanA, Pneumolysin,
PcpA, PsaA, PspA, PspC, sortase, and ZmpB. In addition, a role in patho-
genesis has been suggested for ABC transporters such as the glutamine
transporters, proteases such as HtrA and PrtA, proteins involved in meta-
bolic pathways and regulatory elements such as the transcription factor
RlrA. The importance of individual virulence determinants for pneumococ-
cal pneumonia has further been assessed in in vivo studies.
Pneumococci produce several proteases that are surface-exposed and
implicated in pneumococcal virulence. Intranasal infection experiments
confirmed the significant contribution of IgA1-protease and ZmpB to pneu-
mococcal virulence [89]. ZmpC was characterized in TIGR4 as a bacterial
zinc metallo protease cleaving human matrix metalloproteinase 9 (MMP-9)
and inactivation of the zmpC gene in serotype 19F impaired virulence in
a pneumonia mouse model [90]. The HtrA (high-temperature require-
ment A) functions in a temperature-dependent manner as a molecular
chaperone or heat shock-induced serine protease, and is regulated by the
CiaRH two-component system. HtrA has been shown to be implicated in
resistance against oxidative stress, colonizing the nasopharynx of rats and in
pneumococcal pneumonia. Moreover, htrA knockouts induced lower levels
of inflammatory cytokines IL-6 and TNF-_ in the lungs during pneumonia
compared to the isogenic wild type D39 [91–94]. PrtA is a further surface-
exposed serine protease and prtA knockouts have been shown to be attenu-
ated in an intraperitoneal mouse infection model [95].
The highly variable pneumococcal surface protein A (PspA) is expressed
virtually by all important clinical serotypes and has significant immune pro-
tective potential. Loss of function has been shown to attenuate virulence
and increase complement receptor-mediated clearance of pneumococci
[96]. PspA is a lactoferrin-binding protein [97] and Shaper et al. [98] dem-
onstrated that expression of PspA protects against the bactericidal effect of
apolactoferrin. In the presence of antibodies recognizing PspA this effect
is abrogated, suggesting that binding of apolactoferrin to PspA blocks the
bactericidal activity of apolactoferrin. Regarding the cell wall hydrolases of
S. pneumoniae, loss-of-function of LytB or LytC significantly reduced naso-
pharyngeal colonisation of rats [35].
152 Sven Hammerschmidt et al.

Table 1. Pulmonary virulence factors

Biological activities Refs.


Toxins Pneumolysin inhibition of ciliar mobility, cytotoxic [29, 30, 122]
and cytolytic activities
enzymes/ HtrA resistance against oxidative stress, [91–93]
proteases induction of inflammatory cytokines
Hyaluronidase hydrolysis of hyaluronan [51]
IgA1-protease enhancement of bacterial adherence to [52]
host cells
LytA, LytB, LytC cell wall hydrolases [34, 35]
Neuraminidase cleavage of terminal sialic acids of [46–49]
(NanA) human IgA secretory component, lacto-
ferrin and IgA2
PrtA serine protease [95]
ZmpB impact on pneumococal virulence [89]
ZmpC cleavage of metalloproteinase 9 [90]
(MMP-9)
choline-binding CbpD competence-induced cell lysis by [53, 54]
proteins murein hydrolytic activity, impact in
allolysis
Pce phosphorylcholine esterase [77, 78]
PcpA unknown [36]
PspA binds lactoferrin and protects against [97, 98, 127,
bactericidal effects of apolactoferrin, 128]
inhibition of complement-induced
clearance
PspC pneumococcal adhesin for pIgR [60]
regulatory com- ComD transformation competence factor [5, 211]
ponents RlrA transcriptional regulator [5]
lipoproteins PsaA Mn-transporter and putative adhesin [56]

Regulation of gene expression and virulence

The pneumococcal virulence determinants can be regulated by one-compo-


nent regulatory systems including luxS, rlrA, regM/R, and mgrA, or by one
of the 14 two-component regulatory systems (TCS) that have been identi-
fied in S. pneumoniae [99, 100]. TCS respond to environmental changes and
mediate, therefore, the adaptation of pneumococci to their different micro-
environments. Throup et al. [101] and others have demonstrated the impact
of TCS on pneumococcal virulence in a mouse infection model. The CiaRH
was the first TCS identified (and is required for efficient nasopharyngeal
colonization and important for protecting cells from stress of competence
development [102, 103]). As mentioned above, CiaRH regulates HtrA,
Pathogenesis of Streptococcus pneumoniae infections… 153

which is thought to be the key component in the contribution of CiaRH


to virulence [93]. The major adhesin PspC is upregulated during coloni-
zation [7] and pspC expression has been shown to be influenced by the
TCS RR06/HK06 [104]. However, in vitro and in vivo infections suggested
that this TCS is also important for the regulation of other yet unknown
virulence factors interfering with colonisation and invasion. Recently it
has been shown that the phosphorylated VicR (YycF) response regulator
of the TCS VicRK (YycF) binds to a region upstream of pspA and regu-
lates expression of pspA [105]. As opacity variation accounts for changes
of a number of cell surface-exposed components including capsule, PspC,
PspA and PCho it is implicated in pneumococcal colonization and invasive
diseases. Capsule regulation has been indicated under in vivo and in vitro
conditions. Recombinant exchanges and spontaneous sequence duplications
within type 3- and 8 specific capsule genes cause high-frequency serotype
and phase variations, respectively [106-108]. Proteins such as CpsB and
CpsD which influence production of capsular polysaccacharide regulate
the amount of capsule at the post-translational level. Both have been dem-
onstrateed to modulate the phosphorylation of CpsD or other substrate
molecules [109–112].

The pneumococcus and the innate immunity of the host

Innate immunity covers a diverse array of host defenses including muco-


cilliary clearance, complement, neutrophils and macrophages. It acts as a
non-specific defense able to recognize and respond rapidly against a broad
range of microbes. Unlike adaptive immunity, which involves the clonal
expansion of T- and B-cells specific to the pathogen, innate recognition is
achieved through a limited set of germline encoded receptors and does not
possess immunological memory. The two systems do not however operate
in isolation as the innate immune system plays crucial roles in the initiation,
development and effector stages of adaptive immunity.

Complement and the pneumococcus

The complement system comprises over 30 serum and membrane pro-


teins which, when activated, form a cascade of reactions contributing to
the elimination of invading microorganisms. The binding and activation
of complement components to the surface of a microbe leads to opso-
nophagocytosis and the induction of inflammation. For some organisms,
but not the pneumococcus, complement can destroy the microbe directly
through lysis by the membrane attack complex. Complement contributes
to and links both innate and adaptive immunity. Three pathways of activa-
tion exist (reviewed in [113], in brief these are: 1) The classical pathway
154 Sven Hammerschmidt et al.

activated by antibody: antigen complexes for example, antibody binding to


the microbial surface. Non-antibody-dependent activation can also occur,
such as the binding of acute phase proteins to the microorganism, 2) the
lectin pathway, which is triggered by mannose-binding-lectin recognition
of carbohydrate on the microbial surface, and 3) the alternative pathway
is continuously activated at low levels, but only amplifies on foreign sur-
faces.
The crucial contribution of complement to innate and adaptive
responses to pneumococcal infection has been long established both in
animal models and humans with genetic complement deficiencies (for
recent overview see [114]). However, only recently, through the use of
a panel of gene knock-out mice lacking various complement compo-
nents, has the relative importance of individual activation pathways been
assessed in innate immunity to pneumococcal infection [115]. The classical
pathway of complement activation was found to be the dominant comple-
ment pathway for innate immunity to the pneumococcus in mice [115]; the
specific loss of which resulted in significantly increased disease severity.
Natural IgM antibodies, possibly to C polysaccharide (teichoic acid), con-
tributed to this activation of the classical pathway as shown with the use of
+s–/– knock-out mice which lack such antibodies [115]. However, the activa-
tion of the classical pathway during this innate immune response was only
partially dependent on natural antibodies and other activation pathways
also contributed. These were proposed to include acute phase proteins
such as C-reactive protein or direct binding of complement component
C1q to the pneumococcal surface [115]. The alternative pathway also con-
tributed to protective innate responses, but to a lesser degree than seen
for the classical pathway, while the role for the lectin pathway appeared
negligible. In line with this latter finding, genetic mannose binding lec-
tin deficiency is associated with only a small (but significant) increased
susceptibility to pneumococcal disease in humans [116]. Regardless of
the activation pathway, the deposition and activation of complement
component C3 on the bacterial surface is a key step in the complement
cascade leading to elimination of the microbe. In accordance with this
crucial role of complement in innate immunity, the pneumococcus has
evolved several mechanisms to resist its affects. The capsule is a key factor
in this resistance, acting not only to limit access to cell bound comple-
ment, but also reducing the amount of complement deposited [117]. The
pneumococcal surface protein, PhpA (also called PhtB and BVH-11 [118],
has been found to possess C3 degrading activity [119, 120] and so PhpA
may contribute to preventing complement-mediated clearance. In addi-
tion to its cytolytic activity the toxin pneumolysin, a major pneumococcal
virulence factor, has multiple other biological activities (for review see
[121]). Recently pneumolysin has been shown to confer protection from
complement mediated clearance [122]. Deletion of pneumolysin caused
an increase in C3 deposition on pneumococcal cells in vitro. Showing that
Pathogenesis of Streptococcus pneumoniae infections… 155

this activity of pneumolysin contributed to virulence, absence of comple-


ment in gene knock-out mice reduced the importance of pneumolysin to
pneumococcal virulence. Furthermore, this effect of pneumolysin in vitro
and in vivo was specific to the classical and not the alternative activation
pathway [122]. The mechanisms by which pneumolysin achieves this affect
are as yet unconfirmed. However, this specific evasion of the classical
pathway is in agreement with the ability of pneumolysin to activate this
pathway in the complement; evasion in this instance may ironically be the
result of complement activation rather than inhibition. Released pneumo-
lysin may result in complement activation away from the bacterial cell thus
protecting it and also consuming the available complement components.
In addition, increased complement activation may contribute to host tis-
sue damage thereby promoting bacterial pathogenesis. Furthermore, the
surface proteins, PspA and PspC also contribute to complement resistance.
The PspC molecules are divided into different groups. Interestingly, both
the classical PspC proteins containing a CBD and the PspC-like Hic (PspC
11.4) bind the complement factor H [123]. Hic is produced predominantly
by serotype 3 pneumococcal strains that are negative for SC binding. Hic
contains an LPxTG motif that anchors the protein in a sortase-dependent
manner to the peptidoglycan of the cell wall. Factor H is a fluid phase
regulator of the alternative complement pathway and consists of 20 short
consensus repeats (SCRs). Hic interacts with the SCRs 8-11 and 12-14 of
factor H [124], whereas a role for the SCRs 6 to 10 and 13-15 of factor
H was suggested for the interaction with PspC [67, 125]. Recruitment of
factor H by Hic has been shown to efficiently prevent activation of C3b
and complement mediated opsonophagocytosis of pneumococci [124]. The
improved survival of pneumococci expressing PspC or Hic in a systemic
mouse infection model provides further evidence for the versatility and
importance of PspC in different host niches [126]. PspA contributes to
inhibition of complement receptor-mediated clearance of pneumococci.
Tu et al. [127] have demonstrated a delay of wild type pneumococci clear-
ance compared to the isogenic pspA mutant. It is thought that PspA prob-
ably inhibits the factor B-mediated complement activation and functions
as an inhibitor of C3b deposition [127]. The inhibitory effect by PspA has
been shown for PspA from family 1 and family 2 [128]. In addition, PspA
expression decreases C3 binding on pneumcococi, indicating that PspA
may also inhibit complement deposition via the classical pathway [96].

Pattern recognition receptors

Key components of the innate immune system are so-called pathogen


recognition receptors (PRRs). These can be located on the hosT-cell sur-
face, intracellularly, or be secreted and act to initiate the recognition and
response to microbes and in some cases host products [129]. The microbial
156 Sven Hammerschmidt et al.

components recognized by PRRs are referred to as pathogen-associated


molecular patterns (PAMPs) so-called because they are typically invariant
structures found among many microbes, but absent in eukaryotes. Examples
include lipopolysacchride (LPS) and peptidoglycan from bacteria, double-
stranded RNA from certain viruses and mannan from fungi. C-reactive
protein (CRP) is a well-known example of a PRR involved in the response
to this pneumococcus. This soluble protein binds PCho in the pneumococ-
cal cell wall, inducing complement activation leading to bacterial clearance
[130, 131]. Additional PRRs, important in pneumococcal infection have
recently been described including members of the toll-like receptor (TLR)
family, LBP, and the cytosolic PRR Nod proteins.

Consequences of pneumococcal recognition by Toll-like receptors

The TLR family of PRRs has received much attention due to their impor-
tance in the response to a wide range of microbes (for review see [132,
133]). Their major function is as PRRs to recognize microbes and initiate
an inflammatory response leading to eradication of infection. At least ten
TLRs have been described in humans and mice and several have been
implicated in pneumococcal infection in animal models. Furthermore,
descriptions of genetic defects in TLR signaling associated with increased
susceptibility to pneumococcal disease show these receptors have relevance
to human infection [134, 135].
TLR2 recognizes both pneumococcal lipoteichoic acid (LTA) and cell
wall peptidoglycan [136-139]. Interestingly, despite numerous studies sup-
porting the recognition of bacterial peptidoglycan via TLR2, this interac-
tion has recently been challenged [140]. Knock-out TLR2–/– mice display
increased disease severity and decreased survival times compared with wild
type mice in a pneumococcal meningitis model [141]. This greater suscepti-
bility correlated with heightened bacterial levels in the brain, but appeared
independent of systemic disease as both strains showed similar bacterial
levels in the blood. In agreement with these data, Koedel et al. [142] also
found TLR2–/– mice had enhanced disease and increased bacterial levels in
the brain in their meningitis model, which in turn have contributed to an
enhanced inflammation in the brain later in infection.
The role of TLR2 has also been investigated in experimental pneumo-
coccal pneumonia [143]. Comparison between wild type and TLR2–/– mice
following intranasal infection revealed only a modest contribution for this
receptor in the host response and no changes in bacterial clearance and
morbidity with compared with their wild type counterparts. On the basis
of these data, TLR2 does not appear to play a key role in host resistance
to pneumococcal pneumonia. Interestingly, the stimulation of isolated
alveolar macrophages in vitro to produce TNF-_ in response to heat-killed
pneumococci was entirely dependent on TLR2. However, immunohisto-
Pathogenesis of Streptococcus pneumoniae infections… 157

chemical staining of infected lungs from TLR2–/– mice showed these cells
were producing TNF-_ comparable with wild type mice. Presumably in the
setting of the intact animal other host innate immune factors such as other
PRRs and complement mask the loss of TLR2, rendering its influence
minimal in pneumococcal pneumonia. Following intraperitoneal infection
TLR2 knock-out mice have slightly reduced survival times compared with
wild type [144]. Thus, TLR2 has a protective role in this model of systemic
infection, although, as with pneumonia, the defect was arguably minor.
Furthermore, in a model of nasopharyngeal colonization TLR2 knock-out
mice had impaired clearance of pneumococci [145] showing that TLR2 is of
relevance not only to disease states, but also carriage.
The recognition of pneumococcal peptidoglycan probably involves
interaction between TLR2 and 6 as shown for Staphylococcus aureus pepti-
doglycan [146]. Indeed, confirming a role for this receptor in pneumococcal
recognition, expression of a double negative TLR6 mutant inhibited TNF-_
production in response to stimulation by the pneumococcus in a macrophage
cell line [146]. A role exists for TLR1 in the recognition of pneumococcal
LTA, whereby monoclonal antibodies against this receptor inhibited LTA
induced TNF-_ production from human peripheral blood mononuclear
cells [136]. The importance of these interactions between the pneumococcus
and TLR1 and 6 has not yet been assessed in an infection model.

Recognition of pneumolysin by TLR4

Through the recognition of LPS, TLR4 is a key component of the innate


response to Gram-negative infections. A role for this receptor has also been
extended to the pneumococcus with the finding that the in vitro pro-inflam-
matory effect of pneumolysin on macrophages was TLR4-dependent [147].
Subsequently, pneumolysin has been shown to directly interact with TLR4
[148]. This inflammatory activity was not dependent on the pore-forming or
complement-activating activities of pneumolysin because the PdT pneumo-
lysin mutant, that lacks these properties, was also active in these studies. The
significance of this interaction during colonization was studied by compar-
ing wild type and TLR4 deficient mice in a nasopharyngeal carriage model
[149]. In the absence of functional TLR4, mice were more heavily colonized
and much more likely to develop invasive disease. Thus, through its recogni-
tion of pneumolysin, TLR4 acts in the nasopharynx to limit pneumococcal
proliferation. While the inflammatory response to pneumolysin may con-
tribute to this protection, it has also been shown that pneumolysin-TLR4
signaling can induce hosT-cell apoptosis in vitro and in vivo [148]. This
also appears to be a protective host response as the inhibition of apoptosis
rendered mice more susceptible to death following pneumococcal infection
[148]. Interestingly, a similar model of colonization using the same mouse
strains found no difference in the clearance of pneumococci between wild
158 Sven Hammerschmidt et al.

Fig. 4: Key pathogen recognition receptors involved in the recognition and initiation of the
immune response towards the pneumococcus. Abbreviations: LBP, LPS-binding protein; LTA,
lipoteichoic acid; TLR, toll-like receptor. Reproduced with permission from reference [210].

type and TLR4 deficient mice [145]. The use of different bacterial strains
in these studies may explain this apparent conflict, but this remains to be
tested. In pneumococcal pneumonia TLR4 also plays a protective role [150].
In this experimental model the absence of functional TLR4 rendered mice
more susceptible to morbidity with increased bacterial counts in the lungs.
The effects however, were modest with the effect on death rate only appar-
ent at low doses and with no significant impact on pulmonary inflammation.
Furthermore, the significance of TLR4 in pneumococcal infections appears
restricted to the airway surfaces as earlier work found the absence of TLR4
made no difference to survival rates and blood bacterial counts after intra-
venous infection of mice [151].

TLR signaling

Myeloid differentiation factor 88 (MyD88) is a key adaptor molecule in the


signaling cascade activated by engagement of TLRs or IL-1 family recep-
Pathogenesis of Streptococcus pneumoniae infections… 159

tors [152]. In agreement with a role for TLRs in innate protection against
pneumococcal infection, MyD88–/– mice show enhanced susceptibility to
S. pneumoniae in different infection models [144, 153, 154]. Providing rel-
evance to human infection, deficiency in IL-1 receptor-associated kinase
4 (IRAK4), also a mediator in the TLR/IL-1 receptor signaling pathway,
results in increased susceptibility to pneumococcal disease [135]. As does a
distinct but as yet undefined mutation in this signaling pathway [134].

Pneumococcal cell wall recognition by LBP

In addition to TLR2, the pneumococcal cell wall (PCW) is recognized by


the soluble acute phase protein, LBP (LPS binding protein) [138]. This pro-
tein has previously been found to bind LPS and enhance its inflammatory
activity. It has now also been found to bind purified PCW and whole pneu-
mococci and the addition of LBP accentuated the inflammatory activity of
PCW in vitro [138]. In a mouse meningitis model LBP gene-knock out mice
showed significantly reduced meningeal inflammation following challenge
with purified cell wall or live pneumococci. Recognition of PCW by LBP
appears independent of cell wall phosphorylcholine and teichoic acid, with
the glycan backbone seemingly a crucial structure for this interaction [138].
Significance to human infections was shown by enhanced LBP levels in the
CSF of pneumococcal patients compared with controls. Furthermore, PCW
coprecipated with LBP in the CSF from a patient showing this interac-
tion occurs in human infection [138]. LBP also contributes to the response
to pneumococcal LTA [137]. However, LBP gene knock-out mice do not
have altered susceptibility or responses to pneumococcal pneumonia [155].
Given that LBP acts to facilitate recognition of TLR2 ligands, this pheno-
type is compatible with the modest effect seen with TLR2 knock-out mice
in similar infections [143]. Interestingly, LBP levels in lavage fluid increased
seven-fold during experimental pneumococcal infection [155]. This was a
much greater increase than seen with parallel Klebsiella pneumoniae pneu-
monia [155]. However, unlike pneumococcal pneumonia, endogenous LBP
contributed significantly to protection against K. pneumoniae pneumonia
[155]. Clearly, this highlights that functional importance cannot easily be
predicted merely from expression levels alone.

CD14

Membrane bound and soluble CD14 act as a co-receptor to enhance the


response to LPS [117, 156, 157]. CD14 also contributes to the recognition
of the pneumococcus in vitro [136, 139, 158]. A role in infection has also
been confirmed by the recent finding that CD14 gene knock-out mice show
exacerbated pneumococcal meningitis [159].
160 Sven Hammerschmidt et al.

Nod proteins and the pneumococcus

The cytosolic proteins, Nod1 and Nod2 are additional PRRs acting within
host cells to recognize and respond to microbial products [160]. In addition
to toll-like receptors, a role for Nod proteins in the recognition and response
to pneumococci has also been shown [161]. Transfection with Nod2, but not
Nod1, conferred responsiveness to cells following pneumococcal exposure
as judged by NF-gB activation [161]. In line with the role of Nod proteins in
the response to intracellular material, the recognition of pneumococcus by
Nod2 was dependent on internalization of the bacteria. Nod2 has previously
been shown to be responsive to a muramyldipeptide conserved in multiple
peptidoglycans and this was the suggested mechanism for its recognition
of the pneumococcus [161]. The availability of Nod1 and 2 knock-out mice
will allow a fuller appreciation of these genes in the host response to the
pneumococcus [162–164].

Recognition of capsule

Recognition of the pneumococcal polysaccharide capsule by PRRs has


received much attention of late. Kang et al. [165] showed that the C-type
lectin SIGN-R1 expressed by macrophages, particularly in the marginal
zone of the mouse spleen, bound capsular polysaccharide from several dif-
ferent serotypes as well as whole pneumococcal cells. Lanoue et al. [166]
demonstrated a functional significance of this interaction with the genera-
tion of SIGN-R1 gene knock-out mice. Following intraperitoneal infection
with either serotype 2 or serotype 14 pneumococci, these mice displayed
increased susceptibility to infection compared with their wild type coun-
terparts. Absence of SIGN-R1 caused a decrease in survival rate, shorter
time to death, increased sickness scores and increased bacterial levels in the
blood. A defect in the ability of macrophages in the peritoneum and spleen
to bind and phagocytose the pneumococcus was likely to be a major con-
tributor to the increased susceptibility of the knock-out mice [166].
The role of SIGN-R1 in pneumococcal pneumonia has also been
investigated with the use of SIGN-R1 deficient mice [167]. When infected
intranasally with a serotype 3 strain, the SIGN-R1 knock-out mice showed
increased bacterial levels in the lungs compared with wild type. This was
accompanied by a higher incidence of bacteremia and increased bacterial
counts in the blood and spleen. Interestingly, alveolar macrophages do not
express SIGN-R1 and expression was not induced following pneumococcal
infection [167]. This suggests the protective role of SIGN-R1 in pneumonia
does not occur within the lungs themselves. One potential mechanism for
the increased bacterial growth in the absence of SIGN-R1 was found to be
reduced levels of anti-phosphorylcholine IgM. In addition, systemic disease
in this model was probably exacerbated by defective phagocytosis by mac-
Pathogenesis of Streptococcus pneumoniae infections… 161

rophages in the peritoneum and spleen as described by [166]. Therefore, in


both pulmonary and systemic infections, SIGN-R1 is instrumental in host
resistance to pneumococcus.
Another macrophage receptor, MARCO has also recently been iden-
tified as important in pneumococcal infection [168]. Genetic deletion of
this scavenger receptor rendered mice more susceptible to pneumococcal
pneumonia with impaired bacterial clearance from the lungs and increased
morbidity [168]. Isolated alveolar macrophages from the knock-out mice
were impaired in their ability to bind and phagocyctose the pneumococcus
in vitro and this was likely a key factor in the increased susceptibility to
infection. Interestingly, reduced phagocytosis was not due solely to reduced
bacterial binding and so a role for MARCO appears to exist not only in
binding, but in subsequent bacterial uptake. The pneumococcal ligands rec-
ognized by MARCO have not yet been identified. Previously pneumococcal
capsular polysaccharide has been shown to activate macrophages, [169] an
activity partially dependent on CD14, whether or not it also involves SIGN-
R1 and MARCO remains to be determined.

Surfactant proteins

Pulmonary surfactant is a mixture of lipids and proteins that act to prevent


alveoli from collapsing during expiration. In addition, the surfactant proteins
(SP) SP-A and SP-D play a role in innate immunity against a variety of
pathogens acting by binding microbes and promoting their phagocyctosis or
by modulating immune cell function [170, 171]. In the case of pneumococcal
infection, SP-D knock-out mice show enhanced susceptibility to intranasal
infection [172]. While SP-A has recently been shown to promote phagocycto-
sis of S. pneumoniae by rat and mouse alveolar macrophages in vitro [173].

Innate immunity and interaction between the pneumococcus and


other microbes

For ease of study, most work on the interaction of the pneumococcus with
the innate immune system has employed pure cultures. However, the muco-
sa of the upper respiratory tract is colonized by a diverse array of microbial
species. Indeed, analysis of DNA from human airway surface fluid suggested
the presence of > 500 bacterial species [174]. Concurrent stimulation of the
innate immune system by multiple species appears to have distinct effects
from single species interactions [175, 176] and this has recently been shown
to have relevance to the pneumococcus [177]. Co-stimulation of human
respiratory epithelia cells in vitro by S. pneumoniae and Haemophilus influ-
enzae, also an inhabitant of the upper respiratory tract, resulted in synergis-
tic production of IL-8 [177]. This extended to a mouse colonization model
162 Sven Hammerschmidt et al.

with a synergistic effect on MIP-2 production and inflammatory influx into


the upper airways. This synergy was independent of TLR2 and TLR4 but
involved NF-gB translocation to the nucleus and phosphorylation of p38
MAPK. With regards to the microbial products involved, pneumolysin
could substitute for the pneumococcus, but the PdB pneumolysin toxoid,
lacking cytolytic activity, was inactive. It was therefore speculated that the
pore forming activity of pneumolysin lead to enhanced delivery of microbial
products such as the soluble inflammatory protein, SCF from H. influenzae,
into the host cell where recognition by Nod1 and 2 would result in increased
stimulation [177]. This pro-inflammatory activity of pneumolysin is there-
fore distinct from its effects on macrophages that were mediated through
TLR4 and were independent of pore-forming activity.
The significance of pneumococcal H. influenzae interactions have
recently be examined in a co-colonization mouse model [178]. In contrast
to what might be expected based on in vitro studies with these bacteria [46,
179], co-coloniation in vivo resulted in rapid clearance of the pneumococ-
cus. This effect was dependent on the innate immune system in the form of
neutrophils and complement, with the depletion of either abolishing the
competitive effect. Activation of peritoneal neutrophils with heat-killed H.
influenzae caused an increase in their ability to kill the pneumococcus, but
had no effect on their ability to kill H. influenzae [178]. The basis of this
activity is not yet clear. Thus, interactions with the innate immune system
can have a significant effect during competition between the pneumococcus
and other microbes in the nasopharynx.
Another important microbial interaction is that of the pneumococcus
and influenza A. Subsequent to influenza A outbreaks secondary pneu-
mococcal infection is an important cause of morbidity and mortality. This
heightened susceptibility to pneumococcal disease can be reproduced in
animal models, allowing investigation of the mechanisms involved. While
viral neuraminidase contributes to this phenomenon by exposing pneumo-
coccal receptors [20, 180], alterations in the immune response also seem
to contribute. Prior influenza A infection in mice primes for an exagger-
ated inflammatory response to subsequent pneumococcal infection [181].
Increased levels of IL-10 in this response likely contribute to increased
susceptibility as neutralization of this cytokine improved disease outcome
[181]. In vitro exposure to both influenza A and pneumococcus results in a
synergistic inflammatory response from human middle ear epithelial cells
[176]. Microarray gene expression analysis of these cells following influenza
A infection provides insight into the possible mechanisms behind this syn-
ergy [182]. For example, it was found that tlr2 expression was up-regulated
by influenza A infection. This may make the cell more responsive to stimu-
lation by pneumococcal peptidoglycan and LTA [182].
It is therefore clear that the interaction of the pneumococcus with the
innate immune system is greatly influenced by the presence of other organ-
isms such as H. influenzae and influenza A.
Pathogenesis of Streptococcus pneumoniae infections… 163

CD4 +cells in immunity to the pneumococcus

The function of CD4 + T-cells in adaptive immunity is well established.


Interestingly, they appear also to contribute to early resistance to pneu-
mococcal infection independently of their role in adaptive antigen-specific
responses [183]. Intranasal infection of MHCII knock-out mice, which dis-
play a significant decrease in CD4 + T-cell levels, revealed a key role for
these cells in the early response to pneumococcal pneumonia. These mice
displayed increased susceptibility to infection as evidenced by increased
bacterial counts in the lung and blood compared with their wild type coun-
terparts. Indeed, the increased susceptibility was so great that it resulted in
100% mortality in the knock-out mice by three days post-infection, whereas
all wild type mice survived the challenge. In accordance with previous data
showing T-cell migration to infected lung tissue in pneumococcal pneumonia
[184, 185], purified CD4 + T-cells migrated to the pneumococcus in vitro. This
migration was associated with T-cell activation and interestingly occurred
only in response to in vivo and not to in vitro grown bacteria. Pneumolysin
plays a significant part in this migration as pneumolysin-deficient pneumo-
cocci stimulated significantly less cell migration. How pneumolysin stimu-
lates these T-cells is unclear but the recent description of TLR4 expression
by T-cells may be of relevance [186]. Recently Malley et al. [147] have dem-
onstrated a crucial role for CD4 + T-cells in antibody independent acquired
immunity to pneumococcal colonisation. How CD4 + T-cell migration and
activation in response to the pneumococcus as described in pneumonia
relates to this acquired immunity to colonisation is as yet unclear.

The pneumococcus and the adaptive immunity of the host

During carriage and invasive disease the pneumococcus encounters the pro-
tective effect of host immunoglobulins. The pneumococcal surface is covered
by a capsular polysaccharide which is involved in pneumococcal coloniza-
tion and, more importantly, is a key virulence factor in invasive diseases [23,
187]. A correlation between decrease in pneumococcal carriage with rising
levels of both mucosal and serum antibodies to pneumococcal surface poly-
saccharides has been described [145]. However, capsular polysaccharides
do not yield an anamnestic response, due to the inability of polysaccharides
to recruit cognate CD4 + T-cell help through T-cell receptor recognition of
peptide-major histocompatibility class II complexes (MHCII) on the sur-
face of antigen-presenting cells [188]. McLay and coworkers demonstrated
in mouse infection studies that the lack of memory response by capsular
polysaccharides can be overcome by the use of conjugated vaccines that
elicit a different IgG subclass response to polysaccharides. Co-expression of
surface polysaccharides with proteins has been assumed to mediate cognate
CD4 + T-cell help for polysaccharide-specific B-cells [188]. The development
164 Sven Hammerschmidt et al.

of a seven-valent conjugate vaccine has resulted in high immunogenicity,


but immune protection was restricted only to the seven serotypes, which
were represented by the polysaccharides [189].
Efforts have been made to overcome the problem of variable polysac-
charide determinants and to develop potent protein vaccines that have
the capability to cover most pneumococcal serotypes. A number of pneu-
mococcal cell-surface or secreted components have been shown to induce
opsonophagocytic antibodies or to offer at least partial protection in murine
infection models. The protein antigens currently under investigation include
the phosphorylcholine epitope found on lipoteichoic acid (LTA), PspC,
pneumolysin, PpmA, PsaA, PspA and some surface proteins identified by
whole-genome-approaches [97, 123, 190-198].
Some of the identified protein vaccine candidates have been investigated
in an experimental model of human carriage within the nasopharynx to cir-
cumvent the limitations given by murine models of pneumococcal infections
[196]. In contrast to the low level of antibody response to pneumococcal
polysaccharides, serum IgG and secretory IgA-response was detected to an
N-terminal region of PspA and also to PspC of the inoculum strain during
experimental carriage of type 23F and 6B pneumococci in adults. The spe-
cific antibody titers even exceeded basal levels of humans with pre-existing
antibody response [196, 199]. Former studies described serum IgG titers to
PsaA and pneumolysin emerging with age and exposure to pneumococci. In
contrast, no antibody response to PsaA and pneumolysin was detected in
the carriage studies with adults [196, 200, 201].
A stated problem of the human carriage model concerns the high vari-
ability of pneumococcal surface proteins including PspC that has been
described as strain-to-strain diversity and may account for the low IgG titers
against different serotypes. The functional organization of PspC proteins
among different strains is similar, but the molecular weight of PspC has
been shown to vary between 59 kDa to 105 kDa. PspC is divided into 11
groups due to differences in the N-terminal domain [68, 202]. Further stud-
ies are required to evaluate the impact of the described interaction of PspC
with the secretory IgA and with free secretory component for the immune
status of the host. A further example of the high strain-depending variability
in protein structure and in immunogenicity is PspA, which is divided into
two different protein families and subdivided into six clades [203].
To further explore the mechanism contributing to natural carriage and
clearance of carriage, a mouse model of pneumococcal colonization was
developed. In this model pneumococcal carriage induced a mucosal and
serum antibody response to pneumococci and to PspA. But no correlation
was detected between the density of colonization and amounts of detected
mucosal or serum antibodies [145].
The relationship between systemic and local antibody production and
carriage in children was studied in in vitro studies after antigen stimulation
by Zhang and co-workers [204]. Serum, saliva and cell culture superna-
Pathogenesis of Streptococcus pneumoniae infections… 165

tants of adenoidal mononuclear cells of children undergoing adenectomy


were assayed for antibodies to the pneumococcal proteins: PspC, pneu-
molysin, PsaA, PspA. In this study carriage rates fell with age and serum
levels of anti-PspC, Ply and PspA were rising. The results revealed that
antibody production to PspC and pneumolysin have the potential to pro-
tect children aged 2 years and older against pneumococcal colonization.
Other highly conserved vaccine candidates like LTA, PsaA, and PpmA
were tested, but no antibody response was detected [196]. Further studies
are required to correlate the described data of immunogenicity of vac-
cine candidates during human carriage with protective potential against
colonization.
The development of antibodies against pneumococcal surface associated
proteins including enolase, IgA1.protease, SlrA and PpmA has recently
been investigated in relation to pneumococcal carriage and otitis media in
children [205]. These studies have indicated that enolase, IgA1-protease,
SlrA and PpmA are immunogenic proteins, but no significant correlation
between antibody titers and pneumococcal carriage or infection has been
found. In contrast to the high structural variability of PspC and PspA, eno-
lase and IgA1- are highly conserved proteins that share strong homology
to other bacterial species colonizing the same nasopharyngeal niche like
N. meningitidis and H. influenzae. The presence of cross-reactive epitopes
enhances the basal titer of antibodies and might explain the little impact of
current pneumococcal colonization on serum concentrations of anti-IgA1-
protease and anti-enolase antibodies [205].
Future challenges of vaccine development might be the identification
of a surface exposed pneumococcal protein with serotype-unspecific con-
served immunogenic domains that show no cross reaction with other bacte-
rial or eukaryotic components.

Global analysis of host responses

The advent of microarray technology allows greater insight into the host-
cell response to the pneumococcus. The response of the human monocytic
cell line THP-1 has been assessed by microarray analysis following expo-
sure to S. pneumoniae and an isogenic mutant lacking Pneumolysin [206].
After 3-hour exposure to the pneumococcus, expression differences were
revealed in 182 host genes from the 4133 examined, illustrating the potential
for large-scale expression changes induced by the pneumococcus [206]. Of
these 182 genes, 142 were responsive to pneumolysin showing the dominant
nature of this virulence factor in host responses. While this study will not
be comprehensive in fully documenting the host response it illustrates the
complexity of the interaction between host-cells and the pneumococcus. An
important future challenge will be to understand the significance of these
expression changes in the disease process.
166 Sven Hammerschmidt et al.

Host gene expression changes in an infection model has been investi-


gated in a rat model of otitis media [207]. Twelve hours following pneumo-
coccal challenge, 280 genes in the middle ear (effusion and mucosa) showed
a greater than two-fold change in expression compared with mock infected
controls. This represented ~24% of the genes examined, again showing the
ability of the pneumococcus to induce large-scale changes in host gene
expression. Such data allows the pneumococcal response pathways to be
mapped and for the identification of previously unrecognised responses.
For example, it was found that the transcription factor fra-1, implicated in
bone proliferation was upregulated during experimental otitis media. This
provides a candidate mechanism to explain clinical features of otitis media
involving the bone seen in both human patients and animals [207]. The glob-
al response to nasopharyngeal colonization has also been investigated in a
mouse model [208]. Up-regulation of siderocalin, an iron sequestering host
defense protein was noted in the nasal mucosa. How the pneumococcus
causes this up-regulation of siderocalin is unclear with the effect still seen
in mice deficient for either TLR2 or 4. Interestingly, this response could not
be replicated in vitro, again showing the complexity of the immune response
and the value of whole animal systems [208]. S. pneumoniae is resistant to
siderocalin and its upregulation may be advantageous to the pneumococcus
by inhibiting potential competitors in the nasopharynx [208].

Conclusion

Pneumococcus is a versatile microorganism causing local infections and


severe invasive diseases as well. The dramatic clinical outcome of pneu-
mococcal pneumonia, bacteriaemia and meningitis, respectively, is the
result of massive inflammatory host responses. The infections caused by
this pathogen can be controlled by the innate and antibody-mediated host
defense mechanisms. However, the pneumococcus has developed several
sophisticated mechanisms to overcome the defense mechanism and is able
to subvert host protein functions for its survival and dissemination. Another
threat is the increasing rate of antibiotic resistant isolates that requires the
development and use of cost effective pneumococcal vaccines or new thera-
peutics. A further understanding of the pneumococci-host interaction may
aid to develop a better vaccine against pneumococcal infections.

Acknowledgements

The work in the group of Sven Hammerschmidt is supported by grants of


the German Research Foundation (DFG-SFB 479 to S.H.) and Federal
Ministry of Education and Research (grant 01KI0430 to S.H., Competence
Network CAPNETZ). The work in the group of Tim J. Mitchell is supported
Pathogenesis of Streptococcus pneumoniae infections… 167

by the Wellcome Trust, MRC, BBRSC, the Egyptian government and the
European Union. Our apologies to authors of primary articles we have
failed to discuss in detail or to cite due to limitations on space. The authors
are grateful to Roland Nau (University of Göttingen, Germany) for provid-
ing histopathological micrographs and Manfred Rohde (German Research
Centre for Biotechnology, Braunschweig, Germany) for providing electron
micrographs.

References
1 Cartwright K (2002) Pneumococcal disease in western Europe: burden of dis-
ease, antibiotic resistance and management. Eur J Pediatr 161: 188–195
2 Brown PD, Lerner SA (1998) Empirical therapy for community-acquired pneu-
monia. Ann Intern Med 129: 510
3 Obaro S, Adegbola R (2002) The pneumococcus: carriage, disease and conju-
gate vaccines. J Med Microbiol 51: 98–104
4 Koedel U, Scheld WM, Pfister HW (2002) Pathogenesis and pathophysiology of
pneumococcal meningitis. Lancet Infect Dis 2: 721–736
5 Hava DL, Camilli A (2002) Large-scale identification of serotype 4 Streptococcus
pneumoniae virulence factors. Mol Microbiol 45: 1389–1406
6 Ogunniyi AD, Giammarinaro P, Paton JC (2002) The genes encoding virulence-
associated proteins and the capsule of Streptococcus pneumoniae are upregu-
lated and differentially expressed in vivo. Microbiology 148: 2045–2053
7 Orihuela CJ, Radin JN, Sublett JE, Gao G, Kaushal D, Tuomanen EI (2004)
Microarray analysis of pneumococcal gene expression during invasive disease.
Infect Immun 72: 5582–5596
8 Polissi A, Pontiggia A, Feger G, Altieri M, Mottl H, Ferrari L, Simon D (1998)
Large-scale identification of virulence genes from Streptococcus pneumoniae.
Infect Immun 66: 5620–5629
9 Marra A, Lawson S, Asundi JS, Brigham D, Hromockyj AE (2002) In vivo
characterization of the psa genes from Streptococcus pneumoniae in multiple
models of infection. Microbiology 148: 1483–1491
10 Henrichsen J (1995) Six newly recognized types of Streptococcus pneumoniae.
J Clin Microbiol 33: 2759–2762
11 Gillespie SH (1989) Aspects of pneumococcal infection including bacterial
virulence, host response and vaccination. J Med Microbiol 28: 237–248
12 Bogaert D, van Belkum A, Sluijter M, Luijendijk A, de Groot R, Rumke HC,
Verbrugh HA, Hermans PW (2004) Colonization by Streptococcus pneumoniae
and Staphylococcus aureus in healthy children. Lancet 363: 1871–1872
13 Musher DM (1992) Infections caused by Streptococcus pneumoniae: clinical
spectrum, pathogenesis, immunity, and treatment. Clin Infect Dis 14: 801–807
14 Mulholland K (1999) Strategies for the control of pneumococcal diseases.
Vaccine 17 Suppl 1: S79-S84
15 AlonsoDeVelasco E, Verheul AF, Verhoef J, Snippe H (1995) Streptococcus
pneumoniae: virulence factors, pathogenesis, and vaccines. Microbiol Rev 59:
591–603
168 Sven Hammerschmidt et al.

16 Lloyd-Evans N, O‘Dempsey TJ, Baldeh I, Secka O, Demba E, Todd JE,


McArdle TF, Banya WS, Greenwood BM (1996) Nasopharyngeal carriage of
pneumococci in Gambian children and in their families. Pediatr Infect Dis J 15:
866–871
17 Brueggemann AB, Griffiths DT, Meats E, Peto T, Crook DW, Spratt BG (2003)
Clonal relationships between invasive and carriage Streptococcus pneumoniae
and serotype- and clone-specific differences in invasive disease potential. J
Infect Dis 187: 1424–1432
18 Ghaffar F, Friedland IR, McCracken GH, Jr (1999) Dynamics of nasopha-
ryngeal colonization by Streptococcus pneumoniae. Pediatr Infect Dis J 18:
638–646
19 Weiser JN, Austrian R, Sreenivasan PK, Masure HR (1994) Phase variation in
pneumococcal opacity: relationship between colonial morphology and naso-
pharyngeal colonization. Infect Immun 62: 2582–2589
20 Tong HH, Weiser JN, James MA, DeMaria TF (2001) Effect of influenza A
virus infection on nasopharyngeal colonization and otitis media induced by
transparent or opaque phenotype variants of Streptococcus pneumoniae in the
chinchilla model. Infect Immun 69: 602–606
21 Weiser JN (2000) Phase variation of Streptococcus pneumoniae. In: Fischetti
VA (ed): Gram-positive pathogens. ASM Press, Washington DC, 225–231
22 Kim JO, Weiser JN (1998) Association of intrastrain phase variation in quantity
of capsular polysaccharide and teichoic acid with the virulence of Streptococcus
pneumoniae. J Infect Dis 177: 368–377
23 Magee AD, Yother J (2001) Requirement for capsule in colonization by
Streptococcus pneumoniae. Infect Immun 69: 3755–3761
24 Ring A, Weiser JN, Tuomanen EI (1998) Pneumococcal trafficking across the
blood-brain barrier. Molecular analysis of a novel bidirectional pathway. J Clin
Invest 102: 347–360
25 Hammerschmidt S, Wolff S, Hocke A, Rosseau S, Muller E, Rohde M (2005)
Illustration of pneumococcal polysaccharide capsule during adherence and
invasion of epithelial cells. Infect Immun 73: 4653–4667
26 Adamou JE, Wizemann TM, Barren P, Langermann S (1998) Adherence of
Streptococcus pneumoniae to human bronchial epithelial cells (BEAS–2B).
Infect Immun 66: 820–822
27 Kostrzynska M, Wadstrom T (1992) Binding of laminin, type IV collagen, and
vitronectin by Streptococcus pneumoniae. Zentralbl Bakteriol 277: 80–83
28 van der Flier M, Chhun N, Wizemann TM, Min J, McCarthy JB, Tuomanen EI
(1995) Adherence of Streptococcus pneumoniae to immobilized fibronectin.
Infect Immun 63: 4317–4322
29 Steinfort C, Wilson R, Mitchell T, Feldman C, Rutman A, Todd H, Sykes D,
Walker J, Saunders K, Andrew PW et al (1989) Effect of Streptococcus pneu-
moniae on human respiratory epithelium in vitro. Infect Immun 57: 2006–2013
30 Zysk G, Schneider-Wald BK, Hwang JH, Bejo L, Kim KS, Mitchell TJ,
Hakenbeck R, Heinz HP (2001) Pneumolysin is the main inducer of cytotoxic-
ity to brain microvascular endothelial cells caused by Streptococcus pneumoni-
ae. Infect Immun 69: 845–852
31 Hirst RA, Sikand KS, Rutman A, Mitchell TJ, Andrew PW, O‘Callaghan C
Pathogenesis of Streptococcus pneumoniae infections… 169

(2000) Relative roles of pneumolysin and hydrogen peroxide from Streptococcus


pneumoniae in inhibition of ependymal ciliary beat frequency. Infect Immun 68:
1557–1562
32 Tuomanen EI, Austrian R, Masure HR (1995) Pathogenesis of pneumococcal
infection. N Engl J Med 332: 1280–1284
33 Catterall JR (1999) Streptococcus pneumoniae. Thorax 54: 929–937
34 Lopez R,Garcia E (2004) Recent trends on the molecular biology of pneu-
mococcal capsules, lytic enzymes, and bacteriophage. FEMS Microbiol Rev 28:
553–580
35 Gosink KK, Mann ER, Guglielmo C, Tuomanen EI, Masure HR (2000) Role of
novel choline binding proteins in virulence of Streptococcus pneumoniae. Infect
Immun 68: 5690–5695
36 Sanchez-Beato AR, Lopez R, Garcia JL (1998) Molecular characterization of
PcpA: a novel choline-binding protein of Streptococcus pneumoniae. FEMS
Microbiol Lett 164: 207–214
37 Johnson MK, Geoffroy C, Alouf JE (1980) Binding of cholesterol by sulfhydryl-
activated cytolysins. Infect Immun 27: 97–101
38 Morgan PJ, Hyman SC, Byron O, Andrew PW, Mitchell TJ, Rowe AJ (1994)
Modeling the bacterial protein toxin, pneumolysin, in its monomeric and oligo-
meric form. J Biol Chem 269: 25315–25320
39 Morgan PJ, Hyman SC, Rowe AJ, Mitchell TJ, Andrew PW, Saibil HR (1995)
Subunit organisation and symmetry of pore-forming, oligomeric pneumolysin.
FEBS Lett 371: 77–80
40 Tilley SJ, Orlova EV, Gilbert RJ, Andrew PW, Saibil HR (2005) Structural basis
of pore formation by the bacterial toxin pneumolysin. Cell 121: 247–256
41 Preissner KT, Chhatwal GS (1999) Extracellular matrix (ECM) and host-cell
surfaces: Potential sites of pathogen interaction. Washington DC, ASM Press
42 Geelen S, Bhattacharyya C, Tuomanen E (1993) The cell wall mediates pneu-
mococcal attachment to and cytopathology in human endothelial cells. Infect
Immun 61: 1538–1543
43 Andersson B, Eriksson B, Falsen E, Fogh A, Hanson LA, Nylen O, Peterson H,
Svanborg EC (1981) Adhesion of Streptococcus pneumoniae to human pharyn-
geal epithelial cells in vitro: differences in adhesive capacity among strains iso-
lated from subjects with otitis media, septicemia, or meningitis or from healthy
carriers. Infect Immun 32: 311–317
44 Krivan HC, Roberts DD, Ginsburg V (1988) Many pulmonary pathogenic
bacteria bind specifically to the carbohydrate sequence GalNAc beta 1–4Gal
found in some glycolipids. Proc Natl Acad Sci USA 85: 6157–6161
45 Cundell DR, Tuomanen EI (1994) Receptor specificity of adherence of
Streptococcus pneumoniae to human type-II pneumocytes and vascular endo-
thelial cells in vitro. Microb Pathog 17: 361–374
46 Shakhnovich EA, King SJ, Weiser JN (2002) Neuraminidase expressed by
Streptococcus pneumoniae desialylates the lipopolysaccharide of Neisseria
meningitidis and Haemophilus influenzae: a paradigm for interbacterial com-
petition among pathogens of the human respiratory tract. Infect Immun 70:
7161–7164
47 King SJ, Hippe KR, Gould JM, Bae D, Peterson S, Cline RT, Fasching C, Janoff
170 Sven Hammerschmidt et al.

EN, Weiser JN (2004) Phase variable desialylation of host proteins that bind
to Streptococcus pneumoniae in vivo and protect the airway. Mol Microbiol 54:
159–171
48 King SJ, Whatmore AM, Dowson CG (2005) NanA, a neuraminidase from
Streptococcus pneumoniae, shows high levels of sequence diversity, at least in
part through recombination with Streptococcus oralis. J Bacteriol 187: 5376–
5386
49 Tong HH, Liu X, Chen Y, James M, Demaria T (2002) Effect of neuraminidase
on receptor-mediated adherence of Streptococcus pneumoniae to chinchilla
tracheal epithelium. Acta Otolaryngol 122: 413–419
50 Berry AM, Paton JC (2000) Additive attenuation of virulence of Streptococcus
pneumoniae by mutation of the genes encoding pneumolysin and other puta-
tive pneumococcal virulence proteins. Infect Immun 68: 133–140
51 Chapuy-Regaud S, Ogunniyi AD, Diallo N, Huet Y, Desnottes JF, Paton JC,
Escaich S, Trombe MC (2003) RegR, a global LacI/GalR family regulator,
modulates virulence and competence in Streptococcus pneumoniae 1319
Infect Immun 71: 2615–2625
52 Weiser JN, Bae D, Fasching C, Scamurra RW, Ratner AJ, Janoff EN (2003)
Antibody-enhanced pneumococcal adherence requires IgA1 protease. Proc
Natl Acad Sci USA 100: 4215–4220
53 Kausmally L, Johnsborg O, Lunde M, Knutsen E, Havarstein LS (2005)
Choline-binding protein D (CbpD) in Streptococcus pneumoniae is essential
for competence-induced cell lysis. J Bacteriol 187: 4338–4345
54 Guiral S, Mitchell TJ, Martin B, Claverys JP (2005) Competence-programmed
predation of noncompetent cells in the human pathogen Streptococcus pneu-
moniae: genetic requirements. Proc Natl Acad Sci USA 102: 8710–8715
55 Hermans PW, Adrian PV, Albert C, Estevao S, Hoogenboezem T, Luijendijk
IH, Kamphausen T, Hammerschmidt S (2006) The streptococcal lipoprotein
rotamase A (SlrA) is a functional peptidyl-prolyl isomerase involved in pneu-
mococcal colonisation. J Biol Chem 281: 968–976
56 Dintilhac A, Alloing G, Granadel C, Claverys JP (1997) Competence and
virulence of Streptococcus pneumoniae: Adc and PsaA mutants exhibit a
requirement for Zn and Mn resulting from inactivation of putative ABC metal
permeases. Mol Microbiol 25: 727–739
57 Tseng HJ, McEwan AG, Paton JC, Jennings MP (2002) Virulence of Streptococcus
pneumoniae: PsaA mutants are hypersensitive to oxidative stress. Infect Immun
70: 1635–1639
58 Romero-Steiner S, Pilishvili T, Sampson JS, Johnson SE, Stinson A, Carlone
GM, Ades EW (2003) Inhibition of pneumococcal adherence to human naso-
pharyngeal epithelial cells by anti-PsaA antibodies. Clin Diagn Lab Immunol
10: 246–251
59 Johnson SE, Dykes JK, Jue DL, Sampson JS, Carlone GM, Ades EW (2002)
Inhibition of pneumococcal carriage in mice by subcutaneous immunization
with peptides from the common surface protein pneumococcal surface adhesin
a. J Infect Dis 185: 489–496
60 Hammerschmidt S, Talay SR, Brandtzaeg P, Chhatwal GS (1997) SpsA, a novel
Pathogenesis of Streptococcus pneumoniae infections… 171

pneumococcal surface protein with specific binding to secretory immunoglobu-


lin A and secretory component 13. Mol Microbiol 25: 1113–1124
61 Hammerschmidt S, Tillig MP, Wolff S, Vaerman JP,Chhatwal GS (2000)
Species-specific binding of human secretory component to SpsA protein of
Streptococcus pneumoniae via a hexapeptide motif. Mol Microbiol 36: 726–736
62 Zhang JR, Mostov KE, Lamm ME, Nanno M, Shimida S, Ohwaki M, Tuomanen
E (2000) The polymeric immunoglobulin receptor translocates pneumococci
across human nasopharyngeal epithelial cells. Cell 102: 827–837
63 Elm C, Braathen R, Bergmann S, Frank R, Vaerman JP, Kaetzel CS, Chhatwal
GS, Johansen FE, Hammerschmidt S (2004) Ectodomains 3 and 4 of human
polymeric Immunoglobulin receptor (hpIgR) mediate invasion of Streptococcus
pneumoniae into the epithelium. J Biol Chem 279: 6296–6304
64 Lu L, Lamm ME, Li H, Corthesy B, Zhang JR (2003) The human polymeric
immunoglobulin receptor binds to Streptococcus pneumoniae via domains 3
and 4. J Biol Chem 278: 48178–48187
65 Rosenow C, Ryan P, Weiser JN, Johnson S, Fontan P, Ortqvist A, Masure HR
(1997) Contribution of novel choline-binding proteins to adherence, coloniza-
tion and immunogenicity of Streptococcus pneumoniae 32. Mol Microbiol 25:
819–829
66 Smith BL, Hostetter MK (2000) C3 as substrate for adhesion of Streptococcus
pneumoniae. J Infect Dis 182: 497–508
67 Dave S, Brooks-Walter A, Pangburn MK, McDaniel LS (2001) PspC, a pneu-
mococcal surface protein, binds human factor H. Infect Immun 69: 3435–3437
68 Iannelli F, Oggioni MR, Pozzi G (2002) Allelic variation in the highly polymor-
phic locus pspC of Streptococcus pneumoniae 41. Gene 284: 63–71
69 Luo R, Mann B, Lewis WS, Rowe A, Heath R, Stewart ML, Hamburger AE,
Sivakolundu S, Lacy ER, Bjorkman PJ et al (2005) Solution structure of choline
binding protein A, the major adhesin of Streptococcus pneumoniae. EMBO J
24: 34–43
70 Holmes AR, McNab R, Millsap KW, Rohde M, Hammerschmidt S, Mawdsley
JL, Jenkinson HF (2001) The pavA gene of Streptococcus pneumoniae encodes
a fibronectin-binding protein that is essential for virulence. Mol Microbiol 41:
1395–1408
71 Pracht D, Elm C, Gerber J, Bergmann S, Rohde M, Seiler M, Kim KS, Jenkinson
HF, Nau R, Hammerschmidt S (2005) PavA of Streptococcus pneumoniae
Modulates Adherence, Invasion, and Meningeal Inflammation. Infect Immun
73: 2680–2689
72 Bergmann S, Rohde M, Chhatwal GS, Hammerschmidt S (2001) alpha-Enolase
of Streptococcus pneumoniae is a plasmin(ogen)-binding protein displayed on
the bacterial cell surface. Mol Microbiol 40: 1273–1287
73 Bergmann S, Rohde M, Hammerschmidt S (2004) Glyceraldehyde–3-phos-
phate dehydrogenase of Streptococcus pneumoniae is a surface-displayed plas-
minogen-binding protein. Infect Immun 72: 2416–2419
74 Bergmann S, Rohde M, Preissner KT, Hammerschmidt S (2005) The nine
residue plasminogen-binding motif of the pneumococcal enolase is the major
cofactor of plasmin-mediated degradation of extracellular matrix, dissolution
of fibrin and transmigration. Thromb Haemost 94: 304–311
172 Sven Hammerschmidt et al.

75 Bergmann S, Wild D, Diekmann O, Frank R, Bracht D, Chhatwal GS,


Hammerschmidt S (2003) Identification of a novel plasmin(ogen)-binding
motif in surface displayed alpha-enolase of Streptococcus pneumoniae. Mol
Microbiol 49: 411–423
76 Ehinger S, Schubert WD, Bergmann S, Hammerschmidt S, Heinz DW (2004)
Plasmin(ogen)-binding alpha-enolase from Streptococcus pneumoniae: crys-
tal structure and evaluation of plasmin(ogen)-binding sites. J Mol Biol 343:
997–1005
77 Cundell DR, Gerard NP, Gerard C, Idanpaan-Heikkila I, Tuomanen EI (1995)
Streptococcus pneumoniae anchor to activated human cells by the receptor for
platelet-activating factor. Nature 377: 435–438
78 De Las Rivas B, Garcia JL, Lopez R, Garcia P (2001) Molecular characteriza-
tion of the pneumococcal teichoic acid phosphorylcholine esterase. Microb
Drug Resist 7: 213–222
79 Vollmer W, Tomasz A (2001) Identification of the teichoic acid phosphorylcho-
line esterase in Streptococcus pneumoniae. Mol Microbiol 39: 1610–1622
80 Garau G, Lemaire D, Vernet T, Dideberg O, Di Guilmi AM (2005) Crystal
structure of phosphorylcholine esterase domain of the virulence factor cho-
line-binding protein e from streptococcus pneumoniae: new structural features
among the metallo-beta-lactamase superfamily. J Biol Chem 280: 28591–28600
81 Hermoso JA, Lagartera L, Gonzalez A, Stelter M, Garcia P, Martinez-Ripoll M,
Garcia JL, Menendez M (2005) Insights into pneumococcal pathogenesis from
the crystal structure of the modular teichoic acid phosphorylcholine esterase
Pce. Nat Struct Mol Biol 12: 533–538
82 Amatruda TT, III, Gerard NP, Gerard C, Simon MI (1993) Specific interac-
tions of chemoattractant factor receptors with G-proteins. J Biol Chem 268:
10139–10144
83 Radin JN, Orihuela CJ, Murti G, Guglielmo C, Murray PJ, Tuomanen EI (2005)
beta-Arrestin 1 participates in platelet-activating factor receptor-mediated
endocytosis of Streptococcus pneumoniae. Infect Immun 73: 7827–7835
84 Wang E, Ouellet N, Simard M, Fillion I, Bergeron Y, Beauchamp D, Bergeron
MG (2001) Pulmonary and systemic host response to Streptococcus pneumoni-
ae and Klebsiella pneumoniae bacteremia in normal and immunosuppressed
mice. Infect Immun 69: 5294–5304
85 Bergeron Y, Ouellet N, Deslauriers AM, Simard M, Olivier M, Bergeron MG
(1998) Cytokine kinetics and other host factors in response to pneumococcal
pulmonary infection in mice. Infect Immun 66: 912–922
86 Heumann D, Barras C, Severin A, Glauser MP, Tomasz A (1994) Gram-positive
cell walls stimulate synthesis of tumor necrosis factor alpha and interleukin–6
by human monocytes. Infect Immun 62: 2715–2721
87 Spellerberg B, Rosenow C, Sha W, Tuomanen EI (1996) Pneumococcal cell wall
activates NF-kappa B in human monocytes: aspects distinct from endotoxin.
Microb Pathog 20: 309–317
88 Schmeck B, Zahlten J, Moog K, van Laak V, Huber S, Hocke AC, Opitz B,
Hoffmann E, Kracht M, Zerrahn J et al (2004) Streptococcus pneumoniae-
induced p38 MAPK-dependent phosphorylation of RelA at the interleukin–8
promotor. J Biol Chem 279: 53241–53247
Pathogenesis of Streptococcus pneumoniae infections… 173

89 Blue CE, Paterson GK, Kerr AR, Berge M, Claverys JP, Mitchell TJ (2003)
ZmpB, a novel virulence factor of Streptococcus pneumoniae that induces
tumor necrosis factor alpha production in the respiratory tract. Infect Immun
71: 4925–4935
90 Oggioni MR, Memmi G, Maggi T, Chiavolini D, Iannelli F, Pozzi G (2003)
Pneumococcal zinc metalloproteinase ZmpC cleaves human matrix metal-
loproteinase 9 and is a virulence factor in experimental pneumonia. Mol
Microbiol 49: 795–805
91 Sebert ME, Palmer LM, Rosenberg M,Weiser JN (2002) Microarray-based
identification of htrA, a Streptococcus pneumoniae gene that is regulated by
the CiaRH two-component system and contributes to nasopharyngeal coloni-
zation. Infect Immun 70: 4059–4067
92 Mascher T, Zahner D, Merai M, Balmelle N, de Saizieu AB, Hakenbeck R
(2003) The Streptococcus pneumoniae cia regulon: CiaR target sites and tran-
scription profile analysis. J Bacteriol 185: 60–70
93 Ibrahim YM, Kerr AR, McCluskey J, Mitchell TJ (2004) Control of virulence
by the two-component system CiaR/H is mediated via HtrA, a major virulence
factor of Streptococcus pneumoniae. J Bacteriol 186: 5258–5266
94 Ibrahim YM, Kerr AR, McCluskey J, Mitchell TJ (2004) Role of HtrA in the
virulence and competence of Streptococcus pneumoniae. Infect Immun 72:
3584–3591
95 Bethe G, Nau R, Wellmer A, Hakenbeck R, Reinert RR, Heinz HP, Zysk G
(2001) The cell wall-associated serine protease PrtA: a highly conserved viru-
lence factor of Streptococcus pneumoniae. FEMS Microbiol Lett 205: 99–104
96 Ren B, Szalai AJ, Hollingshead SK, Briles DE (2004) Effects of PspA and
antibodies to PspA on activation and deposition of complement on the pneu-
mococcal surface 14. Infect Immun 72: 114–122
97 Hammerschmidt S, Bethe G, Remane PH, Chhatwal GS (1999) Identification
of pneumococcal surface protein A as a lactoferrin-binding protein of
Streptococcus pneumoniae 60. Infect Immun 67: 1683–1687
98 Shaper M, Hollingshead SK, Benjamin WH Jr, Briles DE (2004) PspA pro-
tects Streptococcus pneumoniae from killing by apolactoferrin, and antibody
to PspA enhances killing of pneumococci by apolactoferrin [corrected]. Infect
Immun 72: 5031–5040
99 Hoskins J, Alborn WE Jr, Arnold J, Blaszczak LC, Burgett S, DeHoff BS,
Estrem ST, Fritz L, Fu DJ, Fuller W et al (2001) Genome of the bacterium
Streptococcus pneumoniae strain R6. J Bacteriol 183: 5709–5717
100 Tettelin H, Nelson KE, Paulsen IT, Eisen JA, Read TD, Peterson S, Heidelberg
J, DeBoy RT, Haft DH, Dodson RJ et al (2001) Complete genome sequence of
a virulent isolate of Streptococcus pneumoniae. Science 293: 498–506
101 Throup JP, Koretke KK, Bryant AP, Ingraham KA, Chalker AF, Ge Y, Marra
A, Wallis NG, Brown JR, Holmes DJ et al (2000) A genomic analysis of two-
component signal transduction in Streptococcus pneumoniae. Mol Microbiol 35:
566–576
102 Guenzi E, Hakenbeck R (1995) Genetic competence and susceptibility to
beta-lactam antibiotics in Streptococcus pneumoniae R6 are linked via a two-
component signal-transducing system cia. Dev Biol Stand 85: 125–128
174 Sven Hammerschmidt et al.

103 Dagkessamanskaia A, Moscoso M, Henard V, Guiral S, Overweg K, Reuter M,


Martin B, Wells J, Claverys JP (2004) Interconnection of competence, stress and
CiaR regulons in Streptococcus pneumoniae: competence triggers stationary
phase autolysis of ciaR mutant cells. Mol Microbiol 51: 1071–1086
104 Standish AJ, Stroeher UH, Paton JC (2005) The two-component signal trans-
duction system RR06/HK06 regulates expression of cbpA in Streptococcus
pneumoniae. Proc Natl Acad Sci USA 102: 7701–7706
105 Ng WL, Tsui HC, Winkler ME (2005) Regulation of the pspA virulence fac-
tor and essential pcsB murein biosynthetic genes by the phosphorylated VicR
(YycF) response regulator in Streptococcus pneumoniae. J Bacteriol 187:
7444–7459
106 Coffey TJ, Enright MC, Daniels M, Morona JK, Morona R, Hryniewicz W,
Paton JC, Spratt BG (1998) Recombinational exchanges at the capsular poly-
saccharide biosynthetic locus lead to frequent serotype changes among natural
isolates of Streptococcus pneumoniae. Mol Microbiol 27: 73–83
107 Waite RD, Struthers JK, Dowson CG (2001) Spontaneous sequence duplica-
tion within an open reading frame of the pneumococcal type 3 capsule locus
causes high-frequency phase variation. Mol Microbiol 42: 1223–1232
108 Waite RD, Penfold DW, Struthers JK, Dowson CG (2003) Spontaneous
sequence duplications within capsule genes cap8E and tts control phase
variation in Streptococcus pneumoniae serotypes 8 and 37. Microbiology 149:
497–504
109 Bender MH, Yother J (2001) CpsB is a modulator of capsule-associated tyrosine
kinase activity in Streptococcus pneumoniae. J Biol Chem 276: 47966–47974
110 Bender MH, Cartee RT, Yother J (2003) Positive correlation between tyro-
sine phosphorylation of CpsD and capsular polysaccharide production in
Streptococcus pneumoniae. J Bacteriol 185: 6057–6066
111 Morona JK, Paton JC, Miller DC, Morona R (2000) Tyrosine phosphorylation
of CpsD negatively regulates capsular polysaccharide biosynthesis in strepto-
coccus pneumoniae. Mol Microbiol 35: 1431–1442
112 Morona JK, Morona R, Miller DC, Paton JC (2003) Mutational Analysis of the
Carboxy-Terminal (YGX)(4) Repeat Domain of CpsD, an Autophosphorylating
Tyrosine Kinase Required for Capsule Biosynthesis in Streptococcus pneu-
moniae. J Bacteriol 185: 3009–3019
113 Walport MJ (2001) Complement. First of two parts. N Engl J Med 344: 1058–
1066.
114 Hostetter MK (2004) Interactions of Streptococcus pneumoniae with proteins
of the complement pathway. In: EI Tuomanen, TJ Mitchell, DA Morrison, BG
Spratt (eds) The Pneumococcus. ASM Press, Washington, DC, 201–210
115 Brown JS, Hussell T, Gilliland SM, Holden DW, Paton JC, Ehrenstein MR,
Walport MJ, Botto M (2002) The classical pathway is the dominant complement
pathway required for innate immunity to Streptococcus pneumoniae infection
in mice. Proc Natl Acad Sci USA 99: 16969–16974
116 Roy S, Knox K, Segal S, Griffiths D, Moore CE, Welsh KI, Smarason A, Day
NP, McPheat WL, Crook DW et al (2002) MBL genotype and risk of invasive
pneumococcal disease: a case-control study. Lancet 359: 1569–1573
117 Abeyta M, Hardy GG, Yother J (2003) Genetic alteration of capsule type but
Pathogenesis of Streptococcus pneumoniae infections… 175

not PspA type affects accessibility of surface-bound complement and surface


antigens of Streptococcus pneumoniae. Infect Immun 71: 218–225
118 Hamel J, Charland N, Pineau I, Ouellet C, Rioux S, Martin D, Brodeur BR
(2004) Prevention of pneumococcal disease in mice immunized with conserved
surface-accessible proteins. Infect Immun 72: 2659–2670
119 Angel CS, Ruzek M, Hostetter MK (1994) Degradation of C3 by Streptococcus
pneumoniae. J Infect Dis 170: 600–608
120 Zhang Y, Masi AW, Barniak V, Mountzouros K, Hostetter MK, Green BA
(2001) Recombinant PhpA protein, a unique histidine motif-containing protein
from Streptococcus pneumoniae, protects mice against intranasal pneumococ-
cal challenge. Infect Immun 69: 3827–3836
121 Mitchell TJ (2004) Pneumolysin and other virulence proteins. In: EI Tuomanen,
TJ Mitchell, DA Morrison, BG Spratt (eds) The pneumococcus. ASM Press,
Washington, DC, 61–74
122 Yuste J, Botto M, Paton JC, Holden DW, Brown JS (2005) Additive inhibition
of complement deposition by pneumolysin and PspA facilitates Streptococcus
pneumoniae septicemia. J Immunol 175: 1813–1819
123 Janulczyk R, Iannelli F, Sjoholm AG, Pozzi G, Bjorck L (2000) Hic, a novel
surface protein of Streptococcus pneumoniae that interferes with complement
function. J Biol Chem 275: 37257–37263
124 Jarva H, Hellwage J, Jokiranta TS, Lehtinen MJ, Zipfel PF, Meri S (2004) The
group B streptococcal beta and pneumococcal Hic proteins are structurally
related immune evasion molecules that bind the complement inhibitor factor
H in an analogous fashion. J Immunol 172: 3111–3118
125 Duthy TG, Ormsby RJ, Giannakis E, Ogunniyi AD, Stroeher UH, Paton JC,
Gordon DL (2002) The human complement regulator factor H binds pneumo-
coccal surface protein PspC via short consensus repeats 13 to 15. Infect Immun
70: 5604–5611
126 Iannelli F, Chiavolini D, Ricci S, Oggioni MR, Pozzi G (2004) Pneumococcal
surface protein C contributes to sepsis caused by Streptococcus pneumoniae in
mice. Infect Immun 72: 3077–3080
127 Tu AH, Fulgham RL, McCrory MA, Briles DE, Szalai AJ (1999) Pneumococcal
surface protein A inhibits complement activation by Streptococcus pneumoni-
ae. Infect Immun 67: 4720–4724
128 Ren B, McCrory MA, Pass C, Bullard DC, Ballantyne CM, Xu Y, Briles DE,
Szalai AJ (2004) The virulence function of Streptococcus pneumoniae surface
protein A involves inhibition of complement activation and impairment of
complement receptor-mediated protection 1. J Immunol 173: 7506–7512
129 Janeway CA Jr,Medzhitov R (2002) Innate immune recognition. Annu Rev
Immunol 20: 197–216
130 Szalai AJ, Agrawal A, Greenhough TJ,Volanakis JE (1997) C-reactive protein:
structural biology, gene expression, and host defense function. Immunol Res 16:
127–136
131 Mold C, Rodic-Polic B, Du Clos TW (2002) Protection from Streptococcus
pneumoniae infection by C-reactive protein and natural antibody requires
complement but not Fc gamma receptors. J Immunol 168: 6375–6381
176 Sven Hammerschmidt et al.

132 Hopkins PA, Sriskandan S (2005) Mammalian Toll-like receptors: to immunity


and beyond. Clin Exp Immunol 140: 395–407
133 Takeda K, Kaisho T, Akira S (2003) Toll-like receptors. Annu Rev Immunol 21:
335–376
134 Currie AJ, Davidson DJ, Reid GS, Bharya S, MacDonald KL, Devon RS,Speert
DP (2004) Primary immunodeficiency to pneumococcal infection due to a
defect in Toll-like receptor signaling. J Pediatr 144: 512–518
135 Picard C, Puel A, Bonnet M, Ku CL, Bustamante J, Yang K, Soudais C, Dupuis
S, Feinberg J, Fieschi C et al (2003) Pyogenic bacterial infections in humans
with IRAK-4 deficiency. Science 299: 2076–2079
136 Han SH, Kim JH, Martin M, Michalek SM,Nahm MH (2003) Pneumococcal
lipoteichoic acid (LTA) is not as potent as staphylococcal LTA in stimulating
Toll-like receptor 2. Infect Immun 71: 5541–5548
137 Schroder NW, Morath S, Alexander C, Hamann L, Hartung T, Zahringer
U, Gobel UB, Weber JR, Schumann RR (2003) Lipoteichoic acid (LTA) of
Streptococcus pneumoniae and Staphylococcus aureus activates immune cells
via Toll-like receptor (TLR)-2, lipopolysaccharide-binding protein (LBP), and
CD14, whereas TLR-4 and MD-2 are not involved. J Biol Chem 278: 15587–
15594
138 Weber JR, Freyer D, Alexander C, Schroder NW, Reiss A, Kuster C, Pfeil D,
Tuomanen EI, Schumann RR (2003) Recognition of pneumococcal peptidogly-
can: an expanded, pivotal role for LPS binding protein. Immunity 19: 269–279
139 Yoshimura A, Lien E, Ingalls RR, Tuomanen E, Dziarski R, Golenbock D
(1999) Cutting edge: recognition of Gram-positive bacterial cell wall compo-
nents by the innate immune system occurs via Toll-like receptor 2. J Immunol
163: 1–5
140 Travassos LH, Girardin SE, Philpott DJ, Blanot D, Nahori MA, Werts C,
Boneca IG (2004) Toll-like receptor 2-dependent bacterial sensing does not
occur via peptidoglycan recognition. EMBO Rep 5: 1000–1006
141 Echchannaoui H, Frei K, Schnell C, Leib SL, Zimmerli W, Landmann R (2002)
Toll-like receptor 2-deficient mice are highly susceptible to Streptococcus
pneumoniae meningitis because of reduced bacterial clearing and enhanced
inflammation. J Infect Dis 186: 798–806
142 Koedel U, Angele B, Rupprecht T, Wagner H, Roggenkamp A, Pfister HW,
Kirschning CJ (2003) Toll-like receptor 2 participates in mediation of immune
response in experimental pneumococcal meningitis. J Immunol 170: 438–444
143 Knapp S, Wieland CW, van ’t Veer C, Takeuchi O, Akira S, Florquin S, van
der Poll T (2004) Toll-like receptor 2 plays a role in the early inflammatory
response to murine pneumococcal pneumonia but does not contribute to anti-
bacterial defense 95. J Immunol 172: 3132–3138
144 Khan AQ, Chen Q, Wu ZQ, Paton JC, Snapper CM (2005) Both innate immu-
nity and type 1 humoral immunity to Streptococcus pneumoniae are mediated
by MyD88 but differ in their relative levels of dependence on toll-like receptor
2. Infect Immun 73: 298–307
145 van Rossum AM, Lysenko ES, Weiser JN (2005) Host and bacterial factors
contributing to the clearance of colonization by Streptococcus pneumoniae in a
murine model. Infect Immun 73: 7718–7726
Pathogenesis of Streptococcus pneumoniae infections… 177

146 Ozinsky A, Underhill DM, Fontenot JD, Hajjar AM, Smith KD, Wilson CB,
Schroeder L, Aderem A (2000) The repertoire for pattern recognition of patho-
gens by the innate immune system is defined by cooperation between toll-like
receptors. Proc Natl Acad Sci USA 97: 13766–13771
147 Malley R, Trzcinski K, Srivastava A, Thompson CM, Anderson PW, Lipsitch M
(2005) CD4 + cells mediate antibody-independent acquired immunity to pneu-
mococcal colonization. Proc Natl Acad Sci USA 102: 4848–4853
148 Srivastava A, Henneke P, Visintin A, Morse SC, Martin V, Watkins C, Paton JC,
Wessels MR, Golenbock DT, Malley R (2005) The apoptotic response to pneu-
molysin is Toll-like receptor 4 dependent and protects against pneumococcal
disease. Infect Immun 73: 6479–6487
149 Malley R, Henneke P, Morse SC, Cieslewicz MJ, Lipsitch M, Thompson CM,
Kurt-Jones E, Paton JC, Wessels MR, Golenbock DT (2003) Recognition of
pneumolysin by Toll-like receptor 4 confers resistance to pneumococcal infec-
tion. Proc Natl Acad Sci USA 100: 1966–1971
150 Branger J, Knapp S, Weijer S, Leemans JC, Pater JM, Speelman P, Florquin S,
van der Poll T (2004) Role of Toll-like receptor 4 in gram-positive and gram-
negative pneumonia in mice. Infect Immun 72: 788–794
151 Benton KA, Paton JC, Briles DE (1997) The hemolytic and complement-acti-
vating properties of pneumolysin do not contribute individually to virulence in
a pneumococcal bacteremia model. Microb Pathog 23: 201–209
152 Yamamoto M, Takeda K, Akira S (2004) TIR domain-containing adaptors
define the specificity of TLR signaling. Mol Immunol 40: 861–868
153 Albiger B, Sandgren A, Katsuragi H, Meyer-Hoffert U, Beiter K, Wartha F,
Hornef M, Normark S, Normark BH (2005) Myeloid differentiation factor
88-dependent signalling controls bacterial growth during colonization and sys-
temic pneumococcal disease in mice. Cell Microbiol 7: 1603–1615
154 Koedel U, Rupprecht T, Angele B, Heesemann J, Wagner H, Pfister HW,
Kirschning CJ (2004) MyD88 is required for mounting a robust host immune
response to Streptococcus pneumoniae in the CNS. Brain 127: 1437–1445
155 Branger J, Florquin S, Knapp S, Leemans JC, Pater JM, Speelman P, Golenbock
DT, van der PT (2004) LPS-binding protein-deficient mice have an impaired
defense against Gram-negative but not Gram-positive pneumonia. Int Immunol
16: 1605–1611
156 Pugin J, Schurer-Maly CC, Leturcq D, Moriarty A, Ulevitch RJ, Tobias PS
(1993) Lipopolysaccharide activation of human endothelial and epithelial cells
is mediated by lipopolysaccharide-binding protein and soluble CD14. Proc Natl
Acad Sci USA 90: 2744–2748
157 Pugin J, Heumann ID, Tomasz A, Kravchenko VV, Akamatsu Y, Nishijima M,
Glauser MP, Tobias PS, Ulevitch RJ (1994) CD14 is a pattern recognition recep-
tor. Immunity 1: 509–516
158 Cauwels A, Wan E, Leismann M, Tuomanen E (1997) Coexistence of CD14-
dependent and independent pathways for stimulation of human monocytes by
gram-positive bacteria. Infect Immun 65: 3255–3260
159 Echchannaoui H, Frei K, Letiembre M, Strieter RM, Adachi Y, Landmann R
(2005) CD14 deficiency leads to increased MIP–2 production, CXCR2 expres-
178 Sven Hammerschmidt et al.

sion, neutrophil transmigration, and early death in pneumococcal infection. J


Leukoc Biol 78: 705–715.
160 Philpott DJ, Girardin SE (2004) The role of Toll-like receptors and Nod pro-
teins in bacterial infection. Mol Immunol 41: 1099–1108
161 Opitz B, Puschel A, Schmeck B, Hocke AC, Rosseau S, Hammerschmidt S,
Schumann RR, Suttorp N,Hippenstiel S (2004) Nucleotide-binding oligo-
merization domain proteins are innate immune receptors for internalized
Streptococcus pneumoniae. J Biol Chem 279: 36426–36432
162 Chamaillard M, Hashimoto M, Horie Y, Masumoto J, Qiu S, Saab L, Ogura Y,
Kawasaki A, Fukase K, Kusumoto S et al (2003) An essential role for NOD1
in host recognition of bacterial peptidoglycan containing diaminopimelic acid.
Nat Immunol 4: 702–707
163 Girardin SE, Boneca IG, Carneiro LA, Antignac A, Jehanno M, Viala J, Tedin
K, Taha MK, Labigne A, Zahringer U et al (2003) Nod1 detects a unique muro-
peptide from gram-negative bacterial peptidoglycan. Science 300: 1584–1587
164 Pauleau AL, Murray PJ (2003) Role of nod2 in the response of macrophages to
toll-like receptor agonists. Mol Cell Biol 23: 7531–7539
165 Kang YS, Kim JY, Bruening SA, Pack M, Charalambous A, Pritsker A, Moran
TM, Loeffler JM, Steinman RM, Park CG (2004) The C-type lectin SIGN-R1
mediates uptake of the capsular polysaccharide of Streptococcus pneumoniae
in the marginal zone of mouse spleen. Proc Natl Acad Sci USA 101: 215–220
166 Lanoue A, Clatworthy MR, Smith P, Green S, Townsend MJ, Jolin HE, Smith
KG, Fallon PG, McKenzie AN (2004) SIGN-R1 contributes to protection
against lethal pneumococcal infection in mice. J Exp Med 200: 1383–1393
167 Koppel EA, Wieland CW, van den Berg VC, Litjens M, Florquin S, van Kooyk
Y, van der Poll T, Geijtenbeek TB (2005) Specific ICAM–3 grabbing noninte-
grin-related 1 (SIGNR1) expressed by marginal zone macrophages is essential
for defense against pulmonary Streptococcus pneumoniae infection. Eur J
Immunol 35: 2962–2969
168 Arredouani M, Yang Z, Ning Y, Qin G, Soininen R, Tryggvason K,Kobzik L
(2004) The scavenger receptor MARCO is required for lung defense against
pneumococcal pneumonia and inhaled particles. J Exp Med 200: 267–272
169 Um SH, Son EW, Kim BO, Moon EY, Rhee DK, Pyo S (2000) Activation of
murine peritoneal macrophages by Streptococcus pneumoniae type II capsular
polysaccharide: involvement of CD14-dependent pathway. Scand J Immunol
52: 39–45
170 Crouch E, Wright JR (2001) Surfactant proteins a and d and pulmonary host
defense. Annu Rev Physiol 63: 521–554
171 Whitsett JA (2005) Surfactant proteins in innate host defense of the lung. Biol
Neonate 88: 175–180
172 Jounblat R, Clark H, Eggleton P, Hawgood S, Andrew PW, Kadioglu A (2005)
The role of surfactant protein D in the colonisation of the respiratory tract and
onset of bacteraemia during pneumococcal pneumonia. Respir Res 6: 126
173 Kuronuma K, Sano H, Kato K, Kudo K, Hyakushima N, Yokota S, Takahashi H,
Fujii N, Suzuki H, Kodama T et al (2004) Pulmonary surfactant protein A aug-
ments the phagocytosis of Streptococcus pneumoniae by alveolar macrophages
Pathogenesis of Streptococcus pneumoniae infections… 179

through a casein kinase 2-dependent increase of cell surface localization of


scavenger receptor A. J Biol Chem 279: 21421–21430
174 Paster BJ, Boches SK, Galvin JL, Ericson RE, Lau CN, Levanos VA,
Sahasrabudhe A, Dewhirst FE (2001) Bacterial diversity in human subgingival
plaque. J Bacteriol 183: 3770–3783
175 Neish AS, Gewirtz AT, Zeng H, Young AN, Hobert ME, Karmali V, Rao AS,
Madara JL (2000) Prokaryotic regulation of epithelial responses by inhibition
of IkappaB-alpha ubiquitination. Science 289: 1560–1563
176 Tong HH, Long JP, Shannon PA, DeMaria TF (2003) Expression of cytokine
and chemokine genes by human middle ear epithelial cells induced by influ-
enza A virus and Streptococcus pneumoniae opacity variants. Infect Immun 71:
4289–4296
177 Ratner AJ, Lysenko ES, Paul MN, Weiser JN (2005) Synergistic proinflamma-
tory responses induced by polymicrobial colonization of epithelial surfaces.
Proc Natl Acad Sci USA 102: 3429–3434
178 Lysenko ES, Ratner AJ, Nelson AL, Weiser JN (2005) The role of innate
immune responses in the outcome of interspecies competition for colonization
of mucosal surfaces. PLoS Pathog 1: e1
179 Pericone CD, Overweg K, Hermans PW, Weiser JN (2000) Inhibitory and
bactericidal effects of hydrogen peroxide production by Streptococcus pneu-
moniae on other inhabitants of the upper respiratory tract. Infect Immun 68:
3990–3997
180 McCullers JA, Bartmess KC (2003) Role of neuraminidase in lethal syner-
gism between influenza virus and Streptococcus pneumoniae. J Infect Dis 187:
1000–1009
181 van der Sluijs KF, van Elden LJ, Nijhuis M, Schuurman R, Pater JM, Florquin
S, Goldman M, Jansen HM, Lutter R, van der Poll T (2004) IL-10 is an impor-
tant mediator of the enhanced susceptibility to pneumococcal pneumonia after
influenza infection. J Immunol 172: 7603–7609
182 Tong HH, Long JP, Li D, DeMaria TF (2004) Alteration of gene expression in
human middle ear epithelial cells induced by influenza A virus and its implica-
tion for the pathogenesis of otitis media. Microb Pathog 37: 193–204
183 Kadioglu A, Coward W, Colston MJ, Hewitt CR, Andrew PW (2004) CD4-T-
lymphocyte interactions with pneumolysin and pneumococci suggest a crucial
protective role in the host response to pneumococcal infection. Infect Immun
72: 2689–2697
184 Jounblat R, Kadioglu A, Mitchell TJ, Andrew PW (2003) Pneumococcal behav-
ior and host responses during bronchopneumonia are affected differently
by the cytolytic and complement-activating activities of pneumolysin. Infect
Immun 71: 1813–1819
185 Kadioglu A, Gingles NA, Grattan K, Kerr A, Mitchell TJ, Andrew PW (2000)
Host cellular immune response to pneumococcal lung infection in mice. Infect
Immun 68: 492–501
186 Komai-Koma M, Jones L, Ogg GS, Xu D, Liew FY (2004) TLR2 is expressed
on activated T-cells as a costimulatory receptor. Proc Natl Acad Sci USA 101:
3029–3034
180 Sven Hammerschmidt et al.

187 Austrian R (1981) Pneumococcal pneumonia and pneumococcal vaccine. Mt


Sinai J Med 48: 532–538
188 Casal J, Tarrago D (2003) Immunity to Streptococcus pneumoniae: Factors
affecting production and efficacy. Curr Opin Infect Dis 16: 219–224
189 Black S, Shinefield H, Fireman B, Lewis E, Ray P, Hansen JR, Elvin L, Ensor
KM, Hackell J, Siber G et al (2000) Efficacy, safety and immunogenicity of
heptavalent pneumococcal conjugate vaccine in children. Northern California
Kaiser Permanente Vaccine Study Center Group. Pediatr Infect Dis J 19: 187–
195
190 Balachandran P, Brooks-Walter A, Virolainen-Julkunen A, Hollingshead SK,
Briles DE (2002) Role of pneumococcal surface protein C in nasopharyngeal
carriage and pneumonia and its ability to elicit protection against carriage of
Streptococcus pneumoniae. Infect Immun 70: 2526–2534
191 Briles DE, Hollingshead SK, King J, Swift A, Braun PA, Park MK, Ferguson
LM, Nahm MH, Nabors GS (2000) Immunization of humans with recombinant
pneumococcal surface protein A (rPspA) elicits antibodies that passively pro-
tect mice from fatal infection with Streptococcus pneumoniae bearing heterolo-
gous PspA. J Infect Dis 182: 1694–1701
192 McDaniel LS, Waltman WD, 2nd, Gray B, Briles DE (1987) A protective mono-
clonal antibody that reacts with a novel antigen of pneumococcal teichoic acid.
Microb Pathog 3: 249–260
193 Ogunniyi AD, Folland RL, Briles DE, Hollingshead SK, Paton JC (2000)
Immunization of mice with combinations of pneumococcal virulence proteins
elicits enhanced protection against challenge with Streptococcus pneumoniae.
Infect Immun 68: 3028–3033
194 Overweg K, Kerr A, Sluijter M, Jackson MH, Mitchell TJ, de Jong AP, de Groot
R, Hermans PW (2000) The putative proteinase maturation protein A of
Streptococcus pneumoniae is a conserved surface protein with potential to elicit
protective immune responses. Infect Immun 68: 4180–4188
195 Wizemann TM, Heinrichs JH, Adamou JE, Erwin AL, Kunsch C, Choi GH,
Barash SC, Rosen CA, Masure HR, Tuomanen E et al (2001) Use of a whole
genome approach to identify vaccine molecules affording protection against
Streptococcus pneumoniae infection. Infect Immun 69: 1593–1598
196 McCool TL, Cate TR, Tuomanen EI, Adrian P, Mitchell TJ,Weiser JN (2003)
Serum immunoglobulin G response to candidate vaccine antigens during
experimental human pneumococcal colonization. Infect Immun 71: 5724–5732
197 Paton JC (1998) Novel pneumococcal surface proteins: role in virulence and
vaccine potential. Trends Microbiol 6: 85–87; discussion 87–88
198 Morrison KE, Lake D, Crook J, Carlone GM, Ades E, Facklam R, Sampson JS
(2000) Confirmation of psaA in all 90 serotypes of Streptococcus pneumoniae
by PCR and potential of this assay for identification and diagnosis. J Clin
Microbiol 38: 434–437
199 McCool TL, Cate TR, Moy G, Weiser JN (2002) The immune response to
pneumococcal proteins during experimental human carriage. J Exp Med 195:
359–365
200 Obaro SK, Adegbola RA, Tharpe JA, Ades EW, McAdam KP, Carlone G,
Sampson JS (2000) Pneumococcal surface adhesin A antibody concentration
Pathogenesis of Streptococcus pneumoniae infections… 181

in serum and nasopharyngeal carriage of Streptococcus pneumoniae in young


African infants. Vaccine 19: 411–412
201 Rapola S, Jantti V, Haikala R, Syrjanen R, Carlone GM, Sampson JS, Briles DE,
Paton JC, Takala AK, Kilpi TM et al (2000) Natural development of antibod-
ies to pneumococcal surface protein A, pneumococcal surface adhesin A, and
pneumolysin in relation to pneumococcal carriage and acute otitis media. J
Infect Dis 182: 1146–1152
202 Brooks-Walter A, Briles DE, Hollingshead SK (1999) The pspC gene of
Streptococcus pneumoniae encodes a polymorphic protein, PspC, which elicits
cross-reactive antibodies to PspA and provides immunity to pneumococcal
bacteremia. Infect Immun 67: 6533–6542
203 Hollingshead SK, Becker R, Briles DE (2000) Diversity of PspA: mosaic genes
and evidence for past recombination in Streptococcus pneumoniae. Infect
Immun 68: 5889–5900
204 Zhang Q, Bernatoniene J, Bagrade L, Pollard AJ, Mitchell TJ, Paton JC, Finn A
(2006) Serum and mucosal antibody responses to pneumococcal protein anti-
gens in children: relationships with carriage status. Eur J Immunol 36: 46–57
205 Adrian PV, Bogaert D, Oprins M, Rapola S, Lahdenkari M, Kilpi T, de Groot
R, Kayhty H, Hermans PW (2004) Development of antibodies against pneu-
mococcal proteins alpha-enolase, immunoglobulin A1 protease, streptococcal
lipoprotein rotamase A, and putative proteinase maturation protein A in rela-
tion to pneumococcal carriage and Otitis Media. Vaccine 22: 2737–2742
206 Rogers PD, Thornton J, Barker KS, McDaniel DO, Sacks GS, Swiatlo E,
McDaniel LS (2003) Pneumolysin-dependent and -independent gene expres-
sion identified by cDNA microarray analysis of THP-1 human mononuclear
cells stimulated by Streptococcus pneumoniae. Infect Immun 71: 2087–2094
207 Chen A, Li HS, Hebda PA, Zeevi A, Swarts JD (2005) Gene expression profiles
of early pneumococcal otitis media in the rat. Int J Pediatr Otorhinolaryngol 69:
1383–1393
208 Nelson AL, Barasch JM, Bunte RM, Weiser JN (2005) Bacterial colonization
of nasal mucosa induces expression of siderocalin, an iron-sequestering compo-
nent of innate immunity. Cell Microbiol 7: 1404–1417
209 Bergmann S, Hammerschmidt S (2006) Versatility of pneumococcal surface
proteins. Microbiology 152: 295–303
210 Paterson GK, Mitchell TJ (2006) Innate immunity and the pneumococcus.
Microbiology 152: 285–293
211 Lau GW, Haataja S, Lonetto M, Kensit SE, Marra A, Bryant AP, McDevitt
D, Morrison DA, Holden D (2001) A functional genomic analysis of type 3
Streptococcus pneumoniae virulence. Mol Microbiol 40: 555–571
Community-Acquired Pneumonia 183
ed. by N. Suttorp, T. Welte and R. Marre
© 2007 Birkhäuser Verlag Basel/Switzerland

Pathogenesis of Mycoplasma pneumoniae infections:


adaptive immunity, innate immunity, cell biology, and
virulence factors

Ken B. Waites1, Jerry W. Simecka2, Deborah F. Talkington3


and T. Prescott Atkinson4
1Department of Pathology, University of Alabama at Birmingham, Department of Pathology,
WP 230, 619 19th St. South, Birmingham, AL 35249, USA; 2Department of Molecular Biology
and Immunology, University of North Texas Health Science Center, Fort Worth, TX 76107,
USA; 3Division of Bacterial and Mycotic Diseases, Centers for Disease Control and Prevention,
Atlanta, GA 30333, USA; 4Department of Pediatrics, University of Alabama at Birmingham,
Birmingham, AL 35249, USA

Abstract
Mycoplasmas represent the smallest self-replicating organisms. They are unique among
bacteria in that they lack a cell wall and require sterols for growth. The limited metabolic
and biosynthetic activities of mycoplasmas have complicated development of accurate
means for laboratory detection and hampered understanding of their roles as human
pathogens. Mycoplasma pneumoniae was first identified and characterized in the 1960s
and shown to be a common cause of upper and lower respiratory disease in children and
adults. Serious infections requiring hospitalization, while rare, occur in persons of all age
groups, and may affect multiple organ systems. Severity of disease appears to be related
to the degree to which the host immune response reacts to the infection. Extrapulmonary
complications involving all of the major organ systems can occur in association with M.
pneumoniae infection as a result of direct invasion and/or autoimmune response. Evidence
is accumulating for this organism’s contributory role in chronic lung conditions such as
asthma. Serology has been the most common means for laboratory detection of M. pneu-
moniae infection due to the slow growth that makes culture impractical. Newer diagnostic
methods utilizing nucleic acid amplification offer the advantages for rapid detection and
are likely to become increasingly important in the future, but these techniques have not
achieved widespread utilization thus far due to the lack of commercially sold products
and non-standardized methodology. Management of M. pneumoniae infections can usu-
ally be achieved with macrolides, ketolides, tetracyclines, or fluoroquinolones. As more is
learned about pathogenesis and immune response elicited by M. pneumoniae, improved
methods for diagnosis and prevention of disease due to this organism are anticipated.
184 Ken B. Waites et al.

Cell biology of Mycoplasma pneumoniae

Mycoplasmas represent the smallest self-replicating organisms capable of


cell-free existence, both in cellular dimensions and genome size. Individual
spindle-shaped cells of M. pneumoniae are 1–2 +m long and 0.1–0.2 +m
wide. Accordingly, the M. pneumoniae cell volume is less than 5% of that of
a typical bacillus. Typical colonies of M. pneumoniae rarely exceed 100 +m
in size and require examination under a stereomicroscope to visualize their
morphologic features. The M. pneumoniae genome was sequenced in 1996
and shown to consist of 816,394 basepairs with 687 genes [1], about one-
sixth the size of Escherichia coli.
The small genome of M. pneumoniae and its limited biosynthetic
capabilities are responsible for many of the biological characteristics and
requirements for complex medium supplementation in order for the organ-
ism to be cultivated in vitro. Mycoplasmas cannot synthesize peptidoglycan
cell walls. Lack of a rigid cell wall makes them pleomorphic and unable to
be classified in the manner of conventional eubacteria. Mycoplasmas are
not found freely living in nature since they depend on a host cell to supply
the necessary nutrients. Another characteristic of the genus Mycoplasma
is the requirement for sterols in artificial growth media, supplied by the
addition of serum. Sterols are necessary components of the triple-layered
mycoplasmal cell membrane providing structural support to the osmotically
fragile organisms.
Although mycoplasmas can flourish within an osmotically stable envi-
ronment in their eukaryotic host, they are extremely susceptible to desicca-
tion. This explains the need for close contact for transmission of infection
from person to person by airborne droplets. Another structural component
that is important for extracellular survival is a protein network that provides
a cytoskeleton to support the cell membrane. M. pneumoniae also produces
capsular material that may have a role in cytadherence.
M. pneumoniae possesses very limited metabolic and biosynthetic activi-
ties for proteins, carbohydrates, and lipids. It scavenges for nucleic acid pre-
cursors and apparently does not synthesize purines or pyrimidines de novo.
Fermentation of glucose to lactic acid by means of substrate phosphoryla-
tion mediated by phosphoglyceric acid kinase and pyruvate kinase activities
are means of ATP generation. M. pneumoniae possesses all reactions of
glycolysis, but the tricarboxylic acid cycle and a complete electron transport
chain containing cytochromes are absent. M. pneumoniae reduces tetrazo-
lium and this property has been used historically to distinguish it from com-
mensal oropharyngeal mycoplasmas. Reproduction occurs by binary fission,
during which the attachment organelle migrates to the opposite pole of the
cell during replication and before nucleoid separation.
Neither genomic analysis nor electron microscopy has demonstrated the
presence of structures such as flagella or pili, suggesting that gliding motility
occurs by an unknown mechanism involving the attachment organelle [2].
Pathogenesis of Mycoplasma pneumoniae infections… 185

In addition to the crossing of the mucus barrier that protects the respiratory
epithelium, gliding motility may contribute to the ability of M. pneumoniae
to travel down respiratory cilia to attach to respiratory epithelial cells.

Epidemiology and transmission of infection

M. pneumoniae infections can involve both the upper and lower respiratory
tract and occur both endemically and epidemically worldwide in persons of
all ages. Climate, seasonality, and geography are not thought to be of major
significance, although most outbreaks in the USA tend to occur in the late
summer and early fall. Foy [3] reported that M. pneumoniae was responsible
for 15-20% of all cases of community-acquired pneumonia (CAP) between
1962 and 1975 in Seattle, Washington, USA. Additional retrospective sero-
logical studies performed in Denmark showed a pattern of M. pneumoniae
infections over a 50-year period from 1946 through 1995 with endemic
disease transmission punctuated with cyclic epidemics every 3 to 5 years
[4]. The long incubation period and relatively low transmission rate have
been implicated in the prolonged duration of epidemics of M. pneumoniae
infections.
A study performed in the USA during the 1990s detected M. pneu-
moniae in 23% of CAP in children 3-4 years of age [5]. Another study
from France [6] documented its occurrence in children less than 4 years
of age without significant differences in infection rates for other children
or adults. These findings may reflect the greater number of young children
who attend day care centers on a regular basis than in previous years, and
the ease with which young children share respiratory secretions with older
household members or contacts. Marston [7] reported that M. pneumoniae
was definitely responsible for 5.4% and possibly responsible for 32.5% of
2,776 cases of CAP in hospitalized adults in Ohio, USA. Extrapolation of
data nationally provides an estimated 18,700 to 108,000 cases of CAP in
hospitalized adults due to M. pneumoniae annually. Since the majority of
CAPs are treated as outpatients, the total number of pneumonias due to M.
pneumoniae is almost certainly many times greater. An additional striking
finding was their observation that mycoplasmal pneumonia in hospitalized
adults increased with age and it was second only to Streptococcus pneu-
moniae in elderly persons.
The P1 adhesin is a 170 kD transmembrane protein that is concentrated
on the adhesive tip of M. pneumoniae and serves an essential function in
cytadherence. Different P1 subtypes may operate in cycling times of M.
pneumoniae epidemics. Gene divergences within the P1 adhesin and devel-
opment of subtype-specific antibodies following initial infection might also
contribute to the frequency of reinfections due to another subtype. Studies
using a variety of genotypic methods to characterize over 200 M. pneu-
moniae isolates collected over several years from multiple countries showed
186 Ken B. Waites et al.

that most of the isolates could be classified into two subtypes based on the
sequences of the P1 adhesin gene, the ORF6 gene, the P65 gene, and by a
typical DNA restriction fragment pattern [8, 9]. One or the other of the two
subgroups tended to predominate in specific regions.
M. pneumoniae is not considered part of the normal flora and its
detection by culture can usually be considered abnormal and of etiologic
significance if detected in a person with a clinical condition known to be
caused by the organism. However, it can persist for variable periods in the
respiratory tract following infections that resolved clinically with appropri-
ate antimicrobial therapy, providing a reservoir for spread of the organism
to others.

Cytadherence and other virulence factors

The initial step in the pathogenesis of mycoplasmal respiratory disease


involves M. pneumoniae adherence to ciliated respiratory epithelium.
This intimate interaction damages respiratory epithelial cells through the
production of toxic substances, such as hydrogen peroxide and superoxide
radicals, leading to oxidative stress. Subsequent development of the host
inflammatory response may actually have the greatest impact on disease. In
fact, the host’s bronchoepithelial cells may contribute to the development
of the inflammatory lesions through the release of cytokines in response to
infection or other stimuli. Release of interleukin (IL)-8, from human bron-
choepithelial cells may occur in response to stimulation with mycoplasma
membranes [10]. Similarly, human lung alveolar type II pneumocytes (A549
cells) infected with M. pneumoniae show an increase IL-8, tumor necrosis
factor-alpha (TNF-_), and IL-1` mRNA [11], supporting the idea that the
adherence to human airway epithelial cells leads to production of cytokines
and recruitment of lymphocytes and other inflammatory cells, and that
these cytokines subsequently modulate the activity of the inflammatory
infiltrates.
The P1 protein adhesin is also immunogenic, and it is the target for
antibodies that develop in the course of natural infection. The host-cell
ligand for mycoplasmal adhesins has not been characterized conclusively,
though sialoglycoconjugates and sulfated glycolipids have been implicated
[12]. At least six other proteins (HMW1, HMW2, HMW3, P90, P40 and
P30) are known to participate in adhesion. All are localized to the terminal
tip of the organism, and most are likely involved in the architecture of the
attachment organelle and localization of P1. P30 appears to be involved
with gliding motility. HMW1, HMW2, and HMW3 are critical in the forma-
tion and stabilization of the attachment organelle, including localization
of other adhesin-related proteins. Once this polar structure is established,
an independently assembled complex of proteins B, C and P1 is drawn to
the structure to complete formation of the functional terminal attachment
Pathogenesis of Mycoplasma pneumoniae infections… 187

Figure 1. Transmission electron micrographs of Mycoplasma pneumoniae grown in culture


demonstrating flask-shaped morphology and the prominent adhesin tip (Courtesy Kristin
Hoek and Leigh Milligan, UAB High Resolution Imaging Facility).

organelle shown as an electron dense region in the flask-shaped organism


by electron micrography (Fig, 1). Two proteins, elongation factor TU and
pyruvate dehydrogenase E1` are involved in binding M. pneumoniae to
fibronectin [13].
Mammalian cells parasitized by M. pneumoniae can exhibit a number
of cytopathic effects as a result of the local damage following cytadher-
ence. Cells may lose their cilia entirely, appear vacuolated, show a reduc-
tion in oxygen consumption, glucose utilization, amino acid uptake, and
macromolecular synthesis, ultimately resulting in exfoliation of all or parts
of the infected cells. These subcellular events result in some of the clinical
manifestations of respiratory tract infection such as the persistent, hacking
cough.
Dallo [14] recently described the ability of M. pneumoniae to survive,
synthesize DNA, and undergo cell replication in artificial cell culture sys-
tems over a 6-month period. Intracellular sequestration could facilitate the
establishment latent or chronic states, circumvent mycoplasmacidal immune
mechanisms, facilitate the ability to cross mucosal barriers and gain access
to internal tissues, and impair efficacy of some drug therapies, accounting
for difficulty in eradicating the mycoplasmas in clinical conditions. However,
the extent to which M. pneumoniae invades and replicates intracellularly in
vivo is not known. High-frequency phase and antigenic variation of surface
adhesin proteins made possible by DNA rearrangements in truncated and
sequence-related copies of the P1 adhesin genes that are dispersed through-
out the genome may also be a means for M. pneumoniae to evade the host
immune response [15].
188 Ken B. Waites et al.

Innate immunity to M. pneumoniae disease

Elements of the host innate immune system are activated by M. pneu-


moniae and the attendant inflammation accounts for many of the initial
signs and symptoms accompanying the early stages of the evolving infec-
tion. Once M. pneumoniae reaches the lower respiratory tract, the organ-
ism may be opsonized by complement or antibodies. Macrophages become
activated, begin phagocytosis, and undergo chemotactic migration to the
site of infection. There is in vitro evidence that the organism is susceptible
to complement-mediated cytolysis, probably through both the alternative
and classical pathways [16, 17]. Therefore, it is possible that complement
plays a significant role in inhibiting growth of the organism, particularly
after inflammation induces an exudate. While there are reports of invasive
infections with M. pneumoniae in hypogammaglobulinemic individuals,
there are none relating similar occurrences in patients with complement
deficiencies alone. A recent study identified a highly significant association
between mannose binding lectin deficiency and invasive infections with M.
pneumoniae in patients with concomitant antibody deficiency [18]. A series
of surfactant proteins (SP-A–SP-D) are also capable of binding and regulat-
ing the growth of microorganisms through lectin-like activity. SP-A exhibits
calcium-dependent binding and growth inhibition of M. pneumoniae [19].
Interaction of M. pneumoniae with mast cells via a sialic acid-dependent
binding mechanism was reported to induce cytokine production, especially
IL-4, IL-5, and TNF-_ [20, 21], and is dependent upon the presence of the P1
adhesin. M. pneumoniae lipoproteins may interact with toll-like receptors 2
(TLR2) and/or 6 (TLR6) of respiratory epithelial or other cells, as found
with other mycoplasmas [22, 23]. As a result of TLR interaction, cells are
stimulated to produce cytokines or begin apoptosis, thus contributing to the
pathogenesis of disease.
M. pneumoniae may induce cellular activation during co-culture with
a variety of other immunologic cells, including lymphocytes, macrophages,
and respiratory epithelial cells. The nonspecific nature of the cellular targets
of activation by M. pneumoniae could explain the broad range of inflamma-
tory and autoimmune phenomena following acute infection.

Adaptive immunity and interaction of M. pneumoniae with host


immune cells

Adaptive immunity, characterized by both B and T lymphocyte responses,


has a major impact on the progression of M. pneumoniae respiratory dis-
ease. While data are limited on the role of lymphoid responses in human M.
pneumoniae infections, studies of other mycoplasmal respiratory diseases in
animals indicate that some immune responses will be beneficial in control-
ling or preventing infection, while others contribute to disease severity.
Pathogenesis of Mycoplasma pneumoniae infections… 189

Local (mucosal) immune responses may be most effective in protect-


ing the host from mycoplasma infection since resistance to M. pneumoniae
seems to be most closely associated with IgA responses, but immunity is
often short-lived, as humans are susceptible to repeated infections, despite
the development of complement fixing and growth inhibiting antibody
responses [15]. Immune responses that develop after infection often fail to
eliminate the mycoplasma, indicating that adaptive immunity apparently
has a limited effect on clearance of an established infection, thus leading to
asymptomatic carriage for variable periods of time. M. pneumoniae infec-
tions in adults may be asymptomatic, perhaps reflecting some degree of
protective immunity against reinfections over time.
Persons with impaired ability to produce antibody, such as in congenital
hypogammaglobulinemia, can suffer from chronic respiratory disease due
to M. pneumoniae, suggesting antibody and other immune responses have
a limited, but significant, role in recovery from disease [24]. In addition,
hypogammaglobulinemic persons are more susceptible to extrapulmonary
complications [25]. These observations suggest that antibody responses are
important in controlling infection and preventing dissemination of myco-
plasmas from the respiratory tract.
Cytokine production and lymphocyte activation may either minimize
disease through the enhancement of host defense mechanisms, or exac-
erbate disease through the development of immunologic hypersensitivity,
worsening damage to the respiratory epithelium. The more vigorous the
cell-mediated immune response and cytokine stimulation, the more severe
the clinical illness and pulmonary injury. Lymphoid infiltration characteristic
of M. pneumoniae disease suggests that lymphocyte activation contributes
to the development of inflammatory responses. M. pneumoniae infection of
laboratory rodents reveal CD4 + T helper (Th) cells within the inflamma-
tory infiltrates in lung [26], and mycoplasma-specific Th cell responses are
found in peripheral blood from humans [27]. CD8 + T cells also increase in
lungs of mice after intranasal infection with M. pneumoniae [28] and T-cell-
depleted hamsters develop less severe M. pneumoniae respiratory disease
[29]. Mycoplasma pulmonis infection of severe combined immunodefi-
ciency (SCID) mice or T-cell-deficient mice results in milder lung lesions
[30], and both T-cell populations modulate disease severity without directly
affecting the number of mycoplasmas in the lungs. CD4 + Th-cells promote
the inflammatory responses in rodent lungs, whereas CD8 + T-cells dampen
these responses [31]. Most likely, a similar dichotomy of T-cell activity
occurs in human M. pneumoniae disease. Depletion of CD4 + T-cells in mice
results in a decreased cytokine responses and lymphocyte accumulation in
the lungs after experimental M. pneumoniae infection [28]. Thus, popula-
tions of CD4 + T- and CD8 + T-cells most likely have contrasting roles in M.
pneumoniae pulmonary disease, with the actions of a population of proin-
flammatory CD4 + Th-cells being suppressed by mycoplasma-specific CD8 +
T-cells. Furthermore, CD4 + T-lymphocyte, B lymphocyte, and plasma cell
190 Ken B. Waites et al.

accumulation in the lung is responsible for the radiographic manifestation


of pulmonary infiltrates and is associated with lymphocyte proliferation,
production of immunoglobulins, and release of TNF-_, interferon gamma
(IFN-a), and various interleukins [15].
Host responses that develop after M. pneumoniae infection likely con-
tribute to extrapulmonary complications. Examples are the association
between M. pneumoniae infection and increased severity of asthma and
production of anti-erythrocyte autoantibodies “cold agglutinins,” once used
as a diagnostic indicator of M. pneumoniae infection.
Overall, adaptive immune responses generated after M. pneumoniae
infections have contrasting impacts on the pathogenesis of infection.
Beneficial effects of host resistance lead to containment of the infection and
ultimately recovery in many persons. Relative persistence of M. pneumoniae
in the host leads to the provocation of ineffective immune-mediated inflam-
matory responses, regulated by opposing T-cell activities. Complicating the
impact of these responses is their contribution to other adverse reactions,
such as asthma and autoimmune-like effects.

Clinical and laboratory aspects of M. pneumoniae infections

Following a 2–3-week incubation period, symptoms may develop over


1–2 days and consist of worsening nonexudative pharyngitis, nasal and
sinus congestion, low-grade fever, and cough. In 1–2 weeks, more seri-
ously affected individuals develop tracheobronchitis and primary atypical
pneumonia (PAP), a syndrome consisting of fever, cough, musical inspira-
tory rhonchi reflecting mucus secretion in large airways, and occasion-
ally bilateral expiratory wheezing. Acute lower respiratory symptoms may
progress to respiratory distress with hypoxemia and necessitate hospital
admission, but such severe cases represent only 5–10% of affected indi-
viduals. Patients with pre-existing pulmonary disease such as asthma or
chronic bronchitis may be more severely affected. Antibiotic therapy with
appropriate drugs results in clinical improvement, but the organism tends
to persist in the airways. The cough, often productive, typically continues
for several weeks and abnormalities in pulmonary hyperresponsiveness can
persist for months.
Diagnosis of PAP is often presumptive because of difficulties in micro-
biological confirmation. Chest radiographs typically reveal only perihilar
and bibasilar streaky infiltrates, findings less impressive than symptoms
would suggest. Small pleural effusions occur in 5-20% of patients. Clinical
presentation may be similar to what is also seen with other pathogens, par-
ticularly Chlamydophila pneumoniae, various respiratory viruses, and even
bacteria such as S. pneumoniae. M. pneumoniae may also be present in the
respiratory tract concomitantly with other pathogens. About one-third of
persons with mycoplasmal infections have leukocytosis and an elevated
Pathogenesis of Mycoplasma pneumoniae infections… 191

erythrocyte sedimentation rate. Sputum gram stain may show mononuclear


cells or neutrophils and normal flora. There are no hepatic or renal abnor-
malities typical of M. pneumoniae infection, although the hemolytic anemia
that develops in some patients may be reflected in the hemogram.

Autoimmunity and extrapulmonary manifestations

Autoimmune reactions are believed to be responsible for many of the


extrapulmonary complications that are associated with as much as 25% of
M. pneumoniae infections. They may occur as a result of molecular mim-
icry in the ascending paralysis of Guillain-Barré Syndrome that results
from autoantibody-mediated destruction of peripheral nerve fibers. These
antibodies may also recognize glycolipids extracted from M. pneumoniae.
Another mechanism is illustrated in hemolytic anemia due to cross-react-
ing antibodies against erythrocyte antigens as seen in cold-agglutinin
disease. The PCR assay has greatly enhanced understanding of how M.
pneumoniae can disseminate throughout the body. The presence of M.
pneumoniae in blood, synovial fluid, cerebrospinal fluid, pericardial fluid,
and skin lesions has been documented by PCR and/or culture. Thus, direct
invasion must always be considered [15]. The frequency of direct invasion
of these sites is unknown because the organism is rarely sought and most
reported extrapulmonary syndromes have been attributed to M. pneu-
moniae infection based on serology. Extrapulmonary manifestations have
been reviewed in depth elsewhere with original citations [15] and are sum-
marized in Table 1.

Pathologic aspects of lung disease due to M. pneumoniae

Acute perivascular and peribronchial cell infiltration results in destruc-


tion of respiratory epithelium. There are also neutrophilic accumulations
within the airways early in disease. At later stages, massive mononuclear
cell infiltration, of which T-cells are a major component, occurs. Ulceration
with destruction of ciliated epithelium of bronchi and bronchioles, edema,
bronchiolar and alveolar infiltrates of macrophages, lymphocytes, and
neutrophils, increased numbers of plasma cells, and deposition of fibrin.
Type II pneumocyte hyperplasia with diffuse alveolar damage and bron-
chiolitis obliterans have been described [15]. Pleura may contain patches
of fibrinous exudates. Pleural effusions sometimes occur in association
with more severe cases complicated by long-term sequelae such as pleural
scarring, bronchiectasis, and pulmonary fibrosis. Lung abscesses may also
occur. Immunosuppressed persons may lack pulmonary infiltrates, further
attesting to the importance of the host immune response in lesion develop-
ment.
192 Ken B. Waites et al.

Table 1. Extrapulmonary manifestations of M. pneumoniae infection resulting from autoim-


mune reaction and/or direct invasion

Organ system Manifestations Comment


Neurologic Ascending paralysis, encepha- Nervous system complications are
litis, aseptic meningitis, trans- among the most common and most
verse myelitis, cerebellar syn- severe manifestations. They often
drome, polyradiculitis, cranial occur within 1–2 weeks of respiratory
nerve palsies, optic neuritis, infection and in some cases without
choreoathetosis, leukoen- any apparent preceding respiratory
cephalopathy, acute psychosis symptoms.
Musculoskeletal Myalgias, arthralgias, polyar- Invasive joint infections occur more
thropathies, septic arthritis, commonly in persons with antibody
reactive (inflammatory nonin- deficiency, but they have also been
fectious) arthritis, osteomyeli- described in immunocompetent per-
tis, rhabdomyolysis sons. Recent data suggest a possible
association with adult and juvenile
rheumatoid arthritis.
Skin Erythematous maculopapular Dermatological disorders are the
and vesicular rashes, Stevens- most common type of extrapulmonary
Johnson Syndrome, conjuncti- manifestations.
vitis, ulcerative stomatitis
Cardiovascular Pericarditis, myocarditis, Cardiac involvement occurs in up to
pericardial effusion, cardiac 8.5% of serologically diagnosed infec-
tamponade tions and is more common in adults
than children.
Hematologic Hemolytic anemia, cold agglu- Cross-reactive antibodies against
tinin disease, aplastic anemia, erythrocyte antigens or plasma von
thrombotic thrombocytopenic Willebrand factor-cleaving protease
purpura, disseminated intra- have been implicated as causative fac-
vascular coagulation tors.
Renal Acute glomerulonephritis, Antibody-mediated pathogenesis is
renal failure, tubulointerstitial believed to predominate, but myco-
nephritis, IgA nephropathy plasma antigen has been detected in
damaged kidney tissue by immunohis-
tochemistry.
Gastrointestinal Nausea, vomiting, diarrhea, These nonspecific complaints can be
and other cholestatic hepatitis, pancreati- associated with respiratory disease.
tis otitis media, myringitis Mechanisms have not been carefully
investigated.

Chronic lung disease associated with M. pneumoniae infection

M. pneumoniae has been isolated in increased frequency from stable asth-


matics. It has been linked with exacerbations of existing asthma, as well as
the subsequent development of asthma in previously healthy persons. Kraft
[32] detected M. pneumoniae by PCR in respiratory secretions of 42% sta-
ble adult asthmatics versus 9% of healthy controls. In another study, throat
cultures for M. pneumoniae were positive in 24.7% of children and adults
Pathogenesis of Mycoplasma pneumoniae infections… 193

with asthma exacerbation, compared with 5.7% of healthy controls [33]. In


another study, 21% of adults having a flare of their asthma were found to
have high levels of M. pneumoniae-specific IgM, suggesting exacerbation
occurred in the context of acute infection [34].
Macrolide treatment of asthma patients in whom M. pneumoniae has
been detected resulted in improvement in pulmonary function tests in
comparison with asthma patients who did not have evidence of the organ-
ism in their airways [35], owing perhaps to both the antibacterial as well as
anti-inflammatory effects of these drugs. Mycoplasmas have been detected
by PCR in airways even when culture and serology are negative, suggest-
ing that low numbers of organisms may evade detection by the immune
system [35]. Lack of a measurable serologic response may also facilitate the
organism’s persistence in the lower respiratory tract.
M. pneumoniae is known to induce a number of the inflammatory
mediators implicated in the pathogenesis of asthma that may play a role in
exacerbations. IgE triggering of mast cell degranulation is a key event in
allergic asthma, and mycoplasma-specific IgE responses may elicit a similar
response [20]. Mycoplasma IL-4 stimulation may enhance the asthma-pro-
moting IgE responses against potential allergens. M. pneumoniae can be
associated with significantly greater numbers of mast cells in humans with
chronic asthma [36], and experimental evidence from a rodent mast cell line
suggests that the organism can induce activation of mast cells [20, 21]. Koh
[37] showed that levels of IL-4 and the ratio of IL-4/IFN-a were significantly
higher in children with M. pneumoniae than those with pneumococcal
pneumonia or uninfected controls, suggesting a TH2-like cytokine response
representing a favorable condition for IgE production.
Gump [38] reported that mycoplasmal infections could be associated
with some cases of chronic obstructive pulmonary disease (COPD) exac-
erbation. Subsequent studies, summarized in a recent review [15] have
reported serologic evidence of acute M. pneumoniae infection in 6-14% of
COPD exacerbations. Evidence of concomitant respiratory pathogens in
some cases complicates understanding of the significance of M. pneumoniae
in this context.
Bacterial infection is the main cause of progressive pulmonary failure
in patients with cystic fibrosis. Serological examinations have been the
sole means of assessing the presence of M. pneumoniae in persons with
cystic fibrosis and the methods employed present some difficulty in proper
interpretation to define a recent infection. Limited findings to date suggest
mycoplasmas may occur but are fairly uncommon causes of respiratory
complications in persons with cystic fibrosis [15]. More work must be done
to clarify the importance of M. pneumoniae in the epidemiology and patho-
genesis of exacerbations of chronic lung diseases using a more comprehen-
sive diagnostic strategy that would include direct tests for the presence of
the organisms by PCR, culture, and serology.
194 Ken B. Waites et al.

Measurement of the immune response as a means for diagnosis

Serology has been the most common laboratory means for diagnosis of M.
pneumoniae infections. Although culture and PCR are also used, persis-
tence of the organism for variable lengths of time following acute infection
makes it difficult to assess the significance of a positive culture or PCR
assay without additional confirmatory seroconversion. The length of time
necessary for culture (sometimes 6 weeks or more) makes it impractical
for patient management and it is not widely available except in specialized
reference laboratories. Detailed information on laboratory diagnosis of M.
pneumoniae infection is available elsewhere [15] and only pertinent sum-
mary information is discussed here.
M. pneumoniae has both lipid and protein antigens which elicit antibody
responses that can be detected after about 1 week of illness, peaking at 3-
6 weeks, followed by a gradual decline, allowing several different types of
serological assays, based on different antigens and technologies. Serology
is a useful epidemiologic tool in circumstances where the likelihood of
mycoplasmal disease is high, but it is less suited for assessment of individual
patients. Its main disadvantage is the need for both acute and convalescent
paired sera collected 2 to 3 weeks apart that are tested simultaneously for
IgM and IgG to confirm seroconversion. This is especially important in
adults over 40 years of age who may not mount an IgM response, presum-
ably because of reinfection. Moreover, IgM antibodies can sometimes per-
sist for several weeks to months, making it risky to base diagnosis of acute
infection on a single assay for IgM alone. Antibody production may also
be delayed in some infections, or even absent if the patient is immunosup-
pressed. False-negative tests for IgM can also occur if serum is collected too
soon after onset of illness. Since M. pneumoniae is a mucosal pathogen, IgA
is typically produced early in the course of infection. Measurement of serum
IgA may therefore be a better approach for diagnosis of acute infection
because of its rapid rise and decline, but very few commercial assays include
reagents for its detection.
Complement fixation (CF) was the first method developed for sero-
logical testing for M. pneumoniae. CF measures mainly the early IgM
response and does not differentiate among antibody classes, which is
desirable to differentiate acute from remote infection. CF suffers from
low sensitivity and specificity because the glycolipid antigen mixture used
may be found in other microorganisms, as well as human tissues, and even
plants. Cross-reactions with Mycoplasma genitalium are well recognized.
In most clinical laboratories CF has been replaced by alternative tech-
niques with greater sensitivity and specificity, many of which have been
developed and sold as commercial kits. Immunofluorescent antibody
(IFA) assays, direct and indirect hemagglutination using IgM capture, and
other particle agglutination antibody assays (PAs) have been developed
to detect antibody to M. pneumoniae. Enzyme immunoassays (EIAs)
Pathogenesis of Mycoplasma pneumoniae infections… 195

have become the most widely used commercial methods for detection
of M. pneumoniae. EIAs are more sensitive for detecting acute infection
than culture, and can be comparable in sensitivity to PCR, providing a
sufficient time has elapsed since infection for antibody to develop and
the patient has a functional immune system. These assays may be qualita-
tive or quantitative, may or may not require specialized equipment for
performing the assay and reading the results, and can be performed with
very small volumes of serum. The need for acute and convalescent sera
has remained the obvious limitation for prompt point-of-care diagnosis.
However, qualitative rapid point-of care serologic assays that detect both
IgM and IgG or IgM alone in an easy-to-read format without the need
for any instrumentation have been developed and shown to compare
favorably with other commercial assays [15]. The variability of results
from comparative studies underscores the need for improved serologi-
cal reagents for detecting acute M. pneumoniae infection [39]. The best
commercial EIA for individual patient diagnosis depends on the age of
the patient, timing of serum collection, whether paired sera are avail-
able, equipment available, and experience of the laboratory personnel.
However, maintaining a large variety of different assays within one labo-
ratory is not practical or cost-effective.

Use of the PCR assay for detection of M. pneumoniae

Gene targets used in PCR assays for M. pneumoniae include 16S rRNA,
P1 adhesin, ATPase operon gene, and tuf gene. Real-time PCR assays
have also been described [15]. The sensitivity of PCR is very high, cor-
responding to a single organism when purified DNA is used. Other
advantages are the potential ability to complete the procedure in one day,
the requirement of only one specimen containing organisms that do not
have to be viable, as well as the ability to detect nucleic acid in preserved
tissues. Comparison of PCR with culture and/or serology has yielded
varied results that are not always in agreement. Positive PCR results in
culture-negative persons without evidence of respiratory disease suggest
inadequate assay specificity, persistence of the organism after infection,
or asymptomatic carriage, perhaps in an intracellular compartment that
does not yield culturable organisms. Quantitative studies may be useful
in drawing conclusions. Positive PCR results in serologically-negative
persons may be due to an inadequate immune response, early success-
ful antibiotic treatment, or to the collection of specimens before specific
antibody synthesis could occur. Negative PCR results in culture or sero-
logically proven infections raise the possibility of inhibitors or other tech-
nical problems with the assay. If antibiotics have been administered, PCR
results may be negative even though serology is positive. Ideally, positive
PCR assays should be confirmed by a second unrelated target gene. Thus
196 Ken B. Waites et al.

far, there are no commercial PCR kits for detection of M. pneumoniae,


but several are in development.

Treatment of M. pneumoniae infections

Administration of antimicrobials to patients with M. pneumoniae infections


will generally produce satisfactory results with a marked reduction in dura-
tion of respiratory symptoms. Management has been guided primarily by
well-known and consistent susceptibilities to a variety of drugs. Macrolides
are the treatments of choice, but tetracyclines and fluoroquinolones are also
effective, as is the new ketolide telithromycin. Most clinical trials evaluating
treatments for CAP identified small numbers of cases proven to be due to
M. pneumoniae by serologic diagnosis, though some recent studies incorpo-
rated culture and/or PCR. Use of serology alone precludes determination as
to whether a treatment regimen actually eradicates the organism, thus very
little data are available regarding microbiological efficacy of any regimen.
A summary of clinical trials evaluating various treatments for CAP in which
outcome data specific for M. pneumoniae were included can be found in a
recent review [15]. A recent study from Japan found that macrolide resis-
tance in M. pneumoniae due to transition mutations in domain V on the 23S
rRNA gene occurred in 6% of 195 clinical isolates from patients with acute
infections [40]. The clinical significance of this resistance is uncertain, but it
suggests the need to monitor clinical isolates for such resistance. High-dose
steroids have been reported to be useful in treatment of encephalitis in chil-
dren with complicated M. pneumoniae infection [41]. Plasmapheresis and
intravenous immunoglobulin therapy might be considered if steroid therapy
is ineffective in these settings.

Summary

M. pneumoniae is a common cause of CAP in both children and adults.


Serious infections requiring hospitalization, while rare, occur in persons of
all age groups, and may affect multiple organ systems. Severity of disease
appears to be related to the degree to which the host immune response
reacts to the infection. Extrapulmonary complications involving all of the
major organ systems can occur in association with M. pneumoniae infection
as a result of direct invasion and/or autoimmune response. Evidence is accu-
mulating for this organism’s contributory role in chronic lung conditions
such as asthma. Effective management of M. pneumoniae infections can
usually be achieved with macrolides, ketolides, tetracyclines, or fluoroquino-
lones. As more is learned about pathogenesis and immune response elicited
by M. pneumoniae, improvement in methods for diagnosis and prevention
of disease due to this organism are anticipated.
Pathogenesis of Mycoplasma pneumoniae infections… 197

Acknowledgments

Portions of the work summarized in this chapter were supported by U.S.


Public Health Service grants HL73907-01A1 to T.P.A. and grants HL069431,
AI42075, and AI055907 to J.W.S.

References
1 Himmelreich R, Hilbert H, Plagens H, Pirkl E, Li BC, Herrmann R (1996)
Complete sequence analysis of the genome of the bacterium Mycoplasma
pneumoniae. Nucleic Acids Res 24: 4420–4449
2 Miyata M (2002) Gliding motility of mycoplasma – a mechanism cannot be
explained by today’s biology. Nippon Saikingaku Zasshi 57: 581–595
3 Foy HM (1993) Infections caused by Mycoplasma pneumoniae and possible
carrier state in different populations of patients. Clin Infect Dis 17 (Suppl 1):
S37–46
4 Lind K, Benzon MW, Jensen JS, Clyde WA, Jr (1997) A seroepidemiological
study of Mycoplasma pneumoniae infections in Denmark over the 50-year
period 1946–1995. Eur J Epidemiol 13: 581–586
5 Block S, Hedrick J, Hammerschlag MR, Cassell GH, Craft JC (1995) Mycoplasma
pneumoniae and Chlamydia pneumoniae in pediatric community-acquired
pneumonia: comparative efficacy and safety of clarithromycin vs. erythromycin
ethylsuccinate. Pediatr Infect Dis J 14: 471–477
6 Layani-Milon MP, Gras I, Valette M, Luciani J, Stagnara J, Aymard M, Lina B
(1999) Incidence of upper respiratory tract Mycoplasma pneumoniae infections
among outpatients in Rhone-Alpes, France, during five successive winter peri-
ods. J Clin Microbiol 37: 1721–1726
7 Marston BJ, Plouffe JF, File TM Jr, Hackman BA, Salstrom SJ, Lipman HB,
Kolczak MS, Breiman RF (1997) Incidence of community-acquired pneumonia
requiring hospitalization. Results of a population-based active surveillance
Study in Ohio. The Community-Based Pneumonia Incidence Study Group.
Arch Intern Med 157: 1709–1718
8 Cousin-Allery A, Charron A, de Barbeyrac B, Fremy G, Skov Jensen J,
Renaudin H, Bebear C (2000) Molecular typing of Mycoplasma pneumoniae
strains by PCR-based methods and pulsed-field gel electrophoresis. Application
to French and Danish isolates. Epidemiol Infect 124: 103–111
9 Dumke R, Catrein I, Pirkil E, Herrmann R, Jacobs E (2003) Subtyping of
Mycoplasma pneumoniae isolates based on extended genome sequencing and
on expression profiles. Int J Med Microbiol 292: 513–525
10 Chmura K, Lutz RD, Chiba H, Numata MS, Choi HJ, Fantuzzi G, Voelker DR,
Chan ED (2003) Mycoplasma pneumoniae antigens stimulate interleukin-8.
Chest 123: 425S
11 Yang J, Hooper WC, Phillips DJ, Talkington DF (2002) Regulation of proin-
flammatory cytokines in human lung epithelial cells infected with Mycoplasma
pneumoniae. Infect Immun 70: 3649–3655
12 Roberts DD, Olson LD, Barile MF, Ginsburg V, Krivan HC (1989) Sialic acid-
198 Ken B. Waites et al.

dependent adhesion of Mycoplasma pneumoniae to purified glycoproteins. J


Biol Chem 264: 9289–9293
13 Dallo SF, Kannan TR, Blaylock MW, Baseman JB (2002) Elongation factor
Tu and E1 beta subunit of pyruvate dehydrogenase complex act as fibronectin
binding proteins in Mycoplasma pneumoniae. Mol Microbiol 46: 1041–1051
14 Dallo SF, Baseman JB (2000) Intracellular DNA replication and long-term
survival of pathogenic mycoplasmas. Microb Pathog 29: 301–309
15 Waites KB, Talkington DF (2004) Mycoplasma pneumoniae and its role as a
human pathogen. Clin Microbiol Rev 17: 697–728
16 Bredt W, Kist M, Jacobs E (1981) Phagocytosis and complement action. Isr J
Med Sci 17: 637–640
17 Loos M, Brunner H (1979) Complement components (C1, C2, C3, C4) in
bronchial secretions after intranasal infection of guinea pigs with Mycoplasma
pneumoniae: dissociation of unspecific and specific defense mechanisms. Infect
Immun 25: 583–585
18 Hamvas RM, Johnson M, Vlieger AM, Ling C, Sherriff A, Wade A, Klein NJ,
Turner MW, Webster AD (2005) Role for mannose binding lectin in the preven-
tion of mycoplasma infection. Infect Immun 73: 5238–5240
19 Phiboonpocanum S, Chiba H, Mitsuzawa H, Martin W, Murphy RC, Harbeck
RJ, Voelker RB (2005) Surfactant protein A binds Mycoplasma pneumoniae
with high affinity and attenuates its growth by recognition of desaturated phos-
phatidylglycerols. J Biol Chem 280: 9–17
20 Hoek KL, Cassell GH, Duffy LB, Atkinson TP (2002) Mycoplasma pneumoni-
ae-induced activation and cytokine production in rodent mast cells. J Allergy
Clin Immunol 109: 470–476
21 Hoek KL, Duffy LB, Cassell GH, Atkinson TP (2005) A role for the Mycoplasma
pneumoniae adhesin P1 in Interleukin (IL)-4 synthesis and release from rodent
mast cells. Microb Pathog 39: 149–158
22 Into T, Nodasaka Y, Hasebe A, Okuzawa T, Nakamura J, Ohata N, Shibata K
(2002) Mycoplasmal lipoproteins induce toll-like receptor 2- and caspases-
mediated cell death in lymphocytes and monocytes. Microbiol Immunol 46:
265–276
23 Takeuchi O, Kaufmann A, Grote K, Kawai T, Hoshino K, Morr M, Muhlradt
PF, Akira S (2000) Cutting edge: preferentially the R-stereoisomer of the myco-
plasmal lipopeptide macrophage-activating lipopeptide-2 activates immune
cells through a toll-like receptor 2- and MyD88-dependent signaling pathway. J
Immunol 164: 554–557
24 Taylor-Robinson D, Webster AD, Furr PM, Asherson GL (1980) Prolonged
persistence of Mycoplasma pneumoniae in a patient with hypogammaglobu-
linaemia. J Infect 2: 171–175
25 Johnston CL, Webster AD, Taylor-Robinson D, Rapaport G, Hughes GR (1983)
Primary late-onset hypogammaglobulinaemia associated with inflammatory
polyarthritis and septic arthritis due to Mycoplasma pneumoniae. Ann Rheum
Dis 42: 108–110
26 Hayakawa M, Taguchi H, Kamiya S, Fujioka Y, Watanabe H, Kawai S, Kobayashi
H (2002) Animal model of Mycoplasma pneumoniae infection using germfree
mice. Clin Diagn Lab Immunol 9: 669–676
Pathogenesis of Mycoplasma pneumoniae infections… 199

27 Tanaka H, Narita M, Teramoto S, Saikai T, Oashi K, Igarashi T, Abe S (2002)


Role of interleukin–18 and T-helper type 1 cytokines in the development of
Mycoplasma pneumoniae pneumonia in adults. Chest 121: 1493–1497
28 Opitz O, Pietsch K, Ehlers S, Jacobs E (1996) Cytokine gene expression in
immune mice reinfected with Mycoplasma pneumoniae: the role of T cell sub-
sets in aggravating the inflammatory response. Immunobiology 196: 575–587
29 Taylor G, Taylor-Robinson D, Fernald GW (1974) Reduction in the severity of
Mycoplasma pneumoniae-induced pneumonia in hamsters by immunosuppres-
sive treatment with antithymocyte sera. J Med Microbiol 7: 343–348
30 Cartner SC, Lindsey JR, Gibbs-Erwin J, Cassell GH, Simecka JW (1998) Roles
of innate and adaptive immunity in respiratory mycoplasmosis. Infect Immun
66: 3485–3491
31 Jones H, Tabor L, Sun X, Woolard M, Simecka JW (2002) Depletion of CD8 + T
cells exacerbates CD4 + Th cell asociated inflammatory lesions during murine
respiratory disease. J Immunol 168: 3493–3501
32 Kraft M, Cassell GH, Henson JE, Watson H, Williamson J, Marmion BP, Gaydos
CA, Martin RJ (1998) Detection of Mycoplasma pneumoniae in the airways of
adults with chronic asthma. Am J Respir Crit Care Med 158: 998–1001
33 Gil JC, Cedillo RL, Mayagoitia BG, Paz MD (1993) Isolation of Mycoplasma
pneumoniae from asthmatic patients. Ann Allergy 70: 23–25
34 Seggev JS, Lis I, Siman-Tov R, Gutman R, Abu-Samara H, Schey G, Naot Y
(1986) Mycoplasma pneumoniae is a frequent cause of exacerbation of bron-
chial asthma in adults. Ann Allergy 57: 263–265
35 Kraft M, Cassell GH, Pak J, Martin RJ (2002) Mycoplasma pneumoniae and
Chlamydia pneumoniae in asthma: effect of clarithromycin. Chest 121: 1782–
1788
36 Martin RJ, Chu HW, Honour JM, Harbeck RJ (2001) Airway inflammation and
bronchial hyperresponsiveness after Mycoplasma pneumoniae infection in a
murine model. Am J Respir Cell Mol Biol 24: 577–582
37 Koh YY, Park Y, Lee HJ, Kim CK (2001) Levels of interleukin-2, interferon-
gamma, and interleukin-4 in bronchoalveolar lavage fluid from patients with
Mycoplasma pneumonia: implication of tendency toward increased immuno-
globulin E production. Pediatrics 107: E39
38 Gump DW, Phillips CA, Forsyth BR, McIntosh K, Lamborn KR, Stouch WH
(1976) Role of infection in chronic bronchitis. Am Rev Respir Dis 113: 465–
474
39 Talkington DF, Shott S, Fallon MT, Schwartz SB, Thacker WL (2004) Analysis
of eight commercial enzyme immunoassay tests for detection of antibodies
to Mycoplasma pneumoniae in human serum. Clin Diagn Lab Immunol 11:
862–867
40 Morozumi M, Hasegawa K, Kobayashi R, Inoue N, Iwata S, Kuroki H,
Kawamura N, Nakayama E, Tajima T, Shimizu K et al (2005) Emergence of
macrolide-resistant Mycoplasma pneumoniae with a 23S rRNA gene mutation.
Antimicrob Agents Chemother 49: 2302–2306
41 Carpenter TC (2002) Corticosteroids in the treatment of etriatal encephalitis
complicating Mycoplasma pneumoniae pneumonia: possible benefit of intrave-
nous immunoglobulin. Pediatr Infect Dis J 20: 534–545
Community-Acquired Pneumonia 201
ed. by N. Suttorp, T. Welte and R. Marre
© 2007 Birkhäuser Verlag Basel/Switzerland

Community-acquired pneumonia: paving the way


towards new vaccination concepts

Pablo D. Becker and Carlos A. Guzmán

Department of Vaccinology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7,


38124 Braunschweig, Germany

Abstract
Despite the availability of antimicrobial agents and vaccines, community-acquired pneu-
monia remains a serious problem. Severe forms tend to occur in very young children
and among the elderly, since their immune competence is eroded by immaturity and
immune senescence, respectively. The main etiologic agents differ according to patient
age and geographic area. Streptococcus pneumoniae, Haemophilus influenzae, respira-
tory syncytial virus (RSV) and parainfluenza virus type 3 (PIV-3) are the most important
pathogens in children, whereas influenza viruses are the leading cause of fatal pneumonia
in the elderly. Effective vaccines are available against some of these organisms. However,
there are still many agents against which vaccines are not available or the existent ones
are suboptimal. To tackle this problem, empiric approaches are now being systematically
replaced by rational vaccine design. This is facilitated by the growing knowledge in the
fields of immunology, microbial pathogenesis and host response to infection, as well
as by the availability of sophisticated strategies for antigen selection, potent immune
modulators and efficient antigen delivery systems. Thus, a new generation of vaccines
with improved safety and efficacy profiles compared to old and new agents is emerging.
In this chapter, an overview is provided about currently available and new vaccination
concepts.

Introduction

The mucosa of the human respiratory tract represents a primary target for
a large number of microbial pathogens. Typically, colonization is an asymp-
tomatic process, resulting from the interplay between bacterial factors and
host clearance mechanisms. Clinical illness may result from either the local
release of bacterial toxins or the systemic dissemination of the pathogen
after breaching the mucosal barrier. In the course of respiratory infections
adaptive immune responses could be significantly impaired. This might
lead to more severe forms of disease or to super-infections, which in turn
complicate the clinical management of the patient. The most severe forms
of respiratory infection tend to occur in very young children and among the
202 Pablo D. Becker and Carlos A. Guzmán

elderly, in whom immune competence is eroded by immaturity or immune-


senescence, respectively. In addition, patients who are immunocompro-
mised, as a result of disease or therapeutic interventions, have the greatest
risk of developing a fatal infection.
Despite the availability of new antimicrobials and effective vaccines,
community-acquired pneumonia remains a common and serious illness. In
fact, it is a leading contributor to the nearly 4 million deaths occurring each
year due to respiratory infections, especially in children from developing
countries [1, 2]. The main causative agents of pneumonia differ according
to the patient age and the geographic area. In addition, there are relatively
few comprehensive studies on the specific aetiology of pneumonia [2] due
to (i) overlaps in the clinical manifestations of the different syndromes, (ii)
difficulties in establishing the precise aetiology, and (iii) frequent occur-
rence of co-infections. However, Streptococcus pneumoniae, Haemophilus
influenzae, respiratory syncytial virus (RSV), and parainfluenza virus type 3
(PIV-3) have been identified as the main agents responsible for acute respi-
ratory infections in children, whereas influenza virus related pneumonia is
the leading cause of disease-related deaths in the elderly. In addition, the
availability of new and more sensitive diagnostic tests have contributed to
the identification of hitherto unknown lower respiratory pathogens, such as
the human metapneumonovirus (hMPV) and novel coronaviruses causing
the severe acute respiratory syndrome (SARS).
Significant efforts have been invested in the last two decades to develop
new diagnostic tools, to elucidate the molecular mechanisms of microbial
pathogenesis and to understand host clearance mechanisms. This resulted in
an improved knowledge on host responses to infection and immuno-patho-
genesis, which in turn have facilitated the establishment of new prophylac-
tic and therapeutic interventions. However, despite our accomplishments
in vaccine development, there are many pathogens for which vaccines or
adequate therapies are not available or the existent ones are suboptimal.
The main approach applied for vaccine development has radically
changed in recent years. Whole cell vaccines are systematically being
replaced by subunit vaccines, in which purified antigens or their coding genes
are exploited in combination with new adjuvants and/or delivery systems. As
a result, many of the vaccines under development will exhibit consistently
improved stability, safety and efficacy profiles. They will also be amenable
for mucosal administration, thereby mimicking natural infections.

Currently available vaccines

Influenza vaccines

Influenza A viruses are the most commonly responsible for severe respira-
tory illness in humans, followed by influenza B. The population’s susceptibly
New vaccination concepts for CAP 203

to infection is renewed annually, because of the rapid antigenic variation of


this virus. The antigenic variation is due to the accumulation of point muta-
tions in the two major surface glycoproteins of the virus, haemagglutinin
(HA) and neuraminidase (NA). This can lead to an antigenic drift of the
virus, which often leaves current influenza vaccines outdated and ineffec-
tive. Antigenic shift can also occur due to the segmented nature of the viral
genome that favours the emergence of re-assorted strains, in which an entire
glycoprotein can be acquired from a different animal influenza virus. Both
types of variation represent a critical bottleneck for the establishment of
a robust vaccination strategy against influenza. In fact, when an influenza
virus with the capacity to spread from person-to-person and a complete new
glycoprotein subtype suddenly emerges, a worldwide pandemic outbreak
can result [3].

Inactivated vaccines against influenza

The earliest vaccines against influenza were whole cell vaccines obtained in
the 1940s by inactivating viruses grown in the allantoic cavity of embryonat-
ed chicken eggs with formalin. While contemporary inactivated influenza
vaccines are still produced in embryonated eggs, improvements in manu-
facturing have resulted in a highly purified and less-reactogenic detergent-
split product. Three viral strains are selected on the basis of the previous
year’s surveillance data on the most prevalent subtypes, therefore, vaccine
composition may vary from year-to-year. Vaccination has a high benefit:cost
ratio, since influenza-related illness (e.g., hospitalizations and deaths) are
effectively prevented [1].
The world’s total vaccine production is approximately 300 million
doses, with a maximum capacity of 900 million doses. However, the World
Health Organization (WHO) estimates that there are about 1.2 billion
people at high risk for severe influenza outcomes (e.g., elderly over 65
years of age, infants, health care workers, children and adults with under-
lying cardiopulmonary disease). Furthermore, the global infrastructure
would not be able to handle the timely manufacturing and distribution
of a vaccine for a pandemic outbreak [4]. One alternative would be to
lower the quantity of antigen per dose and add adjuvants to the vaccine
formulation, but this needs to be tested in clinical trials [5]. Another solu-
tion would be to improve current vaccine production technologies (i.e.,
egg-derived vaccines). However, there is the limited number of egg pro-
ducers and viral strains can emerge, which could not be easily adapted to
embryonated eggs. To overcome these problems, several pharmaceutical
companies have embarked themselves on projects for the development of
vaccines produced by growing the virus on cell lines. The influenza virus
can be adapted to grow on a variety of mammalian cell lines, including
Vero, PER.C-6, and Madin-Darby canine kidney (MDCK) cells [6–8].
204 Pablo D. Becker and Carlos A. Guzmán

This strategy would also improve the possibility of up-scaling vaccine pro-
duction in face of a pandemic spread. Alternatively, it would be possible
to develop a vaccine against any influenza virus, such as the avian H5N1
strain, by using reverse genetics techniques [9] (see below in advances in
vaccinology).

Live attenuated vaccines against influenza

Cold adaptation was found to be a reliable and efficient procedure for the
derivation of live attenuated viral vaccine strains for humans. Cold-adapted
(ca) virus strains can grow in primary chick kidney cells or embryonated
eggs at 25–33°C, however, they exhibit a reduced replication at 37°C. The
process of genetic re-assortment with the transfer of the six internal genes
from a stable attenuated ca master donor strain of influenza A or B to the
new prevailing wild-type epidemic strain has been used to generate attenu-
ated cold-reassorted vaccines with the proper level of attenuation, genetic
stability and immunogenicity, which show low or absent transmissibility
[10]. MedImmune and Wyeth have developed along these lines a trivalent
live ca vaccine (Flumist) for intranasal spray delivery, which was licensed
in 2003. In contrast, to parenterally-administered vaccines, this formulation
triggers immune responses resembling those observed after natural infec-
tions [11]. Despite the moderate hemagglutination-inhibiting antibody titres
observed in vacinees, Flumist showed 92% efficacy over a 2-year period in
children, including protection against antigenic variants that circulated dur-
ing the second year [12–14]. This ca vaccine also stimulated the production
of nasal IgA, as well as T-cell and interferon responses [15]. The cell-medi-
ated immunity against virus matrix and nucleoprotein antigens may favour
viral clearance and early recovery from illness [3]. The Advisory committee
on Immunization Practices has recommended its use only in persons from
5 to 49 years of age, since side-effects were observed in young children
(wheezing, nasal congestion) and there are no data available for elderly
[16, 17]. Despite its remarkable genetic stability, this vaccine has to be kept
at – 18°C. Thus, a new heat stable derivative has recently been developed,
which showed good efficacy in clinical trials [1]. A live vaccine based on
a master virus strain developed at the Institute of Applied Microbiology
(Austria) by growing wild influenza virus in Vero cells at 25°C was also
demonstrated to be safe, well-tolerated and immunogenic after intranasal
immunization in young adults [18].

Subunit and DNA vaccines against influenza

A number of subunit- or DNA-based vaccines are also in various stages


of development. An influenza vaccine formulated in virosomes has been
New vaccination concepts for CAP 205

commercialized by Berna Biotech (Inflexal® V); it contains the surface


spikes of the three currently circulating influenza virus strains inserted
in vesicle membranes of the three corresponding virus types (for more
details see section “Pseudoviruses as antigen delivery systems”) [19].
This company has also developed a virosome-based nasal formulation.
However, it was withdrawn from the market due to the presence of side-
effects (i.e., Bell’s palsy), which was assumed to be linked to the presence
of the Escherichia coli heat labile toxin (LT) as adjuvant. Two companies,
Yeda and BionVax, are also developing a peptide-based influenza vaccine
for nasal administration, which showed protective efficacy in humanized
mice [1]. A subunit vaccine containing recombinant HA protein produced
using a baculovirus system was successfully tested in a phase II trial in
64 to 89-year-old volunteers. An epidermal DNA-based influenza vac-
cine, which contained the HA gene from A/Panama/2007/99 delivered
by particle-mediated epidermal delivery was also tested in humans by
PowderJect [20]. Serum haemagglutination-inhibition antibody responses
were observed in volunteers receiving a single dose of 1, 2 or 4 +g of
DNA, with the strongest and most consistent responses in subjects vac-
cinated with the highest dosage.
Some immunization approaches aim at the development of a univer-
sal vaccine with a broad spectrum of protective activity against different
influenza strains [21]. Among them, the use of the highly conserved trans-
membrane M2 protein of the virion can be mentioned. A recombinant par-
ticulate vaccine has been engineered by genetically fusing copies of the M2
to the hepatitis B core antigen (HBc). The M2-HBc fusion protein sponta-
neously assembled into virus-like particles (VLP), which provided complete
protection against a lethal challenge with influenza virus A in mice [22, 23].
Promising results were also obtained after vaccination with a M2 peptide
conjugated with a Neisseria meningitidis outer membrane protein complex
(OMPC) in monkeys [24].

Vaccines against the parainfluenza virus

The human PIV (hPIV) consist of four serotypes, with hPIV-3 being the
second leading cause of bronchitis and pneumonia in infants. No vac-
cine has been licensed to date against PIV, however, several approaches
are currently under investigation. The initial attempts to provide pro-
tection by using vaccines based on formalin-inactivated viruses failed.
Subsequent work demonstrated that the glycoproteins haemagglutinin-
neuraminidase (HN) and F, which are responsible for virus attachment
and fusion, are able to stimulate the elicitation of neutralizing antibodies
in animals. However, their poor immunogenicity in naïve subjects led to
the currently favoured approach, which is based on the use of live attenu-
ated PIV.
206 Pablo D. Becker and Carlos A. Guzmán

Live attenuated PIV vaccines have been developed from both human
and bovine strains, which are amenable for delivery by the intranasal
route. Candidate vaccines should be able to replicate and induce a protec-
tive immune response in young infants, even in the presence of maternally
acquired antibodies. Two main attenuated strains have been studied in
detail. One is the hPIV-3 strain cp45, which was selected after 45 pas-
sages of the virus in African green monkey cells at low temperature. The
other is a bovine PIV (bPIV)-3 strain, which is antigenically related to the
hPIV-3, and replicates poorly in humans. Both cp45 and bPIV-3 have been
evaluated in phase I/II trials in sero-positive and sero-negative children
and in young infants. They were found to be over-attenuated in sero-posi-
tive children, but immunogenic in sero-negative children and infants [25].
However, the magnitude of the anti-HN response was lower in children
who received the bPIV-3 vaccine [25]. This prompted the engineering of
chimeric bovine/human PIV-3 candidates (e.g., hPIV-Nb strain in which
the human nucleocapsid is replaced by the bovine counterpart, or a bPIV-
3 strain that expresses the F and HN proteins of hPIV-3). Attenuated,
chimeric viruses that contain PIV-3 cp45 internal genes with the F and HN
genes from either PIV-1 or PIV-2 have also been tested in hamsters [26].
Berna Biotech is also developing a virosomal formulation of the PIV-3
[1].

Vaccines against the respiratory syncytial virus

Using the successful approach of the influenza vaccine, a formalin-inacti-


vated candidate against the respiratory syncytial virus (RSV) was tested in
children in the 1960s. The consequence was the hospitalization of 80% of
vaccinees and two deaths [1]. Moreover, vaccinated children also suffered
more severe disease on subsequent exposure to the virus, as compared
to unvaccinated controls [27]. This demonstrated that the elicitation of
a strong immune response is not sufficient to confer protection against
disease, and can even lead to immuno-pathological reactions. Thus, it is
essential to stimulate the “right” type of immune response. In the particu-
lar case of RSV, host responses play an important role in the pathogenesis
of the disease, thereby making the development of a preventive vaccine
extremely difficult. In addition, naturally acquired immunity to RSV is
neither complete nor long-lasting, and recurrent infections often occur
[28]. However, older children and adults are usually protected, suggesting
that protection against severe disease develops after several consecutive
infections. Passive immunization with RSV-neutralizing immune globulins
was also shown to prevent RSV infection in newborns with underlying car-
diopulmonary disease [29]. This demonstrates that antibodies play a major
role in protection against this disease, whereas T-cell immunity targeted to
internal viral proteins appears to contribute to clearance.
New vaccination concepts for CAP 207

Subunit vaccines against RSV

Although live attenuated vaccines seem to be preferable for immunization


of naïve infants, subunit vaccines may be of choice for elderly, high-risk chil-
dren and pregnant women. Candidate subunit vaccines based on purified F
proteins (PFP-1, -2 and -3) were demonstrated to be safe and immunogenic,
even during pregnancy [30]. Maternal immunization using a PFP-based
vaccine could be an interesting strategy to protect infants younger than 6
months of age [25]. However, no significant protection was reported in a
phase III trial performed on children 1–12 years of age with cystic fibrosis
after vaccination with a subunit vaccine based on PFP-3 [31]. A formulation
based on surface glycoproteins F and G together with the virion matrix
protein M from RSV-A was tested in healthy adult volunteers in the pres-
ence of either alum or polyphosphazene as an adjuvant. Short-live neutral-
izing antibody responses to RSV-A and RSV-B were detected in 76–93% of
the vaccinees, suggesting that annual boosting will be needed [32–34]. The
central domain of the G protein of RSV-A is relatively conserved among
viruses from the groups A and B. Thus, a recombinant vaccine candidate,
BBG2Na, was developed by fusing the G2Na domain to the albumin bind-
ing region of streptococcal protein G. This candidate was shown to be mod-
erate immunogenic in adult human volunteers, but its clinical development
was interrupted due to the appearance of purpura in vaccinees [1].

Live attenuated vaccines against RSV

The main two difficulties associated with the generation of live attenuated
vaccines against RSV are over- or under-attenuation of the virus and lim-
ited genetic stability. Temperature-sensitive (ts), ca and cold-passaged (cp)
mutant viral strains have been generated. Despite the attenuation shown in
adults and sero-positive children, cpts mutants still caused moderate conges-
tion in the upper respiratory tract of sero-negative infants (1–2 months old)
[35]. Recombinant RSV vaccines with deletions in non essential genes (e.g.,
SH, NS1 or NS2), which also carry cp and ts mutations in essential genes are
currently being evaluated [1]. Through recombinant DNA technology chi-
meric viruses were engineered, which contain the genes of hPIV-3 surface
glycoproteins F and NH together with those of RSV glycoproteins F and G
in a bPIV-3 genetic background. One of these candidates was found to be
attenuated and able to induce the elicitation of immune responses against
both hPIV-3 and RSV in rhesus monkeys [36]. Similarly, a bPIV-3 genome
was engineered to express hPIV-3 F and HN proteins and either native
or soluble RSV F protein [37]. The resulting strain, which induced RSV
neutralizing antibodies and protective immunity against RSV challenge in
African Green monkeys, needs to be tested for safety and efficacy against
RSV and PIV-3 in infants.
208 Pablo D. Becker and Carlos A. Guzmán

Vaccines against the severe acute respiratory syndrome (SARS)-


associated coronavirus

This emerging disease was originally described in the Guangdong prov-


ince of China in 2002. Even when the global outbreak of SARS was under
control in 2003, new infections were reported in persons who had contacts
with animals in 2003 and 2004 [38]. The typical SARS-CoV-like virus is not
transmitted from animals to humans. However, under certain conditions
the virus can evolve into the early human SARS-CoV, which has the abil-
ity to be transmitted from animals to humans or even humans to humans,
thereby leading to localized outbreaks of mild disease. The early human
SARS-CoV, under selective pressure in humans, may further evolve into
the late human SARS-CoV, which can cause local or global outbreaks of
typical SARS [39].
SARS can be easily grown in cell cultures [38]. Thus, there is an urgent
need for vaccines, not only to prevent naturally occurring epidemic out-
breaks, but also as a tool against the threat of biological weapons. Several
structural proteins are expressed by SARS-CoV, including nucleocapsid,
envelope and spike (S) proteins [38]. The latter is a type I trans-membrane
glycoprotein, which is responsible for virus binding, fusion and entry, and
being the major target of neutralizing antibodies [38, 40]. The extracelullar
domain of the S protein consists of two subunits, S1 and S2 [40]. The S1
subunit possess a receptor-binding domain (RBD), which is responsible for
viral binding to one of its receptors [41, 42]. Vector-based vaccines express-
ing the S protein, as well as DNA vaccines encoding full-length S protein
have been assessed in preclinical studies [43, 44]. When modified vaccinia
virus Ankara (MVA) coding for full-length S protein was administered
by either intranasal or intramuscular route, neutralizing antibodies were
elicited [45]. However, vaccination of ferrets resulted in liver damage after
challenge, raising some concerns about the safety of this approach [46].
Vaccines formulated using different synthetic peptides encompassing lin-
ear B cell epitopes from the S protein, which were identified using sera from
convalescent patients, stimulated high antibody titres. Nevertheless, none
of them triggered the elicitation of neutralizing activity. On the other hand,
some studies demonstrated that although antibodies against S protein of the
late SARS-CoV (Urbani strain) exhibit neutralizing activity, they can also
enhance infection by an early human SARS-CoV isolate (GD03T0013) and
the civet SARS-CoV-like viruses. A derivative of the S protein with a trun-
cation at amino acid (aa) 1153 fails to cause antibody dependent enhance-
ment of infection, but retains the ability to induce neutralizing antibodies.
These findings suggest that the elimination of the putative heptad repeated
2 (HR2, aa 1153-1194), which is implicated in viral fusion, might abrogate
the stimulation of virus infection-enhancing antibodies [47, 48]. The use of
the nucleoprotein of the coronavirus in a DNA vaccination protocol also
led to the stimulation of a protective response [49]. In contrast, protection
New vaccination concepts for CAP 209

was not achieved when a recombinant PIV-3 expressing the nucleoprotein


alone or together with the matrix protein was used [50]. This demonstrates
that the selection of the delivery system and immunization strategy play a
critical role in vaccine efficacy.

Vaccines against adenovirus

The human adenoviruses are divided into six subgroups (A–F). The adeno-
virus can cause large-scale epidemics of acute respiratory disease, and
dissemination is especially favoured under conditions in which persons
are housed communally. The subgroup A viruses, such as Ad31, have been
associated with pneumonia in immunocompromised patients. Neutralizing
antibodies directed against the capsid (hexon and fiber proteins) seems
to be the main effector mechanism to prevent re-infections by adenovirus
[3]. Until 1998, military recruits in USA were administered enteric-coated
capsules containing live viruses from the serotypes 4 and 7. The virus, which
was not attenuated if delivered by respiratory route, was able to replicate
in the gastrointestinal tract without causing disease, thereby stimulating
protective responses in the respiratory tract [51]. When the vaccine went out
of production, outbreaks of respiratory diseases caused by adenovirus re-
emerged among the military recruits [3]. Since serotypes 1, 2, 3 and 5 cause
the 80% of adenovirus associated respiratory disease in young children, the
development of a tetravalent vaccine similar to the above mentioned might
solve the problem in children [52]. However, the implementation of a vac-
cine (live or attenuated) against adenovirus should be carefully evaluated,
since recombinant adenoviruses are proposed both as vaccine vectors and
as tools for the transfer of foreign genes in gene therapy protocols.

Vaccines against Streptococcus pneumoniae

Polysaccharide-based vaccines against S. pneumoniae

In 1945, MacLeod et al. [53] reported the protective efficacy of a capsular


polysaccharide (PS) vaccine in military personnel during an outbreak of
pneumococcal pneumonia. The immunization with purified PS showed a
drastically reduced reactogenicity, in comparison with the previously used
inactivated whole cell vaccines. This was a major breakthrough, not only in
terms of safety, but also because it demonstrated that a specific virulence
factor can be purified and effectively implemented for the prevention of an
infectious disease, thereby paving the road for modern non toxoid-based
subunit vaccines.
Although the serological correlates of immunity are poorly defined,
type-specific anti-capsular antibodies are responsible for protective immu-
210 Pablo D. Becker and Carlos A. Guzmán

nity. However, immunity is serotype specific, rendering extremely difficult


the development of a universal vaccine. This is in part due to the elevate
number of serotypes, the regional variations in dominant serotypes and the
lack of updated sero-prevalence data for certain regions. These problems
have been partially solved by the use of PS-based polyvalent vaccines. The
currently licensed formulations contain 23 serotypes of S. pneumoniae,
which cover approximately 90% of serious pneumococcal disease, but only
in Western industrialized countries. Relatively good antibody responses
(60–70%) are elicited in healthy adults 2–3 weeks after a single intramus-
cular or subcutaneous immunization [54]. Unfortunately, they are poorly
immunogenic in children aged less than 2 years, in immune compromised
individuals (e.g., AIDS patients) and in elderly people with concomitant
disease, and they do not induce good immunological memory. Randomized
controlled trials in healthy elderly and young men also failed to show a ben-
eficial effect against pneumonia [55]. However, vaccination is recommended
for healthy people over 65 years of age to confer protection against inva-
sive disease [54]. PS-based vaccines can be also used in pregnant women to
stimulate the production of antibodies, which are transferred to the foetus
via the placenta or to the newborns by breast-feeding. However, it is still
a matter of controversy whether maternal vaccination can indeed protect
newborns against pneumococcal infections [56].

Conjugate vaccines against S. pneumoniae

The second generation of PS-based conjugate vaccines stimulates stronger


antibody responses, even in infants, young children and immune deficient
individuals, as well as immunological memory. These vaccines also suppress
nasopharyngeal carriage of the pathogen and reduce bacterial transmis-
sion in the community leading to herd immunity, which adds considerable
value to their implementation. The introduction of these vaccines in USA
in 2000 resulted in a dramatic decline in the rates of invasive pneumococcal
disease [1, 57, 58]. A significant reduction in the incidence rates among non
vaccinated individuals was also observed as a result of herd immunity [59,
60]. However, the licensed seven-valent vaccine does not contain some of
serotypes that cause severe disease in developing countries (i.e., serotypes 1
and 5). New conjugate vaccines including more serotypes, such as the nine-
valent vaccine (Wyeth) and two 11-valent vaccines (GlaxoSmithKline and
Sanofi-Pasteur), should provide better serotype coverage.

Protein-based subunit vaccines against S. pneumoniae

New approaches to develop protein-based subunit vaccines against S. pneu-


moniae are currently being pursued by different research groups. This is
New vaccination concepts for CAP 211

expected to enable the generation of a universal vaccine conferring protec-


tive immunity against a large number of serotypes, as well as to avoid the
complexity of manufacturing a conjugate vaccine [61]. There are different
pneumococcal candidate antigens, such as the pneumolysin, neuraminidase,
autolysin, pneumococcal surface protein A (PspA) and adhesin A (PsaA),
which are in an early phase of clinical development [1]. In addition, several
promising candidates have been identified, which are currently being tested
in pre-clinical experimental models [1]. Among them, the two iron uptake
ABC transporters of S. pneumoniae (PiaA and PiuA), which trigger protec-
tive immunity against invasive pneumococcal disease in mice. Through the
screening of S. pneumoniae genomic expression libraries with sera from
convalescent patients, bacterial surface proteins were identified (e.g., BVH-
3 and BVH-11) that promote the elicitation of protective anti-pneumococ-
cal antibodies in mice [1]. A recombinant hybrid protein, BVH3/11V, has
successfully been tested in toddlers and elderly volunteers. This candidate
vaccine should be able to trigger serotype-independent responses, since
the BVH3 and BVH11 antigens are common to all serotypes of S. pneu-
moniae.

Vaccines against typeable and non typeable Haemophilus influenzae

Conjugated Haemophilus influenzae type b vaccines

The major obstacle for developing an effective vaccine against H. influen-


zae capsular PS was related to the inherently poor immunogenicity of this
T-cell-independent antigen. Antibody responses against PS are age-related,
with extremely poor immunogenicity in infants during the first 18 months of
life. Unfortunately, this age group exhibits the highest risk for invasive infec-
tions caused by H. influenzae. A PS-based vaccine against the H. influenzae
type b (Hib) was licensed in the United States in 1985, for children more
than 18 months old [62, 63]. The protective efficacy after licensure studies
showed the inefficacy of this vaccine not only in infants, but also in older
children [64]. This problem was solved by the generation of a conjugate Hib
vaccine. To this end, the Hib PS (i.e., polyribosylribitol phosphate; PRP)
was covalently linked to an immunogenic carrier protein, thereby lead-
ing to T-cell-dependent responses against the PS. Different conjugate Hib
vaccines currently exist. These vaccines are HbOC, PRP-T and PRP-OMP,
which make use of the mutant diphtheria toxin CRM197, the tetanus toxoid
and the outer membrane protein from group B N. meningitidis as carriers,
respectively. All of them trigger similar immune responses at the recom-
mended doses. However, the dynamic of the elicited response may vary for
each of them [65, 66].
Efficacy studies of these vaccines showed that they confer protection
not only against meningitis, but also against pneumonia [67–69]. Although
212 Pablo D. Becker and Carlos A. Guzmán

Hib vaccines are highly effective, their cost is still prohibitive for the world’s
poorest nations. However, with the establishment of the Global Alliance for
Vaccines and Immunization (GAVI), we have moved consistently ahead in
making them also available for developing countries. GAVI has approved
the establishment of a Hib initiative to support countries wishing to sustain
Hib vaccination, as well as those exploring whether their introduction could
be considered a priority in the near future.

H. influenzae typeable and non typeable: vaccination perspectives

Although the introduction of conjugated PS vaccines has significantly


decreased the prevalence of invasive Hib disease, paediatric infections due
to non typeable H. influenzae (NTHi) are still highly prevalent. NTHi is
most often associated with otitis media, sinusitis and bronchitis. In addition,
NTHi is an important cause of lower respiratory infection in adults with
chronic obstructive pulmonary disease (COPD). Thus, the development of
a vaccine against NTHi is considered an important goal in public health. In
contrast to Hib, vaccines against the non-encapsulated NTHi strains must
be directed against alternative virulence factors. The lipoproteins D and
P6 are widely distributed and antigenically conserved among H. influenzae
strains, and also trigger the elicitation of protective immunity in animals
vaccinated by mucosal route [70–73]. Thus, their incorporation in vaccine
candidates might facilitate the generation of a universal vaccine against all
typeable and non typeable H. influenzae.

Vaccines against Bordetella pertussis

Even in the age of vaccine availability, B. pertussis continues to be a major


cause of childhood morbidity and mortality (i.e., approximately 50 million
cases and 300,000 deaths occur annually worldwide). Since the late 1940s,
the incidence of whooping cough has dramatically decreased in most devel-
oped countries, as a result of widespread immunization. The first vaccine
formulations, which are still in use, consist of preparations based on killed
B. pertussis. The frequent incidence of minor adverse effects (e.g., fever,
protracted crying and local erythematous reactions), as well as concerns
raised by reports of serious neurological side-effects, resulted in a decline
in vaccine acceptance and use [74]. This in turn led to a re-emergence of
whooping cough and its complications. This serious problem prompted the
development of a new generation of acellular vaccines.
In 1981 Japan was the first country to successfully introduce acellular
vaccines against whooping cough in its immunisation programme [75], lead-
ing to a consistent reduction in the reported side-effects. In the mid 1980‘s
a major phase III trial of acellular vaccines was undertaken in Sweden, at a
New vaccination concepts for CAP 213

time when the banning of the whole cell vaccine had resulted in a pertussis
epidemic in that country [76]. The first vaccine trials contained chemically
detoxified pertussis toxin (PT) and filamentous haemagglutinin (FHA), or
detoxified PT alone. The results of these trials showed that whilst producing
good antibody responses, the vaccines failed to give an adequate level of
protection in infants. The mono-component vaccine conferred no protec-
tion against infection, whereas the use of the two component candidate only
gave incomplete protection against infection [77]. The results obtained in
Japan and Sweden stimulated vaccine companies in the USA and Europe
to establish vigorous research programmes aimed at the development of a
new generation of acellular vaccines with higher efficacy. Currently avail-
able vaccines have incorporated chemically or genetically inactivated PT
and additional virulence factors, such as FHA, the outer membrane protein
pertactin (PRN) and fimbrial proteins (FIMs).
The efficacy studies of this second generation of acellular vaccines have
demonstrated that they confer levels of protection equivalent to the whole
cell vaccines. The advent of improved techniques for antigenic characterisa-
tion and the introduction of acellular vaccines containing genetically defined
components also resulted in a reduction of lot-to-lot variation in compari-
son with conventional whole cell vaccines and the acellular formulation
originally introduced in Japan. However, despite the wide implementation
of vaccination campaigns in infants and children, the disease continues to
be endemic. In addition, in countries with high vaccine coverage we are now
observing a consistent increment in the cases of pertussis in adolescents
and adults [78–80]. These patients can then transmit the disease to infants,
thereby now representing a primary reservoir for bacterial transmission and
cycling in the community.
The above-mentioned observations can be explained by one or more of
the following factors: (i) improved detection techniques, (ii) major aware-
ness on the possibility that bacteria may affect these age groups, (iii) vaccine-
driven antigenic changes in circulating isolates, and (iv) reduction in vaccine
efficacy over time. In this context, concerns have been raised about genetic
variation between the strains used for vaccine preparation and circulating
isolates. This seems to be true, since the currently used whole cell and acellu-
lar vaccines are prepared with strains that were isolated before mass vaccine
introduction and show clear mismatches with respect to circulating strains.
There is a steady tendency to decrease diversity in recent isolates, together
with clonal expansion during epidemic outbreaks [81, 82]. Over time, at
least two surface proteins (PT and PRN) may have changed sufficiently to
allow for an increase in the incidence of disease. Unfortunately, our global
information on antigenic variation and disease in adults and adolescent is
extremely limited. Thus, despite widespread introduction of pertussis vac-
cines, it is essential to continue surveillance studies and collection of circu-
lating strains. The present view is that successful control of pertussis in the
community may require routine immunization of adolescents and adults
214 Pablo D. Becker and Carlos A. Guzmán

with the new acellular vaccines, perhaps in combination with the diphtheria
and tetanus toxoids (DTaP). This intervention might help in turn to reduce
the burden of disease and transmission to infants.

Vaccines against Chlamydia pneumoniae

Chlamydia pneumonia is an intracellular bacterium transmitted person-to-


person via respiratory droplets. This pathogen is a common cause of pneu-
monia, with infections usually being oligosymptomatic or asymptomatic in
young age groups. However, the rate of asymptomatic carriage in the nor-
mal population is unknown. There is also a tremendous gap in our under-
standing of host response to infections caused by C. pneumoniae. Most of
the studies have been focused on the development of efficient diagnostic
methods. However, less work has been done on vaccine development, and
there is a paucity of knowledge on the microbial components which may
serve as target antigens. In fact, at present there are no licensed vaccines
against C. pneumoniae. However, the potential of different antigens, such as
the major OMP2 [83] have been assessed in experimental animal models.
Nevertheless, mice vaccinated with OMP2 using a protocol based on prim-
ing with DNA and boosting with recombinant VLP showed only partial
protection [84]. Recent studies also suggested that CTL responses play a
role in protection and clearance [85]. Animals immunized with a mini-gene
encoding seven H-2(b)-restricted CTL epitopes fused to a endothelial retic-
ulum-translocation signal showed protection following intranasal challenge
with a virulent C. pneumoniae [85].
The current view is that multi-component vaccine will be required in
order to induce a protective response [86]. Using the promising approach
of reverse vaccinology combined with proteomics (see section “Reverse
vaccinology”), the whole-genome of C. pneumoniae was screened search-
ing for vaccine candidate antigens among exposed and immune accessible
surface proteins [87]. The selected candidates were then expressed in a
heterologous system and used in immunization studies. Approximately 53
proteins were able to trigger the elicitation of C. pneumoniae-binding anti-
bodies. When tested in secondary screenings, six of them were also able to
neutralize bacteria in vitro, and four inhibited systemic dissemination of C.
pneumoniae in a hamster model [86].

Vaccines against Moraxella catarrhalis

Moraxella catarrhalis is the third most common bacterial etiologic agent


of otitis media in children. Furthermore, M. catarrhalis is an important
cause of respiratory infections in patients with COPD. Thus, different stud-
ies have been carried out to characterize potential protective antigens. In
New vaccination concepts for CAP 215

this context, two major OMP (CD and E) have been identified, which are
considered prime candidate antigens for vaccine development. These pro-
teins are expressed on the surface and show a high degree of conservation
among circulating strains. Both OMP triggered the elicitation of bactericidal
antibodies and protective immunity in preclinical models [88]. Additional
candidates are the UspA1 protein [89], which seems to be required for bac-
terial colonization of the human upper respiratory tract, the iron-induced
OMP B1 and LBP, and the iron-repressed OMP B2 [90]. A conjugate vac-
cine based on detoxified lipo-oligosaccharide was also tested in mice by
intranasal route with encouraging results [91, 92]. Some of these candidates
are planned to be tested in clinical studies soon [90].

Vaccines against Mycoplasma pneumoniae

Mycoplasmas are commensal microorganisms, as well as opportunistic


pathogens. Mycoplasma pneumoniae is one of the causative agents of acute
and chronic human respiratory diseases and the main responsible for prima-
ry atypical pneumonia, accounting for approximately 20–30% of all commu-
nity-acquired pneumonia [93]. There is a considerable underreporting for
M. pneumoniae-associated diseases. This is in part due to the wide diversity
of clinical manifestations, the difficulties associated with its cultivation from
clinical specimens and the lack of adequate diagnostic tools. No vaccines
are currently available against this pathogen. However, studies conducted
in human volunteers in the late 1960s demonstrated that a formalin-inacti-
vated whole cell vaccine and an acellular extract were able to confer mod-
erately protective immunity against M. pneumoniae [94]. Unfortunately,
immune pathological reactions were observed following challenge with live
organisms. Therefore, studies are still needed to understand the underlying
mechanisms to the observed autoimmune responses [95]. More specifi-
cally, we need to elucidate the specific role played by humoral and cellular
response in protection against M. pneumoniae. M. pneumoniae is one of the
smallest self-replicating prokaryotic pathogens (approximately 800 kb). The
complete genome sequence is now available. This is expected to expand our
knowledge on the physiological and virulence properties of this agent, as
well as new hints for vaccine development.

Vaccines against Legionella pneumophila

A previously unrecognized bacterium was isolated after the outbreak of


Legionnaires disease in 1976, which was designated Legionella pneumophila
[96, 97]. The spreading of L. pneumophila is increasing due to the use of
air-conditioners and humidifiers, since infections can occur by inhala-
tion of aerosolized contaminated water sources. Several approaches have
216 Pablo D. Becker and Carlos A. Guzmán

been developed in the fight against this facultative intracellular pathogen.


Infection and immunization induce a rapid increase of antibody titres.
However, antibodies do not seem to play a significant role in host resistance,
particularly after aerosol challenge [98–100]. Some authors also suggested
that these antibodies can promote bacterial phagocytosis, thereby favour-
ing invasion and subsequent intracellular replication [101]. In contrast,
cellular responses appear to be important for protection. Different vaccine
candidates were tested in the past. Heat-, acetone- and formalin-killed
L. pneumophila vaccines were not able to confer protective immunity in
guinea pigs, whereas animals immunized with L. pneumophila membranes
survive an aerosol challenge with virulent bacteria [98, 99]. Additional work
demonstrated that also purified antigens, such as the major secretory pro-
tein [98], the major cytoplasmatic membrane protein [102], the peptidogly-
can-associated lipoprotein [103], OmpS [104] and flagella [100] can confer
protection against challenge with virulent L. pneumophila. Finally, different
live attenuated mutants of L. pneumophila were used in animal infection
models with promising results [105].

Vaccines against Pseudomonas aeruginosa

Cystic fibrosis (CF) patients are particularly susceptible to severe bacte-


rial infections of the lung, being Pseudomonas aeruginosa one of the most
prominent etiologic agents. Thus, significant efforts have been invested to
develop a vaccine against this pathogen. Surface PS are among the anti-
gens that were most intensively assessed. Berna Biotech have developed
an octavalent vaccine against the eight most prevalent serotypes based on
O-PS conjugated with the exotoxin A [106–113]. A consistent reduction in
the number of CF patients with chronic P. aeruginosa lung infection was
observed in a cohort receiving the basic immunization protocol, followed by
yearly boosters over a period of 10 years [112, 113]. The conjugate vaccine
induced the production of specific IgG antibodies and increased the number
of IgG memory B cells. It is still unclear if cellular responses might contrib-
ute to the overall protection conferred by this vaccine. However, strong
proliferative responses of lymphocytes with a Th1 phenotype were observed
in vaccinated individuals in response to the carrier exotoxin A protein [113].
Alternative vaccination strategies are currently being tested in clinical tri-
als. Among them, formulations based on a fusion protein between the outer
membrane proteins F and I, which have been administered by parenteral
and mucosal routes [114, 115]. These formulations were demonstrated to
be safe in volunteers and conferred increased protection against P. aerugi-
nosa in CF patients. Cell-surface alginate, flagella, components of the type
III secretion system, inactivated toxins and proteases are other proposed
target antigens [116]. Some of them are already in clinical trials alone or in
combination [116].
New vaccination concepts for CAP 217

New advances in vaccinology

When Pasteur returned from his summer holidays in 1881 to continue with
his studies on chicken cholera, he inoculated chickens with an old culture
of Pasteurella multocida, which was left during the whole summer on his
bench. The animals that received the preparation were protected against
a challenge performed with a fresh isolate. Thus, Pasteur developed the
hypothesis that pathogens could be attenuated by exposure to environmen-
tal insults (e.g., high temperature, oxygen and chemicals) [117]. The strategy
was then successfully extrapolated for developing anthrax vaccines in live-
stock in the 1880s, with significant economic benefits. This was followed by
the generation of attenuated vaccines against rabies and other important
pathogens towards the end of the nineteenth century. Pasteur’s approach
for “attenuating” or “inactivating” a pathogenic organisms still constitutes a
cornerstone in vaccine technology [117]. This exemplifies that until recently
the major achievements in vaccinology have been facilitated by technologi-
cal (e.g., adjuvants, delivery systems, reverse vaccinology, genetic engineer-
ing) rather than immunological advances [117–119]. However, it is expected
that the impressive knowledge accumulated in recent years in the fields of
immunology, immune pathology and microbial pathogenesis will pave the
road to a new golden era in vaccinology, in which knowledge and technol-
ogy will enable rational vaccine design.

New technologies and approaches for vaccine development

Reverse vaccinology

In the 20th century, pertussis vaccines progressed from crude bacterial


preparations to the highly purified antigens used for acellular vaccines. A
similar quantum jump in technology allowed the development of subunit
vaccines against influenza, Hib and S. pneumoniae, as well as the production
of antigens by recombinant DNA techniques (e.g., genetically inactivated
PT). Despite the fact that these techniques enable the production of almost
any foreseeable antigen, the identification of suitable targets still remained
as a main bottleneck for vaccine development [120].
The advent of genomics and its exploitation in the vaccinology field have
rendered possible the implementation of a systematic and holistic approach
for the screening, identification and prioritisation of candidate antigens.
This new approach, called “reverse vaccinology” [121], does not require
cultivation of the original pathogen, thereby being amenable for highly-
pathogenic or non culturable micro-organisms. It is possible to predict and
select the most promising candidates by the analysis of genomic sequences
in silico, which will then be cloned and expressed in heterologous systems.
The resulting proteins are then used to perform immunological and/or
218 Pablo D. Becker and Carlos A. Guzmán

functional studies to select the most promising candidates (e.g., able to


induce the production of microbicidal or neutralizing antibodies, capacity to
confer protective immunity). Flanking studies are usually carried out, such
as molecular epidemiological analysis to assess their degree of conserva-
tion among circulating strains, or transcriptional profiling to evaluate their
expression during natural infections [122].
The time-consuming process in which highly expressed components of
an in vitro cultivable organism are identified (one at a time) and separated
(different components between them) is one of the disadvantages that
reverse vaccinology has solved. The conventional method usually requires
15–20 years to arrive to a clinical trial, whereas reverse vaccinology reduces
the process to approximately 5 years. Reverse vaccinology also allows the
identification of hundreds of potential candidates in a few days, in compari-
son with the small number of antigens that conventional approaches have
provided after decades of research. Moreover, reverse vaccinology offers
the possibility to select potential candidates independent of their expression
levels or purification easiness.
The reverse vaccinology approach has proved its usefulness in the field
for both viral and bacterial pathogens (e.g. hepatitis C virus, Group B
meningococci, group B streptococci) [123, 124]. Reverse vaccinology has
also become an essential tool for several vaccine development projects
against agents causing community-acquired pneumonia (e.g., C. pneumoni-
ae, streptococci). The potential and speed of genomic-based approaches
was also shown when the nucleotide sequence of the coronavirus causing
SARS was made available in less than one month. In addition, the increas-
ing number of available genomes from bacteria and viruses would allow
comparative genomic studies, thereby providing hints on conserved protein
families and/or functional domains. This would facilitate the generation
of vaccines using immunogens covering multiple micro-organisms [125].
Despite the incredible potential of reverse vaccinology, this approach also
has some important limitations (Tab. 1). Among them is the fact that it is
not be possible to identify non-protein antigens (e.g., PS, glycolipids), which
are the cornerstone for many successful vaccines (e.g., pneumococcal and
Hib vaccines).

Reverse genetics

Currently available influenza vaccines (see above) are based on inacti-


vated viruses, and, more recently, attenuated ca viruses and virosomes. All
these vaccines exploit the same starting material (wild-type virus), which
is inactivated or attenuated. The last approach consists in the co-infection
of chicken eggs with the new isolate and a master attenuated strain, and
subsequent selection for re-assorted viruses with the desired genotype/phe-
notype. However, the virulence of certain virus strains, such as the H5N1,
New vaccination concepts for CAP 219

Table 1. Classical vaccine development versus reverse vaccinology

Reverse vaccinology Classical approach


Time required to reach the clinical ~ 5 years ~ 20 years
phase
Type of organisms Culturable and non- Only culturable
culturable
Antigens Only proteins Proteins, lipoproteins,
polysaccharides and gly-
colipids
Genome Necessary Unnecessary
Target genes All Mainly in vitro expressed
Exclusion of known antigens Yes No
Need to handle microorganisms No Yes
(e.g., highly pathogenic)
Surface and structural antigens Yes (only proteins) Yes
Internal antigens Yes (only proteins) Rarely
Antigens with low expression Yes Rarely selected
levels
Number of candidate antigens Many (more than Few
hundred)
Selection of antigens Poorly or highly Mainly highly immunogenic
immunogenic
Antigens identification and Easy Could be difficult
separation
Need for genetic tools during the Not necessary Usually essential (e.g., to
initial discovery process create and complement
mutants)
Need for genetic tools for antigen Necessary for the initial Only in a late phase of
expression phase of development development

renders difficult the implementation of this traditional strategy. The use of


reverse genetics represents a valid alternative for the generation of vaccines
against RNA respiratory viruses, such as the influenza virus, PIV and RSV.
It consists in the production of the virus from cloned DNA [126], thereby
allowing the development of vaccines against any pandemic viral strain.
In some cases (e.g., avian H5N1) an additional mutagenesis step would be
required to attenuate its virulence [127]. Then, the new HA and NA seg-
ments would be transferred into an appropriate influenza A virus master
strain adapted to grow in a cell line. The final re-assorted virus will have the
antigenic specificity of the pandemic strain and the growth characteristics
of the master strain [128, 129].
This technology would also allow production of the influenza vaccine
in cells that are co-transfected with plasmids encoding for different frag-
220 Pablo D. Becker and Carlos A. Guzmán

ments of the virus [130]. Therefore, the complete genome is inside the cell
and virus can be produced and assembled. One of the main advantages is
that a plasmid encoding for HA and NA can be easily replaced. Therefore,
re-assortment and selection become unnecessary. This method would con-
siderably reduce the time for vaccine production, from many months to
only a few weeks. Another advantage would be the simple manipulation of
the genome (contained in plasmids), which would enable detoxification of
specific virulence factors. Similar approaches can be implemented for other
viruses, such as RSV, PIV and SARS-CoV. However, intellectual prop-
erty and liability issues are still obstacles for the industrial development
of reverse-genetics-based vaccines [131]. Furthermore, since the resulting
viruses are considered genetically modified organisms, additional problems
may arise from the regulatory stand point [131].

Mucosal delivery systems

Most of the infective agents are either limited to the mucosal membranes, or
need to transit across them in order to cause disease. Therefore, it is highly
desirable to elicit an efficient immune response at the local site in which the
first line of defence is laid. The stimulation of a pathogen-specific response
at the portal of entry is expected to impair infection (i.e. colonization),
thereby reducing the risk of transmission to susceptible hosts. Parenterally
administered vaccines mainly stimulate systemic responses, whereas vac-
cines given by the mucosal route mimic natural infections, thereby leading
to efficient mucosal and systemic responses. Thus, there is a considerable
interest in the development of mucosal vaccines. However, antigens admin-
istered by this route are usually poorly immunogenic. Different strategies
are being pursued to overcome this bottleneck, among them can be cited the
use of (i) advanced synthetic delivery systems, (ii) live attenuated bacterial
or viral vectors, (iii) bacterial ghosts, (iv) pseudoviruses and (v) mucosal
adjuvants [132–135].

Advanced synthetic mucosal delivery systems


Particulate antigens are more immunogenic than those in solution, due
to their vulnerability to degradation by enzymes and extreme pH. Thus,
it would be helpful to incorporate them into a protective vehicle. Often,
these vehicles do not serve only to protect them, but can also enhance
their uptake, promote targeting to antigen presenting cells and serve as
adjuvants [136]. The most commonly exploited delivery systems are: (i)
gelatine capsules, which are dissolved at alkaline pH in the intestine but
not in the stomach, (ii) muco-adhesive polymers that are highly viscous
inert PS, (iii) eldexomer and carboxymethyl cellulose, which have been
used for oral, nasal and vaginal delivery, (iv) lipid-based structures with
New vaccination concepts for CAP 221

entrapped antigens, such as immune stimulating complexes (ISCOMs)


and liposomes, and (v) biodegradable micro/nano-spheres based on bio-
compatible materials such as starch, copolymers of lactic or glycolic acid
[137, 138]. Some of these approaches are currently being explored to
develop vaccines against agents causing community-acquired pneumonia.
Encouraging results have been obtained, among others, using surface
antigens from S. pneumoniae encapsulated in micro-spheres [139] and a
ISCOM-adjuvanted vaccine obtained by reverse genetics against the influ-
enza virus, in preclinical models [140].

Live attenuated bacterial or viral vectors


Attenuated viruses and bacteria can be used not only as vaccine candidates
per se, but also as delivery systems for heterologous antigens. Thus, many
attenuated microorganisms have been exploited as a scaffold for the devel-
opment of subunit vaccines against other agents, under the premise that the
expression of the recombinant antigen(s) does not increase their pathogenic
potential for humans or animals. The most frequently exploited bacterial
vectors are attenuated derivatives of Salmonella enterica and Shigella spp.,
and the Bacille Calmette-Guérin (BCG). For example, vaccination with an
attenuated Salmonella expressing the OprF-OprI was also shown to be able
to confer protection against P. aeruginosa in a murine experimental infection
model [141]. In addition, it was also demonstrated that a recombinant BCG-
based vaccine expressing the PspA confers protection against S. pneumoni-
ae in an infection animal model [142]. The use of commensals represents an
alternative to attenuated organisms (e.g., lactobacilli). In this context it was
demonstrated that oral administration of Lactobacillus expressing proteins
from coronavirus can protect against a gastric infection [143]. Thus, this
approach has been also proposed to combat SARS. Promising results were
also obtained using x Chlamydia psittaci [144]. On the other hand, different
attenuated viruses, such as MVA, bovine or attenuated hPIV-3 and adeno-
virus can be used as delivery systems for heterologous antigens [25, 145]. In
fact, MVA has recently been exploited for antigens of the SARS associated
coronavirus [146].

Bacterial ghosts
An alternative approach to the use of live attenuated carriers is given by the
use of bacterial ghosts. Ghosts are generated by the conditional expression
of the lethal lysis gene E from bacteriophage PhiX174 in Gram-negative
bacteria [147–151]. This leads to the formation of a trans-membrane tun-
nel through the bacterial cellular envelope [147]. Due to the high internal
osmotic pressure, the cytoplasm content is expelled through the tunnel,
thereby leading to an empty bacterial cell envelope [152]. The presence of
envelope components in the ghosts provides a strong danger signal through
the activation of pattern recognition receptors [153]. In addition, bacterial
222 Pablo D. Becker and Carlos A. Guzmán

ghosts are efficiently taken up by antigen-presenting cells, stimulating their


maturation and activation [154].
Bacterial ghosts retain all morphological, structural, and antigenic fea-
tures of the cell wall and can be used as vaccine candidates per se. Ghosts
can also be externally loaded with purified antigens. Alternatively, ghosts
can be generated from recombinant bacteria expressing heterologous anti-
gens, hence avoiding the difficulties associated with the purification steps.
This technology also offers the possibility to manipulate the topology of
the recombinant antigen (e.g., the antigen can be bound to the inner mem-
brane, secreted into the periplasmic space or associated to the surface).
Encouraging results has been obtained in preclinical models using ghosts
expressing chlamydial antigens [135, 155].

Pseudoviruses as antigen delivery systems


Promising results have been reported using different types of pseudoviruses,
such as virosomes and virus-like particles (VLP), which are non-replicating
viral-like structures. Virosomes are based on the principle of reconstitut-
ing empty viral envelopes through integration of viral envelope proteins in
liposomes. They offer the versatility of liposomes in terms of lipid composi-
tion, with the advantage of including viral membrane proteins. Virosomes
are produced by disassembling the viral membrane envelope with deter-
gents. Then, the viral nucleocapsid is removed by ultracentrifugation before
reconstitution (Fig. 1). In contrast, VLP exploit the capacity of recombinant
viral coat proteins to spontaneously self-assemble, thereby mimicking at
structural level the viral capsid. VLP can be isolated after protein expres-
sion in eukaryotic cells or by in vitro assemblage from subunits produced
in an heterologous system [156]. Their main advantages are the lack the
viral genetic material with an “intact” envelope, and the fact that they are
significantly more immunogenic than soluble proteins. They can be used
as vaccines per se, as well as a delivery system for protein- or nucleic acid
based vaccines, or as carriers for small molecules. Foreign antigens can
be expressed on their surface, or can be simply encapsulated. In addition,
amphiphilic adjuvants can be incorporated into their membranes, thereby
offering the advantage of combining an adjuvant and the antigen in one
entity without a covalent attachment.
Pseudoviruses are especially attractive for mucosal vaccination proto-
cols, since they offer the opportunity to use the natural route of transmis-
sion of the agents. Induction of serum antibodies, secretory IgA, T helper
and CTL responses, and protection against mucosal pathogen challenge
has been reported from studies in animals and humans [157–159]. The
virosomes generated using the influenza virus retain membrane fusion
properties very similar to the naïve virus. Therefore, they are able to deliver
material to the cytosol of target cells, offering the possibility to access the
MHC class I-restricted pathway of antigen presentation to prime CTL activ-
ity [160–162].
New vaccination concepts for CAP 223

Figure 1. Virosomes are reconstituted viral envelopes, which incorporate the cell binding and
fusion proteins of native virus without its viral genetic material. (a) Virosomes are produced
by disassembling the viral membrane envelope with detergents. (b) The viral nucleocapsid is
then removed by ultracentrifugation, and (c) they are reconstituted by removing the detergent
with or without addition of lipids. (d) Electron-microscopy of an influenza virosome kindly
provided by Etna Biotech.
224 Pablo D. Becker and Carlos A. Guzmán

Mucosal adjuvants

Bacterial toxins and their derivatives are among the first molecules that
have been used as mucosal adjuvants. They are characterized by the pres-
ence of an A moiety with enzymatic activity, and a B moiety that medi-
ates toxin binding to the target cells. Cholera toxin and the closely related
Escherichia coli heat-labile toxin showed potent adjuvant activity when
co-administrated with different antigens by the mucosal route [163–165].
However, their use in humans is hampered by their intrinsic toxicity. Thus,
mutated derivatives were developed, in which the A subunit was modi-
fied to remove the ADP-ribosylating activity. The resulting polypeptides
retain their adjuvanticity, in the absence of detectable toxicity [166–168].
However, additional studies have demonstrated that even these deriva-
tives can lead to potential severe side-effects, such as retrograde homing
of adjuvant and antigen to neural tissues [169]. This might explain, at least
in part, the side-effects observed after intranasal vaccination against influ-
enza with a virosomes-based formulation containing heat-labile toxin (i.e.,
Bell’s palsy), which in turn led to its retraction from the market. However,
chimeric derivatives lacking the targeting moiety for neural tissues (i.e., B
subunit) are now available [170]. They might allow the exploitation of the
high potential of these molecules for the development of vaccines against
respiratory pathogens. In fact, preclinical studies provided the proof-of-con-
cept for the usefulness of derivatives of bacterial toxins in the generation
of acellular vaccines against microorganisms, such as S. pneumonia and H.
influenzae [171, 172].
Other bacterial components were also explored for their activity as adju-
vants. The monophosphoryl lipid A retains much of the immune stimulatory
properties of LPS, without the inherent toxicity [165]. On the other hand,
extracellular matrix binding proteins, such as the fibronectin binding protein
I of Streptococcus pyogenes, also exhibit adjuvant activity [173]. This offers
the possibility of using them as dual antigen/adjuvant moieties in the same
formulation. Recent reports also demonstrate that vaccine formulations
containing adamantylamide dipeptide, a non-toxic compound obtained by
linking the L-alanine-D-isoglutamine residue of the muramyl dipeptide to
the antiviral drug amantadine, confer protection against non typeable H.
influenzae in preclinical models [73].
The innate immune system plays a critical early role in host defence
against pathogenic microorganisms through the recognition of pathogen-
associated molecular patterns [174]. This is achieved through the stimula-
tion of pattern-recognition receptors (PRR) that sense a broad range of
exogenous and endogenous danger signals [153, 174]. Toll-like receptors
(TLR) represent the best-characterized family of PRR. Natural and syn-
thetic TLR agonists are being used as immune modulators to optimize
responses after vaccination. Since the identification of the TLR4, many
mammalian TLR homologues have been identified (i.e., 10 in humans
New vaccination concepts for CAP 225

Table 2. Toll-like receptors (TLR) and their ligands

TLR Ligands
TLR1 (with TLR2) Mycobacterial lipoprotein (LP), triacylated lipopeptides, lipotei-
choic acid (LTA)
TLR2 LPS (P. gingivalis), fungal products (zymosan), peptidogy-
can (PGN), LP, GPI anchors (T. cruzi) , lipoarabinomannan,
muramyl dipeptide
TLR3 Viral dsRNA, synthetic Poly (I:C)
TLR4 Gram-negative bacterial products, LPS, respiratory syncytial
virus, synthetic lipid A, E5564, plant products, saturated and
unsaturated fatty acids, murine ß-defensin 2, BCG
TLR5 Flagellin
TLR6 (with TLR2) Mycoplasma LP, LTA, PGN, diacylated LP
TLR7 GU-rich ssRNA, resiquimod, imiquimod
TLR8 GU-rich ssRNA, resiquimod, imiquimod
TLR9 Bacterial and viral DNA, unmethylated CpG-ODN
TLR10 Unknown
TLR11 (in mice) Components from uropathogenic E. coli, and profiling-like from
Toxoplasma gondii

and 13 in mice) [175]. Each TLR member binds specifically to different


ligands (Tab. 2), alone or in combinations (e.g., heterodimers formed by
TLR2 with either TLR1 or TLR6). An example of TLR agonist is bacte-
rial DNA, but not vertebrate DNA, and synthetic oligodeoxynucleotides
containing unmethylated CpG motifs. They act on TLR9, thereby induc-
ing a strong Th1 responses by activation of dendritic cells [176, 177]. CpG
motifs have been successfully used as adjuvants in preclinical studies of
different candidate vaccines against agents causing community-acquired
pneumonia [178-180].
Another important adjuvant with TLR-binding capacities is the
Mycoplasma-derived macrophage-activating lipopeptide MALP-2, which
act a the level of the TLR heterodimer 2/6 [181, 182]. MALP-2 promotes a
global activation of cells from the innate and adaptive immune system [183,
184], such as macrophages, DC, T- and B-lymphocytes [183, 185]. When co-
administered with an antigen by either the parenteral or the mucosal route,
MALP-2 promotes the elicitation of humoral and cellular responses at
systemic and mucosal level [186]. Preclinical studies suggested that MALP-
2 could be exploited in vaccine formulations against the SARS-associated
coronavirus, M. catarrhalis and influenza virus, among others (unpublished
data).
226 Pablo D. Becker and Carlos A. Guzmán

DNA vaccines

DNA vaccination offers some advantage over the normal antigen vacci-
nation, such as the fact that it is not necessary to express any antigen. In
contrast, it is the biosynthetic machinery present in the cells of the vac-
cinees that takes care of this work. Furthermore, since eukaryotic cells are
in charge of protein synthesis, their glycosylation and folding are optimal.
However, the large-scale purification of DNA might be associated with high
costs. This can be solved by the use of attenuated or inactivated bacteria or
viruses as delivery systems [187]. This approach can also lead to an enhanced
induction of antibodies, which is otherwise poor using conventional naked
DNA vaccines. We have recently demonstrated that bacterial ghosts can be
also exploited as a delivery system for DNA vaccines for both in vivo and
ex vivo applications [188].
The potential of this approach is demonstrated by the fact that it is pos-
sible to optimize performance by a broad range of manipulations, such as (i)
choice of optimal promoters, (ii) use of codon optimized genes for expres-
sion in mammalian cells, (iii) addition of nuclear localization signals or ubiq-
uitination signals to improve expression and processing, and (iv) co-delivery
of DNA constructs coding for immune modulatory molecules [189]. In addi-
tion, by the presence of immune stimulatory CpG motifs, the DNA vaccine
constructs has built-in adjuvant properties. This vaccination approach is
particularly suited for the stimulation of cellular immune responses [190].
Interestingly, several reports suggest that DNA vaccines may represent a
valid alternative to prime the neonatal immune system, even in the pres-
ence of passive transferred maternal antibodies [191, 192]. In fact, promis-
ing results were also obtained in preclinical models of community-acquired
pneumonia, such as influenza [193] and S. pneumoniae [194]. Furthermore,
DNA coding for vaccine antigens appears to induce excellent immunologi-
cal memory, which can be reawakened by later immunization or exposure
to the pathogen.

New immunological concepts that need to be addressed to optimise


vaccine design

The knowledge generated in several basic disciplines, such as immunology


and microbial pathogenesis, has allowed the identification of critical bottle-
necks for establishing a successful vaccination strategy. It is expected that
in the coming years we will develop customized approaches to address each
of them, in order to stimulate efficient protection against infective agents
under specific clinical settings (i.e., newborns, aging individuals, immuno-
compromised patients).
New vaccination concepts for CAP 227

The importance of immunological memory

B lymphocytes that have differentiated into plasma cells are the producers
of antigen-specific IgG antibodies. Bone-marrow (BM) plasma cells have a
short life, therefore, the BM reservoir needs to be replenished by the stimu-
lation of memory B cells [195, 196]. The maximal life span of BM plasma
cells is still debated. Only few factors have been identified that control the
differentiation of antigen-specific B cells toward short- or long-life plasma
cells or to memory B cells [119]. Beside the requirement of CD4 + T cells, the
nature of the antigen [197] and the dose are also important. Higher antigen
doses, as well as rapid vaccination schedules (closely spaced vaccine doses)
tend to favour the rapid induction of short-term effectors, whereas lower
doses of antigens preferentially support the induction of immune memory
[198-201].
It was demonstrated that neonatal vaccination (priming) and infant
boosting might be effective even when pathogen exposure occurs very early
in life. In children in whom vaccine-induced Hib antibody titres have fallen
to undetectable levels, memory is readily demonstrated [202]. However,
immune memory per se is not enough to protect against pathogens that
required high levels of neutralizing antibodies. The delay between memory
B-cell reactivation and differentiation may limit the ability to interrupt
pathogen invasion. Therefore, it is important to establish vaccination proto-
cols in which the population is boosted at different ages in order to maintain
the required levels of antibodies. This is particularly important in diseases in
which antibodies play a central role in microbial clearance or toxin neutral-
ization. In the particular case of community-acquired pneumonia, we should
consider that aging individuals are neglected in many vaccination programs.
However, the strategies proposed for elderly would be different from those
used for small children, since the main factors affecting vaccine efficacy are
immune senescence and immaturity, respectively. The attempts to give a
rational solution to this issue are discussed in the next sections.

The immune system in children and elderly

The immune system in children


Immune responses to bacterial and viral antigens usually increase with age
in a stepwise manner [203]. Prompt immunization after birth is required
to induce active immunity against diseases that may occur early in life.
Unfortunately, this strategy is limited by the relative immaturity of the
neonatal and infant immune system. Some factors implicated in this poor
response are the limited switch from IgM to IgG2 antibodies, impaired
complement-mediated reactions and deficient organization of the splenic
marginal zone. Vaccination studies performed in newborn mice suggested
that limited germinal centre reactions may results from the delayed devel-
228 Pablo D. Becker and Carlos A. Guzmán

opment of follicular DC and limit plasma cell differentiation [204]. It was


also showed that the neonatal BM has a limited capacity to support the
establishment of long-life antibody-secreting plasma cells [205].
Thus, the responses to glyco-conjugates and to most T-cell-dependent
antigens are usually affected [119]. Therefore, only few and highly immu-
nogenic vaccines show significant protective efficacy after a single dose in
infants. The limited IgG responses are extended all over the first year of
life. In addition, the immune responses, particularly antibodies, elicited in
the first year of life after vaccination rapidly decline [203]. However, the
problem observed in infants in terms of magnitude and duration of immune
response does not seem to affect efficient priming. In fact, the immune mem-
ory generated in neonates may be recalled later in life [119]. Nevertheless,
strategies to generate strong and long-lasting protective responses in infants
are still needed. This is in part due to the presence of maternal antibodies,
which inactivate and clear the vaccine antigens, thereby rendering difficult
the stimulation of an immature immune system [203]. In addition, the effects
of adjuvants reported in adults cannot be extrapolated to neonates [206]. A
potential strategy to overcome these problems would be to implement vac-
cination during pregnancy, to provide the required antibodies by placenta
and later by maternal feeding [30, 207–209]. This could be complemented
with an early priming of the “immature” immune system of the newborn
by DNA vaccination, followed by a boost during the second half of the first
year or later in life [203].

The immune system in the elderly


Poly-pathology and multiple organ failure is the rule rather than the excep-
tion in aging individuals. Thus, many systems are affected (e.g., endocrine,
cardiovascular), and the immune system is not an exception. The mecha-
nisms involved in the immune senescence process, which in turn may lead to
poor response to vaccination, are not fully understood. However, it is clear
that responses against certain vaccines are more affected by immune senes-
cence than others (e.g., PS-based vaccines against S. pneumoniae) [210]. In
contrast, the responses to a boost dose of the anti-tetanus vaccine are hardly
affected by age [211].
A rapid decline of antibody responses, together with a relative restriction
of the T-cell repertoire is characteristic of the immune senescence process.
This restriction and the reduction in the pool of naïve cells can explain the
poor CD4 + T cell responses against antigens that are cross-reacting with pro-
teins which were seen earlier in life. In contrast, T-cells responses of healthy
elderly individuals to new antigens are often unaffected. Nevertheless, the
overall response to vaccination in the elderly is less efficient than in young
adults, making more vigorous approaches necessary (Fig. 2).
In the case of influenza, the actual strategy is annual re-vaccination.
However, there are concerns regarding the capacity to increase antibod-
ies with proper specificity against re-assorted viruses in aging adults
New vaccination concepts for CAP 229

Figure 2. Factors affecting the responses in young adults and aging individuals after vaccina-
tion. The process of immune senescence impairs host response to both infection and vac-
cination. This critical issue needs to be considered during vaccine design and will require the
development of special approaches.

who have been repeatedly infected or immunized. After exposure to a


new, but cross-reacting antigenic variant, such individuals may respond
by producing antibodies. However, these antibodies could be primarily
directed against influenza strains, which were encountered earlier in life.
230 Pablo D. Becker and Carlos A. Guzmán

For example, individuals previously exposed to the “old” H1N1 influenza


strain (i.e., 50 years ago), may respond differently from naïve adults who
are vaccinated with a “new” H1N1 strain which have accumulated differ-
ent mutations. The former might produce antibodies against the HA of the
“old” H1N1 strain rather than to the cross-reacting epitopes of the new
strain [212]. This is phenomenon is the so-called “original antigenic sin”
[119]. On the basis of this observations, it was proposed that variations
in vaccine efficacy might be due to differences in the antigenic distance
between the vaccine strains and the epidemic strains responsible for
influenza outbreaks [213]. However, this hypothesis was not confirmed by
epidemiologic studies [214]. Even more, individuals aged 65 years or older
who were annually vaccinated showed a significantly reduced mortality
risk. Therefore, until now, it seems that the antigenic sin does not represent
a major practical obstacle in influenza vaccination and additional strate-
gies may not be required.

Concluding remarks

Despite the broad availability of vaccines against agents causing com-


munity-acquired pneumonia, they still represent an important cause of
death, human suffering and economic losses. However, we have dra-
matically expanded our knowledge on the pathophysiology of diseases
caused by respiratory pathogens, their virulence factors and the effector
mechanisms responsible for their clearance. It is becoming clearer which
microbial components are attractive as vaccine targets, as well as the type
of immune response needed to confer protection against disease. Thus,
it is now possible to address vaccine development using rational rather
than empiric approaches. This is facilitated by powerful bioinformatics
tools for the accurate prediction of epitopes and proteasome trimming
[215–217], as well as by the availability of a broad palette of immune
modulators and delivery systems. Therefore, we can predict that new and
improved vaccines against the etiologic agents of community-acquired
pneumonia will considerably reduce the global impact of this disease in
the coming years.

Acknowledgments

This work was supported in part by grants from the DFG (GU482/2-3) and
the BMBF (“PathoGenoMik” – Competence Center for Genome Research
of Pathogenic Bacteria “Pathogenomik”, 031U213B) to CAG. We are par-
ticularly grateful to D. Felnerova from Etna Biotech, who provided us with
a micrograph from a transmission electron microscopy of a virosome, and
to M. Höfle for critical reading of the manuscript.
New vaccination concepts for CAP 231

References
1 Girard MP, Cherian T, Pervikov Y, Kieny MP (2006) A review of vaccine
research and development: Human acute respiratory infections. Vaccine 24:
4692–4700
2 Williams BG, Gouws E, Boschi-Pinto C, Bryce J, Dye C (2002) Estimates
of world-wide distribution of child deaths from acute respiratory infections.
Lancet Infect Dis 2: 25–32
3 Schmid D, Rouse B (2005) Respiratory viral vaccines. In: J Mestecky, M Lamm,
W Strober, J Bienenstock, J McGhee, L Mayer (eds): Mucosal Immunology.
Elsevier Inc., Philadelphia, 923–936
4 Stohr K, Esveld M (2004) Public health. Will vaccines be available for the next
influenza pandemic? Science 306: 2195–2196
5 Kemble G, Greenberg H (2003) Novel generations of influenza vaccines.
Vaccine 21: 1789–1795
6 Palache AM, Scheepers HS, de Regt V, van Ewijk P, Baljet M, Brands R, van
Scharrenburg GJ (1999) Safety, reactogenicity and immunogenicity of Madin
Darby Canine Kidney cell-derived inactivated influenza subunit vaccine. A
meta-analysis of clinical studies. Dev Biol Stand 98: 115–125; discussion 133–
114
7 Kistner O, Barrett PN, Mundt W, Reiter M, Schober-Bendixen S, Eder G,
Dorner F (1999) Development of a Vero cell-derived influenza whole virus
vaccine. Dev Biol Stand 98: 101–110; discussion 111
8 Brands R, Visser J, Medema J, Palache AM, van Scharrenburg GJ (1999)
Influvac: a safe Madin Darby Canine Kidney (MDCK) cell culture-based influ-
enza vaccine. Dev Biol Stand 98: 93–100; discussion 111
9 Bullough PA, Hughson FM, Skehel JJ, Wiley DC (1994) Structure of influenza
haemagglutinin at the pH of membrane fusion. Nature 371: 37–43
10 Belshe R, Maassab H, Mendelman P (2004) Influenza vaccine-live. In: S
Plotkin, W Orenstein (eds): Vaccines. Elsevier Inc., Philadelphia, 371–388
11 Cox RJ, Brokstad KA, Ogra P (2004) Influenza virus: immunity and vaccination
strategies. Comparison of the immune response to inactivated and live, attenu-
ated influenza vaccines. Scand J Immunol 59: 1–15
12 Belshe RB (2004) Current status of live attenuated influenza virus vaccine in
the US. Virus Res 103: 177–185
13 Treanor JJ, Kotloff K, Betts RF, Belshe R, Newman F, Iacuzio D, Wittes J,
Bryant M (1999) Evaluation of trivalent, live, cold-adapted (CAIV-T) and
inactivated (TIV) influenza vaccines in prevention of virus infection and illness
following challenge of adults with wild-type influenza A (H1N1), A (H3N2),
and B viruses. Vaccine 18: 899–906
14 Belshe RB, Nichol KL, Black SB, Shinefield H, Cordova J, Walker R, Hessel C,
Cho I, Mendelman PM (2004) Safety, efficacy, and effectiveness of live, attenu-
ated, cold-adapted influenza vaccine in an indicated population aged 5–49
years. Clin Infect Dis 39: 920–927
15 Belshe RB, Gruber WC, Mendelman PM, Mehta HB, Mahmood K, Reisinger
K, Treanor J, Zangwill K, Hayden FG, Bernstein DI et al (2000) Correlates of
232 Pablo D. Becker and Carlos A. Guzmán

immune protection induced by live, attenuated, cold-adapted, trivalent, intrana-


sal influenza virus vaccine. J Infect Dis 181: 1133–1137
16 Harper SA, Fukuda K, Uyeki TM, Cox NJ, Bridges CB (2004) Prevention
and control of influenza: recommendations of the Advisory Committee on
Immunization Practices (ACIP). MMWR Recomm Rep 53: 1–40
17 Harper SA, Fukuda K, Uyeki TM, Cox NJ, Bridges CB (2005) Prevention
and control of influenza. Recommendations of the Advisory Committee on
Immunization Practices (ACIP). MMWR Recomm Rep 54: 1–40
18 Romanova J, Katinger D, Ferko B, Vcelar B, Sereinig S, Kuznetsov O, Stukova
M, Erofeeva M, Kiselev O, Katinger H et al (2004) Live cold-adapted influenza
A vaccine produced in Vero cell line. Virus Res 103: 187–193
19 Schumacher R, Adamina M, Zurbriggen R, Bolli M, Padovan E, Zajac P,
Heberer M, Spagnoli GC (2004) Influenza virosomes enhance class I restricted
CTL induction through CD4 + T cell activation. Vaccine 22: 714–723
20 Drape RJ, Macklin MD, Barr LJ, Jones S, Haynes JR, Dean HJ (2005)
Epidermal DNA vaccine for influenza is immunogenic in humans. Vaccine 24:
4475–4481
21 Cassetti MC, Couch R, Wood J, Pervikov Y (2005) Report of meeting on
the development of influenza vaccines with broad spectrum and long-lasting
immune responses, World Health Organization, Geneva, Switzerland, 26–27
February 2004. Vaccine 23: 1529–1533
22 Neirynck S, Deroo T, Saelens X, Vanlandschoot P, Jou WM, Fiers W (1999) A
universal influenza A vaccine based on the extracellular domain of the M2
protein. Nat Med 5: 1157–1163
23 Fiers W, De Filette M, Birkett A, Neirynck S, Min Jou W (2004) A “universal”
human influenza A vaccine. Virus Res 103: 173–176
24 Fan J, Liang X, Horton MS, Perry HC, Citron MP, Heidecker GJ, Fu TM, Joyce
J, Przysiecki CT, Keller PM et al (2004) Preclinical study of influenza virus A
M2 peptide conjugate vaccines in mice, ferrets, and rhesus monkeys. Vaccine 22:
2993–3003
25 Durbin AP, Karron RA (2003) Progress in the development of respiratory syn-
cytial virus and parainfluenza virus vaccines. Clin Infect Dis 37: 1668–1677
26 Tao T, Skiadopoulos MH, Durbin AP, Davoodi F, Collins PL, Murphy BR
(1999) A live attenuated chimeric recombinant parainfluenza virus (PIV)
encoding the internal proteins of PIV type 3 and the surface glycoproteins of
PIV type 1 induces complete resistance to PIV1 challenge and partial resis-
tance to PIV3 challenge. Vaccine 17: 1100–1108
27 Kapikian AZ, Mitchell RH, Chanock RM, Shvedoff RA, Stewart CE (1969) An
epidemiologic study of altered clinical reactivity to respiratory syncytial (RS)
virus infection in children previously vaccinated with an inactivated RS virus
vaccine. Am J Epidemiol 89: 405–421
28 Glezen WP, Taber LH, Frank AL, Kasel JA (1986) Risk of primary infection
and reinfection with respiratory syncytial virus. Am J Dis Child 140: 543–546
29 Simoes EA (2002) Immunoprophylaxis of respiratory syncytial virus: global
experience. Respir Res 3 (Suppl 1): S26–33
30 Munoz FM, Piedra PA, Glezen WP (2003) Safety and immunogenicity of respi-
New vaccination concepts for CAP 233

ratory syncytial virus purified fusion protein–2 vaccine in pregnant women.


Vaccine 21: 3465–3467
31 Piedra PA, Grace S, Jewell A, Spinelli S, Hogerman DA, Malinoski F, Hiatt
PW (1998) Sequential annual administration of purified fusion protein vaccine
against respiratory syncytial virus in children with cystic fibrosis. Pediatr Infect
Dis J 17: 217–224
32 Plotnicky H, Siegrist CA, Aubry JP, Bonnefoy JY, Corvaia N, Nguyen TN,
Power UF (2003) Enhanced pulmonary immunopathology following neonatal
priming with formalin-inactivated respiratory syncytial virus but not with the
BBG2NA vaccine candidate. Vaccine 21: 2651–2660
33 Power UF, Nguyen TN, Rietveld E, de Swart RL, Groen J, Osterhaus AD, de
Groot R, Corvaia N, Beck A, Bouveret-Le-Cam N et al (2001) Safety and
immunogenicity of a novel recombinant subunit respiratory syncytial virus vac-
cine (BBG2Na) in healthy young adults. J Infect Dis 184: 1456–1460
34 Power UF, Plotnicky H, Blaecke A, Nguyen TN (2003) The immunogenicity,
protective efficacy and safety of BBG2Na, a subunit respiratory syncytial virus
(RSV) vaccine candidate, against RSV-B. Vaccine 22: 168–176
35 Wright PF, Karron RA, Belshe RB, Thompson J, Crowe JE Jr, Boyce TG,
Halburnt LL, Reed GW, Whitehead SS, Anderson EL et al (2000) Evaluation
of a live, cold-passaged, temperature-sensitive, respiratory syncytial virus vac-
cine candidate in infancy. J Infect Dis 182: 1331–1342
36 Schmidt AC, McAuliffe JM, Murphy BR, Collins PL (2001) Recombinant
bovine/human parainfluenza virus type 3 (B/HPIV3) expressing the respiratory
syncytial virus (RSV) G and F proteins can be used to achieve simultaneous
mucosal immunization against RSV and HPIV3. J Virol 75: 4594–4603
37 Tang RS, MacPhail M, Schickli JH, Kaur J, Robinson CL, Lawlor HA, Guzzetta
JM, Spaete RR, Haller AA (2004) Parainfluenza virus type 3 expressing the
native or soluble fusion (F) Protein of Respiratory Syncytial Virus (RSV)
confers protection from RSV infection in African green monkeys. J Virol 78:
11198–11207
38 Peiris JS, Guan Y, Yuen KY (2004) Severe acute respiratory syndrome. Nat Med
10: S88–97
39 Jiang S, He Y, Liu S (2005) SARS vaccine development. Emerg Infect Dis 11:
1016–1020
40 Holmes KV (2003) SARS-associated coronavirus. N Engl J Med 348: 1948–
1951
41 Li W, Moore MJ, Vasilieva N, Sui J, Wong SK, Berne MA, Somasundaran M,
Sullivan JL, Luzuriaga K, Greenough TC et al (2003) Angiotensin-convert-
ing enzyme 2 is a functional receptor for the SARS coronavirus. Nature 426:
450–454
42 Wong SK, Li W, Moore MJ, Choe H, Farzan M (2004) A 193-amino acid frag-
ment of the SARS coronavirus S protein efficiently binds angiotensin-convert-
ing enzyme 2 J Biol Chem 279: 3197–3201
43 Yang ZY, Kong WP, Huang Y, Roberts A, Murphy BR, Subbarao K, Nabel GJ
(2004) A DNA vaccine induces SARS coronavirus neutralization and protec-
tive immunity in mice. Nature 428: 561–564
44 Wang S, Chou TH, Sakhatskyy PV, Huang S, Lawrence JM, Cao H, Huang
234 Pablo D. Becker and Carlos A. Guzmán

X, Lu S (2005) Identification of two neutralizing regions on the severe acute


respiratory syndrome coronavirus spike glycoprotein produced from the mam-
malian expression system. J Virol 79: 1906–1910
45 Bukreyev A, Lamirande EW, Buchholz UJ, Vogel LN, Elkins WR, St Claire M,
Murphy BR, Subbarao K, Collins PL (2004) Mucosal immunisation of African
green monkeys (Cercopithecus aethiops) with an attenuated parainfluenza
virus expressing the SARS coronavirus spike protein for the prevention of
SARS. Lancet 363: 2122–2127
46 Weingartl H, Czub M, Czub S, Neufeld J, Marszal P, Gren J, Smith G, Jones S,
Proulx R, Deschambault Y et al (2004) Immunization with modified vaccinia
virus Ankara-based recombinant vaccine against severe acute respiratory syn-
drome is associated with enhanced hepatitis in ferrets. J Virol 78: 12672–12676
47 Liu S, Xiao G, Chen Y, He Y, Niu J, Escalante CR, Xiong H, Farmar J, Debnath
AK, Tien P et al (2004) Interaction between heptad repeat 1 and 2 regions in
spike protein of SARS-associated coronavirus: implications for virus fusogenic
mechanism and identification of fusion inhibitors. Lancet 363: 938–947
48 Yang ZY, Werner HC, Kong WP, Leung K, Traggiai E, Lanzavecchia A, Nabel
GJ (2005) Evasion of antibody neutralization in emerging severe acute respira-
tory syndrome coronaviruses. Proc Natl Acad Sci USA 102: 797–801
49 Zhu MS, Pan Y, Chen HQ, Shen Y, Wang XC, Sun YJ, Tao KH (2004) Induction
of SARS-nucleoprotein-specific immune response by use of DNA vaccine.
Immunol Lett 92: 237–243
50 Buchholz UJ, Bukreyev A, Yang L, Lamirande EW, Murphy BR, Subbarao
K, Collins PL (2004) Contributions of the structural proteins of severe acute
respiratory syndrome coronavirus to protective immunity. Proc Natl Acad Sci
USA 101: 9804–9809
51 Howell MR, Nang RN, Gaydos CA, Gaydos JC (1998) Prevention of adenovi-
ral acute respiratory disease in Army recruits: cost-effectiveness of a military
vaccination policy. Am J Prev Med 14: 168–175
52 Schmidt AC, Couch RB, Galasso GJ, Hayden FG, Mills J, Murphy BR, Chanock
RM (2001) Current research on respiratory viral infections: Third International
Symposium. Antiviral Res 50: 157–196
53 Janoff E, Briles D, Rubins J (2005) Respiratory bacterial vaccines. In: J
Mestecky, M Lamm, W Strober, J Bienenstock, J McGhee, L Mayer (eds),
Mucosal Immunology. Elsevier Inc, Philadelphia, 905–921
54 Fedson D, Musher D (2004) Pneumococcal polysaccharide. In: S Plotkin, W
Orenstein (eds): Vaccines. Elsevier Inc., Philadelphia, 529–588
55 Ortqvist A, Hedlund J, Burman LA, Elbel E, Hofer M, Leinonen M, Lindblad
I, Sundelof B, Kalin M (1998) Randomised trial of 23-valent pneumococcal
capsular polysaccharide vaccine in prevention of pneumonia in middle-aged
and elderly people. Swedish Pneumococcal Vaccination Study Group. Lancet
351: 399–403
56 Munoz FM, Englund JA, Cheesman CC, Maccato ML, Pinell PM, Nahm MH,
Mason EO, Kozinetz CA, Thompson RA, Glezen WP (2001) Maternal immu-
nization with pneumococcal polysaccharide vaccine in the third trimester of
gestation. Vaccine 20: 826–837
57 Klugman KP, Madhi SA, Huebner RE, Kohberger R, Mbelle N, Pierce N (2003)
New vaccination concepts for CAP 235

A trial of a 9-valent pneumococcal conjugate vaccine in children with and those


without HIV infection. N Engl J Med 349: 1341–1348
58 Fireman B, Black SB, Shinefield HR, Lee J, Lewis E, Ray P (2003) Impact of the
pneumococcal conjugate vaccine on otitis media. Pediatr Infect Dis J 22: 10–16
59 Eskola D, Black S, Shinefield H (2004) Pneumococcal conjugate vaccine. In: S
Plotkin, W Orenstein (eds): Vaccines. Elsevier Inc., Philadelphia, 589–624
60 Whitney CG, Farley MM, Hadler J, Harrison LH, Bennett NM, Lynfield R,
Reingold A, Cieslak PR, Pilishvili T, Jackson D et al (2003) Decline in invasive
pneumococcal disease after the introduction of protein-polysaccharide conju-
gate vaccine. N Engl J Med 348: 1737–1746
61 Bogaert D, Hermans PW, Adrian PV, Rumke HC, de Groot R (2004)
Pneumococcal vaccines: an update on current strategies. Vaccine 22: 2209–
2220
62 Shapiro ED, Ward JI (1991) The epidemiology and prevention of disease
caused by Haemophilus influenzae type b. Epidemiol Rev 13: 113–142
63 Peltola H, Kayhty H, Virtanen M, Makela PH (1984) Prevention of Hemophilus
influenzae type b bacteremic infections with the capsular polysaccharide vac-
cine. N Engl J Med 310: 1561–1566
64 Robbins JB, Schneerson R, Anderson P, Smith DH (1996) The 1996 Albert
Lasker Medical Research Awards. Prevention of systemic infections, espe-
cially meningitis, caused by Haemophilus influenzae type b. Impact on public
health and implications for other polysaccharide-based vaccines. Jama 276:
1181–1185
65 Decker MD, Edwards KM (1998) Haemophilus influenzae type b vaccines: his-
tory, choice and comparisons. Pediatr Infect Dis J 17: S113–116
66 Mulholland EK, Hoestermann A, Ward JI, Maine N, Ethevenaux C, Greenwood
BM (1996) The use of Haemophilus influenzae type b-tetanus toxoid conjugate
vaccine mixed with diphtheria-tetanus-pertussis vaccine in Gambian infants.
Vaccine 14: 905–909
67 Mulholland K, Hilton S, Adegbola R, Usen S, Oparaugo A, Omosigho C,
Weber M, Palmer A, Schneider G, Jobe K et al (1997) Randomised trial of
Haemophilus influenzae type-b tetanus protein conjugate vaccine [corrected]
for prevention of pneumonia and meningitis in Gambian infants. Lancet 349:
1191–1197
68 Heath PT (1998) Haemophilus influenzae type b conjugate vaccines: a review
of efficacy data. Pediatr Infect Dis J 17: S117–122
69 Lagos R, Horwitz I, Toro J, San Martin O, Abrego P, Bustamante C, Wasserman
SS, Levine OS, Levine MM (1996) Large scale, postlicensure, selective vac-
cination of Chilean infants with PRP-T conjugate vaccine: practicality and
effectiveness in preventing invasive Haemophilus influenzae type b infections.
Pediatr Infect Dis J 15: 216–222
70 Akkoyunlu M, Janson H, Ruan M, Forsgren A (1996) Biological activity of
serum antibodies to a nonacylated form of lipoprotein D of Haemophilus influ-
enzae. Infect Immun 64: 4586–4592
71 Kyd JM, Dunkley ML, Cripps AW (1995) Enhanced respiratory clearance of
nontypeable Haemophilus influenzae following mucosal immunization with P6
in a rat model. Infect Immun 63: 2931–2940
236 Pablo D. Becker and Carlos A. Guzmán

72 Hotomi M, Yamanaka N, Shimada J, Suzumoto M, Ikeda Y, Sakai A, Arai


J, Green B (2002) Intranasal immunization with recombinant outer mem-
brane protein P6 induces specific immune responses against nontypeable
Haemophilus influenzae. Int J Pediatr Otorhinolaryngol 65: 109–116
73 Bertot GM, Becker PD, Guzman CA, Grinstein S (2004) Intranasal vaccina-
tion with recombinant P6 protein and adamantylamide dipeptide as mucosal
adjuvant confers efficient protection against otitis media and lung infection by
nontypeable Haemophilus influenzae. J Infect Dis 189: 1304–1312
74 Baraff LJ, Cody CL, Cherry JD (1984) DTP-associated reactions: an analysis by
injection site, manufacturer, prior reactions, and dose. Pediatrics 73: 31–36
75 Sato Y, Kimura M, Fukumi H (1984) Development of a pertussis component
vaccine in Japan. Lancet 1: 122–126
76 AdHocGroupfortheStudyofPertussisVaccines (1988) Placebo-controlled trial
of two acellular pertussis vaccines in Sweden – protective efficacy and adverse
events. Ad Hoc Group for the Study of Pertussis Vaccines. Lancet 331: 955–
960
77 Storsaeter J, Hallander H, Farrington CP, Olin P, Mollby R, Miller E (1990)
Secondary analyses of the efficacy of two acellular pertussis vaccines evaluated
in a Swedish phase III trial. Vaccine 8: 457–461
78 Guiso N, Boursaux-Eude C, Weber C, Hausman SZ, Sato H, Iwaki M, Kamachi
K, Konda T, Burns DL (2001) Analysis of Bordetella pertussis isolates collected
in Japan before and after introduction of acellular pertussis vaccines. Vaccine
19: 3248–3252
79 Campins-Marti M, Cheng HK, Forsyth K, Guiso N, Halperin S, Huang
LM, Mertsola J, Oselka G, Ward J, Wirsing von Konig CH et al (2001)
Recommendations are needed for adolescent and adult pertussis immunisa-
tion: rationale and strategies for consideration. Vaccine 20: 641–646
80 Weber C, Boursaux-Eude C, Coralie G, Caro V, Guiso N (2001) Polymorphism
of Bordetella pertussis isolates circulating for the last 10 years in France, where
a single effective whole-cell vaccine has been used for more than 30 years. J
Clin Microbiol 39: 4396–4403
81 Denoel P, Godfroid F, Guiso N, Hallander H, Poolman J (2005) Comparison
of acellular pertussis vaccines-induced immunity against infection due to
Bordetellapertussis variant isolates in a mouse model. Vaccine 23: 5333–5341
82 van Amersfoorth SC, Schouls LM, van der Heide HG, Advani A, Hallander
HO, Bondeson K, von Konig CH, Riffelmann M, Vahrenholz C, Guiso N et al
(2005) Analysis of Bordetella pertussis populations in European countries with
different vaccination policies. J Clin Microbiol 43: 2837–2843
83 Penttila T, Vuola JM, Puurula V, Anttila M, Sarvas M, Rautonen N, Makela PH,
Puolakkainen M (2000) Immunity to Chlamydia pneumoniae induced by vac-
cination with DNA vectors expressing a cytoplasmic protein (Hsp60) or outer
membrane proteins (MOMP and Omp2). Vaccine 19: 1256–1265
84 Penttila T, Tammiruusu A, Liljestrom P, Sarvas M, Makela PH, Vuola JM,
Puolakkainen M (2004) DNA immunization followed by a viral vector booster
in a Chlamydia pneumoniae mouse model. Vaccine 22: 3386–3394
85 Pinchuk I, Starcher BC, Livingston B, Tvninnereim A, Wu S, Appella E, Sidney
J, Sette A, Wizel B (2005) A CD8 + T cell heptaepitope minigene vaccine
New vaccination concepts for CAP 237

induces protective immunity against Chlamydia pneumoniae. J Immunol 174:


5729–5739
86 Finco O, Bonci A, Agnusdei M, Scarselli M, Petracca R, Norais N, Ferrari G,
Garaguso I, Donati M, Sambri V et al (2005) Identification of new potential
vaccine candidates against Chlamydia pneumoniae by multiple screenings.
Vaccine 23: 1178–1188
87 Montigiani S, Falugi F, Scarselli M, Finco O, Petracca R, Galli G, Mariani M,
Manetti R, Agnusdei M, Cevenini R et al (2002) Genomic approach for analy-
sis of surface proteins in Chlamydia pneumoniae. Infect Immun 70: 368–379
88 Yang YP, Myers LE, McGuinness U, Chong P, Kwok Y, Klein MH, Harkness
RE (1997) The major outer membrane protein, CD, extracted from Moraxella
(Branhamella) catarrhalis is a potential vaccine antigen that induces bacteri-
cidal antibodies. FEMS Immunol Med Microbiol 17: 187–199
89 Chen D, McMichael JC, VanDerMeid KR, Hahn D, Mininni T, Cowell J,
Eldridge J (1996) Evaluation of purified UspA from Moraxella catarrhalis
as a vaccine in a murine model after active immunization. Infect Immun 64:
1900–1905
90 McMichael JC (2000) Progress toward the development of a vaccine to prevent
Moraxella (Branhamella) catarrhalis infections. Microbes Infect 2: 561–568
91 Jiao X, Hirano T, Hou Y, Gu XX (2002) Specific immune responses and
enhancement of murine pulmonary clearance of Moraxella catarrhalis by intra-
nasal immunization with a detoxified lipooligosaccharide conjugate vaccine.
Infect Immun 70: 5982–5989
92 Yu S, Gu XX (2005) Synthesis and characterization of lipooligosaccharide-
based conjugate vaccines for serotype B Moraxella catarrhalis. Infect Immun
73: 2790–2796
93 Baseman JB, Tully JG (1997) Mycoplasmas: sophisticated, reemerging, and
burdened by their notoriety. Emerg Infect Dis 3: 21–32
94 Smith CB, Friedewald WT, Chanock RM (1967) Inactivated Mycoplasma pneu-
moniae vaccine. Evaluation in volunteers. Jama 199: 353–358
95 Waites KB, Talkington DF (2004) Mycoplasma pneumoniae and its role as a
human pathogen. Clin Microbiol Rev 17: 697–728
96 Brenner DJ, Steigerwalt AG, McDade JE (1979) Classification of the
Legionnaires‘ disease bacterium: Legionella pneumophila, genus novum,
species nova, of the family Legionellaceae, familia nova. Ann Intern Med 90:
656–658
97 McDade JE, Shepard CC, Fraser DW, Tsai TR, Redus MA, Dowdle WR (1977)
Legionnaires‘ disease: isolation of a bacterium and demonstration of its role in
other respiratory disease. N Engl J Med 297: 1197–1203
98 Blander SJ, Horwitz MA (1989) Vaccination with the major secretory protein
of Legionella pneumophila induces cell-mediated and protective immunity in a
guinea pig model of Legionnaires’ disease. J Exp Med 169: 691–705
99 Eisenstein TK, Tamada R, Meissler J, Flesher A, Oels HC (1984) Vaccination
against Legionella pneumophila: serum antibody correlates with protection
induced by heat-killed or acetone-killed cells against intraperitoneal but not
aerosol infection in guinea pigs. Infect Immun 45: 685–691
100 Ricci ML, Torosantucci A, Scaturro M, Chiani P, Baldassarri L, Pastoris MC
238 Pablo D. Becker and Carlos A. Guzmán

(2005) Induction of protective immunity by Legionella pneumophila flagellum


in an A/J mouse model. Vaccine 23: 4811–4820
101 Friedman H, Klein TW, Widen R, Newton C, Blanchard DK, Yamamoto Y
(1988) Legionella pneumophila immunity and immunomodulation: nature and
mechanisms. Adv Exp Med Biol 239: 327–341
102 Blander SJ, Horwitz MA (1993) Major cytoplasmic membrane protein of
Legionella pneumophila, a genus common antigen and member of the hsp
60 family of heat shock proteins, induces protective immunity in a guinea pig
model of Legionnaires‘ disease. J Clin Invest 91: 717–723
103 Yoon WS, Park SH, Park YK, Park SC, Sin JI, Kim MJ (2002) Comparison of
responses elicited by immunization with a Legionella species common lipo-
protein delivered as naked DNA or recombinant protein. DNA Cell Biol 21:
99–107
104 Weeratna R, Stamler DA, Edelstein PH, Ripley M, Marrie T, Hoskin D,
Hoffman PS (1994) Human and guinea pig immune responses to Legionella
pneumophila protein antigens OmpS and Hsp60. Infect Immun 62: 3454–3462
105 Blander SJ, Breiman RF, Horwitz MA (1989) A live avirulent mutant Legionella
pneumophila vaccine induces protective immunity against lethal aerosol chal-
lenge. J Clin Invest 83: 810–815
106 Cryz SJ Jr, Furer E, Cross AS, Wegmann A, Germanier R, Sadoff JC (1987)
Safety and immunogenicity of a Pseudomonas aeruginosa O-polysaccharide
toxin A conjugate vaccine in humans. J Clin Invest 80: 51–56
107 Cryz SJ Jr, Sadoff JC, Ohman D, Furer E (1988) Characterization of the human
immune response to a Pseudomonas aeruginosa O-polysaccharide-toxin A con-
jugate vaccine. J Lab Clin Med 111: 701–707
108 Cryz SJ Jr, Wedgwood J, Lang AB, Ruedeberg A, Que JU, Furer E, Schaad
UB (1994) Immunization of noncolonized cystic fibrosis patients against
Pseudomonas aeruginosa. J Infect Dis 169: 1159–1162
109 Cryz SJ Jr, Lang A, Rudeberg A, Wedgwood J, Que JU, Furer E, Schaad U
(1997) Immunization of cystic fibrosis patients with a Pseudomonas aeruginosa
O-polysaccharide-toxin A conjugate vaccine. Behring Inst Mitt 38: 345–349
110 Schaad UB, Lang AB, Wedgwood J, Ruedeberg A, Que JU, Furer E, Cryz SJ,
Jr (1991) Safety and immunogenicity of Pseudomonas aeruginosa conjugate A
vaccine in cystic fibrosis. Lancet 338: 1236–1237
111 Lang AB, Schaad UB, Rudeberg A, Wedgwood J, Que JU, Furer E, Cryz SJ, Jr
(1995) Effect of high-affinity anti-Pseudomonas aeruginosa lipopolysaccharide
antibodies induced by immunization on the rate of Pseudomonas aeruginosa
infection in patients with cystic fibrosis. J Pediatr 127: 711–717
112 Lang AB, Rudeberg A, Schoni MH, Que JU, Furer E, Schaad UB (2004)
Vaccination of cystic fibrosis patients against Pseudomonas aeruginosa reduces
the proportion of patients infected and delays time to infection. Pediatr Infect
Dis J 23: 504–510
113 Zuercher AW, Imboden MA, Jampen S, Bosse D, Ulrich M, Chtioui H,
Lauterburg BH, Lang AB (2005) Cellular immunity in healthy volunteers
treated with an octavalent conjugate Pseudomonas aeruginosa vaccine. Clin
Exp Immunol 142: 381–387
114 Larbig M, Mansouri E, Freihorst J, Tummler B, Kohler G, Domdey H, Knapp B,
New vaccination concepts for CAP 239

Hungerer KD, Hundt E, Gabelsberger J et al (2001) Safety and immunogenic-


ity of an intranasal Pseudomonas aeruginosa hybrid outer membrane protein
F-I vaccine in human volunteers. Vaccine 19: 2291–2297
115 Gocke K, Baumann U, Hagemann H, Gabelsberger J, Hahn H, Freihorst J, von
Specht BU (2003) Mucosal vaccination with a recombinant OprF-I vaccine of
Pseudomonas aeruginosa in healthy volunteers: comparison of a systemic vs. a
mucosal booster schedule. FEMS Immunol Med Microbiol 37: 167–171
116 Pier G (2005) Application of vaccine technology to prevention of Pseudomonas
aeruginosa infections. Expert Rev Vaccines 4: 645–656
117 Plotkin SA (2005) Vaccines: past, present and future. Nat Med 11: S5–11
118 Plotkin SA (2005) Six revolutions in vaccinology. Pediatr Infect Dis J 24: 1–9
119 Lambert PH, Liu M, Siegrist CA (2005) Can successful vaccines teach us how
to induce efficient protective immune responses? Nat Med 11: S54–62
120 Plotkin SA (2003) Vaccines, vaccination, and vaccinology. J Infect Dis 187:
1349–1359
121 Mora M, Veggi D, Santini L, Pizza M, Rappuoli R (2003) Reverse vaccinology.
Drug Discov Today 8: 459–464
122 Singh U, Shah PH, MacFarlane RC (2004) DNA content analysis on microar-
rays. Methods Mol Biol 270: 237–248
123 De Groot AS, Sbai H, Aubin CS, McMurry J, Martin W (2002) Immuno-
informatics: Mining genomes for vaccine components. Immunol Cell Biol 80:
255–269
124 Sette A, Livingston B, McKinney D, Appella E, Fikes J, Sidney J, Newman M,
Chesnut R (2001) The development of multi-epitope vaccines: epitope identifi-
cation, vaccine design and clinical evaluation. Biologicals 29: 271–276
125 Rappuoli R, Covacci A (2003) Reverse vaccinology and genomics. Science 302:
602
126 Rappuoli R (2000) Reverse vaccinology. Curr Opin Microbiol 3: 445–450
127 Stech J, Garn H, Wegmann M, Wagner R, Klenk HD (2005) A new approach
to an influenza live vaccine: modification of the cleavage site of hemagglutinin.
Nat Med 11: 683–689
128 Ozaki H, Govorkova EA, Li C, Xiong X, Webster RG, Webby RJ (2004)
Generation of high-yielding influenza A viruses in African green monkey kid-
ney (Vero) cells by reverse genetics. J Virol 78: 1851–1857
129 Wood JM, Robertson JS (2004) From lethal virus to life-saving vaccine:
developing inactivated vaccines for pandemic influenza. Nat Rev Microbiol 2:
842–847
130 Subbarao K, Katz JM (2004) Influenza vaccines generated by reverse genetics.
Curr Top Microbiol Immunol 283: 313–342
131 Kaiser J (2004) Influenza: girding for disaster. Facing down pandemic flu, the
world‘s defenses are weak. Science 306: 394–397
132 Holmgren J, Czerkinsky C (2005) Mucosal immunity and vaccines. Nat Med 11:
S45–53
133 Schoen C, Stritzker J, Goebel W, Pilgrim S (2004) Bacteria as DNA vaccine
carriers for genetic immunization. Int J Med Microbiol 294: 319–335
134 Vajdy M, Srivastava I, Polo J, Donnelly J, O’Hagan D, Singh M (2004) Mucosal
240 Pablo D. Becker and Carlos A. Guzmán

adjuvants and delivery systems for protein-, DNA- and RNA-based vaccines.
Immunol Cell Biol 82: 617–627
135 Eko FO, He Q, Brown T, McMillan L, Ifere GO, Ananaba GA, Lyn D, Lubitz
W, Kellar KL, Black CM et al (2004) A novel recombinant multisubunit vaccine
against Chlamydia. J Immunol 173: 3375–3382
136 Duncan J, Gilley R, Schafer D, Moldoveanu Z, Mestecky J (1996) Poly(lactide-
co-glycolide) microencapsulation of vaccines for mucosal immunization. In: H
Kiyono, P Ogra, J McGhee (eds): Mucosal Vaccines. Academic Press, Inc., San
Diego, 159–173
137 Tomasi M, Hearn T (1996) ISCOMs, liposomes, and oil-based vaccine delivery
systems. In: H Kiyono, P Ogra, J McGhee (eds): Mucosal Vaccines. Academic
Press, Inc., San Diego, 175–186
138 Copland MJ, Rades T, Davies NM, Baird MA (2005) Lipid based particulate
formulations for the delivery of antigen. Immunol Cell Biol 83: 97–105
139 Seo JY, Seong SY, Ahn BY, Kwon IC, Chung H, Jeong SY (2002) Cross-pro-
tective immunity of mice induced by oral immunization with pneumococcal
surface adhesin a encapsulated in microspheres. Infect Immun 70: 1143–1149
140 de Wit E, Munster VJ, Spronken MI, Bestebroer TM, Baas C, Beyer WE,
Rimmelzwaan GF, Osterhaus AD, Fouchier RA (2005) Protection of mice
against lethal infection with highly pathogenic H7N7 influenza A virus by using
a recombinant low-pathogenicity vaccine strain. J Virol 79: 12401–12407
141 Arnold H, Bumann D, Felies M, Gewecke B, Sorensen M, Gessner JE, Freihorst
J, von Specht BU, Baumann U (2004) Enhanced immunogenicity in the murine
airway mucosa with an attenuated Salmonella live vaccine expressing OprF-
OprI from Pseudomonas aeruginosa. Infect Immun 72: 6546–6553
142 Langermann S, Palaszynski SR, Burlein JE, Koenig S, Hanson MS, Briles DE,
Stover CK (1994) Protective humoral response against pneumococcal infection
in mice elicited by recombinant bacille Calmette-Guerin vaccines expressing
pneumococcal surface protein A. J Exp Med 180: 2277–2286
143 Ho PS, Kwang J, Lee YK (2005) Intragastric administration of Lactobacillus
casei expressing transmissible gastroentritis coronavirus spike glycoprotein
induced specific antibody production. Vaccine 23: 1335–1342
144 Turner MS, Giffard PM (1999) Expression of Chlamydia psittaci- and human
immunodeficiency virus-derived antigens on the cell surface of Lactobacillus
fermentum BR11 as fusions to bspA. Infect Immun 67: 5486–5489
145 Bender BS, Rowe CA, Taylor SF, Wyatt LS, Moss B, Small PA, Jr (1996) Oral
immunization with a replication-deficient recombinant vaccinia virus protects
mice against influenza. J Virol 70: 6418–6424
146 Czub M, Weingartl H, Czub S, He R, Cao J (2005) Evaluation of modified
vaccinia virus Ankara based recombinant SARS vaccine in ferrets. Vaccine 23:
2273–2279
147 Eko FO, Witte A, Huter V, Kuen B, Furst-Ladani S, Haslberger A, Katinger A,
Hensel A, Szostak MP, Resch S et al (1999) New strategies for combination
vaccines based on the extended recombinant bacterial ghost system. Vaccine
17: 1643–1649
148 Haidinger W, Szostak MP, Jechlinger W, Lubitz W (2003) Online monitoring of
Escherichia coli ghost production. Appl Environ Microbiol 69: 468–474
New vaccination concepts for CAP 241

149 Lubitz W, Witte A, Eko FO, Kamal M, Jechlinger W, Brand E, Marchart J,


Haidinger W, Huter V, Felnerova D et al (1999) Extended recombinant bacte-
rial ghost system. J Biotechnol 73: 261–273
150 Lubitz W (2001) Bacterial ghosts as carrier and targeting systems. Expert Opin
Biol Ther 1: 765–771
151 Szostak MP, Mader H, Truppe M, Kamal M, Eko FO, Huter V, Marchart J,
Jechlinger W, Haidinger W, Brand E et al (1997) Bacterial ghosts as multifunc-
tional vaccine particles. Behring Inst Mitt 98: 191–196
152 Witte A, Wanner G, Sulzner M, Lubitz W (1992) Dynamics of PhiX174 protein
E-mediated lysis of Escherichia coli. Arch Microbiol 157: 381–388
153 Matzinger P (2002) The danger model: a renewed sense of self. Science 296:
301–305
154 Mader HJ, Szostak MP, Hensel A, Lubitz W, Haslberger AG (1997) Endotoxicity
does not limit the use of bacterial ghosts as candidate vaccines. Vaccine 15:
195–202
155 Igietseme J, Eko F, He Q, Bandea C, Lubitz W, Garcia-Sastre A, Black C (2005)
Delivery of Chlamydia vaccines. Expert Opin Drug Deliv 2: 549–562
156 Garcea RL, Gissmann L (2004) Virus-like particles as vaccines and vessels for
the delivery of small molecules. Curr Opin Biotechnol 15: 513–517
157 Guerrero RA, Ball JM, Krater SS, Pacheco SE, Clements JD, Estes MK (2001)
Recombinant Norwalk virus-like particles administered intranasally to mice
induce systemic and mucosal (fecal and vaginal) immune responses. J Virol 75:
9713–9722
158 Niikura M, Takamura S, Kim G, Kawai S, Saijo M, Morikawa S, Kurane I, Li
TC, Takeda N, Yasutomi Y (2002) Chimeric recombinant hepatitis E virus-like
particles as an oral vaccine vehicle presenting foreign epitopes. Virology 293:
273–280
159 Shi W, Liu J, Huang Y, Qiao L (2001) Papillomavirus pseudovirus: a novel vac-
cine to induce mucosal and systemic cytotoxic T-lymphocyte responses. J Virol
75: 10139–10148
160 Arkema A, Huckriede A, Schoen P, Wilschut J, Daemen T (2000) Induction of
cytotoxic T lymphocyte activity by fusion-active peptide-containing virosomes.
Vaccine 18: 1327–1333
161 Bungener L, Huckriede A, Wilschut J, Daemen T (2002) Delivery of protein
antigens to the immune system by fusion-active virosomes: a comparison with
liposomes and ISCOMs. Biosci Rep 22: 323–338
162 Bungener L, Idema J, ter Veer W, Huckriede A, Daemen T, Wilschut J (2002)
Virosomes in vaccine development: induction of cytotoxic T lymphocyte activ-
ity with virosome-encapsulated protein antigens. J Liposome Res 12: 155–163
163 Plant A, Williams NA (2004) Modulation of the immune response by the chol-
era-like enterotoxins. Curr Top Med Chem 4: 509–519
164 Holmgren J, Adamsson J, Anjuere F, Clemens J, Czerkinsky C, Eriksson K, Flach
CF, George-Chandy A, Harandi AM, Lebens M et al (2005) Mucosal adjuvants
and anti-infection and anti-immunopathology vaccines based on cholera toxin,
cholera toxin B subunit and CpG DNA. Immunol Lett 97: 181–188
165 Rharbaoui FG, CA (2005) New generation of immune modulators based on
Toll-like receptors signaling. Curr Immunol Reviews 1: 107–118
242 Pablo D. Becker and Carlos A. Guzmán

166 Pizza M, Giuliani MM, Fontana MR, Monaci E, Douce G, Dougan G, Mills KH,
Rappuoli R, Del Giudice G (2001) Mucosal vaccines: non toxic derivatives of
LT and CT as mucosal adjuvants. Vaccine 19: 2534–2541
167 Sanchez J, Wallerstrom G, Fredriksson M, Angstrom J, Holmgren J (2002)
Detoxification of cholera toxin without removal of its immunoadjuvanticity
by the addition of (STa-related) peptides to the catalytic subunit. A potential
new strategy to generate immunostimulants for vaccination. J Biol Chem 277:
33369–33377
168 Lu X, Clements JD, Katz JM (2002) Mutant Escherichia coli heat-labile entero-
toxin [LT(R192G)] enhances protective humoral and cellular immune respons-
es to orally administered inactivated influenza vaccine. Vaccine 20: 1019–1029
169 van Ginkel FW, Jackson RJ, Yuki Y, McGhee JR (2000) Cutting edge: the
mucosal adjuvant cholera toxin redirects vaccine proteins into olfactory tissues.
J Immunol 165: 4778–4782
170 Gockel CM, Russell MW (2005) Induction and recall of immune memory by
mucosal immunization with a non-toxic recombinant enterotoxin-based chime-
ric protein. Immunology 116: 477–486
171 Areas AP, Oliveira ML, Miyaji EN, Leite LC, Aires KA, Dias WO, Ho PL
(2004) Expression and characterization of cholera toxin B-pneumococcal
surface adhesin A fusion protein in Escherichia coli: ability of CTB-PsaA to
induce humoral immune response in mice. Biochem Biophys Res Commun 321:
192–196
172 Sabirov A, Kodama S, Hirano T, Suzuki M, Mogi G (2001) Intranasal immuni-
zation enhances clearance of nontypeable Haemophilus influenzae and reduces
stimulation of tumor necrosis factor alpha production in the murine model of
otitis media. Infect Immun 69: 2964–2971
173 Medina E, Talay SR, Chhatwal GS, Guzman CA (1998) Fibronectin-binding
protein I of Streptococcus pyogenes is a promising adjuvant for antigens deliv-
ered by mucosal route. Eur J Immunol 28: 1069–1077
174 Janeway CA Jr, Medzhitov R (2002) Innate immune recognition. Annu Rev
Immunol 20: 197–216
175 Roach JC, Glusman G, Rowen L, Kaur A, Purcell MK, Smith KD, Hood LE,
Aderem A (2005) The evolution of vertebrate Toll-like receptors. Proc Natl
Acad Sci USA 102: 9577–9582
176 Chuang TH, Lee J, Kline L, Mathison JC, Ulevitch RJ (2002) Toll-like receptor
9 mediates CpG-DNA signaling. J Leukoc Biol 71: 538–544
177 Wagner H (2004) The immunobiology of the TLR9 subfamily. Trends Immunol
25: 381–386
178 Lee CJ, Lee LH, Gu XX (2005) Mucosal immunity induced by pneumococcal
glycoconjugate. Crit Rev Microbiol 31: 137–144
179 Cooper CL, Davis HL, Morris ML, Efler SM, Krieg AM, Li Y, Laframboise
C, Al Adhami MJ, Khaliq Y, Seguin I et al (2004) Safety and immunogenicity
of CPG 7909 injection as an adjuvant to Fluarix influenza vaccine. Vaccine 22:
3136–3143
180 von Hunolstein C, Mariotti S, Teloni R, Alfarone G, Romagnoli G, Orefici G,
Nisini R (2001) The adjuvant effect of synthetic oligodeoxynucleotide contain-
ing CpG motif converts the anti-Haemophilus influenzae type b glycoconju-
New vaccination concepts for CAP 243

gates into efficient anti-polysaccharide and anti-carrier polyvalent vaccines.


Vaccine 19: 3058–3066
181 Deiters U, Muhlradt PF (1999) Mycoplasmal lipopeptide MALP–2 induces
the chemoattractant proteins macrophage inflammatory protein 1alpha (MIP-
1alpha), monocyte chemoattractant protein 1, and MIP–2 and promotes leuko-
cyte infiltration in mice. Infect Immun 67: 3390–3398
182 Muhlradt PF, Kiess M, Meyer H, Sussmuth R, Jung G (1997) Isolation, struc-
ture elucidation, and synthesis of a macrophage stimulatory lipopeptide from
Mycoplasma fermentans acting at picomolar concentration. J Exp Med 185:
1951–1958
183 Deiters U, Gumenscheimer M, Galanos C, Muhlradt PF (2003) Toll-like recep-
tor 2- and 6-mediated stimulation by macrophage-activating lipopeptide 2
induces lipopolysaccharide (LPS) cross tolerance in mice, which results in
protection from tumor necrosis factor alpha but in only partial protection from
lethal LPS doses. Infect Immun 71: 4456–4462
184 Into T, Kiura K, Yasuda M, Kataoka H, Inoue N, Hasebe A, Takeda K, Akira S,
Shibata K (2004) Stimulation of human Toll-like receptor (TLR) 2 and TLR6
with membrane lipoproteins of Mycoplasma fermentans induces apoptotic cell
death after NF-kappa B activation. Cell Microbiol 6: 187–199
185 Link C, Gavioli R, Ebensen T, Canella A, Reinhard E, Guzman CA (2004) The
Toll-like receptor ligand MALP–2 stimulates dendritic cell maturation and
modulates proteasome composition and activity. Eur J Immunol 34: 899–907
186 Rharbaoui F, Drabner B, Borsutzky S, Winckler U, Morr M, Ensoli B, Muhlradt
PF, Guzman CA (2002) The Mycoplasma-derived lipopeptide MALP–2 is a
potent mucosal adjuvant. Eur J Immunol 32: 2857–2865
187 Gurunathan S, Klinman DM, Seder RA (2000) DNA vaccines: immunology,
application, and optimization*. Annu Rev Immunol 18: 927–974
188 Ebensen T, Paukner S, Link C, Kudela P, de Domenico C, Lubitz W, Guzman
CA (2004) Bacterial ghosts are an efficient delivery system for DNA vaccines.
J Immunol 172: 6858–6865
189 Liu WJ, Zhao KN, Gao FG, Leggatt GR, Fernando GJ, Frazer IH (2001)
Polynucleotide viral vaccines: codon optimisation and ubiquitin conjugation
enhances prophylactic and therapeutic efficacy. Vaccine 20: 862–869
190 Donnelly JJ, Friedman A, Martinez D, Montgomery DL, Shiver JW, Motzel
SL, Ulmer JB, Liu MA (1995) Preclinical efficacy of a prototype DNA vac-
cine: enhanced protection against antigenic drift in influenza virus. Nat Med 1:
583–587
191 Prince AM, Whalen R, Brotman B (1997) Successful nucleic acid based
immunization of newborn chimpanzees against hepatitis B virus. Vaccine 15:
916–919
192 Reddy ST, Ertl HC (1999) The potential use of DNA vaccines for neonatal
immunization. Curr Opin Mol Ther 1: 22–29
193 Ozaki T, Yauchi M, Xin KQ, Hirahara F, Okuda K (2005) Cross-reactive protec-
tion against influenza A virus by a topically applied DNA vaccine encoding M
gene with adjuvant. Viral Immunol 18: 373–380
194 Miyaji EN, Dias WO, Tanizaki MM, Leite LC (2003) Protective efficacy of
PspA (pneumococcal surface protein A)-based DNA vaccines: contribution of
244 Pablo D. Becker and Carlos A. Guzmán

both humoral and cellular immune responses. FEMS Immunol Med Microbiol
37: 53–57
195 Ochsenbein AF, Pinschewer DD, Sierro S, Horvath E, Hengartner H,
Zinkernagel RM (2000) Protective long-term antibody memory by antigen-
driven and T help-dependent differentiation of long-lived memory B cells to
short-lived plasma cells independent of secondary lymphoid organs. Proc Natl
Acad Sci USA 97: 13263–13268
196 Zinkernagel RM (2002) On differences between immunity and immunological
memory. Curr Opin Immunol 14: 523–536
197 Pinschewer DD, Perez M, Jeetendra E, Bachi T, Horvath E, Hengartner H,
Whitt MA, de la Torre JC, Zinkernagel RM (2004) Kinetics of protective anti-
bodies are determined by the viral surface antigen. J Clin Invest 114: 988–993
198 Nicol M, Huebner R, Mothupi R, Kayhty H, Mbelle N, Khomo E (2002)
Haemophilus influenzae type b conjugate vaccine diluted tenfold in diphtheria-
tetanus-whole cell pertussis vaccine: a randomized trial. Pediatr Infect Dis J 21:
138–141
199 Cassidy WM, Watson B, Ioli VA, Williams K, Bird S, West DJ (2001) A random-
ized trial of alternative two- and three-dose hepatitis B vaccination regimens
in adolescents: antibody responses, safety, and immunologic memory. Pediatrics
107: 626–631
200 Ahman H, Kayhty H, Vuorela A, Leroy O, Eskola J (1999) Dose dependency
of antibody response in infants and children to pneumococcal polysaccharides
conjugated to tetanus toxoid. Vaccine 17: 2726–2732
201 Bosnak M, Dikici B, Bosnak V, Haspolat K (2002) Accelerated hepatitis B vac-
cination schedule in childhood. Pediatr Int 44: 663–665
202 Goldblatt D, Miller E, McCloskey N, Cartwright K (1998) Immunological
response to conjugate vaccines in infants: follow up study. BMJ 316: 1570–
1571
203 Siegrist CA (2001) Neonatal and early life vaccinology. Vaccine 19: 3331–3346
204 Pihlgren M, Tougne C, Bozzotti P, Fulurija A, Duchosal MA, Lambert PH,
Siegrist CA (2003) Unresponsiveness to lymphoid-mediated signals at the neo-
natal follicular dendritic cell precursor level contributes to delayed germinal
center induction and limitations of neonatal antibody responses to T-depen-
dent antigens. J Immunol 170: 2824–2832
205 Pihlgren M, Schallert N, Tougne C, Bozzotti P, Kovarik J, Fulurija A, Kosco-
Vilbois M, Lambert PH, Siegrist CA (2001) Delayed and deficient establish-
ment of the long-term bone marrow plasma cell pool during early life. Eur J
Immunol 31: 939–946
206 Ota MO, Vekemans J, Schlegel-Haueter SE, Fielding K, Sanneh M, Kidd
M, Newport MJ, Aaby P, Whittle H, Lambert PH et al (2002) Influence of
Mycobacterium bovis bacillus Calmette-Guerin on antibody and cytokine
responses to human neonatal vaccination. J Immunol 168: 919–925
207 Van de Perre P (2003) Transfer of antibody via mother’s milk. Vaccine 21:
3374–3376
208 Quiambao BP, Nohynek H, Kayhty H, Ollgren J, Gozum L, Gepanayao CP,
Soriano V, Makela PH (2003) Maternal immunization with pneumococcal poly-
saccharide vaccine in the Philippines. Vaccine 21: 3451–3454
New vaccination concepts for CAP 245

209 Daly KA, Toth JA, Giebink GS (2003) Pneumococcal conjugate vaccines as
maternal and infant immunogens: challenges of maternal recruitment. Vaccine
21: 3473–3478
210 Shapiro ED, Berg AT, Austrian R, Schroeder D, Parcells V, Margolis A, Adair
RK, Clemens JD (1991) The protective efficacy of polyvalent pneumococcal
polysaccharide vaccine. N Engl J Med 325: 1453–1460
211 Bjorkholm B, Hagberg L, Sundbeck G, Granstrom M (2000) Booster effect of
low doses of tetanus toxoid in elderly vaccinees. Eur J Clin Microbiol Infect Dis
19: 195–199
212 Powers DC, Belshe RB (1994) Vaccine-induced antibodies to heterologous
influenza A H1N1 viruses: effects of aging and “original antigenic sin”. J Infect
Dis 169: 1125–1129
213 Smith DJ, Forrest S, Ackley DH, Perelson AS (1999) Variable efficacy of repeat-
ed annual influenza vaccination. Proc Natl Acad Sci USA 96: 14001–14006
214 Voordouw AC, Sturkenboom MC, Dieleman JP, Stijnen T, Smith DJ, van der
Lei J, Stricker BH (2004) Annual revaccination against influenza and mortality
risk in community-dwelling elderly persons. Jama 292: 2089–2095
215 Rammensee HG (2003) Immunoinformatics: bioinformatic strategies for bet-
ter understanding of immune function. Introduction. Novartis Found Symp 254:
1–2
216 Rammensee H, Bachmann J, Emmerich NP, Bachor OA, Stevanovic S (1999)
SYFPEITHI: database for MHC ligands and peptide motifs. Immunogenetics
50: 213–219
217 Nussbaum AK, Kuttler C, Hadeler KP, Rammensee HG, Schild H (2001)
PAProC: a prediction algorithm for proteasomal cleavages available on the
WWW. Immunogenetics 53: 87–94
Index

adaptive immunity 183, 188, 190 reinfection 84


adenovirus, vaccines 209 seroprevalence 84
alternative activation pathway 155 virulence factors 95
alveolar macrophage 150 vaccines 212
amantadine 82 Chlamydia, nomenclature 84
amantadine-resistant virus strain 82 Chlamydophila, nomenclature 84
antibiotics 7 ff choline-binding proteins (CBP) 144
penicillins 8 chronic obstructive pulmonary disease
macrolides 8 (COPD) 80, 193
fluorquinolones 9 classical pathway of complement
resistance 5 activation 154
antigen, lipid and protein 194 clathrin-coated vesicle 150
antigenic drift 76 clone, international (pneumococcal) 61
antigenic shift 76 clone, pneumococcal 68
antiviral compounds 82 co-infection with microorganisms
antiviral therapy 31, 33, 36, 45 (influenzavirus) 78
asthma, and M. pneumoniae 192 combined bacterial and viral pneumonia
autoimmune reaction 191 80
complement factor H 155
bacterial ghosts 221 complement fixation (CF) 194
biological characteristics 184 CRB-65 score 5
bird flu virus 77 C-reactive protein 154
blood cultures 6 cytokine production 189
blood-brain barrier (BBB) 149
Bordetella pertussis, vaccines 212 differential fluorescence induction
bronchiectasis 191 (DIF) 151
bronchitis 79, 80 DNA vaccines 226
Dot/Icm type IV secretion system 119
C. pneumoniae see Chlamydia pneumoniae
C3 degrading activity 154 efflux, PmrA 65
CAPNETZ 16 elderly, immune system 228
capsular polysaccharide 142 encephalitis 81
capsule regulation 153 encephalopathy 80
CD4+ 79 enolase 148
CD8+ T-cell responses to influenza 79 enzyme immunoassay (EIA) 194
cell wall hydrolase 144 extrapulmonary complication 190
cellular activation 188 extrapulmonary syndrome 191
chest x-ray 6
Chlamydia pneumoniae 15, 84, 86, 89–95 factor H (complement) 155
detection by PCR 86 failure, treatment 67
differential diagnosis 84 fluoroquinolone 196
immunity 89–93 fowl plague 77
248 Index

GAPDH 148 local (mucosal) immune response 189


gene target 195 LPxTG sequence 144
gene, mosaic 60 lung abscesses 191
ghosts (bacterial) 221 lymphocyte activation 189
G-protein pathway 150 lymphoid infiltration 189
Guillain-Barré syndrome 81, 191
macrolide 196
Haemophilus influenzae 3, 15, 78, 80 macrolide resistance 196
non typeable, vaccines 212 macrolide treatment 193
type b (Hib), conjugated vaccines 212 macrophage receptor, MARCO 161
vaccines 211 manipulation of vesicle traffic 118
healthcare associated pneumonia 2 mast cell degranulation 193
hemagglutinin (HA) 74–76 microimmuno-fluorescence (MIF) 22
hemolytic anemia 191 Moraxella catarrhalis, vaccines 212
hypogammaglobulinemia 189 mouse infection model 146
hypotension 80 mucosal adjuvants 224
mucosal delivery systems 220
IFN-alpha, in influenza 79 multi-locus sequence typing (MLST) 61
Immunoglobulin subclasses, and MurM protein 63
diagnosis 194 Mycoplasma pneumoniae 3, 15, 183, 184
immunologic hypersensitivity 189 adaptive immune response 190
immunological memory 227 adherence to epithelium 186
influenza infection of neonates 80 biological characteristics 184
influenza vaccines 202–204 cell biology 183
influenza virus 73 epidemiology 185
adaptive immune response to 79 genome 184
co-infection with microorganisms 78 pathogenesis 183
influenza, laboratory diagnosis 204 vaccines 215
innate immunity 79, 183, 188 myocarditis 80
interferon antagonism 78 myositis 80
interleukin (IL)-6 79
interleukin (IL)-8 79 naip5 117
natural reservoir of influenza-A-virus 76
ketolide, resistance 65 neuraminidase (NA) 74, 76
neuraminidase (NanA, NanB, NanC) 145
Legionella 15 neuraminidase inhibitors 82
dot/icm genes 112, 120 neuraminidase subtype 76
dot/icm mutants 117 neutralizing antibody directed against the
egress 123 HA 79
entry in host cells 117 nucleic acid amplification assay 15, 20, 195
growth phase 122
immunity 125 opsonophagocytosis 153
in free-living amoebae 113 orthomyxoviridae 74
Naip proteins 117 oseltamivir 82
persistence 114 otitis media 80
Toll-like receptors 125
transient association with amoebae 112 P1 adhesin 185
vesicles 118 pandemic (influenza) 76
Legionella pneumophila, vaccines 215 parainfluenza virus (PIV), vaccines 205
pathogen-associated molecular patterns
Legionnaires’ disease, outbreak 111 (PAMPs) 156
leukocyte recruitment 150 Pce 150
linezolid, resistance 66 PCR assay 15, 20, 195
lipoteichoic acid (LTA) 156 penicillin-binding protein (PBP) 62
Index 249

pericarditis 80 Reye’s syndrome 81


pharmacodynamics 66 ribosomal protein 64
phase variation 141 rimantadine 82
phospholipase C 150 risk stratification 5, 6
phosphorylcholine (PCho) 144
phosphorylcholine esterase 150 secondary bacterial pneumonia 79, 80
photophobia 80 secretory component (SC) 147
PK/PD 67 senescence, immune 229
plasmin(ogen) 148 seroconversion 194
platelet-activating factor (PAF) 148 severe acute respiratory syndrome (SARS),
pleural effusion 191 vaccines 208
pneumococcal clone 61, 68 signature-tagged mutagenesis (STM) 151
pneumococcal colonization 141 Spanish influenza 74, 76
pneumococcal adherence and virulence Staphylococcus aureus 78, 80
factor A (PavA protein) 147, 148 streptococci, viridans 60, 63
pneumococcal pneumonia 142 Streptococcus pneumoniae 3, 5, 15, 78, 80,
pneumococcal serotype 140 209, 210
pneumococcal surface adhesin A vaccines 140, 209, 210
(PsaA) 146 Streptococcus, erm gene 64
pneumococcal surface protein A Streptococcus, mef gene 63
(PspA) 151 streptogramin, resistance 66
pneumococcal surface protein C (PspC,
SpsA, CbpA) 147 telithromycin 196
pneumococcal molecular epidemiology telithromycin, resistance 65
network (PMEN) 61 terminal sialic acid 146
pneumolysin 142 tetracycline 196
polymeric immunoglobulin receptor 147 Toll-like receptor 156
primary atypical pneumonia (PAP), transmission 185
diagnosis 190 transverse myelitis 81
primary viral pneumonia 79, 80 treatment 7ff
progressive pulmonary failure 193 two-component regulatory systems
Pseudomonas aeruginosa 3 (TCS) 152
Pseudomonas aeruginosa, vaccines216
pseudoviruses 222 urinary antigen 6, 20
pulmonary fibrosis 191
pulmonary infiltrates 80 vaccine 140, 202–216
adenovirus 209
quinolone resistance-determining region B. pertussis 212
(QRDR) 64 C. pneumoniae 214
H. influenzae 211, 212
real-time PCR assay 195 influenza 202-204
receptor binding 74 L. pneumophila 215
recombination 60, 65 M. catarrhalis 214
requirement for sterols 184 M. pneumoniae 215
resistance, acquired 60 P. aeruginosa 216
resistance, fluoroquinolone 64 parainfluenza virus (PIV) 205
resistance, macrolides 63 respiratory syncytial virus (RSV) 206,
resistance, tetracycline 64 207
resistant virus variants 33, 35, 36, 39, 40 S. pneumoniae 209, 210
respiratory syncytial virus (RSV), severe acute respiratory syndrome
vaccines 206, 207 (SARS) 208
respiratory viruses 28, 29, 42–44, 48 vaccine design 226
reverse genetics 218 vaccine, pneumococcal 69
reverse vaccinology 217, 219 vectors, live attenuated bacterial 221
250 Index

vectors, live attenuated viral 221


vesicle traffic, manipulation 118
viral pneumonia 27, 32, 33, 37, 41, 42, 47
virosomes 222, 223
virosomes 223
virulence 183
virus detection 42-48
virus-like particles (VLP) 222

wheezing attacks in asthmatics 80

zanamivir 82
zinc metalloprotease 146
The BAID-Series

Birkhäuser Advances in Infectious Diseases

Infectious diseases remain a substantial drain on human well-being and


economies despite the availability of modern drugs. New pathogens emerge
and known pathogens change their geographical distribution and their
susceptibility to the available drugs. An understanding of the structure and
function of infectious disease pathogens is a major scientific challenge with
important potential applications.
This new cross-disciplinary monograph series will provide up-to-date
information on the latest developments in infectious disease research. The
multi-authored volumes will cover basic biology and biochemistry of patho-
gens as well as applied medical aspects and implications for public health
and policy.
The contributions are written by leading infectious disease researchers
and pharmaceutical scientists with a wide range of expertise.
The envisaged readership includes academic and industrial researchers
in medicine and infectious diseases as well as clinicians and others involved
in diagnostics and drug development.

Forthcoming volumes:
Pediatric Infectious Diseases Revisited, H. Schroten, S. Wirth (Editors),
2007

Available volumes:
Coronaviruses with Special Emphasis on First Insights Concerning SARS,
A. Schmidt, M.H. Wolff, O. Weber (Editors), 2005
The Grand Challenge for the Future. Vaccines for Poverty-Related Diseases
from Bench to Field, S.H.E. Kaufmann and P.-H. Lambert (Editors),
2005
Poxviruses, A. Mercer, A. Schmidt, O. Weber (Editors), 2007

View publication stats

You might also like