You are on page 1of 53

Polymyxin Antibiotics From Laboratory

Bench to Bedside Jian Li


Visit to download the full and correct content document:
https://textbookfull.com/product/polymyxin-antibiotics-from-laboratory-bench-to-bedsid
e-jian-li/
More products digital (pdf, epub, mobi) instant
download maybe you interests ...

Advances in Stem Cell Therapy Bench to Bedside 1st


Edition Nagwa El-Badri (Eds.)

https://textbookfull.com/product/advances-in-stem-cell-therapy-
bench-to-bedside-1st-edition-nagwa-el-badri-eds/

Photodynamic medicine from bench to clinic 1st Edition


Herwig Kostron

https://textbookfull.com/product/photodynamic-medicine-from-
bench-to-clinic-1st-edition-herwig-kostron/

Geriatric rehabilitation : from bedside to curbside 1st


Edition Poduri

https://textbookfull.com/product/geriatric-rehabilitation-from-
bedside-to-curbside-1st-edition-poduri/

Global Higher Education Shared Communities: Efforts and


Concerns from Key Universities in China Jian Li

https://textbookfull.com/product/global-higher-education-shared-
communities-efforts-and-concerns-from-key-universities-in-china-
jian-li/
Shaping Education Reform in China Overviews Policies
and Implications Jian Li

https://textbookfull.com/product/shaping-education-reform-in-
china-overviews-policies-and-implications-jian-li/

Transfer Pricing in China: Concepts, Controls,


Practices, and Audit Assessment Jian Li

https://textbookfull.com/product/transfer-pricing-in-china-
concepts-controls-practices-and-audit-assessment-jian-li/

Multisensory Perception From Laboratory to Clinic Krish


Sathian

https://textbookfull.com/product/multisensory-perception-from-
laboratory-to-clinic-krish-sathian/

From Bench to Boardroom The R D Leader s Guide 1st


Edition Clifford L. Spiro (Auth.)

https://textbookfull.com/product/from-bench-to-boardroom-the-r-d-
leader-s-guide-1st-edition-clifford-l-spiro-auth/

Conceptualizing and Contextualizing Higher Education


with Chinese Characteristics Ontological and
Epistemological Dimensions Jian Li

https://textbookfull.com/product/conceptualizing-and-
contextualizing-higher-education-with-chinese-characteristics-
ontological-and-epistemological-dimensions-jian-li/
Advances in Experimental Medicine and Biology 1145

Jian Li
Roger L. Nation
Keith S. Kaye Editors

Polymyxin Antibiotics:
From Laboratory
Bench to Bedside
Advances in Experimental Medicine
and Biology

Volume 1145

Editorial Board
IRUN R. COHEN, The Weizmann Institute of Science, Rehovot, Israel
ABEL LAJTHA, N.S. Kline Institute for Psychiatric Research,
Orangeburg, NY, USA
JOHN D. LAMBRIS, University of Pennsylvania, Philadelphia, PA, USA
RODOLFO PAOLETTI, University of Milan, Milan, Italy
NIMA REZAEI, Children’s Medical Center Hospital, Tehran University of
Medical Sciences, Tehran, Iran
More information about this series at http://www.springer.com/series/5584
Jian Li • Roger L. Nation • Keith S. Kaye
Editors

Polymyxin Antibiotics:
From Laboratory Bench
to Bedside
Editors
Jian Li Roger L. Nation
Biomedicine Discovery Institute Drug Delivery, Disposition and Dynamics
Infection & Immunity Program and Monash Institute of Pharmaceutical
Department of Microbiology Sciences
Monash University, Clayton Campus Monash University, Parkville Campus
Melbourne, VIC, Australia Melbourne, VIC, Australia

Keith S. Kaye
Division of Infectious Diseases
University of Michigan Medical School
Ann Arbor, MI, USA

ISSN 0065-2598     ISSN 2214-8019 (electronic)


Advances in Experimental Medicine and Biology
ISBN 978-3-030-16371-6    ISBN 978-3-030-16373-0 (eBook)
https://doi.org/10.1007/978-3-030-16373-0

© Springer Nature Switzerland AG 2019


This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or
part of the material is concerned, specifically the rights of translation, reprinting, reuse of
illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way,
and transmission or information storage and retrieval, electronic adaptation, computer software,
or by similar or dissimilar methodology now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this
publication does not imply, even in the absence of a specific statement, that such names are
exempt from the relevant protective laws and regulations and therefore free for general use.
The publisher, the authors, and the editors are safe to assume that the advice and information in
this book are believed to be true and accurate at the date of publication. Neither the publisher nor
the authors or the editors give a warranty, express or implied, with respect to the material
contained herein or for any errors or omissions that may have been made. The publisher remains
neutral with regard to jurisdictional claims in published maps and institutional affiliations.

This Springer imprint is published by the registered company Springer Nature Switzerland AG.
The registered company address is: Gewerbestrasse 11, 6330 Cham, Switzerland
Preface

Antibiotics have been a cornerstone of modern medicine and have saved mil-
lions of lives. The use of antibiotics in the clinic has made many complicated
procedures and treatment modalities possible. Unfortunately, resistance to
these ‘magic bullets’ has become widespread and now poses a serious threat
to human health on a global scale. Over the last two decades, Gram-negative
‘superbugs’, in particular Acinetobacter baumannii, Pseudomonas aerugi-
nosa, Klebsiella pneumoniae and Escherichia coli, have been very successful
in developing resistance to most, and in some cases, all currently available
antibiotics. Very often, clinicians have no newer alternatives but an ‘old’ class
of antibiotics, namely, polymyxins, to treat deadly infections caused by these
notorious pathogens. However, polymyxins (i.e. colistin and polymyxin B)
were almost abandoned soon after their approval in the late 1950s and had
never been rigorously evaluated through the modern drug regulatory system.
Therefore, major knowledge gaps exist and have significantly limited the
optimization of their clinical use. Furthermore, the polymyxins have already
been off-patent for several decades and pharmaceutical companies are not
interested in redeveloping both ‘old’ antibiotics.
The onus lies upon a number of academic research groups who have con-
ducted an enormous number of pharmacological, chemical, microbiological
and clinical studies since the late 1990s with the funding mainly from govern-
ments. Now, clinicians are in a much better position with regard to dosing
polymyxins for a variety of types of patients. Excitingly, several promising
candidates are being evaluated in the drug discovery pipeline. Recognizing
these achievements, three international conferences were held on polymyxins
(2013, Prato, Italy; 2015, San Diego, USA; and 2018, Madrid, Spain) with
international opinion leaders and attendees from a large number of countries
around the world. Programmes and slides from the presentations are freely
available at the website of the International Society of Antimicrobial
Pharmacology (https://www.isap.org/index.php/activities/special-meetings).
Polymyxins are arguably one of the most difficult classes of antibiotics to
research for several reasons, including their complex amphiphilic chemical
structures, stickiness to tubes and plates, complicated product composition
and confusing product labelling conventions. We believe that many research-
ers have met significant challenges in developing a sensitive and reliable
assay for the measurement of polymyxins in different biological matrices for
pharmacokinetic and pharmacodynamic studies. To promote the research and
facilitate their clinical use, here, we have brought together the top researchers
in the field to write the first-ever book on the polymyxins. This book com-
v
vi Preface

prises chapters on all major topics in the polymyxin field and reviews the
current progress that has been made in our understanding of the chemistry,
microbiology, pharmacology, clinical use and drug discovery of polymyxins.
We are hopeful that this book will provide readers with a one-stop source
about polymyxin research and help clinicians to improve patient care.
The world is heading towards a potential post-antibiotic era due to the
rapid increase of antibiotic resistance and the lack of commercial interest in
developing new antibiotics. Therefore, every effort must be made to secure
the clinical utility of this last-line defence against Gram-negative pathogens.
Finally, we would acknowledge the many contributions of authors and
reviewers in the creation of this polymyxin book. This book is in memory of
Professors Alan Forrest and Johan Mouton, who made significant contribu-
tions to polymyxin pharmacology. We are also very grateful to our families
and colleagues for their support.
Enjoy the reading!

Monash University, Clayton Campus, Jian Li


Melbourne, VIC, Australia

Monash University, Parkville Campus, Roger L. Nation


Melbourne, VIC, Australia 

University of Michigan Medical School, Keith S. Kaye


Ann Arbor, MI, USA 
Contents

1 Reviving Polymyxins: Achievements, Lessons


and the Road Ahead������������������������������������������������������������������������   1
Jian Li
2 Multidrug-Resistant Gram-­Negative Pathogens:
The Urgent Need for ‘Old’ Polymyxins ����������������������������������������   9
David L. Paterson and Robert A. Bonomo
3 History, Chemistry and Antibacterial Spectrum�������������������������� 15
Tony Velkov, Philip E. Thompson, Mohammad A. K. Azad,
Kade D. Roberts, and Phillip J. Bergen
4 Polymyxins: Mode of Action ���������������������������������������������������������� 37
Zhifeng Li and Tony Velkov
5 Mechanisms of Polymyxin Resistance�������������������������������������������� 55
Jennifer H. Moffatt, Marina Harper, and John D. Boyce
6 Bioanalysis and Stability of Polymyxins���������������������������������������� 73
Robert W. Milne
7 Pharmacokinetics of Polymyxins in Animals�������������������������������� 89
Sandrine Marchand, Nicolas Grégoire, and William Couet
8 In vitro Pharmacodynamics and PK/PD in Animals�������������������� 105
Winnie Lee, Yiying Cai, Tze-Peng Lim, Jocelyn Teo, Sonja
Courtney Chua, and Andrea Lay-Hoon Kwa
9 Polymyxin Susceptibility Testing and Breakpoint Setting ���������� 117
John Turnidge, Katherine Sei, and Johan Mouton
10 Labelling Conventions and Product Package Insert
of Parenteral Polymyxins: Factors Causing Potential
Medication Errors and Impeding Optimal Clinical Use�������������� 133
Jian Li, Kingsley Coulthard, and Roger L. Nation
11 Meta-analysis of Polymyxin Use in Patients���������������������������������� 143
Mical Paul, Oren Zusman, and Leonard Leibovici
12 Use of Colistin in Critically Ill Patients������������������������������������������ 155
Dror Marchaim, Donald Kaye, and Keith S. Kaye

vii
viii Contents

13 Clinical Use of Colistin in Biofilm-­Associated Infections ������������ 181


Jaime Lora-Tamayo, Oscar Murillo, and Javier Ariza
14 Clinical Use of Polymyxin B������������������������������������������������������������ 197
Maria Helena Rigatto, Diego R. Falci,
and Alexandre P. Zavascki
15 Clinical Pharmacokinetics, Pharmacodynamics
and Toxicodynamics of Polymyxins: Implications
for Therapeutic Use�������������������������������������������������������������������������� 219
Roger L. Nation and Alan Forrest
16 Rational Combinations of Polymyxins
with Other Antibiotics �������������������������������������������������������������������� 251
Phillip J. Bergen, Nicholas M. Smith, Tyler B. Bedard,
Zackery P. Bulman, Raymond Cha, and Brian T. Tsuji
17 Toxicity in Patients�������������������������������������������������������������������������� 289
Jason M. Pogue and Vincent H. Tam
18 Mechanisms of Polymyxin-Induced Nephrotoxicity�������������������� 305
Mohammad A. K. Azad, Roger L. Nation, Tony Velkov,
and Jian Li
19 Anti-endotoxin Properties of Polymyxin
B-immobilized Fibers���������������������������������������������������������������������� 321
Tohru Tani, Tomoharu Shimizu, Masaji Tani, Hisataka Shoji,
and Yoshihiro Endo
20 Discovery of Novel Polymyxin-Like Antibiotics���������������������������� 343
Tony Velkov and Kade D. Roberts
21 Conclusion���������������������������������������������������������������������������������������� 363
Roger L. Nation

Index���������������������������������������������������������������������������������������������������������� 365
Reviving Polymyxins:
Achievements, Lessons 1
and the Road Ahead

Jian Li

Abstract Keywords
Antibiotic resistance has become the most sig- Antibiotic resistance · Drug discovery ·
nificant threat to human health across the Polymyxin · Pharmacology · Clinical use
globe. Polymyxins are often used as the only
available therapeutic option against Gram-­
negative ‘superbugs’, namely Acinetobacter
baumannii, Pseudomonas aeruginosa and 1.1 Introduction
Klebsiella pneumoniae. The limited pharma-
cological and clinical knowledge on the poly- One of the most outstanding achievements of
myxins in the old literature substantially modern medicine was the development of antibi-
limited optimizing their clinical use. The cur- otics for treatment of bacterial infections that
rent chapter provides a general introduction to were widely fatal. Antibiotics are regarded as
this first-ever polymyxin book which compre- ‘miracle drugs’ and have significantly decreased
hensively reviews the significant progress over mortality worldwide over the last century [1].
the last two decades in the chemistry, microbi- They have made many complicated surgical pro-
ology, pharmacology, clinical use and drug cedures and treatments possible; unfortunately,
discovery of polymyxins. In particular, recent an increasing number of infections (e.g. pneumo-
pharmacological results have led to the first nia) are becoming more and more difficult to
scientifically-based dosing recommendations treat, as our current antibiotics are losing their
and facilitated the discovery of new-­generation efficacy. Over the last three decades resistance to
polymyxins. Future challenges in polymyxin these ‘magic bullets’ has presented the most sig-
research are highlighted, aiming at improving nificant threat to human health globally. If proac-
the clinical utility of this last-line defence. tive solutions are not found to prevent the
widespread antibiotic resistance, it is estimated
that by 2050 approximately 10 million people per
year will die of antimicrobial-resistant infections,
which is more than the number of people dying
J. Li (*) from any other type of disease (Fig. 1.1) [2].
Biomedicine Discovery Institute, Infection & Antibiotic resistance causes increased mortal-
Immunity Program and Department of Microbiology,
ity, longer hospital stays and higher medical
Monash University, Clayton Campus,
Melbourne, VIC, Australia costs. Globally, the cost of antimicrobial resis-
e-mail: jian.li@monash.edu tance is enormous in terms of the economy and

© Springer Nature Switzerland AG 2019 1


J. Li et al. (eds.), Polymyxin Antibiotics: From Laboratory Bench to Bedside, Advances in
Experimental Medicine and Biology 1145, https://doi.org/10.1007/978-3-030-16373-0_1
2 J. Li

Fig. 1.1 Predicted


global deaths due to
antimicrobial-resistant
infections every year,
compared to other major
diseases [2]

human health [3, 4]. It is predicted that a cumula- value. However, developing new antibiotics is
tive US$100 trillion of economic output by 2050 astonishingly less attractive, as the expected net
is at risk due to antimicrobial resistance [2]. present value for the new antibiotics launched
Based upon the projections of the world economy during 2014–2016 is −$100 million while with a
in 2017–2050, The World Bank Group estimated financial risk of $500 million [9]. The World
that antibiotic resistance could cost the world Health Organization (WHO) has urged all gov-
economy $1 trillion every year by 2050 [5]. A ernment sectors and society to act on antibiotic
recent study showed that the total economic cost resistance. In 2017, WHO identified a list of pri-
of antibiotic resistance in Staphylococcus aureus, ority pathogens which are resistant to the major-
Escherichia coli, Klebsiella pneumoniae, ity of currently available antibiotics and urgently
Acinetobacter baumannii and Pseudomonas require new therapeutic options (Fig. 1.2) [10].
aeruginosa reached $2.8 billion per year in the As shown in the WHO Priority Pathogen List,
US [6]. Furthermore, antibiotic resistance carbapenem-resistant Gram-negative A. bauman-
increases poverty worldwide and affects poorest nii, P. aeruginosa and K. pneumoniae are particu-
countries the most [5]. larly problematic, as efficacious therapeutic
Worryingly, many large pharmaceutical com- options are quickly diminishing against life-­
panies have left the antibiotic market, because the threatening infections caused by these pathogens
development of new antibiotics is scientifically [10]. All three ‘superbugs’ can develop resistance
challenging and not as profitable as for drugs to almost all major classes of antibiotics via mul-
used to treat chronic conditions and lifestyle tiple mechanisms. A recent study investigated
issues [7–9]. A recent report reviewed the major antibiotic resistance in A. baumannii infections
pharmaceutical launches between 2014 and 2016 with inpatients or outpatients from 54 studies (35
across a range of therapeutic areas [9]. It is evi- from the Organisation for Economic Co-operation
dent that in anti-cancer drugs the risk of losing and Development [OECD] countries with 57,188
$450 million on a new molecular entity is easily bacterial isolates and 19 from non-OECD coun-
offset by $8,200 million expected net present tries with 7,395 isolates) by searching Medline,
1 Reviving Polymyxins: Achievements, Lessons and the Road Ahead 3

Fig. 1.2 A list of


priority pathogens
identified by WHO for
research and
development of new
antibiotics [10]

Fig. 1.3 Antibiotic


resistance in A.
baumannii. (a)
Prevalence of multidrug-­
resistance to major
antibiotics except
colistin and tigecycline
during 2000 and 2016 in
the Organisation for
Economic Co-operation
and Development
(OECD) and non-OECD
countries. (b) Increasing
antibiotic resistance in
OECD and non-OECD
countries between 2000
and 2016 [11]. http://
creativecommons.org/
licenses/by/4.0/

Embase, Web of Science, and Cochrane data- is evident in both OECD and non-OECD coun-
bases [11]. Strikingly, a high prevalence of tries, and a faster increase was clearly shown in
multidrug-­resistance in A. baumannii infections OECD countries over the last decade (Fig. 1.3).
4 J. Li

In general, resistance to most commonly used rins and aminoglycosides has reached >50% in
antibiotics in A. baumannii is >70% in both Egypt [13] and a number of eastern European
OECD and non-OECD countries [11]. P. aerugi- countries (Fig. 1.5) [12]. Sadly, few novel antibi-
nosa is intrinsically resistant to many antibiotics otics will become available for these very prob-
and is a major cause of healthcare-associated lematic Gram-negative pathogens in the near
infections globally. The European Centre for future [2, 14, 15]. In many cases, polymyxins
Disease Prevention and Control (ECDC) has one have to be used as the last resort for the treatment
of the most comprehensive antibiotic susceptibil- of life-threatening infections caused by A. bau-
ity surveillance programs in the world. According mannii, P. aeruginosa and K. pneumoniae
to its latest antibiotic surveillance report, the rate [15–20].
of resistance to three or more major classes of Polymyxins (i.e. colistin [also known as poly-
antipseudomonal antibiotics (including piperacil- myxin E] and polymyxin B) entered the clinic in
lin/tazobactam, ceftazidime, carbapenems, fluo- the late 1950s, but their use waned in the 1970s
roquinolones and aminoglycosides) is due to the potential nephrotoxicity and neurotox-
disturbingly high, in particular in eastern and icity [15, 16, 18, 21, 22]. Since the 2000s, how-
south-eastern European countries (Fig. 1.4) [12]. ever, clinicians have had to increasingly use
K. pneumoniae is another major pathogen which colistin and polymyxin B as one of the very few
can become resistant to multiple classes of anti- therapeutic options for Gram-negative ‘super-
biotics and cause serious hospital-acquired infec- bugs’. This chapter serves as an introduction to
tions, such as pneumonia, urinary tract infections this book and provides an overview of the micro-
and bloodstream infections. The resistance rate to biology, chemistry, pharmacology, clinical use,
fluoroquinolones, third-generation cephalospo- and drug discovery of polymyxins.

Fig. 1.4 Antibiotic resistance in P. aeruginosa to three or more classes among piperacillin/tazobactam, ceftazidime,
carbapenem, fluoroquinolones and aminoglycosides in Europe in 2017 [12]
1 Reviving Polymyxins: Achievements, Lessons and the Road Ahead 5

Fig. 1.5 Percentage (%) of K. pneumoniae isolates resistant to fluoroquinolones, third-generation cephalosporins and
aminoglycosides in Europe in 2017 [12]

1.2 Polymyxins: A New ‘Old’ intravenous colistin based on the latest pre-­
Class of Antibiotics clinical and clinical pharmacokinetic, pharmaco-
dynamic and toxicodynamic information; and (3)
Colistin and polymyxin B (Fig. 1.6) were the development of new-generation polymyxins
approved for clinical use in the late 1950s and informed by the newest chemical and pharmaco-
were not subject to contemporary drug logical results. In this book, we invited interna-
­development evaluations and regulatory scrutiny. tional experts to provide comprehensive reviews
As polymyxins have been off patent for many on all of these major topics on polymyxins, with
years and were not widely used between the the aim of assembling the information needed to
1970s and 1990s, most pharmacological infor- facilitate optimizing their clinical use and
mation on colistin and polymyxin B is from the the development of novel, safer polymyxins.
studies conducted in academic research groups This book starts with a comprehensive review
over the last two decades. Figure 1.7 clearly on multidrug-resistance in Gram-negative ‘super-
shows that polymyxins have attracted significant bugs’ (Chap. 2) which highlights the urgent need
research and clinical interest since the early to optimize the clinical use of both polymyxins
2000s, due to increasing need to use them against and minimize any potential emergence of resis-
multidrug-­resistant Gram-negative pathogens. A tance. An in-depth introduction on the history,
number of major achievements have been made antibacterial spectrum and chemistry of polymyx-
in the polymyxin field over the last two decades, ins (Chap. 3) provides key information to under-
including (1) a better understanding of the chem- stand how polymyxins kill bacterial cells (Chap.
istry, structure-­ activity-toxicity relationships, 4) and how bacteria develop resistance (Chap. 5).
and mechanisms of antibacterial activity, resis- To optimize the dosage regimens of polymyxins, it
tance and toxicity of polymyxins; (2) the first is essential to develop sensitive and accurate ana-
scientifically-­based dosing recommendations for lytical methods (Chap. 6), investigate the pharma-
6 J. Li

Fig. 1.6 Structures of polymyxin B and colistin. Dab, α,γ-­diaminobutyric acid

450

400
Number of papers in PubMed

350

300

250

200

150

100

50

0
2011
1947
1949
1951
1953
1955
1957
1959
1961
1963
1965
1967
1969
1971
1973
1975
1977
1979
1981
1983
1985
1987
1989
1991
1993
1995
1997
1999
2001
2003
2005
2007
2009

2013
2015
2017

Year

Fig. 1.7 Number of papers on polymyxins in PubMed by searching with “colistin[title] or polymyxin[title]” on 6 June
2019.
1 Reviving Polymyxins: Achievements, Lessons and the Road Ahead 7

cokinetics in animals (Chap. 7), characterize the option than intravenous administration, because of
pharmacodynamics using in vitro and animal the PK/PD considerations. However, the current
models (Chap. 8), and determine antibacterial sus- dosage regimens of inhaled colistin and polymyxin
ceptibility breakpoints (Chap. 9). The two differ- B are empirical and not based on PK/PD/TD infor-
ent conventions used to describe the dose of mation. The literature on polymyxin combination
colistin, the complex composition of polymyxin therapy versus monotherapy is confusing. Most
products, and the different pharmacopoeial stan- clinical studies evaluating the efficacy of poly-
dards have caused considerable confusion in dif- myxin combinations in the literature have over-
ferent parts of the world, and together with the looked the significant PK/PD issues due to the
outdated product information can affect the ability limited polymyxin exposure in the lungs after intra-
of clinicians to optimize the use of polymyxins in venous administration. PK/PD/TD principles must
patients (Chap. 10). Chapters 11, 12, 13, 14, 15, be considered when optimizing polymyxin combi-
and 16 review the latest achievements in improv- nation therapy, as it is not as simple as dosing mul-
ing the use of colistin, polymyxin B and potential tiple antibiotics together. To achieve synergistic
synergistic combinations in the clinic, which is a killing in vivo, all drugs should achieve optimal
major focus of this polymyxin book. As polymyx- exposure at the infection site at the right timing;
ins have a narrow therapeutic window and nephro- otherwise, polymyxin ‘combination’ therapy is
toxicity is the major dose-­limiting factor [17, 22, essentially monotherapy. As nephrotoxicity can
23], understanding their toxicities (Chap. 17) and occur in patients receiving intravenous polymyx-
mechanisms (Chap. 18) are crucial to ensuring ins, innovative approaches are warranted to
their optimum and safe use in the clinic. In addi- increase their therapeutic indices, thereby improv-
tion, the anti-endotoxin effect of polymyxins has ing the efficacy. Development of new-generation
been extensively evaluated for the treatment of polymyxins is challenging due to the narrow chem-
severe sepsis and septic shock (Chap. 19). Finally, ical space and the complex relations between the
Chap. 20 reviews the latest progress in developing chemical structure, antibacterial activity, different
new-generation polymyxins with better antibacte- resistance mechanisms, toxicity and PK (e.g.
rial activity and safety profiles, which is informed plasma protein binding). Taken together, collective
by the modern polymyxin pharmacology research. efforts are essential to address these challenges in
A number of major challenges and gaps in the coming years.
knowledge have been identified in this book. There
is an imperative to systematically evaluate the clin-
ical efficacy of intravenous colistimethate (an inac- 1.3 Summary
tive prodrug of colistin, see Chap. 3) and polymyxin
B against different types of infections (e.g. blood Almost 60 years after polymyxins were approved
and urinary tract infections) [24, 25]. A large clini- for clinical use, clinicians are now in a much bet-
cal PK/PD/TD study on intravenous polymyxin B ter position to determine appropriate dosage regi-
is being conducted in critically-ill patients funded mens for intravenous polymyxins in patients,
by the National Institutes of Health (ClinicalTrials. which is the result of extensive preclinical and
gov Identifier: NCT02682355). Hopefully, scien- clinical pharmacological investigations over the
tifically-based dosing recommendations will be last two decades. Since 2013, three international
available in the near future for intravenous poly- conferences have been held with the contribu-
myxin B in different types of patients. Considering tions of distinguished speakers worldwide, most
the narrow therapeutic window, prospective studies of whom are authors in this book, the first-ever
with therapeutic drug monitoring and adaptive on polymyxins. It is very encouraging that sub-
feedback control are needed for optimizing the use stantial progress has been made across all major
of both polymyxins in patients. For the treatment of areas of polymyxin research, and the list of high-­
MDR Gram-negative respiratory tract infections, priority issues and challenges identified at the
inhalation of polymyxins is very likely a better international polymyxin conferences becomes
8 J. Li

shorter. In this ‘Bad Bugs, No Drugs’ era, poly- 11. Xie R, Zhang XD, Zhao Q et al (2018) Analysis of global
prevalence of antibiotic resistance in Acinetobacter bau-
myxins will continue to play an important role in mannii infections disclosed a faster increase in OECD
the treatment of life-threatening infections caused countries. Emerg Microbes Infect 7:31
by Gram-negative ‘superbugs’. 12. European Centre for Disease Prevention and Control
(ECDC) (2018) Surveillance of antimicrobial resis-
tance in Europe – annual report of the European
antimicrobial resistance surveillance network (EARS-­
References net) 2017. ECDC, Stockholm
13. World Health Organization (WHO) (2017) Global
1. Armstrong GL, Conn LA, Pinner RW (1999) Trends antimicrobial resistance surveillance system (GLASS)
in infectious disease mortality in the United States report: early implementation 2016–2017. Geneva.
during the 20th century. JAMA 281:61–66 ISBN: 978-92-4-151344-9. http://apps.who.int/iris/
2. O’neill J (2015) Tackling a global health crisis: initial bitstream/handle/10665/259744/9789241513449-eng.
steps. Wellcome Trust, London. https://amr-review. pdf;jsessionid=9F6FFF4F4DA947B2346DFEAD918
org/sites/default/files/Report-52.15.pdf. Last accessed 80B04?sequence=1. Last accessed 12 April 2019
12 April 2019 14. Bush K, Courvalin P, Dantas G et al (2011) Tackling
3. Hofer U (2019) The cost of antimicrobial resistance. antibiotic resistance. Nat Rev Microbiol 9:894–896
Nat Rev Microbiol 17(1):3 15. Landman D, Georgescu C, Martin DA et al (2008)
4. Organisation for Economic Co-operation and Polymyxins revisited. Clin Microbiol Rev 21:449–465
Development (OECD) (2018) Stemming the super- 16. Li J, Nation RL, Turnidge JD et al (2006) Colistin:
bug tide. http://www.oecd.org/els/health-systems/ the re-emerging antibiotic for multidrug-resistant
Stemming-the-Superbug-Tide-Policy-Brief-2018.pdf. gram-negative bacterial infections. Lancet Infect Dis
Last accessed 12 April 2019 6:589–601
5. World Bank Group (2016) By 2050, drug-resistant 17. Nation RL, Garonzik SM, Thamlikitkul V et al (2017)
infections could cause global economic damage on Dosing guidance for intravenous colistin in critically-­
par with 2008 financial crisis. http://www.worldbank. ill patients. Clin Infect Dis 64:565–571
org/en/news/press-release/2016/09/18/by-2050-drug- 18. Poirel L, Jayol A, Nordmann P (2017) Polymyxins:
resistant-infections-could-cause-global-econo- antibacterial activity, susceptibility testing, and resis-
mic-damage-on-par-with-2008-financial-crisis. Last tance mechanisms encoded by plasmids or chromo-
accessed 12 April 2019 somes. Clin Microbiol Rev 30:557–596
6. Shrestha P, Cooper BS, Coast J et al (2018) 19. Velkov T, Roberts KD, Nation RL et al (2013)
Enumerating the economic cost of antimicrobial Pharmacology of polymyxins: new insights into an
resistance per antibiotic consumed to inform the eval- ‘old’ class of antibiotics. Future Microbiol 8:711–724
uation of interventions affecting their use. Antimicrob 20. Zavascki AP, Goldani LZ, Li J et al (2007) Polymyxin
Resist Infect Control 7:98 B for the treatment of multidrug-resistant patho-
7. Cooper MA, Shlaes D (2011) Fix the antibiotics pipe- gens: a critical review. J Antimicrob Chemother
line. Nature 472:32 60:1206–1215
8. Infectious Diseases Society of America (IDSA) (2004) 21. Li J, Nation RL, Milne RW et al (2005) Evaluation
Bad bugs, no drugs. Infectious Diseases Society of of colistin as an agent against multi-resistant gram-­
America, Alexandria, VA. https://www.idsociety.org/ negative bacteria. Int J Antimicrob Agents 25:11–25
globalassets/idsa/policy%2D%2Dadvocacy/current_ 22. Nation RL, Li J, Cars O et al (2015) Framework for
topics_and_issues/antimicrobial_resistance/10x20/ optimisation of the clinical use of colistin and poly-
statements-manually-added/070104-as-antibiotic- myxin B: the Prato polymyxin consensus. Lancet
discovery-stagnates-a-public-health-crisis-brews.pdf. Infect Dis 15:225–234
Last accessed 12 April 2019 23. Nation RL, Velkov T, Li J (2014) Colistin and poly-
9. The Boston Consulting Group (BCG) for the myxin B: are they like peas in a pod or chalk and
German Federal Ministry of Health (2017) Breaking cheese? Clin Infect Dis 59:88–94
through the wall: a call for concerted action on 24. Cheah SE, Wang J, Nguyen VT et al (2015) New
antibiotics research and development. Follow-up pharmacokinetic/pharmacodynamic studies of sys-
report for the German GUARD initiative. https:// temically administered colistin against Pseudomonas
www.bundesgesundheitsministerium.de/fileadmin/ aeruginosa and Acinetobacter baumannii in mouse
Dateien/5_Publikationen/Gesundheit/Berichte/ thigh and lung infection models: smaller response
GUARD_Follow_Up_Report_Full_Report_final.pdf. in lung infection. J Antimicrob Chemother
Last accessed 12 April 2019 70:3291–3297
10. World Health Organization (WHO) (2017) WHO 25. Landersdorfer CB, Wang J, Wirth V et al (2018)
priority pathogens list for R&D of new antibiotics. Pharmacokinetics/pharmacodynamics of systemi-
https://www.who.int/medicines/publications/WHO- cally administered polymyxin B against Klebsiella
PPL-Short_Summary_25Feb-ET_NM_WHO.pdf. pneumoniae in mouse thigh and lung infection mod-
Last accessed 12 April 2019 els. J Antimicrob Chemother 73:462–468
Multidrug-Resistant Gram-­
Negative Pathogens: The Urgent 2
Need for ‘Old’ Polymyxins

David L. Paterson and Robert A. Bonomo

Abstract Penicillin-resistant bacteria were detected within


Antibiotic resistance has presented a major the first decade of use of this antibiotic. More
health challenge in the world and many iso- than 70 years later, antibiotic use for hospitalized
lates of Enterobacteriaceae, Acinetobacter patients has switched to agents such as carbapen-
baumannii and Pseudomonas aeruginosa ems, quinolones, aminoglycosides and tigecy-
become resistant to almost all current antibiot- cline. Unfortunately, the epidemiology of
ics. This chapter provides an overview on the infections has changed so that bacteria resistant
mechanisms of antibiotic resistance in these to some or all of these antibiotics are now com-
Gram-negative pathogens and outlines the for- monplace in many institutions. Typically, units
midable problem of the genetics of bacterial with compromised patients and heavy antibiotic
resistance. Prevalent multidrug-resistance in use, such as intensive care units, hematology and
Gram-negative bacteria underscores the need transplant wards, and long-term stay units are
for optimizing the clinical use of the last-line those in which multidrug-resistant bacteria are
polymyxins. most common. Although attention in the past
focused on antibiotic-resistant Gram-positive
Keywords organisms (such as methicillin-resistant
Antibiotic resistance · Enterobacteriaceae · Staphylococcus aureus or vancomycin-resistant
Acinetobacter baumannii · Pseudomonas Enterococcus faecium), the problem bacteria
aeruginosa · polymyxin today are the Gram-negative bacteria. The
Enterobacteriaceae, Acinetobacter spp. and
Pseudomonas aeruginosa are common causes of
healthcare-acquired infections [1] and are fre-
quently resistant to commonly used antibiotics.
The purpose of this chapter is to outline the
mechanisms of resistance in these Gram-negative
pathogens, as a means of outlining the formida-
D. L. Paterson (*) ble problem of the genetics of bacterial resis-
University of Queensland Centre for Clinical tance. It underscores the need for polymyxins,
Research, Brisbane, QLD, Australia since this class of pathogens is not susceptible to
e-mail: paterson1@uq.edu.au
beta-lactams and related resistance mechanisms
R. A. Bonomo so frequently seen today.
Louis Stokes Cleveland VA Medical Center,
Cleveland, OH, USA

© Springer Nature Switzerland AG 2019 9


J. Li et al. (eds.), Polymyxin Antibiotics: From Laboratory Bench to Bedside, Advances in
Experimental Medicine and Biology 1145, https://doi.org/10.1007/978-3-030-16373-0_2
10 D. L. Paterson and R. A. Bonomo

2.1 Enterobacteriaceae producers of NDM and other MBLs are typically


resistant to carbapenems, penicillins, cephalo-
Carbapenem antibiotics have typically been sporins, fluoroquinolones and aminoglycosides.
regarded as highly stable to beta-lactamases, for This explains the necessity to use polymyxins in
example via extended-spectrum beta-lactamases significant infections due to NDM producers.
(ESBLs). However, over the last decade, many of The NDM-type β-lactamase was first isolated
the Enterobacteriaceae have become resistant to in 2009 from a Swedish patient returning from
carbapenems by way of production of carbapen- India [9]. The patient was infected with NDM
emase enzymes [2]. producing K. pneumoniae, resistant to multiple
Early reports of carbapenem-resistant antibiotics including all carbapenems. The blaNDM
Enterobacteriaceae were as a result of expression gene has now spread to all inhabited continents
of AmpC type beta-lactamases or ESBLs plus and is carried by multiple Gram-negative species
loss of outer-membrane proteins in K. pneu- [10]. NDM producing organisms have been
moniae [3]. However, carbapenem resistance in strongly linked with the Indian subcontinent
the Enterobacteriaceae has emerged over the last (India, Pakistan, Bangladesh and Nepal) [11].
15–20 years due to production of beta-lactamases China is also known to be a reservoir country for
known as carbapenemases [4]. In the United NDM producers, although surveillance data is not
States and some parts of Europe, the most fre- yet complete – at this stage it does not appear that
quently observed type of carbapenemase is the NDM producers are as widely prevalent in China
KPC type [5, 6]. KPC-producing strains are typi- as in the Indian subcontinent. The Balkan states
cally multidrug-resistant, being resistant to car- (for example, Serbia, Montenegro, and Bosnia-
bapenems, penicillins, cephalosporins, Herzegovina) may also be considered as a reser-
fluoroquinolones and aminoglycosides [6]. voir area for blaNDM acquisition since a number of
Therefore, polymyxins are one of the few options cases have been reported with no travel history to
available for use against KPC producers. A single Asia [10]. In general, travel appears to be the
clone of KPC-producing K. pneumoniae (known major means by which NDM producing bacteria
as ST 258 by multilocus sequence typing have spread throughout the world. Europe pro-
(MLST)), has been found in the United States, vided the first case in 2009 in Sweden and shortly
Israel and some parts of Europe (particularly after, many other countries began reporting travel
Greece and Italy) [7, 8]. This indicates that a related NDM acquisition from the Indian Sub-
hospital-­adapted clone of KPC-producing K. continent or the Balkan states. Unlike the case
pneumoniae was transferred from person to per- with KPC-producing K. pneumoniae, various K.
son as a result of breakdown in infection control pneumoniae sequence types (STs) have been
measures. A new beta-lactamase inhibitor, avi- reported to harbor blaNDM [10].
bactam, does have activity against the KPC beta-­ A variety of other MBLs have been found to
lactamase. However, data are limited as to its lead to carbapenem resistance in the
clinical effectiveness against KPC producers, and Enterobacteriaceae. These include the VIM-type
it remains to be seen as to whether it will replace (with worldwide distribution, but particularly
polymyxins as drug of choice for KPC-producing noteworthy in Greece), the IMP-type (with IMP-4
organisms. particularly prominent in Australia) and the SPM-
Although KPC has been found in China and type (almost exclusively found in Brazil).
other parts of Asia, resistance of the Standard susceptibility testing may sometimes
Enterobacteriaceae to carbapenems in Asia is categorize KPC- or MBL-producing
more frequently due to production of Enterobacteriaceae as susceptible to carbapen-
metallo-beta-lactamases (MBLs) than due to ems. This issue is particularly pertinent to another
KPC. A variety of MBLs have been detected. group of carbapenemases found in the
Foremost amongst these is the NDM beta-­ Enterobacteriaceae – OXA-48, and related
lactamase [9]. Like the KPC-type beta-lactamase, enzymes [12]. The OXA-48 like carbapenemases
2 Multidrug-Resistant Gram-Negative Pathogens: The Urgent Need for ‘Old’ Polymyxins 11

are widespread in North Africa, the Middle East Carbapenem resistance in Acinetobacter spp.
and India [12, 13]. There has now been signifi- is a common indication for polymyxin use. As is
cant spread to Europe [12]. the case with the Enterobacteriaceae, carbape-
The genes encoding the carbapenemases fre- nem resistance is typically mediated by produc-
quently reside on mobile genetic elements (such tion of carbapenemases.
as plasmids), which are capable of transferring The OXA-type beta-lactamases (especially
resistance genes from one bacterial cell to another OXA-23) are the most common mechanisms of
[10]. Other resistance genes which can be co-­ carbapenem resistance in Acinetobacter.
harbored on the same genetic elements as car- Although the KPC type carbapenemases are
bapenemases include ESBLs, AmpC, quinolone widely found in the Enterobacteriaceae, they are
resistance mechanisms, aminoglycoside modify- rarely found in Acinetobacter. However, the
ing enzymes and 16S ribosomal RNA methyl- OXA-type carbapenemases, especially OXA-23,
ases. Chromosomally encoded mechanisms may predominate. OXA-23 was first isolated in
also occur in strains with mobile genetic ele- Acinetobacter spp. in the United Kingdom in
ments. For example, quinolone resistance in 1985 [14, 15]. This carbapenemase is typically
Enterobacteriaceae is usually due to chromosom- found in an internationally prevalent clone
ally encoded alterations in target enzymes (DNA termed international clone (IC) 2. OXA-27 and
gyrase and/or topoisomerase IV) or to impaired OXA-49 are closely related enzymes that make
access to the target enzymes, occurring either up the blaOXA-23 gene cluster in A. baumannii
because of changes in porin expression or because [14]. Other OXA-type genes may have carbapen-
of efflux mechanisms. emase activity including blaOXA-24- (OXA-24,
The end-result of the proliferation of this mul- -25, -26, -40) and the blaOXA-58-like [14] car-
titude of resistance mechanisms is truly bapenemase genes. Additionally, a chromosom-
multidrug-­resistant Enterobacteriaceae. It is not ally encoded gene, blaOXA-51, is intrinsic to A.
surprising, therefore, that in settings where baumannii – its contribution to carbapenem
carbapenem-­ resistant Enterobacteriaceae are resistance is dependent on the presence of an
highly prevalent, polymyxin use becomes a insertion sequence, ISAba1 [16]. In the absence
necessity. of this insertion sequence, blaOXA-51 does not lead
to carbapenem resistance [14].
MBLs have also been well described in
2.2 Acinetobacter spp. Acinetobacter spp., although they are not as fre-
quent a cause of resistance as OXA-23 [14].
A. baumannii and newly described species, A. However, the carbapenem hydrolyzing activity of
pittii and A. nosocomialis, also possess a wide the MBLs is typically much more potent than that
array of antimicrobial resistance mechanisms. of the OXA-type carbapenemases [14]. The
Intrinsically, Acinetobacter species are resistant NDM type MBLs are particularly noteworthy
to first and second generation cephalosporins, since they may have originated within the genus
aztreonam and ertapenem, due to a combination Acinetobacter [10]. As noted previously, the
of poor permeability to these antibiotics and NDM enzymes are prominent in the Indian sub-
intrinsic beta-lactamase production. Antibiotics continent but have now spread widely. IMP, VIM
with activity against wild-type strains of and SIM MBLs have also been found in
Acinetobacter include sulbactam, meropenem, Acinetobacter spp. [17]. Additionally,
ciprofloxacin, aminoglycosides, tetracyclines, Acinetobacter isolates may co-produce both
tigecycline and trimethoprim/sulfamethoxazole MBL and OXA type carbapenemases [14]. Most
[14]. Acquired resistance mechanisms frequently commonly, MBLs produced by Acinetobacter
originate from Pseudomonas spp., E. coli and are encoded within integrons, especially class 1
other Gram-negative species, and may be local- integrons. These genetic structures typically
ized to large resistance islands [14]. encode a wide variety of resistance genes, and
12 D. L. Paterson and R. A. Bonomo

contribute to the multidrug resistance typical of produced by P. aeruginosa, while OXA-type


Acinetobacter [14]. beta-lactamases are rarely seen [10, 19].
Carbapenemase-producing Acinetobacter iso- Quinolone resistance in P. aeruginosa is typi-
lates are usually resistant to all beta-lactam anti- cally found to be due to mutations in the chromo-
biotics. Aminoglycoside resistance is also somally encoded QRDRs [19]. First-step
common, via the production of aminoglycoside mutations occurring in the gyrA QRDR appear to
modifying enzymes or 16S rRNA methylases. be the most significant in causing quinolone
Ciprofloxacin resistance is typically mediated by resistance, while subsequent mutations are
changes in the chromosomally encoded quino- believed to further decrease susceptibility levels.
lone resistance determining regions (QRDRs). Aminoglycoside resistance in P. aeruginosa is
Upregulated efflux pumps may also contribute to associated with efflux by the MexXY-OprM
aminoglycoside, quinolone, tigecycline and beta-­ transporter, aminoglycoside modifying enzymes
lactam resistance. Finally, the sul gene may lead or 16S rRNA methyltransferases. Typically,
to sulfamethoxazole resistance. The end-result of mechanisms of quinolone and aminoglycoside
this multiplicity of resistance genes may be resistance occur in isolates with OprD loss and/or
Acinetobacter strains resistant to all antibiotics. production of carbapenemases leading to multi-
Hence, polymyxins play a major role in the arma- drug or extensively drug resistant (XDR) strains.
mentarium against the carbapenem-resistant Given that tigecycline is ineffective against P.
Acinetobacter isolates commonly observed in aeruginosa by way of intrinsic efflux mecha-
clinical practice. Unfortunately, an A. baumannii nisms, the polymyxins are one of the very few
strain has now been described which is resistant treatment options available for multidrug resis-
to polymyxins and all commercially available tant strains.
antibiotics [18].

2.4 Conclusions
2.3 Pseudomonas aeruginosa
A wide variety of issues have contributed to the
P. aeruginosa is an organism with enhanced viru- problem of multidrug resistance in Gram-­
lence characteristics and is the sixth most com- negative bacilli. Pharmaceutical company disin-
monly isolated organism responsible for vestment in antibiotic discovery has led to fewer
hospital-acquired infections [1]. Unlike the case new options for clinical use. At the same time,
with Enterobacteriaceae or Acinetobacter spp., proliferation of genetic elements encoding resis-
the most common mechanism of carbapenem tance has continued unabated. Exacerbators of
resistance in P. aeruginosa appears to be muta- this problem have included heavy agricultural
tional loss of the OprD porin [19]. The primary use of antibiotics, over the counter availability of
function of OprD is importation of arginine, but it antibiotics and environmental contamination by
is also the major entry point for carbapenems antibiotics themselves as well as antibiotic resis-
[19]. Mutations in oprD can result in loss of porin tant organisms in water, food and hospital envi-
function. Thus, uptake of carbapenems by P. ronments. In particular, the recent discovery of
aeruginosa is substantially reduced and typically plasmid-mediated polymyxin resistance via mul-
confers resistance to this antibiotic class. Efflux tiple mcr genes indicate that polymyxin resis-
mechanisms such as MexAB-OprM, MexXY-­ tance exists in food animals and patients [20–22].
OprM, and the regulator MexZ may play a con- The polymyxins are not perfect treatment options
tributory role in carbapenem resistance, as may by any means. However, they have become the
carbapenemase production. MBLs (such as VIM only option for many patients in this current era
or NDM) and KPC-type beta-lactamases may be of resistance.
2 Multidrug-Resistant Gram-Negative Pathogens: The Urgent Need for ‘Old’ Polymyxins 13

References 11. Castanheira M, Deshpande LM, Farrell SE, Shetye S,


Shah N, Jones RN (2013) Update on the prevalence
and genetic characterization of NDM-1-producing
1. Magill SS, Edwards JR, Bamberg E et al (2014)
Enterobacteriaceae in Indian hospitals during 2010.
Multistate point prevalence survey of healthcare-­
Diagn Microbiol Infect Dis 75:210–213
associated infections. N Engl J Med 370:1198–1208
12. Poirel L, Potron A, Nordmann P (2012) OXA-48-like
2. Peleg AY, Hooper DC (2010) Hospital-acquired infec-
carbapenemases: the phantom menace. J Antimicrob
tions due to Gram-negative bacteria. N Engl J Med
Chemother 67:1597–1606
362:1804–1813
13. Zowawi HM, Balkhy HH, Walsh TR, Paterson DL
3. Bradford PA, Urban C, Mariano N, Projan SJ, Rahal
(2013) β-Lactamase production in key gram-negative
JJ, Bush K (1997) Imipenem resistance in Klebsiella
pathogen isolates from the Arabian Peninsula. Clin
pneumoniae is associated with the combination of
Microbiol Rev 26:361–380
ACT-1, a plasmid-mediated AmpC beta-lactamase,
14. Peleg AY, Seifert H, Paterson DL (2008)
and the loss of an outer membrane protein. Antimicrob
Acinetobacter baumannii: emergence of a success-
Agents Chemother 41:563–569
ful pathogen. Clin Microbiol Rev 21:538–582
4. World Health Organization (WHO) (2017) WHO prior-
15. Paton R, Miles RS, Hood J, Amyes SG, Miles RS,
ity pathogens list for R&D of new antibiotics [Online].
Amyes SG (1993) ARI 1: beta-lactamase-mediated
Available: http://www.who.int/medicines/publica-
imipenem resistance in Acinetobacter baumannii. Int
tions/WHO-PPL-Short_Summary_25Feb-ET_NM_
J Antimicrob Agents 2:81–87
WHO.pdf?ua=1. Accessed 28 Apr 2019
16. Turton JF, Ward ME, Woodford N, Kaufmann ME,
5. Yigit H, Queenan AM, Anderson GJ et al (2001)
Pike R, Livermore DM, Pitt TL (2006) The role of
Novel carbapenem-hydrolyzing beta-lactamase, KPC-­
ISAba1 in expression of OXA carbapenemase genes
1, from a carbapenem-resistant strain of Klebsiella
in Acinetobacter baumannii. FEMS Microbiol Lett
pneumoniae. Antimicrob Agents Chemother
258:72–77
45:1151–1161
17. Walsh TR, Toleman MA, Poirel L, Nordmann P
6. Tumbarello M, Losito AR, Giamarellou H (2018)
(2005) Metallo-beta-lactamases: the quiet before the
Optimizing therapy in carbapenem-resistant
storm? Clin Microbiol Rev 18:306–325
Enterobacteriaceae infections. Curr Opin Infect Dis
18. Gottig S, Gruber TM, Higgins PG, Wachsmuth M,
31:566–577
Seifert H, Kempf VA (2014) Detection of pan drug-­
7. Satlin MJ, Chen L, Patel G, Gomez-Simmonds A,
resistant Acinetobacter baumannii in Germany.
Weston G, Kim AC, Seo SK, Rosenthal ME, Sperber
J Antimicrob Chemother 69:2578–2579
SJ, Jenkins SG, Hamula CL, Uhlemann AC, Levi
19. Bonomo RA, Szabo D (2006) Mechanisms of
MH, Fries BC, Tang YW, Juretschko S, Rojtman AD,
multidrug resistance in Acinetobacter spp. and
Hong T, Mathema B, Jacobs MR, Walsh TJ, Bonomo
Pseudomonas aeruginosa. Clin Infect Dis 43(Suppl
RA, Kreiswirth BN (2017) Multicenter clinical and
2):S49–S56
molecular epidemiological analysis of bacteremia due
20. Liu YY, Wang Y, Walsh TR, Yi LX, Zhang R,
to carbapenem-resistant Enterobacteriaceae (CRE) in
Spencer J, Doi Y, Tian G, Dong B, Huang X, Yu
the CRE Epicenter of the United States. Antimicrob
LF, Gu D, Ren H, Chen X, Lv L, He D, Zhou H,
Agents Chemother 61: pii: e02349–16
Liang Z, Liu JH, Shen J (2016) Emergence of
8. Navon-Venezia S, Leavitt A, Schwaber MJ et al
plasmid-mediated colistin resistance mechanism
(2009) First report on a hyperepidemic clone of KPC-­
MCR-1 in animals and human beings in China: a
3-­producing Klebsiella pneumoniae in Israel geneti-
microbiological and molecular biological study.
cally related to a strain causing outbreaks in the United
Lancet Infect Dis 16:161–168
States. Antimicrob Agents Chemother 53:818–820
21. Schwarz S, Johnson AP (2016) Transferable resis-
9. Yong D, Toleman MA, Giske CG et al (2009)
tance to colistin: a new but old threat. J Antimicrob
Characterization of a new metallo-β-lactamase gene,
Chemother 71:2066–2070
blaNDM-1, and a novel erythromycin esterase gene car-
22. Wise MG, Estabrook MA, Sahm DF, Stone GG,
ried on a unique genetic structure in Klebsiella pneu-
Kazmierczak KM (2018) Prevalence of mcr-type
moniae sequence type 14 from India. Antimicrob
genes among colistin-resistant Enterobacteriaceae
Agents Chemother 53:5046–5054
collected in 2014–2016 as part of the INFORM
10. Wailan AM, Paterson DL (2014) The spread and
global surveillance program. PLoS One
acquisition of NDM-1: a multifactorial problem.
13:e0195281
Expert Rev Anti-Infect Ther 12:91–115
History, Chemistry
and Antibacterial Spectrum 3
Tony Velkov, Philip E. Thompson,
Mohammad A. K. Azad, Kade D. Roberts,
and Phillip J. Bergen

Abstract mannii, and Klebsiella pneumoniae. This


Polymyxins are naturally occurring cyclic chapter covers the history, chemistry and anti-
lipopeptides that were discovered more than bacterial spectrum of these very important
60 years ago. They have a narrow antibacterial last-line lipopeptide antibiotics.
spectrum, which is mainly against Gram-­
negative pathogens. The dry antibiotic pipe- Keywords
line, together with the increasing incidence of Discovery of polymyxins · Aerosporin ·
bacterial resistance in the clinic, has been Chemical structure · Methanesulphonate ·
dubbed ‘the perfect storm’. This has forced a Antibacterial spectrum
re-evaluation of ‘old’ antibiotics, in particular
the polymyxins, which retain activity against
many multidrug-resistant (MDR) Gram-­
negative organisms. As a consequence, poly- 3.1 History
myxin B and colistin (polymyxin E) are now
used as the last therapeutic option for infec- 3.1.1 Discovery
tions caused by ‘superbugs’ such as
Pseudomonas aeruginosa, Acinetobacter bau- The polymyxins are a family of chemically dis-
tinct antibiotics produced by the widely distrib-
T. Velkov uted Gram-positive spore-forming soil bacterium
Department of Pharmacology and Therapeutics, Paenibacillus polymyxa (previously known as
University of Melbourne, Melbourne, Australia Bacillus polymyxa) (Table 3.1). They were first
P. E. Thompson identified in the 1940s simultaneously by three
Medicinal Chemistry, Monash Institute of different research groups working independently
Pharmaceutical Sciences, Monash University, in the field of antibiotic discovery [1–3]. Initially,
Melbourne, Australia
Benedict and Langlykke at the Northern Regional
M. A. K. Azad · K. D. Roberts (*) Research Laboratories in the United States pub-
Monash Biomedicine Discovery Institute, Monash
University, Melbourne, Australia lished a paper in July of 1947 describing the ant-­
e-mail: kade.roberts@monash.edu bacterial properties of crude liquid cultures of
P. J. Bergen (*) Paenibacillus polymyxa [1]. Later that month
Centre for Medicine Use and Safety, Monash Institute Stansley, Shepherd and White at the Stamford
of Pharmaceutical Sciences, Monash University, Research Laboratories of the American Cyanamid
Melbourne, Australia
Company in the United States published a paper
e-mail: phillip.bergen@monash.edu

© Springer Nature Switzerland AG 2019 15


J. Li et al. (eds.), Polymyxin Antibiotics: From Laboratory Bench to Bedside, Advances in
Experimental Medicine and Biology 1145, https://doi.org/10.1007/978-3-030-16373-0_3
16 T. Velkov et al.

Table 3.1 The chemical structures of the naturally occurring polymyxins

Polymyxin Fatty-acyl group Pos 3 Pos 6 Pos 7 Pos 10


A1 (S)-6-methyloctanoyl D-Dab D-Leu L-Thr L-Thr
A2 6-methylheptanoyl D-Dab D-Leu L-Thr L-Thr
B1 (S)-6-methyloctanoyl L-Dab D-Phe L-Leu L-Thr
B2 6-methylheptanoyl L-Dab D-Phe L-Leu L-Thr
B3 octanoyl L-Dab D-Phe L-Leu L-Thr
B4 heptanoyl L-Dab D-Phe L-Leu L-Thr
B5 nonanoyl L-Dab D-Phe L-Leu L-Thr
B6 3-hydroxy-6-methyloctanoyla L-Dab D-Phe L-Leu L-Thr
B1-Ile (S)-6-methyloctanoyl L-Dab D-Phe L-Ile L-Thr
(Circulin A)
B2-Ile 6-methylheptanoyl L-Dab D-Phe L-Ile L-Thr
(Circulin A)
Dab3-B1 (S)-6-methyloctanoyl D-Dab D-Phe L-Leu L-Thr
Dab3-B2 6-methylheptanoyl D-Dab D-Phe L-Leu L-Thr
C1† 6-methyloctanoylb L/D-Dab D-Phe L-Thr L-Thr
C2† 6-methylheptanoyl L/D-Dab D-Phe L-Thr L-Thr
D1 (S)-6-methyloctanoyl D-Ser D-Leu L-Thr L-Thr
D2 6-methylheptanoyl D-Ser D-Leu L-Thr L-Thr
E1 (S)-6-methyloctanoyl L-Dab D-Leu L-Leu L-Thr
(Colistin A)
E2 6-methylheptanoyl L-Dab D-Leu L-Leu L-Thr
(Colistin B)
E3 octanoyl L-Dab D-Leu L-Leu L-Thr
E4 heptanoyl L-Dab D-Leu L-Leu L-Thr
E7 7-methyloctanoyl L-Dab D-Leu L-Leu L-Thr
E1-Ile (S)-6-methyloctanoyl L-Dab D-Leu L-Ile L-Thr
E1-Val (S)-6-methyloctanoyl L-Dab D-Leu L-Val L-Thr
E1-Nva (S)-6-methyloctanoyl L-Dab D-Leu L-Nva L-Thr
E2-Ile 6-methylheptanoyl L-Dab D-Leu L-Ile L-Thr
E2-Val 6-methylheptanoyl L-Dab D-Leu L-Val L-Thr
E8-Ile 7-methylnonanoyl L-Dab D-Leu L-Ile L-Thr
F† 6-methyloctanoylb L/D-Dab D-Leu/D-Ile L-Leu/L-Ile/L-Ser L-Leu/L-Ile/L-Ser
F† 6-methylheptanoyl L/D-Dab D-Leu/D-Ile L-Leu/L-Ile/L-Ser L-Leu/L-Ile/L-Ser
F† octanoyl L/D-Dab D-Leu/D-Ile L-Leu/L-Ile/L-Ser L-Leu/L-Ile/L-Ser
M1 (S)-6-methyloctanoyl D-Dab D-Leu L-Thr L-Thr
(Mattacin)
M2 6-methylheptanoyl D-Dab D-Leu L-Thr L-Thr
(Mattacin)
P1 (S)-6-methyloctanoyl D-Dab D-Phe L-Thr L-Thr
P2 6-methylheptanoyl D-Dab D-Phe L-Thr L-Thr
S1 6-methyloctanoylb D-Ser D-Phe L-Thr L-Thr
T1 6-methyloctanoylb L-Dab D-Phe L-Leu L-Leu
T2 6-methylheptanoyl L-Dab D-Phe L-Leu L-Leu
L-Dab = L-2,4-diaminobutyric acid, D-Dab = D-2,4-diaminobutyric acid, D-Phe = D-phenylalanine, L-Leu = L-Leucine,
L-Ile = L-Isoleucine, L-Val = L-Valine, L-Nva = L-Norvaline, L-Ser = L-Serine, D-Ser = D-Serine, L-Thr = L-Threonine
a
stereochemistry at C3 and C6 not confirmed, † position of amino acid residues is speculative
b
stereochemistry at C6 not confirmed
3 History, Chemistry and Antibacterial Spectrum 17

describing the isolation and partial purification of a new species isolated from a soil sample in Japan
an antibiotic substance from Paenibacillus poly- [18]. Colistin was originally thought to be dis-
myxa which they designated ‘Polymyxin’ [2]. tinct from polymyxins, although the striking
This organism produced on agar a wide zone of pharmacological and chemical similarities of
inhibition of the Gram-negative pathogen colistin to the entire polymyxin group of antibiot-
Salmonella schottmuelleri. The ‘polymyxin’ ics were recognized from the outset [19–21]. It
entity was unique in its remarkable specificity for was eventually determined that colistin was
Gram-negative bacteria, which distinguished it structurally identical to polymyxin E and that
from all antibiotics previously reported. In they were in fact the same compound [22–24];
August of 1947 Brownlee and co-workers at the colistin, however, was the name ultimately
Wellcome Physiological Research laboratory in adopted in the literature. During this period the
England published their work on the identifica- exact chemical structures of the polymyxins
tion of an antibiotic substance from an organism remained speculative [12–14, 25]. It was known
identified as Bacillus aerosporus, isolated from that that they were peptides and possibly cyclic in
the soil of a market garden in Surry in 1946 [3]. nature. Individual amino acid residues had been
They initially called this antibiotic ‘Aerosporin’ identified and it was also established that they
and like the antibiotic ‘Polymyxin’, Aerosporin contained a fatty acyl group that had been identi-
had selective antimicrobial activity against fied as the S-6-methyloctanoyl acyl group. In
Gram-­negative bacteria. Brownlee and Bushby 1954, Hausmann and Craig made the discovery
went on to further identify the chemotherapeutic that polymyxin B was in fact composed of two
and pharmacological properties of ‘Aerosporin’ individual peptide components that differed only
showing that the substance they had isolated was in the structures of the fatty-acyl groups they con-
a basic peptide [4]. Subsequently, researchers at tained [26]. These two peptide components were
both the Stamford and Wellcome labs determined labelled polymyxin B1 and B2 (Table 3.1). It was
that the three groups were working with different soon established that the presence of multiple
strains of P. polymyxa and that the antibiotic peptide components with variations in their struc-
called ‘Polymyxin’ was also a basic peptide that tures, primarily their fatty-acyl component, was a
was chemically distinct from ‘Aerosporin’ yet feature common to all of the polymyxin groups.
had a very similar antimicrobial spectrum and In 1963, Suzuki and co-workers at the Osaka
biological activity. It was concluded that the two Univeristy in Japan finally determined the abso-
antibiotics belonged to the same family of antibi- lute chemical structures for polymyxin B1, poly-
otic compounds [5–15]. By international agree- myxin B2 and colistin A (polymyxin E1) followed
ment the generic name of ‘polymyxin’ was by colistin B (polymyxin E2) in 1964 (Table 3.1)
adopted for all the antibiotics derived from P. [27]. They went on to also confirm the structures
polymyxa and a nomenclature was developed that of polymyxin D1 and D2 (Table 3.1) [28]. These
described the chemically distinct groups of anti- polymyxins were all identified as being cyclic
biotics, which comprise the polymyxin family lipopeptides. Since the initial discovery of the
[16, 17]. With this new nomenclature ‘Aerosporin’ polymyxin A, B, C, D and E groups of
became known as polymyxin A, while lipopeptides, five other groups of polymyxins
­
‘Polymyxin’ became known as polymyxin containing multiple unique lipopeptide compo-
D. Three other chemically distinct antibiotics iso- nents have been identified from P. polymyxa
lated from P. polymyxa strains by researchers at strains which include the polymyxin F [29], M
the Wellcome labs during this period became [30, 31], P [32, 33], S [34, 35] and T [34, 36]
known as polymyxin B, C and E [11]. Colistin groups (Table 3.1). The structures and chemistry
(polymyxin E) was first described in 1950 and of the polymyxins are discussed in more detail in
obtained from Bacillus (Aerobacillus) colistinus, the next section of this chapter.
18 T. Velkov et al.

3.1.2 Adoption into Clinical Practice toxicity than the parent antibiotic. Subsequent
studies demonstrated similar results with the sul-
Although the polymyxin compounds were recog- phomethylated derivatives of both colistin and
nized to exhibit similar antimicrobial activity, polymyxin B [21, 39]. Interestingly, Stansly et al.
there were striking differences in their potential [2] also reported substantially less painful irrita-
for eukaryotic cell toxicity [5, 6, 37–39]. For tion at subcutaneous or intramuscular injection
example, Brownlee et al. [37] demonstrated sites with the sulphomethylated derivative than
severe though reversible renal toxicity in rats with the unsubstituted lipopeptide, a common
with polymyxin A, C and D, and likewise with problem with the polymyxins initially considered
polymyxin A in rabbits and dogs (polymyxins C by some to be more significant than the potential
and D not tested); polymyxin B and especially renal toxicities. This is exemplified by Barnett
colistin (polymyxin E), which were tested in all et al. [40] who in 1964 commented that “In the
species, produced significantly less nephrotoxic- literature much value has been attached to the
ity in all cases. Interestingly, in contrast to what reduction in acute intravenous toxicity achieved
is now known about the nephrotoxicity of both by the sulphomethylation of the polymyxins, but
polymyxin B and colistin, the authors in that with these antibiotics this toxicity is of no thera-
study commented that this “lends support to the peutic importance because even in the unsubsti-
view that it [i.e. colistin] has little nephrotoxic tuted form they have a satisfactory therapeutic
activity”. Early reports such as this indicating index. The use of the polymyxins has, however,
substantially reduced renal toxicity from colistin been much affected by the pain that develops at
and polymyxin B are likely the reason that of the the site of intramuscular injection and by an
five polymyxin antibiotic groups initially discov- undeserved reputation for nephrotoxicity. The
ered, only these two were further developed and painful reactions are undoubtedly avoided by
adopted into clinical practice. Nevertheless, using the sulphomethylated derivatives.” Indeed,
while the prevailing view at the time was that sulphomethylation was applied by Koyama [42]
colistin and polymyxin B were generally safe in 1957 specifically to overcome this problem
compounds the potential for toxicity, especially with colistin. As will be discussed below colistin
renal toxicity, was well recognized [39, 40]. is still administered in the clinic intravenously as
Subsequently, research was undertaken to exam- its sulphomethylated derivative.
ine ways to reduce further their toxicity.

3.1.4 Commercial Preparations


3.1.3 Sulphomethyl Derivatives
The polymyxins colistin and polymyxin B
The reaction of a primary amine with an alde- became available clinically in the late 1950s and
hyde and sodium sulphite to convert a basic sub- early 1960s [43, 44]. Presently, ‘colistin’ is com-
stance to labile alkane sulphonic acids was mercially available in two different forms,
introduced into drug synthesis in the early 1900s namely colistin sulphate [1264-72-8, CAS
in a successful attempt to reduce the toxicity of ­registry number], hereafter referred to as colistin,
phenetidine without loss of antipyretic activity and its sulphomethylated derivative, sodium
[41]. The reaction is equally applicable to basic colistin methanesulphonate [8068-28-8] (CMS,
polypeptides such as the polymyxins (the chem- also known as colistimethate sodium, sodium
istry of which is discussed later in this chapter), colistimethate, penta-sodium colistimethanesul-
and the treatment of polymyxins with formalde- phate and sulphomethyl colistin); polymyxin B is
hyde and sodium bisulphite was first reported by only available as polymyxin B sulphate [1405-
Stansly et al. [2]. These investigators showed that 20-5] [45]. Colistin, which is poorly absorbed
a sulphomethyl derivative of ‘Polymyxin’ (later from the gastrointestinal tract and through skin
shown to be polymyxin D) produced less acute [21, 37, 46], has been formulated as an oral prep-
3 History, Chemistry and Antibacterial Spectrum 19

aration (indicated for bowel decontamination) practice, colistin was marketed as offering greater
and topical preparations (indicated for bacterial or equal antibacterial potency as compared with
skin, eye and ear infections), but is not used par- polymyxin B and, as the methanesulphonate (i.e.
enterally due to its high potential to elicit toxicity CMS), was said to lack serious toxic effect in
upon intravenous administration (median lethal patients [19, 21, 39, 53–57]. It was demonstrated
dose (LD50) = 5.46 mg/kg in mice) [21]. CMS is that larger doses of CMS were required for effec-
poorly absorbed from the adult gastrointestinal tiveness and thus the rate of nephrotoxicity
tract [47] and its sodium salt, in lyophilized form, approximated that of polymyxin B [39]; this,
is the form of ‘colistin’ that is administered par- together with the noted reduction of pain at injec-
enterally, most commonly intravenously [48, 49]. tion sites with the sulphomethylated derivatives,
However, it may also be administered intramus- may explain why the use of CMS was adopted far
cularly, intrathecally, intraventricularly, and via more widely than polymyxin B. Interestingly, in
inhalation, the latter a common route of adminis- 1961 the sodium salt of a sulphomethyl deriva-
tration for patients with cystic fibrosis. Although tive of polymyxin B was administered in large
CMS can be administered intramuscularly at the doses intramuscularly and intraventricularly in
same doses as intravenously, intramuscular five children with secondary meningitis due to
administration is not commonly used in clinical Pseudomonas pyocyanea (now Pseudomonas
practice because of variable absorption and aeruginosa) [43]. This was done in an attempt to
severe pain at the injection site [50]. reduce the meningeal irritant and nephrotoxic
It is important not to use the terms colistin and properties of polymyxin B. With all five patients
CMS interchangeably, as the chemistry, antibac- cured and no toxicity observed, the authors rec-
terial activity, toxicity and pharmacokinetics of ommended this derivative of polymyxin B for
these two entities differ substantially. future use in the treatment of such infections.
Unfortunately, despite the urging of Goodwin However, for reasons, which may never be
[51] who as early as 1969 pointed out the poten- known, the sulphomethylated derivative of poly-
tial confusion that may arise when the general myxin B was never adopted into regular clinical
term ‘colistin’ is used in reference to either colis- practice. At present there is greater worldwide
tin sulphate or CMS (as was common practice at use of colistin compared to polymyxin B. Notably,
the time; for examples, see Kunin [52], and a survey across 56 different countries revealed
Schwartz et al. [21]), authors to this day still formulations of polymyxins used were CMS
occasionally report and discuss ‘colistin’ in (48.6%), colistin (sulfate) (14.1%), both (1.4%),
generic terms which makes determination of polymyxin B (1.4%), and unknown [58]; respon-
even the preparation used (colistin sulphate or dents from 11 countries had no access to poly-
CMS) difficult. For the purposes of this and all myxins. Intravenous formulations were used by
remaining discussions, colistin sulphate will 84.2% of respondents, aerosolised or nebulised
hereafter be referred to as colistin. colistin by 44.4%, and oral colistin for selective
gut decontamination by 12.7% [58].
Despite the early belief that colistin and poly-
3.1.5 Clinical Use myxin B were relatively safe drugs, and the use
of less toxic CMS as the parenteral form of
In terms of their clinical use, the only difference ‘colistin’, clinical reports began to emerge which
between polymyxin B and the two commercially suggested a high incidence of nephrotoxicity and
available forms of ‘colistin’ (colistin sulphate neurotoxicity following intravenous administra-
and CMS) is that polymyxin B is not indicated tion in a considerably large number of patients
for oral use. Otherwise, polymyxin B sulphate [59–67]. As a consequence, use of polymyxins
can be administered via intravenous, intramuscu- declined in the 1970s with the arrival of poten-
lar, inhalational, intrathecal or topical routes [45]. tially less toxic antimicrobials such as the amino-
With the introduction of polymyxins to clinical glycosides, which possessed the same or broader
20 T. Velkov et al.

antibacterial spectra. However, a resurgence in polymyxin scaffold and are always of the
their use began in the late 1980s when colistin L-configuration. Position 2 of the polymyxin
(the most commonly used polymyxin) was rein- scaffold always contains a conserved hydrophilic
troduced to manage infection or colonisation by L-threonine residue. Position 3 sees variation and
P. aeruginosa in patients with cystic fibrosis [68]. can contain either a D or L-Dab residue or a
More recently, with the emergence of multidrug-­ D-serine residue. Position 6 always contains a
resistant (MDR) Gram-negative ‘superbugs’ conserved hydrophobic residue that is of the
resistant to almost all other available antibiotics D-configuration and varies between phenyala-
[69–72], and a lack of novel antimicrobial agents nine, leucine. Position 7 sees the greatest varia-
in the drug development pipeline for Gram-­ tion and can either contain one of several
negative infections [70–76], the place of poly- hydrophobic residues including leucine, isoleu-
myxins in therapy is presently being re-evaluated. cine, valine, norvaline or the hydrophilic residue
With no new antibiotics to treat these infections threonine. The stereochemistry at position 7 is
to become available in the foreseeable future [71, always of the L-configuration. Position 10 in
74], ‘old’ polymyxins are often the only available most cases has an L-threonine residue but in at
therapeutic options. As a consequence the use of least one case contains an L-leucine residue. In
polymyxins, especially CMS, has increased dra- regards to the N-terminal fatty-acyl group, six
matically over the last decade [48, 49, 68, 77– chemically distinct fatty acyl groups that vary in
83]. The growing importance of polymyxins as a length from 7 to 9 carbons have been identified to
treatment option for MDR Gram-negative infec- date. These include (S)-6-methyloctanoyl,
tions is exemplified by the growing problem of 6-methylheptanoyl, octanoyl, heptanoyl, non-
New Delhi metallo-β-lactamase (NDM)- anoyl and 3-hydroxy-6-methyloctanoyl. Like
producing Enterobacteriaceae. Since the first many other antimicrobial peptides, this mixture
identification on the Indian subcontinent in of lipophilic and hydrophilic groups makes them
December 2009 of NDM-1-producing Klebsiella amphipathic, a chemico-physical property which
pneumoniae [84], NDM-producing is essential for their activity [88]. This also allows
Enterobacteriaceae (mainly K. pneumoniae and them to be readily water soluble (e.g. logP values
E. coli) have spread rapidly to more than 20 coun- for colistin A and colistin B are −3.15 and −3.68,
tries in all continents [85–87]. Many of these respectively) [89]. The relationship between
NDM-producing MDR isolates are only suscep- these structural features and the activity of the
tible to polymyxins. polymxyins is discussed in detail in Chap. 20:
Discovery of Novel Polymyxin-Like Antibiotics.
Examination of the literature to date reveals
3.2 Chemistry that 37 unique polymyxin lipopeptides have been
isolated and structurally identified from the P.
From a chemical perspective, the polymyxins are polymyxa species [27–33, 35, 36, 90–96]. The
non-ribosomal cyclic lipopeptides and the gen- chemical structures of these individual lipopep-
eral structure is illustrated in Table 3.1. They are tides are illustrated in Table 3.1. These have been
decapeptides containing an intramolecular cyclic classified into 10 different groups (A, B, C, D, E,
heptapeptide amide-linked loop between the F, M, P, S and T) with each group being structur-
amino group of the side chain of the diaminobu- ally defined and loosely classified by the pres-
tyric acid (Dab) residue at position 4 and the car- ence of unique amino acid residue(s) or amino
boxyl group of the C-terminal threonine residue. acid stereochemistry in their amino acid sequence
They also have several other distinguishing struc- at positions 3, 6, 7 and 10 (Table 3.1). These dis-
tural features, which include four or five non-­ tinct groups of polymyxins have each been
proteogenic Dab residues, which are charged at labelled with a letter. Each group can contain sev-
physiological pH. Four of these Dab residues are eral individual lipopeptide components which
always found at positions 1, 5, 8 and 9 in the differ from one another in the chemical structure
3 History, Chemistry and Antibacterial Spectrum 21

of the fatty-acyl group they present at their B products, no less than 80% of total content is to
N-terminus and in some cases the residue pre- consist of polymyxin B1, B2, and two minor
sented at position 7. The individual lipopeptide components. Notably, similar composition limits
components of each ‘polymyxin’ group are for colistin or polymyxin B are absent from the
labelled with a number. This nomenclature is United States Pharmacopoeia (USP) [102]. The
demonstrated in Table 3.1. It is important to note remaining discussion will focus only on the
here that the use of this classification system to chemical structures of the lipopeptide compo-
label newly discovered polymyxins has not nents of these two groups of polymyxins.
always been consistent as evident with the label-
ling of the individual components of the poly-
myxin E group (Table 3.1). In the case of the 3.2.1  hemistry of the Polymyxin B
C
polymyxin C and F lipopeptides, the amino acid Lipopeptides
residue and fatty acyl composition of the lipopep-
tides in these two groups have been identified; Structurally, the lipopeptides of the polymyxin B
however, the stereochemistry and exact positions group are generally defined by the presence of a
of the amino acids are yet to be unambiguously D-phenylalanine residue at position 6, an
determined. Therefore, in Table 3.1 the position L-leucine residue at position 7 and an L-Dab resi-
of the amino acid residues for the individual lipo- due at position 3. To date, seven individual poly-
peptides in these two groups is speculative and myxin B lipopeptide components have been
based on the structural trends observed in the identified (Table 3.1) [92, 95, 96]. Of these seven
other polymyxin groups. To date no examples lipopeptides, six contain structurally different
have been reported in the literature of individual branched and non-branched N-terminal fatty-acyl
polymyxin producing P. polymyxa strains pro- groups varying in length from 7 to 9 carbons,
ducing ‘cross mixtures’ containing lipopeptides which have been labelled polymyxin B1 to B6.
from the different polymyxin groups. Furthermore The 6-methyloctanoyl fatty-acyl group of poly-
the polymyxins are always produced as mixtures myxin B1 and B1-Ile has a stereo-centre at C6,
of the individual lipopeptide components of that which has been identified as being the (S)-
group and never as a single lipopeptide compo- configuration. Polymyxin B6 is unique in that its
nent [90, 92–95, 97]. The relative abundance of fatty-acyl group contains a hydroxyl group at C3,
the individual components produced does vary which is not present in the fatty acyl chains of the
from strain to strain and in the commercial manu- other polymyxin B lipopeptides. This unique
facture of polymyxins from the same strain, fatty acyl group also has two stereo-centres at C3
batch-to-batch variation can be observed [92, 93, and C6, however the absolute stereochemistry of
98, 99]. Of the different ‘polymyxin’ groups these two stereo-centres is yet to be reported.
identified to date, only the lipopeptide compo- Interestingly, polymyxin B1-Ile, the seventh poly-
nents of the polymyxin B and E (Colistin) groups myxin B lipopeptide is almost identical to poly-
have undergone extensive structural analysis myxin B1 except that it contains an isoleucine
[92–95]. This is a reflection of the fact that only residue at position 7, but is still considered part of
‘mixtures’ of individual polymyxin B lipopep- the polymyxin B group. Although isoleucine is
tides as well as ‘mixtures’ of individual poly- only a structural isomer of leucine it is still a
myxin E lipopeptides are used therapeutically in structurally distinct residue. In terms of relative
the clinic. The European (Ph. Eur.) and British abundance of individual components found in
Pharmacopeias (BP) have established limits on polymyxin B mixtures, polymyxin B1 and B2 are
the minimum amount of certain components always the major lipopeptide components.
required in colistin and polymyxin B products Notably, the proportion of the different lipopep-
[100, 101]. For colistin products, colistin A and B tide components in polymyxin B can vary
together with three minor components must con- between different brands and even between dif-
stitute ≥ 77% of the total content; for polymyxin ferent batches from the same manufacturer [99].
Another random document with
no related content on Scribd:
¹ Hebrew daughters. ² In Joshua xvi. 7, Naarah.

³ Many MSS. read, Ayyah.

28. Beth-el] the southern boundary. Beth-el is the modern Beitîn,


ten miles north of Jerusalem (Bädeker, Palestine⁵, p. 217). The city
was on the border of Ephraim and Benjamin and in Joshua xviii. 22
is assigned to Benjamin, but it was originally conquered by Ephraim
(Judges i. 22), and during the division of the kingdom it belonged to
the North: compare 1 Kings xii. 29, 32; 2 Chronicles xiii. 19, note.

Naaran] the eastern boundary. In Joshua xvi. 7, Naarath


(Revised Version Naarah).

Gezer ... Shechem] On Gezer the western, and Shechem the


northern boundary—see the note on vi. 67.

Azzah] or Ayyah, has not yet been identified. Probably, like


Shechem, it serves to define the northern border between Ephraim
and Manasseh.

²⁹and by the borders of the children of


Manasseh, Beth-shean and her towns,
Taanach and her towns, Megiddo and her
towns, Dor and her towns. In these dwelt the
children of Joseph the son of Israel.
29. For Manasseh four important towns are enumerated: Beth-
shean on the east in the valley of the Jordan, Taanach and Megiddo
in the plain of Esdraelon or Megiddo, and Dor on the Mediterranean
coast, south of Mt Carmel.

Beth-shean] In 1 Samuel xxxi. 10, 12 spelt Beth-shan. It is the


Greek Scythopolis, the modern Beisan.
Taanach] See vi. 70, note on Aner.

Megiddo] Judges v. 19; 2 Kings xxiii. 29; Zechariah xii. 11.

Dor] modern Tantura. Compare Joshua xvii. 11.

30‒40.
The Genealogy of Asher.

³⁰The sons of Asher; Imnah, and Ishvah,


and Ishvi, and Beriah, and Serah their sister.
30. The sons of Asher] The names in verses 30, 31 are derived
from Genesis xlvi. 17 (compare Numbers xxvi. 44‒46). There is no
variation in the Hebrew spelling of the names, but Ishvah is missing
in Numbers Either Ishvah or Ishvi must be regarded as an error of
dittography.

Beriah] Beriah is mentioned above, verse 23, as a clan of


Ephraim, and appears also as a family of Benjamin, viii. 13, 16.

³¹And the sons of Beriah; Heber, and Malchiel,


who was the father of Birzaith. ³²And Heber
begat Japhlet, and Shomer ¹, and Hotham, and
Shua their sister. ³³And the sons of Japhlet;
Pasach, and Bimhal, and Ashvath. These are
the children of Japhlet.
¹ In verse 34, Shemer.

31. Heber, and Malchiel] The antiquity of these two names seems
to be attested by the mention of “Habiri and Malchiel” in the Amarna
tablets (circa 1400 b.c.).
Birzaith] probably the name of a place, “The well of the olive-
tree.”

³⁴And the sons of Shemer ¹; Ahi, and Rohgah,


Jehubbah, and Aram. ³⁵And the sons ² of
Helem his brother; Zophah, and Imna, and
Shelesh, and Amal. ³⁶The sons of Zophah;
Suah, and Harnepher, and Shual, and Beri,
and Imrah; ³⁷Bezer, and Hod, and Shamma,
and Shilshah, and Ithran, and Beera. ³⁸And
the sons of Jether; Jephunneh, and Pispah,
and Ara. ³⁹And the sons of Ulla; Arah, and
Hanniel, and Rizia.
¹ In verse 32, Shomer. ² Hebrew son.

34, 35. Shemer ... Helem] Read perhaps Shomer ... Hotham, to
agree with verse 32.

⁴⁰All these were the children of Asher, heads


of the fathers’ houses, choice and mighty men
of valour, chief of the princes. And the number
of them reckoned by genealogy for service in
war was twenty and six thousand men.
40. twenty and six thousand] In xii. 36 the men of war of Asher
are reckoned at forty thousand (compare Numbers i. 41, xxvi. 47,
where still higher reckonings are given). The numbers here and in
verses 5, 7, 9, 11 (as well as in verse 2, which see) are perhaps
supposed to refer to the time of David. The numbers may be based
on family traditions, but no important conclusions ought to be drawn
from them.
Chapter VIII.
1‒40 (compare vii. 6‒12).
The Genealogy of Benjamin.

1‒40. Various indications combine to show that the names in this


list reflect post-exilic conditions. It has generally been compared with
the “Benjamite” genealogy in vii. 6‒12 which was supposed to
express the relationships and strength of the tribe at the time of
David. If, however, according to the view adopted in this volume, the
passage vii. 6‒12 is in reality a genealogy of Zebulun, comparison
between it and this list is futile. Such parallels as can justly be made
between the names in the two lists are due to the Benjamite
colouring which has been imparted to vii. 6‒12 after the initial error in
vii. 6 turned the “sons of Zebulun” into “Benjamin.”

This, the real genealogy of Benjamin, unfortunately presents not


a few problems for which as yet no convincing solution can be
offered. The difficulties are due in large measure to the corrupt state
of the text in several verses: especially verses 6‒14.

¹And Benjamin begat Bela his firstborn,


Ashbel the second, and Aharah the third;
1. Benjamin begat ...] Compare Genesis xlvi. 21.

firstborn] = Becher in Genesis xlvi. 21. In the unvocalised Hebrew


text the noun and proper name are represented by the same letters,
BKR.

Ashbel] literally “man of Baal.” Compare note on Eshbaal, verse


33.
²Nohah the fourth, and Rapha the fifth.
2. Nohah ... Huram] the list is assuredly based on Genesis xlvi.
21 and Numbers xxvi. 38‒40, despite the surface divergences.
Several of the changes are due to textual errors, e.g. Aharah and
Ahoah are probably both variants of Ahiram (Genesis xlvi. 21).

³And Bela had sons, Addar ¹, and Gera, and


Abihud; ⁴and Abishua, and Naaman, and
Ahoah;
¹ In Genesis xlvi. 21, Ard.

3. Abihud] read perhaps (a slight change in the Hebrew) Gera,


father of Ehud.

⁵and Gera, and Shephuphan ¹, and Huram.


¹ In Numbers xxvi. 39, Shephupham.

5. Shephuphan, and Huram] See vii. 12, note on Shuppim.

6‒28. Apparently a list of five post-exilic families [Elpaal (verses


11, 18), Beriah (verses 13, 16), Shema (verses 13, 21), Shashak
(verses 14, 25), and Jeroham (verses 14, 27)], whose genealogy
seems to be traced from Ehud, and whose descendants reside in
Jerusalem (so verse 28, but see note ad loc.). The uncertainty on the
former point is the inevitable consequence of the corrupt state of the
text in verses 6‒14.

⁶And these are the sons of Ehud; these are


the heads of fathers’ houses of the inhabitants
of Geba, and they carried them captive to
Manahath:
6. Ehud] Ehud (the deliverer of Israel from Moab) was descended
from Gera (verse 5; Judges iii. 15).

Geba] Compare vi. 60.

they carried them captive] an utterly obscure phrase, most


probably due to textual error. It is a plausible suggestion that the
phrase is a corruption of proper names commencing the list which
we should expect to follow the preceding words: “these are the
heads of,” etc. Hogg, Jewish Quarterly Review xi. 102 ff., therefore
conjectured the names “Iglaam and Alemoth”; and similarly in verse
7, in place of the equally obscure words “he carried them captive;
and he,” he would read “and Iglaam begat.”

⁷and Naaman, and Ahijah, and Gera, he


carried them captive; and he begat Uzza and
Ahihud.
7. Naaman, and Ahijah, and Gera] perhaps to be deleted, as a
repetition of verse 5.

⁸And Shaharaim begat children in the field of


Moab, after he had sent ¹ them away; Hushim
and Baara were his wives. ⁹And he begat of
Hodesh his wife, Jobab, and Zibia, and
Mesha, and Malcam; ¹⁰and Jeuz, and
Shachia, and Mirmah. These were his sons,
heads of fathers’ houses.
¹ Or, sent away Hushim and Baara his wives.

8, 9. Again the Hebrew text appears to be in disorder, and the


verses in consequence are so obscure that conjectures are all
precarious.
Hushim] is elsewhere the name of a man. Hence verse 11 below
should perhaps read And Hushim begat....

¹¹And of Hushim he begat Abitub and Elpaal.


11. Abitub] no sons of his are recorded.

¹²And the sons of Elpaal; Eber, and Misham,


and Shemed, who built Ono and Lod, with the
towns thereof:
12. sons of Elpaal] Elpaal’s sons are given also and more fully in
verses 17, 18; and, as the three names in the present verse appear
to be transcriptional variants of three mentioned in 17, 18, it is
probable that this verse is a marginal note which has crept into the
text.

who built Ono and Lod] the subject is not Shemed, but Elpaal;
“built,” i.e. entered into possession of. Ono and Lod (= Lydda), some
seven and eleven miles respectively south of Jaffa, are referred to in
Nehemiah vii. 35, xi. 35, and Ezra ii. 33. The Targum adds, which the
sons of Israel laid waste and burnt with fire, when they made war in
Gibeah with the tribe of Benjamin.

¹³and Beriah, and Shema, who were heads of


fathers’ houses of the inhabitants of Aijalon,
who put to flight the inhabitants of Gath;
13. Aijalon] compare Joshua x. 12. It was situated near the Jaffa
road, about thirteen miles from Jerusalem.

who put ... Gath] an interesting remark, which should be


compared with vii. 21, 23—note the name Beriah in both passages.
The relation of the two passages is, however, uncertain.
¹⁴and Ahio, Shashak, and Jeremoth; ¹⁵and
Zebadiah, and Arad, and Eder; ¹⁶and Michael,
and Ishpah, and Joha, the sons of Beriah;
¹⁷and Zebadiah, and Meshullam, and Hizki,
and Heber; ¹⁸and Ishmerai, and Izliah, and
Jobab, the sons of Elpaal; ¹⁹and Jakim, and
Zichri, and Zabdi;
14. And Ahio, Shashak, and Jeremoth] Read, following LXX.,
And their brethren Shashak and Jeremoth. The pronoun of
course refers to Beriah and Shema (verse 13), and to Abitub and
Elpaal (verse 11)—these four, with Shashak and Jeremoth, being
sons of Hushim, if verse 11 be emended and verses 12, 13 be
regarded as a marginal addition, as is suggested above.

²⁰and Elienai and Zillethai, and Eliel;


20. Elienai] Read, perhaps, Elioenai, a name meaning “My eyes
look towards Jehovah,” compare iii. 23.

²¹and Adaiah, and Beraiah, and Shimrath, the


sons of Shimei ¹; ²²and Ishpan, and Eber, and
Eliel; ²³and Abdon, and Zichri, and Hanan;
¹ In verse 13, Shema.

21. Shimei] = Shema (verse 13).

²⁴and Hananiah, and Elam, and Anthothijah;


²⁵and Iphdeiah, and Penuel, the sons of
Shashak; ²⁶and Shamsherai, and Shehariah,
and Athaliah;
24. Anthothijah] The name is a trace of an ancient Egyptian war-
goddess ‘Anath, apparently associated with Jehovah in the Jewish
temple at Elephantine (see ‘Anath-bethel in the papyri). Compare
also Anathoth near Jerusalem.

²⁷and Jaareshiah, and Elijah, and Zichri, the


sons of Jeroham.
27. Jeroham] = Jeremoth (verse 14).

²⁸These were heads of fathers’ houses


throughout their generations, chief men: these
dwelt in Jerusalem.
28. these dwelt in Jerusalem] i.e. in the writer’s day the heads of
families enumerated in verses 15‒27 dwelt in Jerusalem. Compare
ix. 2, 3; Nehemiah xi. 1‒8. But the words may be a gloss brought in
from ix. 34 along with the following verses (see below).

29‒38 (= chapter ix. 35‒44).


The Genealogy of the house of Saul.

29‒38. These verses, which set forth the ancestors and


descendants of Saul, are found also in ix. 35‒44, where they serve
as the introduction to the account of Saul’s death in ch. x. The latter
passage would naturally seem to be the original place of these
verses, but the arguments in favour of that view are not conclusive,
and the point must be allowed to be doubtful.

²⁹And in Gibeon there dwelt the father of


Gibeon, Jeiel, whose wife’s name was
Maacah:
29. Gibeon] some six miles north of Jerusalem, was apparently
the residence in post-exilic days of families which claimed descent
from the house of Saul. Compare 2 Chronicles i. 3.

Jeiel] added in accordance with ix. 35.

³⁰and his firstborn son Abdon, and Zur, and


Kish, and Baal, and Nadab;
30. and Baal] Add with LXX. (A) and ix. 36 and Ner. LXX. (B)
shows that a word is missing after Baal for it reads Βααλακαίμ (=
Βαὰλ καὶ Ν....?).

³¹and Gedor, and Ahio, and Zecher ¹.


¹ In chapter ix. 37, Zechariah.

31. and Zecher] Read with ix. 37, and Zechariah, and Mikloth.

³²And Mikloth begat Shimeah ¹. And they also


dwelt with their brethren in Jerusalem, over
against their brethren.
¹ In chapter ix. 38, Shimeam.

32. with their brethren, etc.] i.e. with some of their brethren in
Jerusalem over against other of their brethren in Gibeon and other
places. “They” would seem to refer to Mikloth and Shimeah, but the
clause is far from clear, and it may be noted that verse 32b looks like
the heading of a list that has been lost.

³³And Ner begat Kish; and Kish begat Saul;


and Saul begat Jonathan, and Malchi-shua,
and Abinadab ¹, and Eshbaal ².
¹ In 1 Samuel xiv. 49, Ishvi.

² In 2 Samuel ii. 8, Ishbosheth.

33. begat Kish] here and in ix. 39, read begat Abner—as in 1
Samuel xiv. 51, etc.

Jonathan ... Abinadab] Slain with Saul on Mt Gilboa; x. 2; 1


Samuel xxxi. 2.

Eshbaal] In 2 Samuel ii. 8 called Ish-bosheth. In the (more


generally read) Samuel text the offensive name Eshbaal, “Man (i.e.
worshipper) of Baal,” has been changed to Ishbosheth, “Man of the
Shameful-thing” (i.e. of the idol), but it has been left standing in the
less-used text of Chronicles The title Baal (“Lord”) was applied in
early days (e.g. in the days of Saul) to the national God of Israel, but
in later days the prophets objected to it because of its general use in
designation of the heathen gods also. Hosea (ii. 17), for example,
declares that the true worshippers of Jehovah must no longer call
him “My Baal” (Baali). Thus to Saul and Samuel the name Eshbaal
was acceptable as meaning “Man of the Lord,” i.e. of Jehovah, but to
the late reviser of the book of Samuel it was offensive as signifying
“Man of Baal,” i.e. of one of the gods worshipped by the old
Canaanite peoples or by the neighbouring nations. Since the text of
Chronicles has retained such forms as Eshbaal (here), Ashbel (verse
1), it seems that the conscientious alterations of such forms in the
books of Samuel, Kings, etc., are later than the time of the
Chronicler.

³⁴And the son of Jonathan was Merib-baal ¹;


and Merib-baal begat Micah.
¹ In 2 Samuel iv. 4, ix. 6, 10, Mephibosheth.
34. Merib-baal] A name meaning “Baal pleadeth”; in chapter ix.
40b (Hebrew) it is written Meri-baal, i.e. “Man of Baal.” The person
meant seems to be Mephibosheth (2 Samuel ix. 6, 12).

³⁵And the sons of Micah; Pithon, and Melech,


and Tarea ¹, and Ahaz.
¹ In chapter ix. 41, Tahrea.

35. Tarea] In ix. 41, Tahrea.

³⁶And Ahaz begat Jehoaddah ¹; and


Jehoaddah begat Alemeth, and Azmaveth,
and Zimri; and Zimri begat Moza:
¹ In chapter ix. 42, Jarah.

36. Jehoaddah] In ix. 42, Jarah.

³⁷and Moza begat Binea; Raphah ¹ was his


son, Eleasah his son, Azel his son: ³⁸and Azel
had six sons, whose names are these;
Azrikam, Bocheru, and Ishmael, and
Sheariah, and Obadiah, and Hanan. All these
were the sons of Azel. ³⁹And the sons of
Eshek his brother; Ulam his firstborn, Jeush
the second, and Eliphelet the third. ⁴⁰And the
sons of Ulam were mighty men of valour,
archers, and had many sons, and sons’ sons,
an hundred and fifty. All these were of the
sons of Benjamin.
¹ In chapter ix. 43, Rephaiah.

37. Raphah] In ix. 43, Rephaiah.


Chapter IX.
1‒17 (compare Nehemiah xi. 1‒19).
The Heads of the Families which dwelt in Jerusalem.

Verses 2‒17 contain the lists of the heads of families of Judah


(3‒6), of Benjamin (7‒9), of the priests (10‒13), of the Levites (14‒
16), and of the porters (17), who dwelt in Jerusalem at some period
after the Return (compare note on verse 2). A similar list (with some
variations which are recorded in their places in the following notes)
occurs in Nehemiah xi. 3‒19. The partial agreement coupled with the
partial divergence of the two lists may be explained by supposing
that both are extracts independently made from the same document,
and have been inserted, one in Chronicles, the other in Nehemiah,
lest the peculiarities of either list should be lost. We may conclude
from Nehemiah xi. 1, 2 that both lists represent the population of
Jerusalem, after Nehemiah had taken measures for increasing it.
Another way of accounting for the divergences in the two lists is to
suppose that the present list represents the Jerusalem of a later
period than the list in Nehemiah See also verse 17.

¹So all Israel were reckoned by


genealogies; and, behold, they are written in
the book of the kings of Israel: and Judah was
carried away captive to Babylon for their
transgression.
1. in the book of the kings of Israel] See Introduction § 5, B (3).
The LXX., however, reads “in the book of the kings of Israel and
Judah.” With the LXX. reading, all Israel must be taken as subject of
the verb was carried away, but of course the phrase must still be
taken as meaning an “Israel” = Judah.

²Now the first inhabitants that dwelt in their


possessions in their cities were, Israel, the
priests, the Levites, and the Nethinim.
2. the first inhabitants] It has been thought that the word “first”
here refers to pre-eminence (compare Nehemiah xi. 3), and that the
list which follows (verses 4 ff.) is a list of chief men. It is better,
however, to take “first” in a temporal sense, meaning “pre-exilic,” and
to suppose that the Chronicler or whoever placed this chapter here
mistakenly imagined this list to be a pre-exilic register. That it is not
really pre-exilic is certain by reason of its vital connection with the
post-exilic list in Nehemiah xi. 3‒19. The suggestion that the
resemblances are due to the continuity of population in Jerusalem
before and after the exile is utterly improbable.

in their cities] The phrase is apparently an abridgment of words in


Nehemiah xi. 3, and is really meaningless in the present context. In
Nehemiah it signifies “townships in Judah” where certain persons,
who now elected to dwell in Jerusalem, had formerly resided.

Israel] i.e. laymen as distinguished from men of Levitical descent.


According to verse 3 Israel included at least Judah, Benjamin,
Ephraim, and Manasseh (compare Psalms lxxx. 2, where Judah—
the speaker—associates Ephraim, Benjamin, and Manasseh with
herself in her appeal to the God of Israel, See also note on 2
Chronicles xxx. 18). This is a totally different usage from that of
earlier times, when Israel meant the Northern kingdom, and Judah
the Southern.

Nethinim] These were a class of Temple servants reckoned as


inferior to the Levites. Perhaps they were of foreign extraction and
included the Gibeonites (compare Joshua ix. 23). They are
mentioned nowhere else in the Old Testament except in the books of
Ezra and Nehemiah.
³And in Jerusalem dwelt of the children of
Judah, and of the children of Benjamin, and of
the children of Ephraim and Manasseh;
3. of Ephraim and Manasseh] See note on 2 Chronicles xxx. 18.

4‒6 (compare Nehemiah xi. 4‒6).


The Sons of Judah.

⁴Uthai the son of Ammihud, the son of Omri,


the son of Imri, the son of Bani, of the children
of Perez the son of Judah.
4. Uthai] In Nehemiah Athaiah. The two words are more alike in
Hebrew than in English and are perhaps various readings of one
name.

Perez] compare ii. 4, 5. We have here (verses 4‒6) a threefold


division of the tribe of Judah into the descendants of Perez, Shelah,
and Zerah, just as in Numbers xxvi. 20.

⁵And of the Shilonites; Asaiah the firstborn,


and his sons.
5. Shilonites] Or Shelanites as Numbers xxvi. 20; they were
descendants of Shelah, who is mentioned as a son of Judah in ii. 3.
For other descendants, see iv. 21 f., and Nehemiah xi. 5.

Asaiah] In Nehemiah xi. 5 Maaseiah, a kindred name.

⁶And of the sons of Zerah; Jeuel, and their


brethren, six hundred and ninety.
6. Jeuel] In Nehemiah xi. 5 the “sons of Zerah” are missing.
six hundred and ninety] Compare Nehemiah xi. 6 (four hundred
threescore and eight sons of Perez) where Perez may be an error for
Zerah.

7‒9 (compare Nehemiah xi. 7‒9).


The Sons of Benjamin.

⁷And of the sons of Benjamin; Sallu the son of


Meshullam, the son of Hodaviah, the son of
Hassenuah;
7. Sallu] His genealogy is differently stated in Nehemiah xi. 7, but
see next note.

the son of Hodaviah, the son of Hassenuah] Read perhaps


Judah, the son of Hassenuah (compare Nehemiah xi. 9). Hodaviah
and Judah could easily be confused in Hebrew.

⁸and Ibneiah the son of Jeroham, and Elah the


son of Uzzi, the son of Michri, and Meshullam
the son of Shephatiah, the son of Reuel, the
son of Ibnijah;
8. Ibneiah, Elah, Meshullam] Not mentioned in Nehemiah xi.

⁹and their brethren, according to their


generations, nine hundred and fifty and six. All
these men were heads of fathers’ houses by
their fathers’ houses.
9. nine hundred and fifty and six] 928 in Nehemiah xi. 8.

10‒13 (compare Nehemiah xi. 10‒14).


The Priests.
¹⁰And of the priests; Jedaiah, and Jehoiarib,
and Jachin;
10. Jehoiarib] Spelt Joiarib in Nehemiah xi. 10. Jehoiarib and
Jedaiah occur as names of the first and second courses of the
priests in xxiv. 7; Nehemiah xii. 6, 19. The Maccabees were of the
course of Joarib (= Jehoiarib); 1 Maccabees ii. 1.

Jachin] The name of the twenty-first course; xxiv. 17.

¹¹and Azariah ¹ the son of Hilkiah, the son of


Meshullam, the son of Zadok, the son of
Meraioth, the son of Ahitub, the ruler of the
house of God;
¹ In Nehemiah xi. 11, Seraiah.

11. Azariah] In Nehemiah xi. 11, Seraiah.

the ruler of the house of God] This title could perhaps be borne
by the high-priest (2 Chronicles xxxi. 10, 13), but in any case it was
not confined to him (2 Chronicles xxxv. 8, where several such
“rulers” are mentioned; compare also Jeremiah xx. 1; Acts iv. 1).

¹²and Adaiah the son of Jeroham, the son of


Pashhur, the son of Malchijah, and Maasai the
son of Adiel, the son of Jahzerah, the son of
Meshullam, the son of Meshillemith, the son of
Immer;
12. Malchijah] The name of the fifth course; xxiv. 9.

Maasai] The reading of Nehemiah xi. 13 Amashsai is corrupt.


The form given in Chronicles is open to suspicion. Probably the true
reading is lost.

Adiel] In Nehemiah Azareel.

Immer] The name of the sixteenth course; xxiv. 14.

¹³and their brethren, heads of their fathers’


houses, a thousand and seven hundred and
threescore; very able men for the work of the
service of the house of God.
13. a thousand and seven hundred and threescore] Only the five
“courses” of priests mentioned above (viz. Jedaiah, Jehoiarib, and
Jachin, verse 10, and Malchijah and Immer, verse 12) seem to be
included in this reckoning. Some commentators, however, regard
Azariah (= Seraiah) in verse 11 as the name of a new course, which
took the place of one of the courses reckoned in xxiv. 7‒18. If this be
right we have here the sum of six courses.

In Nehemiah xi. 12‒14 the number of the priests is given on a


different plan; eight hundred and twenty-two “did the work of the
house”; two hundred and forty-two were “chiefs of fathers’ houses”;
an hundred and twenty-eight were “mighty men of valour.” The total
falls far short of the thousand and seven hundred and threescore of
Chronicles We have not sufficient data on which to base any
explanation of the different totals.

very able men] The Hebrew is the same as in Nehemiah xi. 14


and is usually rendered mighty men of valour. The sense, however,
is no doubt correctly given by Revised Version very able, or efficient.
Compare 2 Chronicles xxvi. 17.

14‒16 (compare Nehemiah xi. 15‒18).


The Levites.
¹⁴And of the Levites; Shemaiah the son of
Hasshub, the son of Azrikam, the son of
Hashabiah, of the sons of Merari;
14. of the sons of Merari] In Nehemiah the sons of Bunni, which
is probably a corruption of the reading of Chronicles Otherwise of the
three great Levitical families, Merari, Asaph, and Jeduthun,
mentioned here, only the last two appear in Nehemiah.

¹⁵and Bakbakkar, Heresh, and Galal, and


Mattaniah the son of Mica, the son of Zichri ¹,
the son of Asaph;
¹ In Nehemiah xi. 17, Zabdi.

15. Bakbakkar, Heresh, and Galal] The reading appears to be


corrupt, for the analogy of the latter half of the verse as well as of
verses 14, 16 leads us to expect something more than bare names.
Neither the LXX. nor the Vulgate gives any real help for emending
the clause. The corresponding words in Nehemiah (xi. 17) are
Bakbukiah the second among his brethren.

¹⁶and Obadiah ² the son of Shemaiah ³, the son


of Galal, the son of Jeduthun, and Berechiah
the son of Asa, the son of Elkanah, that dwelt
in the villages of the Netophathites.
² In Nehemiah xi. 17, Abda.

³ In Nehemiah xi. 17, Shammua.

16. Obadiah the son of Shemaiah] In Nehemiah Abda the son of


Shammua. Which was the reading of the original document cannot

You might also like