Tuberculosis in Infancy and Childhood 4th Ed (PPS)
Tuberculosis in Infancy and Childhood 4th Ed (PPS)
IN INFANCY
AND CHILDHOOD
4th edition
2016
ii
PHILIPPINE PEDIATRIC SOCIETY, INC.
TUBERCULOSIS
IN INFANCY
AND CHILDHOOD
4th edition 2016
COMMITTEE ON HANDBOOK
ON CHILDHOOD TUBERCULOSIS
iii
MESSAGE FROM THE PRESIDENT OF THE
PHILIPPINE PEDIATRIC SOCIETY
I would like to congratulate the contributors and editors of the Handbook on Tu-
berculosis in Infancy and Childhood, ably led by Dr. Cleotilde How, Chair of the
PPS Committee on TB Handbook. After almost two years of painstaking and
diligent work, the 4th edition of Tuberculosis in Infancy and Childhood has come
to fruition. This edition continues a line of excellent TB handbooks designed to
equip pediatricians and health practitioners dedicated to the study and manage-
ment of childhood and infant tuberculosis with up to date clinical information and
current approaches to prevention, detection, diagnosis, and treatment.
iv
MESSAGE FROM THE PAST PRESIDENT OF THE
PHILIPPINE PEDIATRIC SOCIETY
The chapters of the TB Handbook will walk us through the Epidemiology local
and international data, the clinical presentations of the disease, its diagnosis
and the treatment the TB disease.
Most important to the success of this 4th edition is the dedication and hard work
of the committee members and the team leader Dr Telly How, whose commit-
ment to excellence is unparalleled; your committee members got the right mix of
dedication and enthusiasm.
The Philippine Pediatric Society congratulates the committee and indeed grate-
ful to you all for a job well done!
Mabuhay!
Milagros S. Bautista, MD
Immediate Past President
Philippine Pediatric Society, Inc.
v
FOREWORD
The 4th edition of Tuberculosis in Infancy and Childhood has a history that dates back
to its first publication in 1993, to serve as a supplement of the already established PPS
Handbook on Infectious Diseases, marking its 25th year then. The idea for that maiden
issue came about during the First National Consensus in Tuberculosis in 1989 by the
Tri-Chest organizations. That 1st edition was preceded in 1975 by the Manual on Tuber-
culosis in Filipino Children by Dr Mita Pardo de Tavera, whose work in AKAP (Alay
Kapwa Kilusang Pangkalusugan) exemplified what community-based TB control can
do.
Several key publications coming out in 2014 provided the impetus for this new edition
of TBIC and to consult local guidelines for agreement in best practices. Among these
are the WHO Guidance for national tuberculosis programmes on the management of
tuberculosis in children (Second edition) , the International Standards for Tuberculosis
Care (ISTC, 3rd edition), the Department of Health National Tuberculosis Control Pro-
gram (DOH-NTP) Manual of Procedures (MOP, 5th edition) and an earlier WHO Rapid
advice: treatment of tuberculosis in children in 2010/2011. The Universal Health Care
policy of the government and the new TB Law present a challenge for the multidiscipli-
nary health sector that leads the effort for TB prevention and control.
The issue over what age range constitutes a ―child‖ or an ―adolescent‖ needs mention
here as it follows operational lines in TB control. Although the WHO definition of a
―child‖ general falls under the age of 19 years, in the WHO Guidance a ―child‖ refers to
the 0-10 age group. A child over 10 years is expected to expectorate for sputum smear
and culture. Further, the approach to management of TB in adolescents has followed
the standards set in the care of adults. The Manual of Procedures sets the age of 15 as
the cut-off in application of diagnostic criteria. The early teens falls somewhere in the
middle of the overlap. In computing for dose requirements of anti-TB drugs, it is not the
age but the weight of 25 kg that arbitrarily divides the child from the adult.
At the doorstep are some marketed molecular/NAAT tests like FluoroType MTB, the
GeneChip, LiPA Z (line probe assay for PZA resistance), many more LPAs,
PureLamp (manual NAAT), MBio Array System (measures 57 MTN ag-ab reactions)
and CAD 4TB (digital CXR for TB screening). While new data are still forthcoming,
WHO still does not recommend LPAs on smear-negative samples. Candidate bi-
omarkers like LAM (lipoarabinomannan), CRP (C-reactive proteins), TAM-TB (T-cell
activation marker-tuberculosis), RNA and gene expression signatures are being ex-
plored to detect active TB and be able to differentiate active TB from latent infection.
Quality of tests and affordability still have to come hand-in-hand, particularly in high-
burden low-resource countries.
A long time coming is the development and arrival of the new fixed-dose-
combinations (HRZ, HR ) as child-friendly dispersible tablets; a single dispersible
ethambutol as fourth drug in the intensive phase and a single dispersible isoniazid for
IPT. Ongoing Phase 3 clinical trials in children for second-line drugs (bedaquiline and
delamanid) are expected to be completed to join the race to combat MDR-TB. Not
only is there a push for early clinical trials in children (and pregnant/lactating women),
there is equally a surge for repurposed drugs (eg, clofazimine, PAS) to supplement
the dearth of anti-TB drugs.
A new chapter on Adjunctive Therapy includes the role of corticosteroids, surgery and
nutrition. The search for new vaccines has not yielded a likely candidate. The direc-
tion of vaccine research is not the search for a replacement for BCG, given the short-
fall in vaccine trials. There is a change in perspective, taking a closer look at the
mechanism of immunity, which has traditionally centered on cell-mediated immunity,
but looking elsewhere for new targets to measure the effects of new candidate vac-
cines. An abbreviated International Classification of Diseases (ICD-10): Tuberculosis
is reproduced on Appendix 3 to supplement use of this system in hospital audits.
The chapter on prevention and control reemphasizes the value of contact investiga-
tion, active case-finding and isoniazid preventive therapy (IPT). Transmission reduc-
tion (eg, cough etiquette) and simple hygiene are not as technology-bound but essen-
tial for the business of infection control. Success relies more on implementation, re-
cording and reporting, quite a culture shock for the busy private practitioner. To deal
with TB, even he/she has to transform into a public health practitioner and join the
team. Integration is key at all levels: search-and-control TB operations means going
to both pediatric and adult clinics, in Maternal and Child Health units, in IMCI and EPI
vii
clinics, Well-Child/Sick-Child units, that is, everywhere you find children and adults who
are at risk for TB. TB and HIV, TB and diabetes, TB and smoking, other co-morbidities
that matter. Integration also involves partnerships with common advocacies – in com-
munication, education, livelihood, nutrition, shelter, cleanwater.
Acknowledgments
The Board of Trustees led by Dr Mila S Bautista wholeheartedly supported this project
from the time it was conceived in late 2014 until the change of hands under the leader-
ship of Dr Alexander O Tuazon, when the manuscript was launched in the 2016 annual
convention.
The members of the panel of editors (How CH, Paje-Villar EB, Carlos JC, Ong-Lim AL)
have worked hard in deliberating on the manuscripts with the able assistance of Dr Sa-
rah Makalinaw. Dr Paje-Villar, who was editor of the first edition, has kept her constant
presence and scrutiny up to this latest work. The set of contributors per chapter have
similarly met the challenge of bringing the data and information up to par, with constant
revisions and verification of sources. The group is extremely grateful to Ms Thess Victo-
ria and staff from PPS and PIDSP for arranging meetings and venues, keeping track of
members, providing materials needed and coordination work.
Our respective families, for the chunks of time spent away from them; they will be re-
lieved, until the next work comes along. We thank them in the words of Francis Fukuya-
ma ―simply by being there‖.
viii
PHILIPPINE PEDIATRIC SOCIETY, INC.
BOARD OF TRUSTEES 2014-2016 BOARD OF TRUSTEES 2016-2018
Officers Officers
MILAGROS S BAUTISTA, MD ALEXANDER O TUAZON, MD
President President
Members Members
CYNTHIA P DANIEL, MD FRANCIS XAVIER M DIMALANTA,
RHODORA D DIAZ, MD MD
BENJAMIN T LIM, MD BENJAMIN T LIM, MD
RENIE M MAGUINSAY, MD CESAR M ONG, MD
MAY B MONTELLANO, MD WINSTON FELIX J. QUEBEC, MD
CESAR M ONG, MD WILFREDO R SANTOS, MD
FLORENTINA U TY, MD NELLY S SIMON, MD ALFREDO A.
YAP, MD
ix
COMMITTEE ON TB HANDBOOK IN CHILDREN
x
LIST OF EDITORS AND CONTRIBUTORS
EDITORS
CLEOTILDE H HOW, MD, FPPS, FPSECP
Retired Professor of Pharmacology and Clinical Pediatrics, UP College of Medicine
xi
CONTRIBUTORS
MYRA R ASUNCION-SON, MD, DPPS, DPAPP
Consultant, St. Luke's Medical Center Global City, Parañaque Doctors Hospital
xiii
Children‘s Medical Center
xiv
GLOSSARY: DEFINITIONS
Acquired Resistance – refers to the bacterial resistance in patients with some record of previ-
ous treatment
BCG – stands for Bacillus Calmette-Guerin, is an attenuated vaccine strain of M. bovis used to
immunize against tuberculosis
Exposed person – a person who has had recent contact with another person with suspected or
confirmed contagious pulmonary tuberculosis disease and who has a negative tuberculin (or
IGRA) reaction, normal physical examination findings, and chest radiographic findings that are
not compatible with tuberculosis
Incubation period – time interval from exposure to the mycobacterium to the development of
delayed type hypersensitivity reaction as manifested by a positive TST (or IGRA)
Mantoux test – a skin test for tuberculosis infection, using tuberculin PPD (purified protein de-
rivative) administered intradermally/intracutaneously at a specified tamount and positive result
read as induration between 48 to 72 hours of injection
Poly-resistance – resistance to more than one antituberculosis drug, other than isoniazid and
xv
rifampicin
Primary complex – composed of the primary focus, lymphangitis, localized pleural effusion and
regional lymphadenitis, demonstrable by radiographic study
Primary resistance – bacterial resistance present in patients who have not received prior treat-
ment with antituberculosis drugs
Scrofuloderma – TB of the skin overlying a caseous lymph node that has ruptured to the out-
side, leaving an ulcer or a sinus
Source case – the person, usually with pulmonary TB, who has transmitted infection with Myco-
bacterium tuberculosis
Tuberculin – a protein extracted from Mycobacterium tuberculosis that is used in a skin test to
determine if a person has been exposed to tuberculosis and infected with the tuberculoprotein
from the organism
Tuberculin Skin Testing (TST) – a diagnostic test to determine the presence of delayed-type
hypersensitivity reaction to tuberculoproteins, either from natural infection with the organism or
in reaction to the mycobacterium antigen in BCG. A positive reaction will manifest as an indura-
tion; cut-off size determined by conditions set
Tuberculosis – a highly contagious infection and disease caused by exposure to the virulent
bacterium Mycobacterium tuberculosis, progressing from infection to disease, when signs and
symptoms become manifest, either from pulmonary or extrapulmonary involvement
xvi
GLOSSARY: ABBREVIATIONS
AFB acid-fast bacilli FM fluorescence microscopy
xvii
GLOSSARY: ABBREVIATIONS
MTBC Mycobacterium tuberculosis PTB pulmonary tuberculosis
complex
R rifampicin
MPT multiple puncture test
RFLP Restriction fragment length
NAAT nucleic acid amplification test polymorphism
xviii
TABLE OF CONTENTS
Message 1 iv
Message 2 v
Foreword vi
Glossary xv
Abbreviations xvii
Table of Contents
Chapter 1 Highlights 1
Estrella Paje-Villar, MD, Josefina Carlos, MD,
Ana Liza Duran, MD
Chapter 2 Epidemiology 9
Luis Chan Jr., MD, Ana Liza Duran, MD
Chapter 3 Microbiology 19
Jaime Santos,MD, Mary Antonette Madrid,MD
Chapter 5 Pathogenesis 39
Maria Anna Banez, MD, Cynthia De Ocampo,MD
Chapter 7 Diagnosis 69
Cleotilde How,MD
xix
Chapter 8 Tuberculin Skin Test 83
Luis Chan Jr.,MD, Cleotilde How,MD
xx
I
1
Highlights
Estrella B Paje-Villar, MD
Josefina C Carlos, MD
Ana Liza H Duran, MD
1910 Founding of the Philippine Islands Anti-Tuberculosis Society now known
as the Philippine Tuberculosis Society Inc (PTSI), a private agency with
government subsidy
1949 A ―miracle drug‖, streptomycin (S), was first used for TB treatment
1958 Establishment of the Bureau of Disease Control which included the Divi-
sion of TB by virtue of Executive Order no. 288
2
1973 Launching of the Domiciliary Care Program
1974 Offering of the Anti – TB treatment using H, S and Ethambutol (E) for 18
months
1981-1983 Conduct of the First National TB Prevalence Survey by the National In-
stitute for Tuberculosis (NIT)
1984 Tuberculosis all forms as the 3rd leading cause of mortality 52.6/100,000
population (Philippine Health Statistics)
1986 Creation of the TB Control Service with EO no. 119, under the Office of
Public Health Services (Ministry of Health which became the Department
of Health)
3
1988 Publication of the National TB Program Manual of Procedures (2nd
edition), which marked the adoption of the Short Course Chemotherapy
(SCC) for the management of TB cases under the National TB Preva-
lence; presented the results of the 1981- 1983 First National TB Preva-
lence Survey (NPS); introduced the itinerant team approach to selected
regions/ provinces and intensified the training, monitoring/supervision in
low performing areas.
First reference to the Public-Private Mix (PPM) for TB Control in the Phil-
ippine NTBP manual
1990 Financial and technical support to the NTBP from the Italian government
and World Bank for improving TB control in Regions V, VIII, X and in
other cities and provinces
1994 Field testing of new NTBP policies and guidelines and establishment of
First TB Reference Laboratory in Cebu under the DOH-JICA Project
4
banan)
1996 The 3rd Annual Convention of PhilCAT, in collaboration with the PPS and
the Pediatric Infectious Disease Society of the Philippines (PIDSP) with
the theme: Childhood Tuberculosis, a Concern for All
5
Creation of the Task Force on Childhood Tuberculosis through De-
partment Order No 248-H s. 1998 (amended in 2001): a DOH initiative in
cooperation with PPS and other agencies.
Formal initiation by the WHO of PPM for TB control during the first global
assessment of PPM in TB control
First National Drug Resistance Survey supported by WHO and JICA be-
came the basis for the policy to address multi – drug resistant (MDR) TB
6
PPS
2006 – 2010 Inclusion of the PPMD component in the National Strategic Plan to stop
TB in the Philippines
7
136/100,000 population; TB - all forms with morbidity rate at
152/100,000 population (Philippine Health Statistics National Epidemiol-
ogy Center, DOH)
2007 – 2008 Establishment of 220 PPMD units nationwide covering 40% of the popu-
lation and contributing 90% in the national case detection
2012 PPS co-sponsored the 2012 World TB Day (24 March 2012) at the V.
Luna General Hospital Gymnasium, as well as the 16th PhilCAT Annual
Convention at the Crowne Plaza (17-17 August 2012)
2013 WHO organized external NTP review – Joint Program Review (JPR) of
the PhilPACT
2016 Republic Act No. 10767 ―Comprehensive Tuberculosis Elimination Plan Act‖
was signed into law on April 26, 2016.
8
2
Epidemiology
Luis T Chan, Jr, MD
Ana Liza H Duran, MD
OUTLINE
1 Overview
5 Drug-Resistant TB
7 Mode of Transmission of TB
9
OVERVIEW
Tuberculosis (TB) ranks along HIV as a leading cause of death from an infectious dis-
ease1. Mycobacterium tuberculosis (MTB) causes this infectious disease and primarily
affects the lungs and other sites as well. TB is seen in different parts of the world, and
death from the disease is still high.
TUBERCULOSIS KEY FACTS1,2:
In 2013, the treatment success rate continued to be high at 86% among All New TB
cases.
Between 2000 and 2014, TB treatment alone saved an estimated 35 million lives
among HIV- people.
In 2014, 9.6 million people are estimated to have fallen ill with TB: 5.4 million men,
3.2 million women and 1.0 million children.
6 million TB cases were reported to WHO. Of these, 5.7 million were newly di-
agnosed and another 0.4 million were already on treatment.
Among pulmonary TB cases, 58% were bacteriologically confirmed.
There were an estimated 700,000 TB deaths among HIV- men and 340,000
among HIV- women
TB killed 1.5 million people: 1.1 million HIV negative and 0.4 million are HIV
positive.
The death toll comprised 890,000 men, 480,000 women and 140,000 children.
10
The number of incident TB cases relative to population size (the incidence rate) varies
widely among countries. The lowest rates are found predominantly in high-income
countries, including most countries in Western Europe, Canada, United States of Amer-
ica, Japan, Australia, and New Zealand. In these countries, the incidence rate is less
than 10 cases/100,000 population.
Globally, the incidence rate was relatively stable form 1990 up until around 2000, and
then started to fall, achieving the MDG target ahead of the 2015 deadline. Between
2000 and 2013, the average rate of decline per year was 1.5%.
There were an estimated 13 million prevalent cases (range, 11 million to 14 million) of
TB in 2014, equivalent to 174 cases/100,000 population. By the end of 2015, it is esti-
mated that the prevalence rate will have fallen 42% globally since 1990, missing the
target. However, two regions met the target before 2015 (the Region of the Americas
and the Western Pacific Region) and the Southeast Asia Region reached the target
(according to the best estimate) in 2015. TB prevalence is falling in all of the other three
regions.
The number of TB deaths per 100,000 population averaged 16 globally in 2014, and 21
when TB deaths among HIV positive people are included. There is considerable varia-
tion among countries, ranging from under 1 TB death per 100,000 population (Western
Europe, Canada, USA, Australia and New Zealand) to more than 40 deaths per
100,000 population in much of the African Region as well as 3 High Burden Countries
(HBCs) in Asia (Bangladesh, Cambodia and Myanmar), and the 2 HBCs in the 2 East-
ern Mediterranean Region (Afghanistan and Pakistan).
Globally, the TB mortality rate (excluding deaths among HIV+ people) fell by 47% be-
tween 1990 and 2015, narrowly missing the target of a 50% reduction.
Western Pacific Region1,2,3
The Western Pacific Region, where the Philippines belongs, has an estimated popula-
tion of about 2 billion. The incidence of TB, including HIV positive patients, is 85 per
100,000 population. Mortality rate, excluding HIV+TB, is presently at 4.8 per 100,000
population while that of HIV+TB patients is 0.27/100,000 population. The prevalence
rate is 116 per 100,000 population in 2014. The estimated number of notified new and
relapse cases is close to 1.4 million, while the Case Detection Rate, all forms, is 85%.
(Table 2.1)
11
Table 2.1. Estimates of TB Burden 2014
NUMBER (Thousands) RATE (per 100,000
population)
Mortality (excludes 88 (81-95) 4.8 (4.4-5.1)
HIV+TB)
Mortality (HIV+TB only) 5 (4-6) 0.27 (0.23-0.31)
Prevalence (includes 2,100 (1,900-2,400) 116 (102-128)
HIV+TB)
Incidence (includes 1600 (1500-1600) 85 (80-89)
HIV+TB)
Incidence (HIV+TB only) 31 (28-35) 1.7 (1.5-1.9)
Case detection, all forms 85 (81-90)
(%)
National Profile1,2,3
Tuberculosis is the sixth leading cause of morbidity and mortality in the Philippines;
the country is ninth out of the 22 highest TB-burden countries in the world and has one
of the highest burdens of multi-drug resistant TB3. In 2014, the estimated incidence of
TB including HIV positive TB patients is 290,000 and TB deaths totaled 10,000 exclud-
ing HIV with TB. The WHO data of 2014 showed the Philippine TB incidence rate at
288/100,000 population, while the prevalence rate is at 417/100,000 population. The
percentage of the incidence of TB cases that are HIV positive was at 2.6/100,000 pop-
ulation in 2014. (Table 2.2).
Table 2.2. Estimates of TB Burden 2014
NUMBER RATE
(thousands) (per 100,000 population)
Mortality (excludes 10 (9-11) 10 (9.1-11)
HIV+TB)
Mortality (HIV+TB only) 0.08 (0.055-0.11) 0.08 (0.06-0.11)
Prevalence (includes 410 (360-470) 417 (367-471)
HIV+TB)
Incidence (includes 290 (250-320) 288 (254-324)
HIV+TB)
Incidence (HIV+TB only) 2.5 (2-3.2) 2.6 (2-3.3)
Case detection, all forms 85 (76-97)
(%)
TB IN CHILDREN
At least half a million children become ill with TB each year. Seventy to eighty percent
12
of children with TB, have the disease in their lungs (Pulmonary TB). The rest are af-
fected by TB disease in other parts of the body (Extrapulmonary TB). In 2014, children
aged <15 years of age accounted for 6.5% of notified cases. An estimated 550,000
children became ill with TB and 80,000 children who were HIV negative died from TB
in 2013. There were an additional 81,000 (range: 69,000-93,000) TB deaths among
HIV- children, equivalent to 27% of total HIV- TB deaths. In 2014, 359,000 new and
relapse cases among children were reported, an increase of about 30% compared with
2013. The largest increases were in India (about 30,000) and the Philippines (about
10,000).1
13
Table No. 2.4. Reported cases of RR-/MDR-TB 2014
NEW RETREATMENT TOTAL
Cases tested for RR-/MDR-TB 4,415 (5%) 20,196 (67%) 27,287
Laboratory-confirmed RR-/MDR 3,000
-TB cases
Patients started on MDR-TB 2,680
treatment
INFECTIOUSNESS OF TB PATIENTS4
The infectiousness of a person with TB disease is directly related to the number of tu-
bercle bacilli expelled into the air. A person is considered more infectious when more
tubercle bacilli are expelled. Infectiousness of TB patients also depends on a variety of
factors: susceptibility (immune status) of the exposed individual, environmental factors
that affect the concentration of M. tuberculosis organisms and the proximity, frequency,
and duration of exposure to an infectious source. In general patients who have suspect-
ed or confirmed TB disease should be considered infectious if:
1. They are coughing, undergoing cough-inducing procedures, or have positive
sputum smear results for acid-fast bacilli (AFB), presence of pulmonary cav-
ity;
and
2. They are not receiving adequate anti-tuberculosis therapy, have just started
therapy, or have a poor clinical or bacteriologic response to therapy.
The risk of TB infection among child contacts is associated with the proximity
and duration of contact and the degree of lung involvement and/or sputum smear posi-
tivity of the index case, which is usually from an infectious adolescent or adult.7-10 Child-
hood TB arises most often as a result of the inhalation of droplet nuclei approximately 1
to 5 micra in size containing M. tuberculosis bacilli expectorated by a sputum smear-
positive individual with pulmonary TB. TB infected infants and postpubertal adolescents
are at increased risk for progression to TB disease, and children aged less than five
years are at increased risk for disseminated disease.11
17
REFERENCES:
1. World Health Organization. Global Tuberculosis Report 2015
2. http://www.who.int/tb/publication/2015/factsheet_tb_2015 update November
2015.pdf
3. World Health Organization_HQ_Reports TB Country Profile
4. Centers for Disease Control and Prevention, National Center for HIV, STD and TB
Prevention Division of Tuberculosis Elimination. Core Curriculum on Tuberculosis,
What the Clinician Should Know. 6th ed. 2013
5. Frampton MW. An outbreak of tuberculosis among hospital personnel caring for a
patient with a skin ulcer. Ann Intern Med. 1992;117:312.
6. Haas DW. Mycobacterial diseases. Philadelphia, Churchill Livingstone. 2000. p.
2576-2607. (GL Mandell, Douglas, JE Bennett, editors. Principles and Practice of
Infectious Diseases. 5th ed.).
7. Grzybowski S, Barnett GD, Styblo K. Contacts of cases of active pulmonary tuber-
culosis. Bull Int Union Tuberc. 1975;50:90–106.
8. Marks SM, Taylor Z, Qualls NL, et al. Outcomes of contact investigations of infec-
tious tuberculosis patients. Am J Respir Crit Care Med. 2000;162:2033–8.
9. Kenyon TA, Creek T, Laserson K, et al. Risk factors for transmission of Mycobacte-
rium tuberculosis from HIV-infected tuberculosis patients, Botswana. Int J Tuberc
Lung Dis. 2002;6:843–50.
10. Lienhardt C, Sillah J, Fielding K, et al. Risk factors for tuberculosis infection in chil-
dren in contact with infectious tuberculosis cases in The Gambia, West Africa. Pedi-
atrics. 2003;111:e608–14.
11. American Academy of Pediatrics, Committee on Infectious Diseases. Tuberculosis.
Elk Grove Village, IL: American Academy of Pediatrics. 2003. p. 642–60.
(Pickering LK, editor. 2003 Redbook: report of the Committee on Infectious Diseas-
es. 26th ed.).
12. National Tuberculosis Controllers Association, Center for Disease Control. Guide-
lines for the Investigation of Contacts of Persons with Infectious Tuberculosis.
MMWR Weekly. 2005 December 16;54:RR-15.
18
3
Microbiology
Jaime A Santos, M.D
Mary Antonette C Madrid, M.D
OUTLINE
1 Overview
5 Laboratory Diagnosis
19
OVERVIEW
Mycobacterium tuberculosis, classified under the M. tuberculosis complex, is the agent
that causes tuberculosis in humans. Tuberculosis continues to be a subject of interest
in the medical field as there are still a lot to be investigated on the disease agent‘s viru-
lence and new information derived from the M. tb genome can contribute to the devel-
opment of therapeutic and preventive strategies. The availability of Xpert MTB/Rif as-
say, a real time-DNA based test, has revolutionized TB case detection by providing
early results. This chapter discusses the morphology, taxonomy, growth characteristics,
virulence factors, laboratory diagnosis, and the M. tb genome sequence.
20
based on growth rate and pigmentation. This scheme is becoming less important due to
increasing availability of new methods of rapid detection of mycobacteria. A rapidly
growing or pigmented isolate of Mycobacteria is not to be mistaken as M. tuberculosis 7
Among the important members of NTM are (1) M. avium complex (MAC), M. kansasii
and M. xenopi that can cause pulmonary disease; (2) M. avium and M. scrofulaceum
can cause lymphadenitis particularly in children, and, (3) MAC and M. kansasii are
causes of disseminated disease among HIV patients.8
Unlike members of the MTBC, NTMs have a wide range of pathogenicity and are not
transmissible from one human to another. The NTMs are also more difficult to treat be-
cause they are relatively drug resistant compared to M. tuberculosis.8
The phylogeographically relevant clonal distribution of the M. tuberculosis complex can
be accessed in the SpolDB4database.9,10
A more recent characterization of M. tb diversity and molecular epidemiology utilizing
the mycobacterial interspersed repetitive units–variable number of DNA tandem re-
peats (MIRU–VNTRs) method described another international database, the SITVIT-
WEB. This database has genotyping information on more than 62,582 isolates from
153 countries compared to only spoligotyping information on 39,295 isolates from 122
countries contained in the previous version of the SpolDB4 database.11,12
LABORATORY DIAGNOSIS
TB bacilli have a slow doubling time of 18 to 24 hours.2 Although the newly developed
techniques like PCR give earlier results, demonstration of the organism by culture is
still considered the gold standard for the diagnosis of TB.3
Specimen
Specimens include pulmonary secretions, urine, genital discharge, CSF, blood, pleural
fluid, and other biologic fluids. Demonstration of the bacilli in children is limited by the
fact that childhood TB (primary tuberculosis) is often paucibacillary and that very young
children cannot raise any sputum. For these reasons, a gastric aspirate specimen as-
sumes greater importance for children compared to adults. (For collection and handling
of specimens see chapter 8 on diagnostic tests)
The quality of specimens submitted for microbial investigation affects the performance
of the laboratory test. Specimens should reflect the site of infection and preferably col-
lected aseptically15
Microscopy
Acid-fast staining techniques:
a. The ZN technique (or the hot method) uses carbol fuchsin, which is taken up
by the cell even after treatment with acid alcohol. Mycobacteria will appear
as red rods against a blue background. Heat is applied in this method.
b. Kinyoun or cold stain is a modification of the ZN method and does not in-
volve application of heat. It uses a higher concentration of phenol with carbol
fuchsin. Mycobacteria appear as above.
c. Auramine-O stain makes use of auramine instead of carbol fuchsin. TB bacil-
li are seen fluorescing under the fluorescence microscope. The use of fluo-
rescent stain increases sensitivity of microscopy.15
Culture
Non-sterile specimens contaminated with commensal organisms undergo decontami-
nation prior to inoculation onto culture media. This is achieved thru the use of either
NaOH method or Nalc-NaOH method. 15
Culture Media15
The following are the suggested media for cultivation of Mycobacteria from clinical
specimens:
1. Solid Media is either egg-based or agar-based. Among the egg-based media, LJ is
the most popular
Agar-based: Middlebrook 7H10, Middlebrook 7H11, Mitchison's selective 7H11
Egg-based: Wallenstein, Lowenstein-Jensen (LJ) with RNA, LJ with pyruvic ac-
22
id; Ogawa16
2. Liquid Media15
Middlebrook 7H9 broth
Dubos Tween Albumin broth
Commercial liquid-based mycobacterial culture systems:
Bactec 460 TB
Bactec Mycobacterial Growth Indicator Tube (MGIT) 960 system
VersaTREK (ESP II) culture system
MB/BacT system
Of these culture media, the currently available ones in some local laboratories are Low-
enstein-Jensen, Middlebrook, Ogawa and Bactec.
Culture Method
1. Conventional
Conventional culture of mycobacteria involves inoculation on solid media. If this is the
only method used, inoculation of both the egg-based and agar-based media is recom-
mended. For specimens such as sputum that are contaminated with normal bacterial
flora, a selective medium containing antimicrobial agents, such as Mitchison's selective
7H11 agar, should also be inoculated.1
Conventional culture media should be examined for growth twice a week, for the first
four weeks, starting from day 3 to 5 postinoculation, and thereafter, once a week until
the eighth week. Positive cultures should be identified to the level of species using ei-
ther biochemical or molecular methods.16
The major advantage of conventional culture is that it allows visualization of colony
morphology and pigmentation. Disadvantages of conventional solid media are pro-
longed time of growth of mycobacteria (3 to 4weeks) and low sensitivity. 1,6,17,18 This is
because the doubling time (every 12-24 hours) of M. tuberculosis is extremely slow
compared to most bacteria.16
While solid media are still being used by most centers, the use of liquid media such as
Middlebrook 7H9 and Dubos Tween broths offers the advantage of more rapid growth
of mycobacteria as well as a higher isolation rate, than on solid media.15
2. BACTEC TB System and other commercial liquid-based culture systems
The most widely used system is the semi-automated BACTEC TB system which utilizes
broth media (one for blood and another for all other specimen types) that contain 14-C-
labeled palmitic acid substrate. Each specimen (5.0 ml of blood or 0.5 ml of all other
specimen types) is inoculated into one vial and an antibiotic mixture is added to speci-
mens other than blood. Bacilli multiply in the broth and utilize the 14-C-labeled sub-
strate, releasing CO2 into the head space above the broth. The amount of CO2 is
measured by the BACTEC 460, which calculates the growth index (GI). A GI greater
than ten suggests that mycobacteria are present. Vials containing mycobacteria are
23
sub-cultured to a solid medium, and direct tests for identification are done. 1 Bactec 460
(Beckton Dickinson) was the first commercial liquid-based mycobacterial culture system
used. It has been considered a standard against which most other systems were com-
pared15
There are advantages to the BACTEC TB system: rapid detection of growth (5 to 12
days); increased sensitivity compared with solid media; the ability to distinguish MTBC
from other mycobacterial species; and rapid susceptibility testing of isolates of M. tuber-
culosis. The major disadvantage of the BACTEC system is its radioactivity; the dispos-
al of which is expensive and may be problematic for some institutions1, 6, 17 However,
there are now non-radiometric BACTEC systems which provide a viable alternative to
the original radiometric BACTEC.18
Other available commercial liquid-based mycobacterial culture systems include Bactec
MGIT 960 culture system, Versa TREK culture system, and MB/BacT system. Bactec
MGIT is a system that uses a modified 7H11 broth containing an indicator that fluo-
resces under UV light. As oxygen in the medium is consumed, the indicator will fluo-
resce. Fluorescence is detected either manually using a Wood‘s lamp, or automatically
and continuously if Bactect 960 instrument is available. MB/BacT uses a colorimetric
sensor that changes color as increased amounts of C02 produced by the growing or-
ganism is detected. VersaTREK system monitors gas-related pressure changes that
occurs during bacterial multiplication. This was previously marketed as ESP II culture
system. 15
DNA Fingerprinting
Analysis of the M. tuberculosis genome is made through DNA fingerprinting, which
uses the restriction fragment-length polymorphism (RFLP) method. DNA fingerprinting
differentiates M. tuberculosis from other members of the MTBC and other atypical iso-
lates; it also permits the identification of patterns of spread of a particular M. tuberculo-
sis strain among individuals, which makes it possible to obtain the source of infection,
track down changes in antimicrobial susceptibility and allows for differentiation between
human and bovine isolates.6, 18, 19, 20 This technique utilizes restriction enzymes or en-
donucleases that cut DNA at specific "recognition" sites and which in turn results in
short segments or restriction fragments of DNA with lengths that vary according to the
particular enzyme used. The DNA fingerprints with unequal lengths and specific nucle-
otide sequences can be compared, in a map-like fashion, with the DNA patterns of M.
tuberculosis organisms from other individuals. If the patterns are the same, they sug-
gest a common source of exposure.6, 19
Nucleic acid amplification test (NAAT)
Using a nucleic acid probe, M. tuberculosis-specific nucleic acid sequences located on
regions of differences between mycobacterial DNA are amplified, allowing direct detec-
tion of organisms in clinical specimens. NAATs can also distinguish among MTBC or-
ganisms, can identify NTM, and has a role in rapid detection of drug resistance. It is
regarded as a complementary tool to the conventional laboratory approach to TB diag-
nosis, giving accurate and rapid results. Limitations to its use include diminished sensi-
tivity in smear-negative and non-respiratory samples, and cost.21 A commonly used
NAAT is PCR.
24
Xpert MTB/RIF assay
This is a new rapid diagnostic tool that simultaneously detects the presence of M. tu-
berculosis complex and rifampicin resistance, giving results in as early as 2 hours from
test run. It is a nucleic acid amplification test using the GeneXpert platform (developed
by Cepheid, Sunnyvale CA, U.S.). Sample purification, nucleic acid amplification and
detection of target sequence are combined in a single Xpert MTB/RIF cartridge. A rea-
gent that comes with the assay is added to the collected sample, then mixed and incu-
bated, before it is placed in the cartridge and loaded in the instrument. Following sam-
ple loading, all succeeding steps are fully automated. There is thus very minimal opera-
tor training needed. In addition to this, safety concerns on handling biohazardous mate-
rials are minimized because the assay‘s reagent is tuberocidal. These characteristics
make the test suitable for use outside a central or reference laboratory. The instrument,
however, will need an uninterrupted, stable electrical supply, temperature control and
yearly calibration.22, 23
Rifampicin resistance can been used to represent multidrug resistance since rifampicin
monoresistance is uncommon,24 and most isolates that are resistant to rifampicin are
also resistant to INH. The use of molecular methods for rapid detection of rifampicin is
feasible since >95% of resistance is associated with mutations in a limited region of the
M. tuberculosis rpoB gene (the gene that encodes the beta subunit of RNA polymerase
to which rifampicin binds under wild-type conditions), while INH resistance involves at
least four different genes and would be more difficult to detect. 21 Xpert MTB/Rif makes
use of molecular beacon technology to identify rifampicin resistance. 23 Molecular bea-
cons are nucleic acid hybridization probes that bind to target DNA sequences in regions
(such as the rpoB gene) where there are resistance mutations. The probes will fluo-
resce when bound to a target. Presence of mutation will prevent fluorescence. 21
XPERT MTB/RIF assay amplifies MTBC-specific sequence of the rpoB gene, then mu-
tations are searched for in the rifampicin resistance determining region (RRDR) using 5
molecular beacon probes (Probes A to E). 25
The test will determine if MTBC was detected or not detected in the sample. An
―invalid‖ result is also possible for which the test should be repeated. If MTBC was de-
tected, it will also indicate the following: 1) Rifampicin resistance detected, 2) Rifampic-
in resistance not detected, or, 3) Rifampicin resistance indeterminate. 22 Detailed steps
involved in the test are discussed in number of web sources.23, 25, 26
Based on WHO‘s Current Policies and Guidance, Xpert MTB/RIF is being recommend-
ed as an initial diagnostic test in suspected cases of MDR-TB or HIV-associated TB, as
well as a follow-on test for smear-negative specimens. WHO‘s recommendation on the
use of XpertMTB/Rif apply to the following specimens: processed or unprocessed spu-
tum, gastric lavage or aspirate, CSF, lymph node and other tissues. There are still inad-
equate data on the utility of the test for stool, blood and urine specimens.23
Conventional culture methods for TB detection take around 2-6 weeks before growth is
detected and an additional 3 weeks waiting time is needed for drug resistance results to
become available. The availability of XpertMTB/Rif assay sets an important milestone
in TB diagnosis and control as it offers rapid means of detecting MTBC and drug re-
sistance. Unnecessary regimens are avoided, appropriate drugs are provided, and in-
25
fection control is timely implemented.
For more information on the use of this test for TB diagnosis, please refer to
chapter 10.
26
REFERENCES:
1. Woods, Gail. Mycobacteria. In: Henry‘s clinical diagnosis and management
by laboratory methods. 22nd ed. / [edited by] Richard A. McPherson, Mat-
thew R. Pincus. Saunders Elsevier Ltd. PA. 2011, p. 1145.
2. Baron S. Mycobacteria and Nocardia. In Medical Microbiology. 4 th ed. UTMC;
1996.
3. Grange JM, Yates MD, de Kantor IN. Guidelines for specification within the
Mycobacterium tuberculosis complex. WHO/EMC/ZOO/96.4.
4. Grange, GM. The genus Mycobacterium and Mycobacterium tuberculosis
complex. In: Tuberculosis A Comprehensive Clinical Reference. Zumla A.I.
and Schaaf H.S. eds. Saunders Elsevier Ltd., Oxford, UK, 2009, pp. 44-59.
5. Grange JM. Pathogenesis of tuberculosis: pathway to apical localization. Tu-
bercle and Lung Disease. 1995;76:276-277.
6. Baron EJ, et. al. Tuberculosis. In: Bailey and Scott‘s Diagnostic Microbiology.
9th ed. p. 590-633.
7. Mycobacterium. In: Medical Microbiology 7th ed. Murray PR, Rosenthal KS,
Pfaller MA. eds. Saunders, Elsevier Inc. Philadelphia, PA. 2013, pp. 235-247
8. Daley CL, Heifets L. Other mycobacteria causing human disease. In: Tuber-
culosis A Comprehensive Clinical Reference. Zumla A.I. and Schaaf H.S.
eds. Saunders Elsevier Ltd., Oxford, UK, 2009, pp.60-74.
9. SpolDB4database web link: http://www.pasteur-guadeloupe.fr/tb/
bd_myco.html or http://www.pasteur-guadeloupe.fr:8081/SITVITDemo
10. Brudley et al. Mycobacterium tuberculosis complex genetic diversity: mining
the fourth international spoligotyping database (SpolDB4) for classification,
population genetics and epidemiology. BMC Microbiol 2006 Mar 6;6 (1) : 23.
11. Demay et al. A publicly available international multimarker database for stud-
ying Mycobacterium tuberculosis genetic diversity and molecular epidemiolo-
gy. Infect Genet Evol. vol 12, issue 4, 2012 June.
12. http://www.pasteur-guadeloupe.fr:8081/SITVIT_ONLINE
13. Geisel RE, Sakamoto K, Russell DG, et al. In vivo activity of released cell
wall lipids of Mycobacterium bovis bacillus Calmette-Gue ́rin is due principal-
ly to trehalose mycolates. J Immunol 2005;174:5007–5015.
27
14. Kang PB, Azad AK, Torrelles JB, et al. The human macrophage mannose
receptor directs Mycobacterium tuberculosis lipoarabinomannan-mediated
phagosome biogenesis. J Exptl Med 2005;202:987–999.
15. Whitelaw AC and Sturm WA. Microbiological testing for Mycobacterium tu-
berculosis. In: Tuberculosis A Comprehensive Clinical Reference. Zumla A.I.
and Schaaf H.S. eds. Saunders Elsevier Ltd., Oxford, UK, 2009, pp. 169-
178.
16. Palomino JC, Leão SC, Ritacco V. Tuberculosis 2007: From basic science
to patient care. 1st ed. Available from: www.TuberculosisTextbook.com
17. Luquin M, et. al. Comparison of a biphasic non-radiometric system with Low-
enstein-Jensen and Bactec-460 system for recovery of mycobacteria from
clinical specimens. Tubercle and Lung Disease. 1996;77:449-453.
18. Behr H, Small PM. Molecular fingerprinting of Mycobacterium tuberculosis:
How can it help the clinician? Clinical Infectious Diseases. 1997;25:806-810.
19. Nogueira JM, et. al. Correspondence: Molecular approach to identifying
route of M. tuberculosis transmission in a village. The Int. Journal of Tuber-
culosis and Lung Diseases. 2000;4(1):91-93.
20. Rigouts L, et. al., Use of DNA restriction fragment typing in the differentiation
of M. tuberculosis complex isolates from animals and humans in Burundi.
Tubercle and Lung Diseases. 1996;77:264-268.
21. Daniel Brodie and Neil W Schluger. Nucleic acid amplification for detection
of Mycobacterium tuberculosis. In: Tuberculosis A Comprehensive Clinical
Reference. Schaaf and Zumla (Eds). pp 197-204
22. CDC. A new diagnostic tool to diagnose TB. The Xpert MTB/RIF Assay.
Downloaded from: http://www.cdc.gov/tb/publications/factsheets/pdf/
xpertmtb-rifassayfactsheet_final.pdf
23. World Health Organization. Automated real-time nucleic acid amplification
technology for rapid and simultaneous detection of tuberculosis and rifampic-
in resistance: Xpert MTB/RIF assay for the diagnosis
of pulmonary and ex-
trapulmonary TB
in adults and children. Policy update.
24. Cruz AT, Starke J. Tuberculosis. In: Feign and Cherry’s Textbook of Pediat-
ric Infectious Diseases. Feign RD, Cherry JD, Demmler-Harrison GJ and
Kaplan SL (eds). Philadephia USA Saunders Elsevier, 7th ed, 2014, 1335-
1380
28
25. Xpert MTB/RIF package insert. Cephaid, Sunnyvale CA, USA. Revised Feb-
ruary 2015. Downloaded from: http://www.cepheid.com/administrator/
components/com_productcatalog/library-
files/2cc115cd6ba267207c04661a3d0fcfc8-Xpert-MTB-RIF-ENGLISH-
Package-Insert-301-1404-Rev-B-February-2015.pdf
26. http://www.finddiagnostics.org/programs/tb/find_activities/
automated_naat.html
27. Genome sequence and analysis. The Institute for Genomic Research. Avail-
able from: www.tigr.com
28. Cole ST, Brosch R, et. al. Deciphering the biology of Mycobacterium tuber-
culosis from the complete genome sequence. Nature. 1998;393;537-544.
29. Lew, JM, Kapopoulou A, Jones LM, Cole ST. TubercuList—10 years after.
Tuberculosis (Edinb). 2011 Jan;91(1):1-7.
30. TubercuList - Mycobacterium tuberculosis Database: tuberculist.epfl.ch/
31. Li L, Bannantine JP, Zhang Q, et. al. The complete genome sequence of My-
cobacterium avium subspecies paratuberculosis. Proc Natl Acad Sci (USA).
2005;102:12344-9.
32. Maari PR, Bannantine JP, Golding GB. Comparative genomics of metabolic
pathways in Mycobacterium species: gene duplication, gene decay and lat-
eral gene transfer. FEMS Microbiol Rev. 2006;30:906-25.
33. Young DB. Blueprint for the white plague. Nature. 1998;393:515-516.
29
4
Immune Responses
Jaime A Santos, M.D
Mary Antonette C Madrid, M.D
OUTLINE
1 Overview
30
OVERVIEW
The healthy tuberculin reactor and a patient with severe miliary tuberculosis (TB) repre-
sent the two ends of the spectrum of TB infection and disease—extremely effective and
poor immune responses to M. tuberculosis. A relatively good immune response is seen
in a patient with TB pleuritis (as evidenced by few bacilli in pleural fluid, resolution of
disease without treatment and preserved tuberculin skin test responses), while a rela-
tively poor immune response leads to advanced, progressive pulmonary disease. While
immune responses do not entirely account for the clinical spectrum of TB infection as it
does in leprosy, an enhanced understanding of the immunology of TB coupled with the
knowledge gained from the sequencing of the M. tuberculosis genome could signifi-
cantly lead to breakthroughs in vaccine development and TB immunotherapy.1, 2,3
Although there are differences between murine models of TBs and human TB disease,
there is extensive agreement and experimental support for the following: macrophages
and T lymphocytes are the primary defenses against TB; antibodies and B lympho-
cytes have little role, if any, in protecting one against TB; and. the role of neutrophils
and natural killer (NK) cells is unclear.
31
suppress IFN-gamma responses to protective antigens, such as HBHA; this suggests
another escape mechanism of M. tuberculosis.13
Mycobacterial Antigens
In mice, protective immunity against TB can be induced by live, but not dead, mycobac-
teria while delayed-type hypersensitivity (DTH) can be induced by either dead or live
organisms or by cell wall components. This phenomenon suggests that protection and
tuberculin reaction are dissociable; that secreted antigens are important relative to cell-
bound or structural antigens, and that the way the antigen is presented to the immune
system is crucial for immunity. Among the prime candidates of protective antigens are
ESAT-6,15 and the 30 kDa a or 85 B antigen.3 Another protective antigen that has come
to prominence is heparin-binding hemagglutinin (HBHA), a mycobacterial antigen that
can induce mycobacterial aggregation. Presence of antibodies against HBHA (anti-
HBHA antibodies) inhibits hemagglutination, mycobacterial aggregation, as well as at-
tachment to epithelial cells.16 HBHA has also been shown to stimulate secretion of high
levels of IFN-γ by peripheral blood lymphocytes among subjects with latent TB infec-
tion.17,18
32
Cells mediating the tuberculin skin test (delayed-type hypersensitivity) are CD4-
positive, producing mostly Th1 cytokines. DTH response is associated with, but not
identical to, cell-mediated immunity. The reasons may be: (1) DTH responses are a
function of CD4 T cells, whereas protective immunity involves CD8 T cells (cytotoxic
T lymphocytes) and unconventional T-cell subsets as well; (2) PPD does not contain
all the antigens derived from M. tuberculosis; and (3) in TB patients who have granulo-
ma formation but have negative tuberculin test, T cells may be trapped in granulomas
reflecting their accumulation at the site of defense.21
A recognized subset of T-helper cells, called Th17 cells, were found to be an important
modulator of inflammation and CD4+ T-cell recall or memory response. Th17 cells se-
crete IL-17, can recruit neutrophils, monocytes and CD4+ T-cells producing IFN-
gamma, and stimulate chemokine expression.22
Anergy, indicated by a negative tuberculin test, reflects suppression of DTH from pro-
cesses which interfere with Th1 response i.e. malnutrition, HIV infection, steroid thera-
py and severe tuberculosis itself (as a result of production of inhibitory cytokines like
TGF-b). Anergy is often transient and reverts to normal when underlying causes are
resolved.23
The possibility of modifying deleterious immune responses or shunting the immune
response to a direction favorable to the patient is among the driving forces behind re-
search on cytokines and tuberculosis.
Cytolytic or Cytotoxic T Lymphocytes
Cytolytic or cytotoxic T cells kill cells harboring intracellular pathogens including M. tu-
berculosis. These cells either exhibit granzyme/perforin-mediated killing of target cells
(in which the cell membrane is destroyed as an initial step) or induce apoptosis (in
which destruction of target cell DNA occurs followed by lysis of the cell). Although CD8
T cells are known as cytotoxic T cells, they are also capable of lysing monocytic cells
infected with M. tuberculosis, mostly through apoptosis.2, 15 CD8 T cells have likewise
been shown to be cytotoxic against M. bovis-infected macrophages in-vitro.24 The rela-
tive contribution of cytolytic T cells to the total protective immune response to TB is un-
known but is expected to be significant.
33
determinants and may be related to the largely unexplored relationship between histo-
compatibility leukocyte antigen (HLA) complex and TB. Studies on the completed hu-
man genome sequence could give leads in predicting human susceptibility genes.3,27
34
infection identified possible reasons for the development of disease and these include:
1) impaired antigen-specific CD4þ T-lymphocyte responses, 2) decreased capacity of
neonatal T-lymphocytes to secrete IFN gamma and TNF alpha, 3) cytokine production
leans towards a Th2 response, and 4)suboptimal response of CD8þ T-
lymphocytes.35,36.37
35
REFERENCES
1. Stead WW : Pathogenesis of Tuberculosis: Clinical and epidemiologic perspective. Reviews
of Infectious Diseases 1989; 11 (supplement 2): S366-8.
3. Ellner JJ: The Immune Response in Human Tuberculosis – Implications for Tuberculosis
Control. The Journal of Infectious Diseases 1997; 176:1351-9.
4. Riley LW: Phagocytosis of M. tuberculosis in Rom WN, Garay S (ed): Tuberculosis, Boston,
New York, Little, Brown and Company, 1996; 281-9.
10. Vergne I, Singh S, Roberts E, Kyei G, Master S et al. Autophagy in immune defense against
Mycobacterium tuberculosis. Autophagy 2006; 2: 175–8
11. Barnes PF and Rom WN. Cytokine Production in Tuberculosis in Rom WN and Garay S
(ed.): Tuberculosis, Boston, New York, Little Brown and Company, 1996; 291-303.
13. Hougardy JM, Place S, Hildebrand M, Drowart A, Debrie AS et al. Regulatory T cells de-
press immune responses to protective antigens in active tuberculosis. Am. J. Respir. Crit.
Care Med. 2007; 176: 409–16.
14. Rook GA. Th2 cytokines in susceptibility to tuberculosis. Curr.Mol. Med. 2007; 7: 327–37.]
16. Menozzi FD, Rouse JH, Alavi M, Laude-Sharp M, Müller J, et al. (1996) Identification of a
36
heparin-binding hemagglutinin present in mycobacteria. J Exp Med 184: 993–1001.
17. Masungi C, Temmerman S, Van Vooren JP, Drowart A, Pethe K, et al. (2002) Differential T
and B cell responses against Mycobacterium tuberculosis heparin-binding hemagglutinin
adhesin in infected healthy individuals and patients with tuberculosis. J Infect Dis 185: 513–
520.
19. Zhang M, Lin Y, Iyer DV et al. T-cell cytokine responses in human infection with Mycobacte-
rium tuberculosis, Infection and Immunity, 1995, 63: 3231-34.
20. Barnes PF, Fong SJ, Brennan PJ et al. Local production of TNF and IFN- in tuberculous
pleuritis, Journal of Immunology, 1990, 145: 149-154.
21. Kauffmann SHE. Immunity to Extracellular Bacteria in. Paul WE (ed.) Fundamental Immu-
nology, Philadelphia, Lippincott-Raven, 1999; 1335-72.
22. Khader SA, Bell GK, Pearl JE, Fountain JJ, Rangel-Moreno J et al. IL-23 and IL-17 in the
establishment of protective pulmonary CD4+ T cell responses after vaccination and during
Mycobacterium tuberculosis challenge. Nat. Immunol. 2007; 8: 369–77
23. Karim M and Ellmer JJ. Mechanisms of Anergy in Rom WN and Garay S (ed.): Tuberculo-
sis, Boston, New York, Little Brown and Company, 1996; 343-51.
24. Turner J and Dockrell HM, Stimulation of Human peripheral blood mononuclear cells with
live Mycobacterium bovis BCG activates cytolytic CD84 T cells in vitro, Immunology, 1996;
87: 339-42.
25. Medina E, North RJ, Evidence inconsistent with a role for the BCG gene (Nramp1) in re-
sistance of mice to infection with virulent Mycobacterium tuberculosis., Journal of Experi-
mental Medicine, 1996; 183: 1045-51.
26. Soborg C, Andersen AB, Madsen HO, Kok-Jensen A, Skinhoj P, Garred P. Natural-
Resistance-Associated Macrophage Protein 1 Polymorphisms are Associated with Micros-
copy-Positive Tuberculosis. The Journal of Infectious Disease. 2002;186:577-21
27. Initial sequencing and analysis of the human genome. The genome international sequenc-
ing consortium. Nature, 2001; 409: 860-921
28. Palomino JC, Leão SC, Ritacco V. Tuberculosis 2007 From basic science to pa-
tient care TuberculosisTextbook.com First Edition
29. Parrish NM, Dick JD, Bishai WR. Mechanism of latency in Mycobacterium tubercu-
losis. Trends Microbiol 1998; 6: 107-12.
30. Voskuil, M. I., Schnappinger, D., Harrell, M. I., Visconti, K. C., Dolganov, G. M.,
Sherman, D. R. & Schoolnik, G. K. (2002). Inhibition of respiration by nitric oxide
induces a Mycobacterium tuberculosis persistence program. J Exp Med 198, 705–
713.
37
31. Arriaga AK, Orozco EH, Aguilar LD, Rook, GAW, Hernandez-Pando R. Immunologi-
cal and pathological comparative analysis between experimental latent tuberculosis
infection and progressive pulmonary tuberculosis. Clin Exp Immunol 2002; 128: 229
-37.
32. Flynn JL, Scanga CA, Tanaka KE, Chan J. Effects of aminoguanidine on latent mu-
rine tuberculosis. J Immunol 1998; 160: 1796-806.
33. Farel LS, Lowell AM, Meador MP. Extrapulmonary tuberculosis in the United
States. Am J Epidemiol 1979; 109: 205-17.
34. Hopewell PC. Overview of clinical tuberculosis. In: Bloom BR. Ed. Tuberculosis:
pathogenesis, protection, and control. Washington DCASM Press 1994; pp. 25-46.
35. Eley BS and Beatty DW. The basic immunology of tuberculosis. In: Tuberculosis: A
Comprehensive clinical reference. Schaaf HS and Zumla AI (eds). Saunders Else-
vier 2009, 75-86.
36. Lewinsohn DA, Gennaro ML, Scholvinck L, et al. Tuberculosis immunology in chil-
dren: diagnostic and therapeutic challenges and opportunities. Int J Tuberc Lung
Dis 2004;8:658–674.
37. Smith S, Jacobs RF, Wilson CB. Immunobiology of childhood tuberculosis: a win-
dow on the ontogeny of cellular immunity. J Pediatr 1997;131:16–26
38
5
Pathogenesis
Maria Anna Pablo-Baňez, MD
Cynthia Cecilia J de Ocampo, MD
OUTLINE
1 Overview
2 Source of Infection
5 Incubation Period
7 Timetable of Tuberculosis
39
OVERVIEW
A series of events occurs from exposure to M. tuberculosis, its transmission and the
development of infection and disease. A good grasp of the natural history enables one
to understand the broad spectrum of disease and the approach to management.
SOURCE
Children are usually infected by a tuberculous adult or adolescent in the immediate
household, but exposure can occur in a school, day care, or other closed settings. The
risk of infection is greatest with a household exposure to a sputum smear-positive
source.1,7 Contributory environmental factors, which increase the probability of trans-
mission, include overcrowding and poor ventilation.2 ,7
According to the World Health Organization (WHO), the following are the key risk fac-
tors for TB:1
INCUBATION PERIOD
Incubation period or the time between entry of the tubercle bacilli into the body and the
development of tissue hypersensitivity manifested as a reactive tuberculin test, ranges
from 3 to12 weeks8,10. It may be shorter when the inoculum is large.8,9
41
strained replication. Some macrophages carry the bacilli from the lung to regional (hilar
and mediastinal) lymph nodes, from which they may be disseminated via lymphatics or
bloodstream to other sites, including kidneys, meninges, epiphyseal plates of long
bones, vertebrae and apical segments of the upper lobes of the lungs.
Eventually, the logarithmic increase in the number of bacilli is inhibited by the develop-
ment of both cell-mediated immunity (CMI) and delayed-type hypersensitivity (DTH).
Infected macrophages present tuberculous antigens to T lymphocytes.
T lymphocytes get sensitized, to produce a progeny of similarly reactive cells that se-
crete lymphokines (IFN gamma and TNF) and activate macrophages, thereby increas-
ing their concentration of lytic enzymes and greatly enhancing their capacity to kill the
bacilli. These lytic enzymes in the activated macrophages, which include epitheliod
macrophages and Langhans giant cells, however, may also cause tissue necrosis if
released into surrounding tissue.
The development of a population of activated lymphocytes responsive to M. tuberculo-
sis antigen constitutes the delayed-type hypersensitivity response (DTH) to the organ-
ism. The related development of activated macrophages with increased capacity to in-
gest and destroy the bacilli comprises the cell-mediated immunity (CMI).These re-
sponses work together to contain the infection.
The classic lesion in the lung of primary tuberculosis is a caseous granuloma called the
Ghon focus. It is characteristically located peripherally in any part of the lungs, close to
the pleura. A granuloma has a soft, semi-soild core surrounded by epithelioid macro-
phages, Langhans giant cells and lymphocytes.
Dissemination of the bacilli from the primary granuloma in the lungs to the hilar nodes
causes a granulomatous lymphadenitis. The combination of granulomatous hilar lym-
phadenitis and Ghon focus forms the Ghon complex.
In most instances, the primary infection is controlled with a reactive tuberculin skin test
as the only evidence of infection. In both the lungs and lymph nodes,the lesions of the
Ghon complex heal by fibrosis,but subsequent calcification may occur. While most of
the bacilli die, a few may remain viable and reactivate later, if immune mechanisms
wane or fail.11,12,13,14
Secondary (Adult-type) TB
This occurs following re-infection with M.tuberculosis or reactivation of dormant bacilli in
the Ghon focus or complex. Reactivation is usually brought about by immunosuppres-
sion from whatever cause. The lungs, particularly in the apices, are most often affected
due to higher oxygen tension and poor lymphatic drainage, but infection may be widely
disseminated.
In secondary TB, tissue destruction is brought about by progressive, frequently exten-
sive caseous necrosis due to inability to contain the bacteria. The course is variable.
The localized inflammation may undergo organization and healing into a fibrocalcified
nodule, progress into caseating granulomas with cavitation, or disseminate to other or-
gans via rupture into a bronchus, pulmonary artery, pulmonary veins or lymphat-
ics.8,12,14
TIMETABLE OF TUBERCULOSIS
Wallgren‘s timetable of TB previously described the early course and its progression
over time (see figure 5.1)8,19. Complications of tuberculosis occur during the first five
years after the initial infection. An arrested or silent lesion may reactivate, heralding the
complications.
43
Figure 5.1 Wallgren’s Timetable of Tuberculosis8,19
The end of the initial asymptomatic incubation period, around 3-12 weeks after the pri-
mary infection is characterized by hypersensitivity reactions such as fever, erythema
nodosum, positive TST response, and primary complex on chest x-ray. Hypersensitivi-
ty to tuberculoprotein antigens once developed after infection is expected to remain re-
active for life.
It is estimated that an immunocompetent adult without treatment has a 5-10% risk to
develop the disease during their lifetime.8,10,16 On the other hand, serious, life-
threatening forms of the disease will develop within 1 to 2 years in as many as 40 per-
cent of immunocompetent infants with untreated tuberculosis infection.8,10
One to three months after the primary infection, following the occult hematogenous
spread during the incubation, is the highest risk for TB meningitis and disseminated
(miliary) TB. However, TB meningitis and military tuberculosis may also occur after any
time interval.
Around 3 to 7 months after the primary infection is the period of secondary airway in-
volvement due to infected lymph nodes in children younger than 5 years of age. Large
reactive pleural effusions can also occur during this time but more often among children
older than 5 years of age.
44
Approximately 1 to 3 years after the primary infection is the period of osteoarticular TB
in children under 5 years of age, and adult type of disease among adolescents. The risk
of TB progression following primary infection is much less by the time calcification ap-
pears. In adolescents, adult type disease may have a delayed clinical onset after calcifi-
cation, ranging from as early as 3 months to as late as 20 years.
More than 3 years after the primary infection, when the calcification is completed, the
highest risk period is said to have passed. However, some of the very late manifesta-
tions of TB, including primary pulmonary reactivation may occur. Renal involvement
appears much later, which is usually 5 to 25 years after the initial infection.7
With regards to TB-related mortality, infants have the highest risk following primary in-
fection at 5 to 10% as they are more likely to develop more severe forms of TB. The
risk decreases to 1% between 1 to 4 years old and with the lowest risk at < 0.5% at 5 to
14 years old. The risk again increases to more than 2% after 15 years of age. 7 In a re-
port by Schaaf et al17 it was estimated that 13% of children under 3 months with TB
died. Among children below 10 years old, majority of mortality occurred within the 1 st
year following the primary infection, but for older children who developed adult type
cavitary disease, mortality declined more slowly.7 TB-related mortality resulting from a
sputum smear-positive household source case is almost twice as high compared to an
infection from an unknown source case. Mortality rate is three times increased among
TST-positive reactors than those who are TST-negative reactors.18
45
REFERENCES
1. WHO Guidance for national tuberculosis programmes on the management of tu-
berculosis in children .2nd ed.2014:p21
2. Morcillo N. Tuberculosis in children. In: Palomino JC, Leao SC, Ritacco V.editors.
Tuberculosis 2007: From basic science to patient care. 1st ed.p527-528. Retrieved
from: www.TuberculosisTextbook.com.
3. Stop TB Partnership Childhood TB Subgroup. Chapter 1: Introduction and diagno-
sis of tuberculosis in children.Int J Tuberc Lung Dis. 2006;10:1091-1097
4. Schluger NW. The pathogenesis of tuberculosis. Am J Respir Cell Mol Biol.
2005;32:251–256.
5. Smith I. Mycobacterium tuberculosis Pathogenesis and Molecular Determinants of
Virulence. Clinical Microbiology Reviews. 2003;16:463–496.
6. Pope AS, Sarwell, Zacks D. Development of Tuberculosis in Infected Children. Am
J Public Health .1939; 29: 1318-1325
7. Marais BJ and Donald PR.The Natural History of Tuberculosis Infection and Dis-
ease in Children. In: Schaaf HS., Zumla, AI.editors. Tuberculosis: A Comprehen-
sive Clinical Reference. Saunders Elsevier Ltd., Oxford, UK; 2009:133-145
8. Starke JR and Cruz AT. Tuberculosis.In: FeiginRD,Cherry JD,editors.Textbook of
Pediatric Infectious Diseases. 7thed.WB.Saunders Co;2014,1335-1380.
9. American Academy of Pediatrics.Tuberculosis.In:Kimberlin DW,Brady MT. et al edi-
tors.Red Book: 2015 Report of the Committee on Infectious Diseases,30th ed.:805-
831
10. Hatzenbuehler LA and Starke JR. Tuberculosis.In: Kliegman RM, Stanton BF,et al,
editors.Nelson Textbook of Pediatrics.20th ed.Elsevier;2016:1445-1461
11. DannenbergAM,Jr.PathogenesisandImmunology:BasicAspects.In:Schlossberg
D,editor.Tuberculosis.3rd ed. Springer-Verlag; 1994:17-27.
12. Rubin E, Farber JL, editors. Textbook of Pathology. 4th ed. Philadelphia: Lippincott-
Raven; 2005 : 421– 424.
13. Adler JJ, Rose DN. Transmission and Pathogenesis of Tuberculosis. In: Rom WN
and Garay SM,editors.Tuberculosis. Boston: Little Brown and Company; 1996: 129-
140.
14. Bezuidenhout J. and Scheider J.W. Pathology and Pathogenesis of Tuberculosis.
In: Schaaf HS., Zumla, AI,editors. Tuberculosis: A Comprehensive Clinical Refer-
ence. Saunders Elsevier Ltd., Oxford, UK; 2009: 117-128
15. Bernard J. (2015). Epidemiology and Pathology of Extrapulmonary and Miliary tu-
46
berculosis.In:UpToDate, ,von Reyn CF and Baron EL.(Eds), Retrieved
www.uptodate.com/.../epidemiology-and-pathology-of-extrapulmonary-.and miliary-
tuberculosis ..Aug 2015
16. Maher D. The natural history of Mycobacterium tuberculosis infection in adults.In:
Schaaf HS., Zumla, AI,editors.Tuberculosis: A Comprehensive Clinical Reference.
Saunders Elsevier Ltd., Oxford, UK; 2009:129-132
17. Schaaf HS, Gie RP, et al.Tuberculosis in infants less than 3 months of age.Arch Dis
Child .1993;69:371-374
18. Marais BJ, Gie RP, Schaaf HS, et al. The Clinical Epidemiology of Childhood Pul-
monary tuberculosis: a critical review of literature from the pre-chemotherapy era.
Int J Tuberc Lung Dse. 2004; 8:278-285
19. Wallgren, A. Primary pulmonary tuberculosis in children. Am J Dis
Child.1935;49:1105-1136
47
6
Clinical Forms
Gyneth Lourdes G Bibera, MD,
Mary Ann C Bunyi, MD,
Michelle C Cuvin, MD,
Fatima I Gimenez, MD
OUTLINE
1 Overview
2 Definition of Terms
3 Pulmonary TB
4 Extrapulmonary TB
48
OVERVIEW
Mycobacterium tuberculosis can produce infection and disease in almost every tissue
and organ in the body. The disease is usually the result of dissemination from an initial
primary focus. Since infection usually takes place by way of the respiratory tract, the
lung is the first organ involved and it is here that the initial major manifestations of dis-
ease occur.
Distinction must be made between infection and disease. With primary infection, pa-
tients are asymptomatic. When disease is present, early manifestations are mild with
nonspecific systemic symptoms of low grade fever, lassitude, easy fatigability, anorexia,
weight loss, malaise, and night sweats which are common to many illnesses in child-
hood. With progression and chronicity, symptoms may be respiratory or other mean
depending on the main site.
DEFINITION OF TERMS
TB Exposure – a condition in which a child is in close contact with an active adult TB
cases, but without any signs and symptoms of TB, with negative TST reaction, and no
radiologic and laboratory findings suggestive of TB.1
TB Infection or Latent TB infection (LTBI) – a condition in which a child has no signs
and symptoms presumptive of TB nor radiologic or laboratory evidence, but has a posi-
tive TST reaction1
TB disease – a presumptive TB who after clinical and diagnostic evaluation (TST and/
or radiology) is confirmed to have TB. Classification is either based on the bacteriologi-
cal status or on the anatomical site.1
Bacteriological status1
Bacteriologically confirmed – biological specimen is positive by smear mi-
croscopy, culture or rapid diagnostic tests (such as Xpert MTB/RIF)
Clinically diagnosed – does not fulfil the criteria for bacteriological confirma-
tion but has been diagnosed with active TB by a clinicial or other medical
practitioner who has decided to give the patient a full course of TB treat-
ment. Cases diagnosed on the bases of Xray abnormalities or suggestive
histology, and extrapulmonary cases without laboratory confirmation are also
included.
Anatomical Site2
Pulmonary TB (PTB) – refers to a case of tuberculosis involving the lung pa-
renchyma. A patient with both pulmonary and extrapulmonary TB should be
classified as a case of pulmonary TB
Extrapulmonary TB (EPTB) – refers to a case of tuberculosis involving or-
gans other than the lungs (eg. Larynx, pleura, lymph nodes, abdomen, geni-
tourinary tract, skin, joints and bones, meninges). Histologically diagnosed
EPTB through biopsy of appropriate sites will be considered clinically diag-
49
nosed TB. Laryngeal TB, through likely sputum smear-positive, is consid-
ered an extrapulmonary case in the absence of lung infiltrates on chest x-
ray.
PULMONARY TB (PTB)
Primary Disease
This is the initial of children who inhale the Mycobacterium tuberculosis. it contains 3
elements namely: the Ghon focus, lymphadenitis, and lymphangitis. In 95% of cases,
it heals by fibrosis and/or calcification.
Primary TB in children is asymptomatic in up to 65% of patients.2 There are no striking
clinical manifestations. Signs and symptoms can vary substantially in the population
although infants and young children are likely to have more signs and symptoms than
school-aged children. Occasionally, the initiation of the infections is marked by several
days of low grade fever and mild cough.3 Fever alone or sometimes with pleuritic or ret-
rostenal pain of gradual onset can also be seen. Fever can be low grade in character
which can last for an average of 14 to 21 days.4
Progressive Primary Tuberculosis
This is a condition which develops when initial infection fails to heal and continues to
progress over a period of months or years, causing further pulmonary and even distant
organ involvement
Secondary (Reactivation) Tuberculosis2
A condition which represents reactivation of an old, possibly subclinical infection. This
occurs in less than 10% of the cases of primary infection. This tends to produce more
damage to the lungs than does primary tuberculosis
This is more common in adolescents than in younger children. Clinical manifestations
include chronic or persistent cough, prolonged fever, chest pain, hemoptysis, and su-
praclavicular adenitis. Most of these features may however improve within several
weeks after starting effective treatment, but the cough may persist for several months.
Miliary Tuberculosis
This refers to clinical disease resulting from the hematogenous dissemination of Myco-
bacterium tuberculosis. It can arise as a result of progressive primary infection or via
reactivation of latent form with subsequent spread. It is considered to be the most com-
mon clinically significant form of disseminated tuberculosis which occurs when massive
numbers of tubercle bacilli are released into the bloodstream causing disease in two or
more organs.2 Originally a pathologic and then a radiographic description, the term mili-
tary tb is now used to denote all forms of progressive, widely disseminated hematog-
enous tuberculosis.4
The clinical presentation of military TB is protean and non-specific, depending on the
load of organisms and where they lodge.3 Manifestations can be acute and explosive
and the patient may become gravely ill in several days3 but are more likely to be sub-
50
acute or chronic. This can also present with relatively acute onset and rapid clinical
course such as multiorgan failure, septic shock and acute respiratory distress syn-
drome (ARDS). The subacute or chronic presentations of military TB include failure to
thrive, fever of unknown origin and dysfunction of one or more organ systems including
the brain.5,6 The most common extrapulmonary sites of disease include the lymphatic
system, bones and joints and the liver. In children with military tuberculosis, clinical
presentation is similar to that in adults. Chills, night sweats, hemoptysis, and produc-
tive cough have been reported less frequently in the pediatric population while periph-
eral lymphadenopathy and hepatomegaly are more common in children compared with
adults. Signs or symptoms or peritonitis are found in 20 to 40% of patients with ad-
vanced disease.3
EXTRAPULMONARY TB
Endobronchial tuberculosis
This condition can be a complication of primary tuberculosis which results to enlarge-
ment of peribronchial lymph nodes with subsequent compression or intrabronchial,
which is the nodal extension into the bronchus. Such obstruction can result to sudden
death due to asphyxia, obstructive emphysema (partial obstruction with hyperaeration
of a lobar segment), or atelectasis of a distal segment (complete obstruction of the lu-
men). The right middle lobe is more vulnerable by its anatomical feature and drainage.
Signs and symptoms of progression include moderately high fever, anorexia, night
sweats, loss of weight and paroxysmal cough ending in cyanosis. Crepitant rales and
expiratory wheezes are likewise noted, often mistaken for pertussis or bronchial asth-
ma.
Cervical Lymph Nodes (Scrofula), adenitis
Tuberculous lymphadenitis is the most common form of extrapulmonary TB and proba-
bly the most common cause of chronic lymhadenitis in children. TB lymphadenitis in
the cervical region is known as scrofula.7,8 Narang et al reported the prevalence of this
disease in children up to 14 years of age is approximately 4.4 per 1000 cases in India. 9
In the Philippines, a study by Santos reported that TB of the lymph nodes accounts for
6.1% of extrapulmonary tuberculosis with mean age of 5.8 years.10 The disease is pre-
sent at all ages but more frequently at ages 10 to 18 years (39.1%). The most common
location was the anterior cervical space in children (49.4%), followed by the axillary
and supraclavicular areas.11,12
TB lymphadenitis may occur during primary tuberculous infection or as a result of reac-
tivation of dormant foci or direct extension from a contiguous focus. The infection may
spread from primary focus to regional lymph nodes and continue to spread via the lym-
phatic system to other nodes or may pass through the nodes to reach blood stream,
from where it can spread to virtually all organ of the body.13
The most common presentation is unilateral single or multiple slowly growing non-
tender lymphadenopathy which can be present for up to 12 months before diagnosis.
51
Early in the course, the nodal enlargement is usually firm, painless, rubbery, discrete or
matted noted fixed to surrounding structures; the overlying skin may be indurated. The
patient may have low-grade fever and sometimes acute respiratory infections precipi-
tate or aggravate superficial tuberculous lymphadenitis.7,12 Other systemic clinical signs
such as weight loss and tiredness may be present. Concomitant primary pulmonary
tuberculosis is also reported in several studies.2,11
Fistula formation is seen in nearly 10% of tuberculous cervical lymphadenitis. Lymph
node abscess may burst infrequently leading to a chronic non-healing sinus and ulcer
formation. Direct extension of the tuberculosis into the skin from the underlying struc-
tures or by contact exposure to tuberculosis can also be seen as scrofuloderma.13
Cervical lymph nodes may be palpable in normal young children. Enlargement can be
caused be infections other than TB, such as carious teeth, infected tonsils, acute upper
respiratory tract infection and Hodgkin‘s disease. Hence, diagnosis of TB as a cause of
lymphadenitis should include a careful history of exposure to infectious tuberculosis,
tuberculin skin test and chest radiography.
The use of fine-needle aspiration, combined with new, rapid molecular diagnostic tests,
can improve the diagnosis of tuberculous lymphadenopathy and provide valuable infor-
mation for appropriate treatment.12,15,16
Central Nervous System
Various forms of CNS tuberculosis include tuberculous meningitis, serous meningitis,
tuberculoma or TB brain abscess or spinal tuberculous leptomeningitis.
Tuberculous meningitis
Tuberculous meningitis (TBM) is the most severe form of extrapulmonary tuberculosis
and causes substantial morbidity and mortality in children and adults. 17,18 A local study
by Santos noted meninges are the most common organ involved in extrapulmonary tu-
berculosis with mean age of 6.2 years.19 However, deaths due to tuberculous meningi-
tis worldwide each year are unknown due to difficulties in diagnosis and under reporting
in children, especially when infection causes early death.14 This disease has been esti-
mated to develop in one of every 300 untreated primary infections, most frequently in
children younger than 6 years but uncommon in infants younger than 4 months. TBM
usually appears within 2 to 6 months after initial infection, and accompanies miliary tu-
berculosis in approximately 50 percent of cases.14,20
Tubercle bacilli are distributed during the lymphohematogenous spread into all parts of
the body including the central nervous system. However, they do not multiply as well in
nervous system, as in other areas as the lungs. Thus, CNS tuberculosis, although an
early manifestation of infection, usually does not appear simultaneously with miliary
spread. Caseous foci in the brain and meninges may remain dormant for many years
and may discharge tubercle bacilli into the subarachnoid space. A thick gelatinous exu-
date develops around the basal cisterns, Sylvian fissure and the brainstem causing ob-
struction leading to hydrocephalus and papilledema. Vasculitis can result to thrombosis
and subsequent infarction.14,21
52
1. Clinical Manifestations and Diagnosis
TB meningitis is typically subacute with broad and may be nonspecific clinical course
making early diagnosis difficult.17,21,22 Also, the low yield of microbiological confirmation
complicates diagnosis. A high index of suspicion is needed to make an early diagnosis
and the key factors to diagnose TBM is the persistence of symptoms, detection of pul-
monary or other forms of tuberculosis and identification of a recent close contact with
infectious tuberculosis (within the past 12 months). 3,11,12,13
Onset of tuberculous meningitis is usually gradual occurring over a period of approxi-
mately 3 weeks. The clinical course is divided into three stages: The first stage is char-
acterized by personality changes, irritability, anorexia, listlessness, and some fever.
After 1 or 2 weeks, the second stage begins with signs of increased intracranial pres-
sure and cerebral damage appear: drowsiness, stiff neck, cranial nerve palsies, ine-
quality of the pupils, vomiting, tache cérébrale, absence of the abdominal reflexes, and
convulsions that may be tonic or clonic, focal or generalized. The third stage is charac-
terized by coma, irregular pulse and respirations, and rising fever.20
A history of contact with an adult who has infectious tuberculosis aids in establishing
the diagnosis. However, the family history of tuberculosis may be negative and the con-
tagious adult may not be identified.20
The evidence of TB infection or disease outside the CNS can significantly increase the
probability or possibility that a child with cerebral signs and symptoms has TBM. How-
ever, a great number of patients, especially when HIV negative and the very young,
may present with normal chest radiography.18,28,29 Chest radiograph findings may re-
veal typical of primary TB or it may have a miliary pattern21, but in many cases, by the
time the meningitis develops, the original focus in the lungs may no longer be demon-
strable by plain radiographic studies.30
The tuberculin skin test (TST) is of limited value and with variable sensitivities. It is
very useful if positive; if negative, it does not rule out TBM. An HIV infection in patients
with TBM reduces the likelihood of positive TST results.18
The diagnosis of TBM is difficult and may be based only on clinical and preliminary cer-
ebrospinal fluid (CSF) findings. Lumbar puncture should be done and characteristics
CSF findings noted. CSF may be clear but with increased opening pressure. It is usual-
ly opalescent and contain 50 to 500 white blood cells/mm3, with polymorphonuclear leu-
kocytes predominant very early in the disease and lymphocytes predominant later.
CSF glucose level may be at the lower limits of normal early in the second stage and is
very low by the third stage. The protein content may be normal initially but rises to a
very high concentration, at which pellicle forms on standing.6,14,16 CSF sample should
be sent for acid fast smear but a single sample has low sensitivity, 20-40%. CSF cul-
ture also has low sensitivity (~ 40-80%) and can take several weeks for the result.23
Neuroimaging can also contribute in the diagnosis of TB meningitis. The most com-
mon brain computed tomography (CT) or magnetic resonance (MR) features in children
with TBM are hydrocephalus in 80% and basal meningeal enhancement in 75% of
young children.28,32 Hypodensities due to cerebral infarcts, cerebral edema and nodular
53
enhancing lesions may also be seen.23
A variety of newer diagnostic methods such as polymerase chain reaction (PCR), ELI-
SA, latex agglutination have been recently developed and used to search for mycobac-
terial antigens in the spinal fluid. These tests are valuable aids in the rapid diagnosis of
TBM, particularly in the pediatric age group.33,34
Empirical antituberculosis therapy should be started promptly in all patients suspected
with TB meningitis because treatment delay is strongly associated with death. 35 Adjunc-
tive treatment with corticosteroids improves survival but does not prevent severe disa-
bility.36
The most important determinant of outcome is the stage of illness at which the diagno-
sis is made and appropriate treatment is given.37 Despite treatment, childhood tubercu-
lous menigitis has very poor outcome particularly with stage 3 TB meningitis.25,38
2. Other Forms (Tuberculoma, Brain Abscess, Spinal leptomeningitis )
Other forms of central nervous system tuberculosis include tuberculoma, TB brain ab-
scess and spinal TB leptomeningitis.
Tuberculomas are enlarged granulomatous foci within the brain parenchyma and TB
brain abscess lacks the giant cell and granulomatous reaction associated with tubercu-
loma. Both are manifested clinically as space occupying lesion. Tuberculomas occur
most often in children younger than 10 years old and often located infratentorial or at
the base of the brain around the cerebellum. In contrast, tuberculomas in adults are
supratentorial.20,39,40 Clinical manifestations include headaches, seizures, papilledema
and focal deficits may all occur. The presentation of brain abscess is more subacute (1
week to 3 months after infection), but much slower than pyogenic brain abscesses. Tu-
berculomas may present months to years after infection.22
Careful evaluation, knowledge of exposure to tuberculosis, a tuberculin skin test, chest
radiography, and computed tomography help recognized the disease in time to begin
appropriate chemotherapy before neurosurgical intervention is needed.
Tuberculous spinal meningitis may manifest as an acute, subacute, or chronic form
which is often characterized by myelopathy, with progressive ascending paralysis. Pa-
tients may develop acute paraplegia with sensory deficits and urinary retention which
often mimics transverse myelitis or Guillain-Barré syndrome. The subacute form is of-
ten dominated by myeloradiculopathy, with radicular pain and progressive paraplegia or
tetraplegia. A less virulent chronic form might mimic a very slowly progressive spinal
cord compression or a non-specific arachnoiditis. The dorsal cord is affected most com-
monly, followed by the lumbar and the cervical regions.
Bones and Joints
Bones and joints tuberculosis accounts for approximately 10-15% of the extrapulmo-
nary tuberculosis and only 2% of all cases of tuberculosis.40,41,42 A local study by San-
tos noted 6.1% of cases with extrapulmonary tuberculosis have bone involvement with
mean age of 6.2 years.10 It can be seen in 1 to 5 percent of children with untreated pri-
54
mary infection and becomes symptomatic one to three years after initial pulmonary in-
fection. Tubercle bacilli are disseminated to skeletal structures during lymphohematog-
enous spread during the initial infection or may be develop as a direct extension from a
caseous regional lymph node or by extension from a neighboring infected bone. Very
young children are more vulnerable to suffer from this form than older children because
of increased blood flow through the growing bones. The lesion usually starts as an area
of endarteritis in the metaphysis of the long bone, where the blood supply is more
abundant.20,44
Skeletal tuberculosis should be considered immediately in any child who is known to
have been infected with tubercle bacilli and in whom a bone or joint lesion develops
and in any child with a persistent, not otherwise explained bone or joint lesion.
TB of the Spine (Pott’s Disease)
The spine (especially the vertebrae) is the most common skeletal site affected by tu-
berculosis, followed by the hip and knee.43,45 Vertebral tuberculosis or tuberculous
spondylytis constitutes about 50% of all cases.43,46 Often, a history of trauma is present
and contributes either in activation of an underlying lesion or simply to draw attention to
the process.20
In spinal tuberculosis, there is destruction of the intervertebral disk space and adjacent
vertebral bodies, collapse of the spinal elements, and anterior wedging leading to the
characteristic angulation and gibbus (palpable deformity because of involvement of
multiple vertebrae) formation. The upper lumbar, lower thoracic and lumbosacral spine
are most frequently involved sites. Multiple vertebrae are typically affected more fre-
quently the vertebral body. Distortion of spinal column leads to severe kyphosis (Pott‘s
disease).43,20,45,46
Back pain is the most frequent symptom of spinal tuberculosis and progression is slow
and insidious.48 Duration of illness varies with average duration ranging from 4 to 11
months. Usually, patients seek advice only when there is severe pain, marked deformi-
ty, or neurological symptoms.43 Signs and symptoms include ―night cries‖ and restless
sleep, daily low-grade fever, and peculiar position (such as torticollis with cervical le-
sions) or gait. Sensory disturbance and bowel and bladder dysfunction are also com-
mon in children.46 Physical examination findings include marked ―guarding‖ because of
dorsal muscle spasm, local pain and tenderness, gibbus deformity or reflex changes
including clonus.14,45
Serious complications such as paravertebral abscess (Pott‘s abscess), retropharyngeal
abscess, psoas abscess and neurologic lesions are seen in 10-30% of the TB of the
spine. Neurologic involvement such as paraplegia and even paresis range from 25-
39% in foreign literature and 17-44% in the Philippines. These result from inflammation
of the spinal cord secondary to an adjacent cold abscess, granuloma in the extradural
space, or spinal vessel thrombosis. 14,30
TB of the Joints
Tuberculosis of the hip is second to spine as the common site of skeletal tuberculosis.
Other weight bearing joints such as knee, shoulders and elbows are also frequently in-
55
volved.44 Insidious onset of pain, stiffness and limp on walking or refusal to walk were
the main clinical features in children. The outcome of tuberculosis of the hip in children
depends on the extent of the disease. Tuberculosis of the femoral head and neck may
lead to various deformities.49,50
Tuberculous arthritis is rare in children and monoarticular in 90% of cases. Weight
bearing joints and only exceptionally the joints of the upper extremities are primarily
involved.20 TB arthritis result from direct invasion of tubercle bacilli into a joint space or
as a consequence of an aseptic reactive polyarthritis (Poncet's disease).51
Gastrointestinal/Abdominal Tract
Gastrointestinal tuberculosis or abdominal tuberculosis includes infection of the perito-
neum, hollow or solid abdominal organs (intestinal tract, liver, spleen, pancreas, mes-
entery) and abdominal lymph nodes.52,2,53,54 Although uncommon in children, there
have been reports that it is second in frequency after pulmonary disease and a signifi-
cant rise has also been observed to occur in association with HIV infection. 53,55 In India
and Tunisia, the most common forms of abdominal TB are nodal involvement, peritoni-
tis and intestinal disease53,55 while liver (6.1%), ileum (1.5%), perineum (1.5%), and
spleen (1.5%) are the abdominal organs involved in a local study done over a 4-year
period in a tertiary government hospital.10
The clinical presentation is both varied and non-specific (Table 6.1). Common present-
ing symptoms are abdominal enlargement, abdominal pain and weight loss.2 Malnutri-
tion is a common feature. Abdominal pain is described in various ways but is most of-
ten cramping in nature and can be diffuse or localized in either the right upper or lower
quadrant.30 Most children often appear systemically ill and anemic with low grade fever.
Distended abdomen, ascites, abdominal mass typically located in the right lower quad-
rant and hepatomegaly are the most common signs.
The reported prevalence of pulmonary TB in children with abdominal TB ranges from
20%-60%. Most commonly, the onset of abdominal tuberculosis is insidious although
some children may present with acute symptoms.2
The diagnosis is oftentimes delayed and is usually made through a combination of radi-
ologic, endoscopic, microbiologic, histologic, and molecular techniques. The absence of
radiologic or clinical signs of pulmonary disease does not exclude the diagnosis. Ab-
dominal TB should be considered in the differential diagnosis of children presenting
with acute abdomen, particularly in areas with high TB incidence. Prognosis is poor in
the presence of severe malnutrition, disseminated TB disease and presence of co-
morbidities.2
56
Table 6.1 of Clinical Presentations of abdominal tuberculosis in children*2,53,54 (in
percentage)
Signs Tinsa, Shah, Saczek, Talwar, Veeragan- John- Da-
and et.al et.al et.al. et.al dham son vies
Symp- (2010) (2010) (2001) (2000) (1996) (1987) (198
toms N= 13 N= 32 N = 45 N = 125 N = 26 N = 59 2)
Ab- 77 53 51 100 71 44 60
dominal
pain
Ab- 62 - 64 38 35 92 42
dominal
disten-
tion
Growth - 47 56 58 69 78 30
failure,
weight
loss
Fever 31 75 47 74 50 - 34
Loss of - 41 - 54 15 - 24
appetite
Ascites - 20 44 35 44 7
Ab- 15 - 47 12 23 56 -
dominal
mass
Extra- - - 49 n/a 19.2 47 18
ab- (extraintestin
dominal al TB)
lymph
node en-
largemen
t
57
signs to look out for, include vague abdominal pain, intussusceptions, blood in the stool
and sinus formation after an uncomplicated appendectomy.56 Although rectal bleeding
and hematemesis are rare in children, intestinal TB should be considered in the differ-
ential diagnosis of children with an intestinal bleed, particularly in high TB incidence
countries.2 Enlarged caseous and calcified mesenteric lymph glands (―tabes mesenter-
ica‖) are often accidentally discovered on roentgenogram of the abdomen as ―shadows
of increased density‖ which incites local inflammatory reaction. These mesenteric
nodes become matted and result in adhesions interfering with intestinal motility, thus
producing intestinal obstruction or compression of the portal vein, giving rise to ascites
and dilatation of the superficial abdominal veins.
TB peritonitis
Tuberculous peritonitis is commonly due to rupture of a caseous abdominal lymph node
and less frequently from a focus in the intestine or fallopian tube. Clinically, it is classi-
fied into ―plastic‖ and serous types: the former being less common and characterized by
tender abdominal masses and a doughy abdomen and the latter by ascites and signs of
peritonitis. Symptoms of fever, abdominal pain, weight loss, anorexia and clinical evi-
dence of ascites are common.60
In a clinical study in Mexico involving autopsied cases of children dying from tuberculo-
sis (TB), 95.2% (20/21) of children with evidence of peritoneal tuberculosis at autopsy
had evidence of disease arising from a site other than the peritoneum. The sites were
either secondary to a pulmonary lesion or as an extension to the peritoneum from pri-
mary or secondary intestinal lesions. This would suggest that primary peritoneal tuber-
culosis is not common. The presence of ascites is a constant indicator of peritoneal dis-
ease, and the cases referred to in literature as ―dry‖ or ―sclerotic‖ peritoneal tuberculo-
sis are very seldom seen in children.55
The presence of ascites usually warrants aspiration and analysis of ascetic fluid. Analy-
sis of ascitic fluid often shows exudative features with lymphocytic predominance and
serum ascitic fluid albumin gradient less than 1.1 g/dL57. The protein content is usually
elevated (> 25g/L).2 The ascitic fluid-blood glucose ratio may be helpful in differentiating
tuberculous peritonitis from other causes of ascites. In the study by Wilkins, the ratio
was less than 0.96, and in patients with non-tuberculous ascites the ratio was greater
than 0.96, the difference being statistically significant.56,58
Hepatobiliary TB
A subset of patients with extrapulmonary TB have the infection confined solely and pre-
dominantly in the liver or biliary tract. Majority of patients present with non-specific
symptoms such as fever, malaise, fatigue, night sweats, anorexia and weight loss.
Those who have prominent hepatic complaints may manifest with jaundice, abdominal
pain, hepatosplenomegaly and ascites. This form of infection is referred to as ―Primary
Miliary TB of the Liver‖ by some authors.
Khan, et al described three forms of hepatic tuberculosis. The most common form seen
in 50% to 80% of patients dying of pulmonary tuberculosis is diffuse hepatic involve-
ment with pulmonary or miliary tuberculosis. The second form is diffuse hepatic infiltra-
tion without recognizable pulmonary involvement. The third very rare form presents as
58
focal or local tuberculoma or abscess. Isolated liver tuberculosis (ILT) is the rarest form
of local hepatic TB. The reported overall incidence for ILT is 0.3%. Hepatic tuberculo-
sis lesions that appear as masses larger than 2mm in diameter are referred to as mac-
ronodular and pseudotumoral tuberculosis. These lesions are indistinguishable from
other focal lesions in the liver such as hepatocellular carcinoma and Hodgkin‘s disease
by imaging techniques. Most of the time, histopathologic diagnosis is necessary.59
TB of the pancreas
This is a rare condition and occurs secondarily to generalized TB and occasionally in
far advanced cases. There is scarcity of literature on pancreatic tuberculosis with most
published material being case reports.
Cutaneous Tuberculosis
Cutaneous tuberculosis is a chronic granulomatous disease caused by Mycobacterium
tuberculosis and rarely by Mycobacterium bovis. Most published studies in childhood
skin tuberculosis are from developing countries where Southeast Asia contributing
much of the epidemiological data. In India, the reported prevalence of childhood skin
tuberculosis varies from 18% to 54%. Pakistan has recorded the highest prevalence of
childhood skin tuberculosis, with 82% of all skin tuberculosis being reported in chil-
dren.65
Cutaneous tuberculosis manifests in several clinical forms:
1. lesions by inoculation from an exogenous source or after inoculation with BCG vac-
cine;
2. lesions resulting from hematogenous dissemination;
3. lesions arising from an exogenous source; and
4. erythema nodosum52
A simple classification of cutaneous tuberculosis includes true primary and secondary
skin lesions and tuberculids (see Table 6.2). Scrofuloderma is the most common form
of childhood cutaneous TB. Scrofuloderma indicates TB of the skin overlying a caseous
lymph node that has ruptured to the outside, leaving an ulcer or a sinus. Frequently, the
lesions are in the cervical area but may also involve the inguinal, submandibular and
axillary groups.56,65
Table 6.2 Classification of Skin Tuberculosis in Children
PRIMARY SECONDARY TUBERCULIDS
59
Ocular Tuberculosis
Ocular TB is reported occasionally in tertiary hospitals that admit the more serious or
complicated forms of TB. In literature, it frequently involves the conjunctiva and the cor-
nea in the form of conjunctivitis and phylctenular keratoconjunctivitis.56 Ocular TB may
occur at any age – during primary infection or reactivation of latent disease and may be
seen in 14-60% of patients with systemic TB. Patients may be immunocompetent or
may have varying degrees of immunosuppression. The rise in HIV/AIDS cases has also
led to an increase in ocular TB.2 In 2007, Gupta,et.al. updated the clinical spectrum,
laboratory investigation, and diagnostic criteria that would serve as guide in the diagno-
sis of presumed or confirmed intraocular TB so that anti-tuberculous therapy (ATT) can
be initiated on a rational basis.66
The conjunctiva can serve as the portal of entry for the bacilli, especially after trauma. A
local reaction is induced in the form of unilateral lacrimation and reddening, with subse-
quent formation of yellowish-gray nodules on the palpebral conjunctivae. Enlargement
of the preauricular, submandibular and cervical lymph nodes are commonly noted. In
patients with pulmonary and systemic TB, choroiditis is the most common ocular mani-
festation.
Tubercles of the choroid, frequently multiple, often have been found in 70% of patients
with miliary TB and in children with uncomplicated TB. These heal slowly with deposi-
tion of retinal segment.56
In a local study done in a tertiary government hospital involving 108 patients with tuber-
culosis, 6.8% had signs of ocular inflammation. There was unilateral presentation of all
patients labelled as presumed ocular TB (POTB), concurring with the reports of some
authors that ocular TB is usually unilateral. Posterior segment lesions were the predom-
inant finding in this series which is logical since choroidal tubercles and retinal vasculitis
hematogenous seeding of bacilli. Another common ocular lesion found in the study was
non-granulomatous anterior uveitis with posterior synechiae.66
A tuberculin skin test (TST) and biopsy with culture can be performed to confirm the
diagnosis. The specificity of the TST for Mycobacterium tuberculosis increases with
larger skin reaction and with a history of exposure to an active case of TB.
60
Genitourinary Tract Tuberculosis
Renal TB is an uncommon complication of primary TB occurring very late, up to 15 to
20 years after primary infection. In a local study done on extrapulmonary TB, genitouri-
nary TB constituted 3%. Genitourinary involvement were mostly seen in children older
than 7 years of age.10 However, TB bacilli can be recovered from the urine of patients
with miliary TB and in some cases with pulmonary TB. Hematogenous spread can give
rise to tubercles in the glomeruli, with resultant caseating sloughing lesions which dis-
charge TB bacilli into the tubules. Infection can be unilateral or bilateral and can spread
caudad to involve the bladder. The disease can be insidious in onset and has strikingly
few specific symptoms. In 75% of patients, however, they would present with symptoms
related to urinary tract inflammation such as dysuria, hematuria, sterile pyuria, and flank
pain. Renal tuberculosis should be suspected in the presence of destructive pulmonary
TB with persistent, painless, sterile pyuria associated albuminuria and hematuria. Re-
peated cultures of the urine are advisable and for those with persistent pyuria, a com-
plete urologic investigation is mandatory. Children whose urine reveal presence of tu-
bercle bacilli are considered to be highly infectious and should be isolated until their
urine is sterile.56
Genital tuberculosis is uncommon in both sexes before puberty. It may arise either as a
metastatic lesion during lymphohematogenous spread or by direct extension from an
adjacent lesion of the bone, gut or the urinary tract. Frequently, other forms of TB ac-
counts with initial infection, such as pleural effusion, are also present. In females, the
fallopian tubes are the most frequently involved (90 to 100% of cases), followed by the
endometrium (50%), ovaries (20 to 30%), and cervix (2 to 4%). Signs and symptoms
include lower abdominal pain, amenorrhea, a lower abdominal mass and free peritone-
al fluid. These are to be considered in mothers when managing newborns suspected of
infection (e.g., congenital TB) in the perinatal period. TB of the external genitalia has
been seen as a manifestation of child abuse. Males may develop primary TB of the pe-
nis after circumcision manifesting as massive inguinal lymphadenopathy. Epididymitis
or epididymo-orchitis can likewise occur characterized by gradual onset of nodular,
painless swelling of the scrotum with a dragging pain in the groin.56
Proposed routes of spread mentioned in literature are: hematogenous, usually from dis-
tant sites; direct extension from adjacent structures; external, from perforation of the
tympanic membrane; passage through an infected genital canal; and via the placenta
in congenital infection.60,61
61
Characteristic features include painless otorrhea, preauricular lymphadenopathy, oto-
scopic findings of tympanic membrane perforation, granulomatous tissue and bone ne-
crosis. However, in the review by Sens et al., the more common clinical findings were
of significant otalgia, serous otitis media which could become purulent from secondary
bacterial infection and profound hearing loss. Complications arise from delay in diagno-
sis. Facial paralysis is one reported complication that is seen more frequently in chil-
dren than adults.61
Perinatal/Postnatal Tuberculosis
Congenital TB left unrecognized and untreated may result in significant morbidity and
mortality. Often mentioned as a rare occurrence, published figures may be an underes-
timate particularly in endemic areas where diagnosis may have been missed or under
reported.
For infection to occur, possible routes include: hematogeneous spread from the umbili-
cal vein, ingestion in utero or intrapartum of infected amniotic fluid, or postpartum inha-
lation from a source case which is usually the mother. Ingestion via breastmilk is not a
known mode of transmission.62
63
REFERENCES
1. National Tuberculosis Control Program. Manual of Procedures. 5th Ediction. 2013
2. Schaaf, HS. Tuberculosis, 2009
3. Starke, JR. Tuberculosis in children. Semin Respir Crit Care Med 2004; 25 (3)
4. Pozniak, A. Clinical Manifestations and Complications of pulmonary tuberculosis.
In: UpTodate Post TWG( Ed.). UpToDate, Waltham MA ( Accessed March, 2016)
5. Bernardo, J. Epidemiology and pathology of military and extrapulmonary tuberculo-
sis. In : UpToDate Post TW(Ed.). UpToDate, Waltham, MA ( Accessed May 15,
2016)
6. Lessnau, KD. Miliary tuberculosis. In: Medscape. Aug,03, 2015
7. Fontanilla JM, et.al. Current Diagnosis and management of peripheral tuberculous
lymphadenitis. Clin Infect Dis. 2011: 53(6): 555.
8. Jawahar, MS. Scrofula revisited: An update on the diagnosis and management of
tuberculosis of superficial lymph nodes. Indian J Pediatr. 2000. Feb: 67 ( 2
suppl.).S28-33.
9. Narang,P et.al. Prevalence of tuberculous lymphadenitis in children in Wardha dis-
trict, Maharashka State, India. Inst. J Tuber Lung Dis., 2005; 9 (2): 188.
10. Santos, Regie S. Clinical Profile of Extrapulmonary Tuberculosis Cases Admitted
and Diagnosed in a Tertiary Government Hospital from Jan 2006 to June 2010.
PIDSP Journal 2013 Vol 13 # 2.
11. Ileana, P. et.al. Diagnosis of Tuberculous lymphadenitis in Children. Pediatrics
2008; 120 s. 130.
12. Minime-Lingoupou, Fanny,et.al. Fine-needle aspiration for diagnosis of tuberculous
lymphadenitis in children in Bangui, Central African Republic. BMC Pediatrics 2012,
12:191
13. Prasanta Raghab Mohaptra, Ashok Kumar Janmeja. Review Article: Tuberculous
Lymphadenitis. J Assoc Physicians India. 2009 Aug: 57: 585 – 90
14. Starke, Jeffrey R. Feigin and Cherry‘s Textbook of Pediatric Infectious Diseases.
6th Ed. 2009.
15. Sangeeta, Sharma et.al. Clinical Profile and treatment outcome of tuberculous lym-
phadenitis in children using DOTS strategy. Indian J Tuberc 2010; 57: 4 – 11
16. Marais BJ, et.al. Tuberculous lymphadenitis as a cause of persistent cervical lym-
phadenopathy in children from a tuberculosis-endemic area. Pediatr Infect Dis J.
2006 Feb; 25(2): 142-6.
64
17. Galimi R. Extrapulmonary Tuberculosis: Tuberculous meningitis new develop-
ments. Europe Rev Med Pharmacol Sci. 2011 Apr; 15(4): 365-86.
18. Suzaa, Marais,et.al. Tuberculous meningitis: a uniform case definition for use in
clinical research. Lancet Infect Dis. 2010; 10: 803 – 12
19. Graham, Stephen M , Peter R. Donald. Death and disability: the outcomes of tuber-
culous meningitis. www.thelancet.com/infection. Vol 14. October 2014.
20. Van de Bos, F. et.al. Tuberculous meningitis and military tuberculosis in young chil-
dren. Tropical Medicine and International Health. Volume 9 no. 2 pp.309 – 313.
Feb, 2004.
21. Sandip, Chatterjee. Brain tuberculomas, tubercular meningitis, and post-tubercular
hydrocephalus in children. J Pediatr Neurosci. 2011 Oct; 6 ( Suppl 1): S96 – S100.
Doi:10. 4103/1817 – 1745. 85725.
22. Marx, Grace E., Edward D. Chan. Tuberculous Meningitis: Diagnosis and Treat-
ment Overview. Tuberculosis Research and Treatment. Vol 2011.
Doi:10.1155/2011/798764.
23. Suzuki, Y. Tuberculosis infection in the nervous system. Nihon Rinsho. 2011 Aug;
69(8): 1422-6.
24. Chiang, Silvia S, et.al. Treatment outcomes of childhood tuberculous meningitis: a
systemic review and meta-analysis. Lancet Infect Dis. 2014; 14: 947-57.
25. Torok, ME. Tuberculous meningitis: advances in diagnosis and treatment. Br.
Med Bull. 2015 Feb 18. Pii:idv003. (Epub ahead of print)
26. Yaramis,A1,et.al. Central nervous system tuberculosis in children: a review of 214
cases. Pediatrics 1998 Nov.102(5): E49.
27. Principi,N.,susanna Esposito. Diagnosis and therapy of tuberculous meningitis in
children. Tuberculosis 92 (2012) 377e383.
28. Solomons RS, et.al. Chest radiograph findings in children with tuberculous menin-
gitis. Int J tuberc Dis. 2015 Feb; 19 (2): 2004-4. Doi: 10-5588/ijtld.14.0634.
29. PPS Inc. Tuberculosis in Infancy and Childhood. 3rd ed. 2010
30. Donald, Peter R., Johan F. Schoeman. Perspective: Tuberculous Meningitis. N
Eng J Med 351; 17. www.NEJM. ORG. Oct. 2004
31. Etlik, Omer, et.al. Radiologic and clinical findings in tuberculous meningitis. Eur J
Gen Med 2004; 1 (2): 19-24.
32. Sagarika, Haldar, et.al. Efficient diagnosis of tuberculous meningitis by detection of
Mycobacterium tuberculosis DNA in cerebrospinal fluid filtrates using PCR. Journal
of Medical Microbiology (2009), 58, 616-624.
65
33. Rafi, A. Naghily B. Efficiency of polymerase chain reaction for the diagnosis of tu-
berculous meningitis. Southeast Asigan J Trop Med Public Health 2003 Jun; 34(2):
357-60.
34. Thwaites G,et.al. British Infection Society guidelines for the Diagnosis and treat-
ment of tuberculosis of the central nervous system in adults and children. J Infect
2009 Sept; 59(3): 167 – 87. Doi: 10. 1016/j.inf.2009.06.011. Epub 2009 Jul 4.
35. Guy E. Thwaites,et.al. Dexamethasone for the Treatment of Tuberculous meningi-
tis in adolescents and adults. N Engl J Med 2004; 351: 1741 – 51.
36. Lee,LV. Neurotuberculosis among Filipino children: an 11 years experience at the
Philippine Children‘s Medical Center. Brain Dev. 2000 Dec; 22(8):
37. Buonsenso D, et.al. Management of central nervous system tuberculosis in chil-
dren: light and shade. Eur Rev Med Pharmacol Sci. 2010 Oct; 14 (10): 845-5.
38. Bathla,G.et.al. Manifestations of cerebral tuberculosis. Singapore Med J 2011; 52
(2): 124.
39. Ning, Guo,et.al. Infratentotiral Tuberculoma Mimics tumor in immunocompetent
adults: an analysis of 11 patients. Open Journal of Modern Neurosurgery. 2013, 3,
36-40 http://dx.doi.org/10.4236/0jmn.2013.33008 P
40. Gautam, MP,et.al. Pott‘s spine and paraplegia. JNMA J Nepal Med Assoc 2005 Jul
-Sept; 44 (159): 106-15
41. Hosalkar, HS, et.al. Skeletal tuberculosis in children in the Western world: 18 new
cases with a review of literature. J Child Orthop (2009) 3: 319-324. DOI 10. 1007/s
11832-009-0184 – 7
42. Kumar R. Spinal tuberculosis: with reference to the children of Northern India.
Child‘s Nerv Syst 2005 Jan; 21 (1): 19 – 26. Epub 2004 spt 30
43. Haider Abdul-Lateef Moussa. Bones and Joints Tuberculosis. Bahrain Medical
Bulletin., Vol 29, No.1, March 2007.
44. Ravindra Kumar Garg, Dilip singh Somvanshi. Spinal tuberculosis: a review. The
Journal of Spinal Cord Medicine 2011 vol. 34.
45. Benzagmout, Mohammed, et.al. Pott‘s Disease in children. Surg Neurol Int. 2011;
2: 1 doc: 10.4103/2152-7806.75459.
46. Myung-Sang, Moon,et.al. Spinal tuberculosis in children: retrospective analysis of
124 patients. Indian J Orthop. 2012 Mar-Apr
47. Honywood, Eisen S. Shingadia D. Novelli. Spinadl tuberculosis in children. Arch
Dis Child 2012; 97: 724 – 729.
48. Shyam Kumar Saraf and Surendra Mohan Tuli. Tuberculosis of hip: a current con-
66
cept review. Indian J Orthop. 2015 Jan-Feb; 49(1). Doi: 10. 4103/0019-
5413.143903
49. MAF Mohideen, MN Rasool. Tuberculosis of the hip joint region in children. SA
Orthopaedic Journal Autumn 2013/ Vol 12. No 1
50. Rajakumar, Derek and Alan M Rosenberg. Case Report: Mycobacterium tubercu-
losis monoarthritis in a child. Pediatric Rheumatology 2008, 6:15 doi: 10.
1186/1546-0096-6-15
51. Sheer,TA and Walter Coyle. Gastrointestinal Tuberculosis. Current Gastroenterol-
ogy Reports 2003, 5: 273 – 278
52. Shah, I. Clinical profile of abdominal tuberculosis in children. Indian J Med Sci
2010 May; 64(5) 204-9
53. Tinsa, F.et.al. Abdominal tuberculosis in children. J Pediatr Gastroenterol Nutr.
2010 Jun; 50(6). 654-8.
54. Ridaura-Sanz,C.et.al. Intestinal/Peritoneal Tuberculosis in Children: An Analysis of
Autopsy Cases. Tuberculosis Research and Treatment. Vol 2012 Article ID
230814.
55. Cherry, J.et.al. Feigin & Cherry‘s Textbook of Pediatric Infectious Diseases, 7th ed
56. Dinler,G. et.al. Tuberculous peritonitis in children: Reports of nine patients and re-
view of the literature. World J Gastroenterol vol. 14 (47); 2008 Dec 21.
57. Wilkins EG. Tuberculous peritonitis: diagnostic value of the ascetic/blood glucose
ratio. Tubercle. 1984; 65: 47-52.
58. Khan, S. et.al. Hepatiec tuberculosis mimicking metastasis in a case of carcinoma
sigmoid colon. J Lab Physicians 2015 Han-June; 7(1Z) 64-66.
59. Adhikari P. Tuberculous Otitis Media: A Review of Literature. The Internet Journal
of Otorhinolaryngology. Vol 9. No.1
60. Sere, Patricia Maria et.al. Tuberculosis of the Ear, a professional disease. Rev
Blas Otorhinolaryngology. 2008: 621-7.
61. Mittal,et.al. Management of Newborn Infant born to mother suffering from TB: Cur-
rent gaps in knowledge. Indian Journal Med Res. 140. July 2014, pp. 32 – 35.
62. Diar H. and Velaphi S. Congenital tuberculosis as a proxy to maternal TB. A case
report. Journal of Perinatology (2009) 29, 709 – 711.
63. Schaaf,et.al. Tuberculosis at extremes of age. Respirology (2010), 15. 747 – 763.
64. Sethuraman G, Rameck V. Cutaneous tuberculosis in children. Pediatr Dermatol
2013. Jan-Feb; 30 (1) 7-16 (Pubmed)
67
65. Lara, Leron Paelo and Vicente Ocampo Jr. Prevalence of presumed ocular tuber-
culous among pulmonary tuberculous patients in a tertiary hospital in the Philip-
pines. J ophthalmic infect 2013; 3: 1
68
7
Diagnosis
Cleotilde H How, MD
OUTLINE
1 Overview
2 Approach to Diagnosis
5 Classification
69
OVERVIEW
APPROACH TO DIAGNOSIS
Marais BJ, et al6 suggest a rational framework integrating old and new concepts to meet
these diagnostic challenges through the use of accurate case definitions (e.g., differenti-
ating primary infection from active disease); risk stratification (age less than 2 years
having the greatest risk of progression to disease); and accurate disease classification.
An earlier review by Daniel7 approaches the diagnosis of mycobacterial disease based
on recognition of either the infecting agent or the host‘s response to the agent.
For those tests based on recognition of the agent, there are direct methods that do not
employ prior amplification, such as direct sputum (AFB, acid-fast bacilli) smear, rapid
culture (BACTEC) and tuberculostearic acid detection. Those that employ amplification
include sputum smear after short-term culture, ultraviolet microscopy with fluorescence
stains, short-term culture with antigen detection by immunoassay, and polymerase
chain reaction such as the nucleic acid amplification tests (NAATs) exemplified by the
Xpert MTB/RIF.
Marais and Pai8 outline the traditional and new approaches to the diagnosis of mycobac-
terial disease. They cite important WHO landmark developments that focus on essential
tools to aid in reaching an accurate diagnosis, particularly in resource-limited TB-
endemic settings. The authors included symptom-based screening9, along with tubercu-
lin skin test (TST), chest radiography and tuberculosis (TB) culture. Of these, it is the
identification of the tubercle bacilli that offers positive confirmation of TB disease.
When the presenting symptom is nonspecific, it may not be of value in recognizing ac-
tive disease. With more clearly-defined clinical features in the refined symptom-based
screening10 there is greater basis for making a diagnosis and early treatment. A symp-
tom screen is also necessary for assessment of an asymptomatic child considered vul-
nerable in a high-risk environment. A child below 3 to 5 years of age who is asympto-
matic but exposed to a documented case of pulmonary tuberculosis calls for prompt
preventive chemotherapy. See Chapter 15 (Prevention and Control)
Recognition of host response is best illustrated in the TST, chest radiology and serolo-
gy. The introduction of enzyme-linked immunosorbent assay (ELISA) and the develop-
70
ment of more specific antigens (recognized by monoclonal antibody) have tremendous-
ly improved serodiagnosis. Those new modalities for detection of the organism and its
products include pathogen-based and immune-based assays.8
Cuevas et al11 reviewed the TB diagnostics for children in high-burden countries, noting
the lack of sufficient number of research studies and a dearth of evidence of test perfor-
mance in children. Novel markers for T-cell activation with proof of concept12 have
emerged to develop more advanced assays for the diagnosis of active TB in children,
prospectively ushering in tests that can differentiate active disease from infection men-
tioned in the 2016 WHO Pipeline Report.13
These diagnostic tests are further discussed in Chapters 8 (Tuberculin Skin
Test), 9 (Radiologic Findings) and 10 (Diagnostic Tests/Laboratory Findings).
71
Vulnerability of the Young
Young children are most vulnerable among household members who are exposed to
an adult index case or source case, who remains untreated or even with treatment, fails
to complete therapy. These children are at greatest risk to develop infection, disease
and its complications, with progression, dissemination or even death. More operational
research is needed to address the need for more data in low-income countries.16 See
Chapter 2 (Epidemiology), Chapter 3 (Microbiology) and Chapter 4 (Immune Re-
sponses)
Approach to Management
In a child to be assessed for TB, the resources for diagnosis are often limited, not just
in confirmatory tests but even in the basic clinical skills to recognize TB. The clinician in
such a situation has to optimize skills to process the clinical picture presented and aim
for a clinical diagnosis, or ideally, a bacteriologic diagnosis.
Use of a symptom-based screening tool9 and a ―refined symptom-based approach‖10
referred to earlier are designed to improve the diagnosis of pulmonary TB in children.
The recommended approach to diagnosis is summarized from the guidelines adopted
for this edition.1,2,3,4
Careful history, including history of TB contact and symptoms consistent with TB;
Clinical examination, including growth assessment;
Tuberculin skin test
Bacteriological confirmation whenever possible
HIV testing
Investigations relevant for suspected pulmonary or extrapulmonary TB
72
Table 7.1. Spectrum of TB exposure, infection and (pulmonary) disease. (modifiedfrom
DOH/NTP Table17)
TB Exposure TB Infection TB Disease
Exposure positive positive positive
Signs & symptoms negative negative positive
Tuberculin skin test negative positivea positivea
Chest X-ray negative negative (may be positive)b
Direct sputum smear negative negative (may be positive)c
microscopy (DSSM)
Other diagnostics negative (may be positive)d (may be positive)d
a
may be false negative in many children
b
may be positive, showing hilar adenopathy and the Ghon complex
c
may be negative due to paucibacillary nature in children and difficulty of specimen collection
d
may be positive in immunology-based tests like IGRA
CLASSIFICATION OF TB DISEASE1-5
Classification of TB disease is based on bacteriological status, anatomical site, history
of previous treatment, HIV status and drug susceptibility testing of clinical isolates.
Treatment outcome definitions make a distinction according to whether a patient has
been treated for drug-susceptible or drug-resistant TB; discussed in Chapter 11
(Management of TB).
Classification Based on Bacteriological Status
Bacteriologically-confirmed TB case is one from whom a biological specimen is posi-
tive by smear microscopy, culture or WHO-approved rapid diagnostic tests (such as
Xpert MTB/RIF).
Clinically-diagnosed TB case is one who does not fulfill the criteria for bacteriological
confirmation but has been diagnosed with active TB by a clinician or a medical practi-
tioner who has decided to give the patient a full course of TB treatment. This definition
includes cases diagnosed on the basis of X-ray abnormalities (i.e., not solely) or sug-
gestive histology, and extrapulmonary cases without laboratory confirmation.
Classification Based on Anatomical Site
Pulmonary TB (PTB) refers to bacteriologically-diagnosed or clinically-diagnosed
case of TB involving the lung parenchyma or the tracheobronchial tree. A patient with
both pulmonary and extrapulmonary TB should be classified as a case of pulmonary
TB.
PTB is characterized by the formation of lesions mainly in the lungs. To establish a di-
agnosis of PTB, a DSSM is of great value when a child can expectorate and the DSSM
is performed. Therefore, a child over ten years or those below who are able to expec-
torate sputum, can be categorized into PTB, sputum smear-positive, or, PTB, spu-
73
tum smear-negative. Culture studies should be done when indicated, particularly
when drug resistance is confirmed or suspected.
The course varies according to whether or not there is progression of the disease. In
the Chapter on TB in the local competency-based textbook of pediatrics4, there are two
forms of TB classified as:
Other Classifications
The International Classification of Diseases (ICD)-1018 provides a more detailed system
of classification that is site-specific, as well as, descriptive of the level of diagnostic
work-up done.
Presumptive TB refers to any person whether adult or child with signs and/or symp-
toms suggestive of TB whether pulmonary or extra-pulmonary, or those with CXR find-
ings suggestive of active TB.5 Clinical signs and symptoms are sought to arrive at a
presumptive diagnosis in children below 15 years and in those 15 years and above.
Epidemiologic Considerations
Every effort must be made to establish a history of exposure to TB. In a great majority
75
of childhood TB, the source of infection is the parent, grandparent or caregiver. Howev-
er, because the Filipino family is characterized as an extended one, inquiry must be
made on all possible case contacts to include all relatives and close friends who were
in contact with the patient.
For young children, the more probable source of infection is in the home—among close
household contacts. Many older children are infected by outside sources such as
teachers, group leaders or young adults. Widespread infection may develop from a sin-
gle active case at school or a day-care center as basis for the clinical question on ex-
posure or history of contact.
The PPS Evidence-based Clinical Practice Guidelines20 made recommendations on
the diagnosis, treatment and prevention of TB in children. New information in the last
decade have added more evidence to strengthen established procedures or modify the
approach to management.
Clinical Manifestation
Many children with primary TB are asymptomatic in the early stage of the disease or
may present with minor constitutional manifestations such as low grade fever, lassi-
tude, easy fatigability, anorexia, weight loss, malaise and night sweats, which are all
protean and common to many illnesses.
The DOH/NTP Manual of Procedures3 outlines the criteria for initiating investigation
among children below 15 years and those 15 years and above, to launch more tests
that will help confirm the diagnosis of TB. These are reproduced below, formatted with
highlights.
76
ii.. Unexplained cough of any duration in:
a close contact of a known active TB case;
high-risk clinical groups (e.g., HIV/AIDS, diabetes, end-stage renal disease, can-
cer connective tissue diseases, autoimmune diseases, silicosis, patients who un-
derwent gastrectomy or solid organ transplantation and patients on prolonged
systemic steroids); and,
high risk populations (e.g., elderly, urban poor, inmates and other congregate set-
tings).
b. For patients below 15 years old, a presumptive PTB has any of the following:
i. At least three (3) of the following clinical criteria:
coughing/wheezing of 2 weeks or more, especially if unexplained;
unexplained fever of 2 weeks or more after common causes such as malaria or
pneumonia have been excluded;
loss of weight/failure to gain weight/weight faltering/loss of appetite;
failure to respond to 2 weeks of appropriate antibiotic therapy for lower respiratory
tract infection;
failure to regain previous state of health 2 weeks after a viral infection or exanthe-
ma (e.g., measles); and,
fatigue, reduced playfulness, or lethargy (e.g., child has lost his/her normal ener-
gy).
ii. Any one (1) of the above signs and symptoms (e.g., clinical criteria) in a child
who is a close contact of a known active TB case.
c. CXR findings suggestive of PTB, with or without symptoms, regardless of
age.
d. Presumptive extra-pulmonary TB may have any of the following:
gibbus, especially of recent onset (resulting from vertebral TB);
non-painful enlarged cervical lymphadenopathy with or without fistula formation;
neck stiffness (or nuchal rigidity) and/or drowsiness suggestive of meningitis that
is not responding to antibiotic treatment, with a sub-acute onset or raised intracra-
nial pressure;
pleural effusion;
pericardial effusion;
distended abdomen (i.e., big liver and spleen) with ascites;
non-painful enlarged joint; and
signs of tuberculin hypersensitivity (e.g., phlyctenular conjunctivitis, erythema
nodosum).
77
3. Ask and verify the following:
a. History of previous anti-TB treatment and its details
b. Exposure to active TB cases or presumptive TB (including MDR-TB, if applicable)
within the workplace, family or household
c. Presence of clinical or other high-risk factors (e.g., HIV/AIDS, diabetes, end-stage
renal disease, cancer, connective tissue diseases, autoimmune diseases, silicosis,
patients who underwent gastrectomy or solid organ transplantation and patients on
prolonged systemic steroids)
4. Once identified as a presumptive TB, record the patient in Form 1. Presumptive TB
Masterlist.
Physical findings are also nonspecific and localizing symptoms may be absent even in
the presence of extensive disease. There are, however, clinical findings compatible
with TB such as relatively painless lymphadenitis, meningitis with insidious onset, gib-
bus, skin granuloma, erythema nodosum, and phlyctenular conjunctivitis. The presence
of such findings suggests TB disease and warrant work-up to confirm the diagnosis.
The next step in the investigation is to decide to treat as active TB if the child has any
three (3) of the following criteria, (1997 PPS Consensus).19
i. Positive exposure to an adult/adolescent with active TB disease;
ii. Positive tuberculin test (a positive TST confirms TB infection after exposure);
iii. Positive signs and symptoms suggestive of TB;
iv. Abnormal chest radiograph suggestive of TB;
v. Laboratory findings suggestive or indicative of TB.
In a low-resource country, early detection of active TB can prevent delays in interven-
tion and control of TB infection and disease. Application of a systematic screening par-
ticularly in high-risk groups avoids more delays in treatment and control.21,22 A simple
but ―refined symptom-based approach‖ to diagnose pulmonary TB in children found a
high degree of accuracy by risk-stratification with the combined presence of three well-
defined symptoms of cough, weight loss and fatigue.10 Emphasis on careful history and
clinical exam including growth assessment are paramount.1
See Chapter 6 (Clinical Forms and Features)
Immunologic Evidence of Infection
According to Dr Mita Pardo de Tavera,23 ―the tuberculin test is the most important diag-
nostic tool in tuberculosis, be it recent or remote, active or inactive. In the time-table of
tuberculosis it is this biochemical alteration in the tissues of the host that precedes, by a
comfortable margin of time ranging from several months to even years, the clinical, the
bacteriologic and radiologic evidence of the disease.‖
The only means of making a diagnosis of TB infection without disease (i.e., in latent
78
tuberculosis infection or LTBI) is through a TST and IGRA test. The TST is based on
the fact that infection with M. tuberculosis produces sensitivity to certain components of
this organism (antigens or specifically, tuberculins). The reaction to intracutaneously
injected tuberculin is that of a delayed cellular hypersensitivity.
A significant reaction means that hypersensitivity to mycobacterial protein has devel-
oped, but does not necessarily signify the presence of disease. Clinically, a delayed
hypersensitivity reaction to tuberculin is nearly always a manifestation of previous infec-
tion with either M. tuberculosis, a variety of nontuberculous mycobacteria, or a previous
BCG vaccination; it can antedate isolation of M. tuberculosis from sputum or other
specimens.
Characteristic features of a delayed hypersensitivity reaction include: a delayed
course, reaching its peak 48 up to 72 hours after the administration of tuberculin; an
indurated character largely because of its cellular infiltration; and occasionally, occur-
rence of vesiculation and necrosis. Interpretation of TST reaction is dictated by the pur-
pose for which the test was given and on the consequences of false classification. A
variety of factors—host, antigen used, method of administration, reading and record-
ing—can cause decreased ability to respond to tuberculin or can yield a false negative
test.
Mantoux Test1,2
The current standard for TST is the Mantoux test which is the intradermal administra-
tion of 0.1 ml of solution containing 0.1 ug of 5 tuberculin units (TU) of purified protein
derivative (PPD)-S or 2 TU of PPD-RT 23 with Tween 80.
The Tine (multipuncture) Test, the Two-Step Test and the BCG Test (accelerated BCG
reaction) were discussed in the previous editions of TBIC. These tests are no longer
part of current practice.
Immunologic-based Testing2
Immunologic-based testing with interferon-gamma release assay (IGRA) such as
QuantiFERON-TB Gold, Gold In-Tube and T-Spot are fairly specific to M. tuberculosis
complex thus it is not affected by previous BCG vaccination. Like TST, however, it can-
not distinguish between latent infection and disease.
The sensitivity, specificity, positive and negative predictive values of the tuberculin skin
test, its rationale, procedure and interpretation - and its comparison with immunologic-
based testing like IGRAs - are discussed more thoroughly in Chapter 8 (Tuberculin Skin
Test).
Radiologic Findings
There are no pathognomonic radiographic findings in childhood TB. Neither the pres-
ence nor absence of the primary disease can be determined conclusively from the
chest film alone. Perhaps the only radiographic finding that may be highly suggestive of
TB in infants and children is the uniform stippling of both lungs found in miliary TB.
79
To appreciate the primary complex on chest radiograph, a lateral projection is neces-
sary. With frontal projections alone, the different components of the primary complex
may be obscured by the heart and other structures. Partially calcified mediastinal nodes
sometimes lie in the midsagittal thoracic plane and may be visible only on lateral films.
A variety of diseases and conditions may be mistaken for TB. Sarcoidosis and mycotic
infections produce shadows in the lung that may resemble TB. Tuberculous lobar and
lobular consolidations may be indistinguishable from that due to pneumoccal or strepto-
coccal disease.
See Chapter 9 (Radiologic Findings).
Laboratory Examinations
Bacteriologic studies are discussed in Chapter 3 (Microbiology of Mycobacterium tuber-
culosis). Laboratory findings suggestive of TB by other diagnostic tests and histological,
cytological, biochemical, and immunological and/or molecular tests are discussed sepa-
rately in Chapter 10 (Diagnostic Tests).
80
REFERENCES
1. Guidance for national tuberculosis programmes on the management of tuberculosis
in children. Geneva, World Health Organization, 2014. (WHO/HTM/TB/2014.03)
2. TB Care I. International Standards for Tuberculosis Care, Edition 3, TB CARE I,
The Hague, 2014.
3. Manual of Procedures of the National Tuberculosis Control Program, 2014.
4. Villar EP, Santos JA, Madrid MAC. Chapter 28 Tuberculosis pp679—737.
In:.Fundamentals of Pediatrics: competency-based (Eds Navarro XR, Bauzon AF,
Aguilar JS, Malanyaon OQ). C & E Publishing, Quezon City, 2014.
5. Definitions and reporting framework for tuberculosis – 2013 revisions. World Health
Organization, 2013 (WHO/HTM/TB/2013.2).
6. Marais BJ, Gie RP, Schaaf HS et al. Childhood pulmonary tuberculosis. Old wis-
dom and new challenges. Am J Respir Crit Care Med. 2006;173:1078-1090.
7. Daniel TM. The rapid diagnosis of tuberculosis: A selective review. J Lab Clin
Med.1990;1:277-282.
8. Marais BJ, Pai M. Recent advances in the diagnosis of childhood tuberculosis. Arch
Dis Child. 2007;92:446-452.
9. Marais BJ, Hesseling AC, Gie RP et al.. The burden of childhood tuberculosis and
the accuracy of routine surveillance data in a high-burden setting. Int J Tuberc
Lung Dis, 2006; 10: 259-63.
10. Marais BJ, Gie RP, Hesseling CH et al. A Refined Symptom-based Approach to
Diagnose Tuberculosis in Children, Pediatrics, 2006: 1350-59.
11. Cuevas LE, Petrucci R, Swaminathan S. Tuberculosis diagnostics for children in
high-burden countries: what is available and what is needed. Paediatrics and Child
Health 2012, 32: S2-30-37. DOI 10.1179/2046904712Z.00000000076.
12. Portevin D, Moukambi F, Clowes P, Bauer A, Chachage M, Ntinginya NE, et al As-
sessment of the novel T-cell activation marker-tuberculosis assay for diagnosis of
active tuberculosis in children: a prospective proof-of-concept study.
www.thelancet.com/infection.PublishedonlineSeptember1,2014. http://
dx.doi.org/10.1016/S1473-3099(14)70884-9.
13. Clayden P, Collins S, Frick M, Harrington M, Horn T, Jefferys R, Lessem E, McKen-
na L. 2016 Pipeline Report. HIV and TB. Drugs, Diagnostics, Vaccines, Preventive
Technologies, Research Toward a Cure, and Immune-Based and Gene Therapies
in Development. TAG i-base, www.pipelinereport.org.
14. Enarson PM, Enarson DA, Gie R. Management of tuberculosis in children in low
81
income countries. Int J Tuberc Lung Dis. 2005;9:1299-1304.
15. Marais BJ, Schaaf HS, Donald PR. Management of Tuberculosis in Children and
New Treatment Options. Infectious Disorders – Drug Targets, 2011; 11:144-156.
16. Zachariah R, Harries AD, Ishikawa N, Rieder HL, Bissell K, Laserson K, Massaquoi
M, Van Herp M, Reid T. Operational research in low-income countries: what, why
and how? Lancet Infect Dis. 2009;9:711-17.
17. Training Modules for TB in Children, 2008. Department of Health/National TB Pro-
gram.
18. World Health Organization International Classification of Diseases (ICD) 10.
www.who.int/classifications/icd/en.
19. National Consensus on Childhood Tuberculosis. Philippine Pediatric Society. 1997.
20. Evidence-Based Clinical Practice Guidelines. Childhood Tuberculosis. Philippine
Pediatric Society,. 2008.
21. Lonnroth K, Corbett E, Golub J, Godfrey-Faussett, Uplekar M, Weil D, Raviglione
M. Int J Tuberc Lung Dis, 2013; 17: 289-298. http://dx.doi.org/10.5588/ijtld.12.0797.
22. World Health Organization. WHO Systematic screening for active tuberculosis:
Principles and recommendation. (WHO/HTM/TB/2013.04). http://www.who.int/tb/
tbscreening
23. de Tavera, P. M. Tuberculosis in Filipino Children.1975.
82
8
Tuberculin Skin Test
Cleotilde H How, MD
Luis T Chan, Jr, MD
OUTLINE
1 Overview
2 The Procedure
3 Interpretation
4 Special Considerations
83
OVERVIEW
Since Koch‘s discovery of the tuberculin in the year 1890, the Mantoux tuberculin skin
test (TST) has been used as the standard method in demonstrating Mycobacterium
tuberculosis (MTB) infection. In 2001 the US-FDA initially approved and recognized the
interferon-y gamma release assay blood test (IGRA) as an alternative diagnostic tool to
the TST for M. tuberculosis infection.1 The TST and IGRAs are indirect tests that meas-
ure immunologic responses that suggest past or present exposure to the TB bacilli.
While both tests are designed to detect tuberculosis (TB) exposure and latent TB infec-
tion (LTBI); it may also be used as a presumptive test for TB disease2,3 in conjunction
with other criteria4,5,6 or with bacteriologic confirmation7. See Chapter 7(Diagnosis)
A diagnosis of LTBI requires that active TB be excluded by history and physical exami-
nation to check for suggestive signs and symptoms, chest X-ray, and or bacteriologic
evidence - before treatment is initiated. The goal of testing for LTBI is to identify individ-
uals who are at increased risk for the development of active tuberculosis (TB) and
therefore benefit from treatment of latent TB infection.8
In the absence of bacteriologic evidence, a positive TST is one of the five (5) criteria
set by the Philippine Pediatric Society in making a presumptive diagnosis of TB dis-
ease in children9. The TST is also an important tool in TB control and prevention. The
recognition of latent TB infection or LTBI (i.e. TB infection without disease) helps in the
decision-making for preventive chemotherapy; especially in the under-fives, who are at
increased risk of infection progressing to TB disease. LTBI in the young is also consid-
ered a sentinel event, reflecting recent transmission. Thus the TST is important as an
epidemiological tool for tuberculin surveys in assessing the annual risk of TB infection
(ARTI).10
Tuberculin reactivity provides a general measure of a person‘s cellular immune respon-
siveness.. It is based on a delayed (cellular) hypersensitivity to certain antigens of the
TB organism contained in extracts of culture filtrates known as ―tuberculin‖11,12. The de-
layed (cellular) hypersensitivity reaction is manifested clinically as a type IV immune
response mediated by sensitized T lymphocytes and is characterized by an indurated
response to the intradermal injection of protein from the cell wall of the tubercle bacil-
lus.
A prior infection with the Mycobacterium tuberculosis (M. tuberculosis) or tuberculopro-
teins from BCG vaccine results in T-cell sensitization that releases lymphokines at the
site of injection. These lymphokines then induce local vasodilation, edema, fibrin depo-
sition, and recruitment of other inflammatory cells to the area resulting in induration.
Features of the reaction include:12 (1) its delayed course, reaching a peak in 48 – 72
hours after injection of the antigen; (2) its indurated character; and (3) its occasional
vesiculation and necrosis. Reaction to the tuberculin starts five to six hours after injec-
tion, in which maximal induration is noted within 48 to 72 hours post-injection and sub-
sides over a period of days.
In most children the delayed-type hypersensitivity response to tuberculin, which is the
estimated interval between TB exposure and detectable skin test reactivity (referred to
84
as the window period) ranges from two to twelve weeks.13,14 Analysis of data collected
from studies indicates that eight weeks is the outer limit of this window period. A nega-
tive test result obtained less than eight weeks after exposure is therefore considered
unreliable for excluding TB infection. The Centers for Disease Control and Prevention
(CDC) recommends a TST in individuals with history of contact with an infectious
source, between eight to ten weeks after the most recent TB exposure and if initial TST
result is negative among individuals with history of contact with an infectious
source.13,15 The Department of Health - National TB Program (DOH – NTP) recom-
mends a repeat TST at three months after a negative TST in a TB symptomatic child or
three months after a negative TST in children four years of age and below.16
THE PROCEDURE4
The Mantoux test is the standard and recommended method of giving the tuberculins
for TB screening. It employs an introduction of a small amount of tuberculoprotein
(purified protein derivative or PPD) into the intradermal tissues with a small-gauge nee-
dle.
Infants, children, pregnant women, HIV – infected individuals or persons vaccinated
with BCG vaccine can have a TST. It is contraindicated for persons who have had a
severe reaction (e.g., necrosis, blistering, anaphylactic shock or ulcerations) to a previ-
ous TST.13
Preparations of PPD for Mantoux Tuberculin Skin Testing
Purified protein derivative (PPD) or tuberculin, which is used for most skin testing, is
isolated from culture filtrate by protein precipitation. Tuberculin units are defined on a
weight basis: one tuberculin unit (1 TU) equals 0.02 microgram (ug) in 0.1 mL of tuber-
culin PPD-RT 23 SSI. The standard test dose of a commercial PPD preparation is de-
fined as the dose of the product that is biologically equivalent to that contained in five
tuberculin units (5 TU) of PPD-S (i.e. it elicits reactions of equivalent size + 20%)17,18,19.
PPD–RT 23
The World Health Organization (WHO) and the International Union Against Tuberculo-
sis and Lung Disease (IUATLD) recommend the 2 tuberculin units (2 TU) PPD-RT 23,
as the standardized dose for Mantoux TST surveys14. The PPD–RT 23 is the most
widely used TST in the world10,16. This is equivalent to two-fifths the concentration of
antigen determined to be bioequivalent to 5 TU of PPD-S, the standard tuberculin prep-
aration.12,17,20 Therefore, a dose of 0.1 ml of 2 TU PPD-RT 23 is biologically equivalent
to 0.1 mL of 5 TU PPD-S.
PPD–S
The American Thoracic Society and the Centers for Disease Control and Prevention
(CDC) in the US endorse the 5 TU of PPD-S as the standard dose for TST in North
America. Newly manufactured batches of tuberculin are bioassayed, and the 5 TU
standard is the amount of material which produces results equal to those produced by 5
TU PPD-S = 0.1 microgram of PPD-S.18,19
85
Using 10 mm as the positive cut-off point for tuberculin reactivity, a simultaneous study
involving 202 health workers comparing the PPD reactivity between 2 TU PPD-RT 23
and 5 TU PPD-S was found to be comparable13. The skin test reaction sizes with the
two antigens (i.e. 2 TU PPD-RT 23 and 5 TU PPD-S) did not differ statistically, based
on age, sex or prior BCG vaccination. As a rule of thumb, 0.1 ml of the 2 TU of RT 23
will have a tuberculin reactivity similar to 0.1 mL of the 5 TU of PPD-S11,12.
Administration, Reading, and Recording of the Mantoux TST11,12,20
A valid TST entails standardization of procedures, training and supervision of person-
nel, and practice. Only trained health care providers/workers are advised to administer
and read the Mantoux TST.
Administration of the Test
Locate the site of injection, about 2 inches below the elbow joint in the volar aspect of
the forearm. Clean the preferred site with alcohol swab and air dry. For one time test,
use a disposable tuberculin syringe with gauge 25 to 27 short bevel needle (i.e. ¼- to ½
-inch). Aspirate 0.1 mL of either the 2 TU of PPD-RT 23 or the 5 TU of PPD-S and in-
ject the needle bevel up intradermally. A pale wheal of six to ten mm in diameter should
be evident after injection; otherwise, repeat the test on an area at least two inches
away from the original site.
Untoward reactions to tuberculin are uncommon. Some delicately sensitive individuals
may have vesicular or ulcerating local reactions to skin testing. Much less encountered
are regional adenopathy and fever. Advise subjects not to scratch or apply any oint-
ments or cream on the injected site.
Reading and Recording of the Test
The TST should be read between 48 to 72 hours after administration. Positive TST re-
actions can be measured accurately for up to seven days, while negative TST reactions
can be read accurately up to 72 hours only21.
Under good light, visually inspect the injected area for any reaction to tuberculin which
is the induration (palpable, raised hardened area) and not the erythema. The diameter
of the indurated area should be measured across the forearm (perpendicular to the
long axis). Using the fingertips, palpate and locate for the widest diameter of induration
and mark both edges of the widest induration across the forearm using a ball pen. With
a standard ruler and using the mm part, place the ―0‖ ruler line inside left dot edge and
read ruler line inside the right dot edge.
The size of induration should be recorded in millimeters. If no induration is found, ―0
mm‖ should be recorded. Do not record as ―positive‖ or ―negative‖.
INTERPRETATION
The interpretation of a PPD reaction should be based on the purpose for which the test
is given, the prevalence of TB infection in the population being tested, and the conse-
quences of false classification. An accurate interpretation of the TST requires
86
knowledge of the antigen used (tuberculin), the proper technique in the administration
and reading of the test, results of epidemiological and clinical experience with the test,
and conditions that can bring about the false positive and false negative interpretations
of the tests.
Factors that May Affect the TST Reaction
False-positive reactions21
Certain individuals may have a positive reaction to the TST even though they are not
infected with M. tuberculosis. The causes of these false-positive reactions may include,
but are not limited to the following:
Infection with nontuberculous mycobacteria
Previous BCG vaccination
Incorrect method of TST administration
Incorrect measurement and interpretation of reaction
Incorrect strength of antigen used
A history of BCG vaccine is not a contraindication for tuberculin skin testing. Post-BCG
tuberculin reactions develop six to 12 weeks after vaccination22,23. BCG immunization
and other nontuberculous mycobacteria (NTM) can bring about tuberculin reactivity.
Reaction sizes caused by nontuberculous mycobacteria are usually £ 10 mm of indura-
tion.21
However, the tuberculin reaction believed to be affected by BCG wanes after five years
from immunization. About 80% to 90% of children who received BCG in their infancy
have non-reactive6 TST at five years of age.22,23,24 Studies have shown that infants, chil-
dren, and adults from countries with intermediate and high TB rates have the same
prevalence of significant tuberculin reactions, regardless of BCG status.25,26,27
A positive PPD reaction in individuals residing in areas highly endemic for TB, e.g.
>100/100,000 population, is most likely due to exposure from natural infection caused
by M. tuberculosis, rather than those caused by non-tuberculous mycobacteria (NTM)
or BCG immunization, since most patients who receive BCG vaccination during infancy/
childhood lose their cutaneous hypersensitivity reaction to tuberculin within five
years.11,12,18 Therefore, a significant reaction more likely represents true exposure to TB
especially in the setting of a recent exposure.
False-negative reactions
Some individuals may have a negative reaction to the TST even if they are infected
with the M. tuberculosis. Factors that may cause false negative reactions to the Man-
toux test are enumerated in Table 8.1.
87
Table 8.1 Factors that may cause false negative reactions to the Mantoux test
Factors related to the person being tested
infections: viral - measles, mumps, chickenpox; bacterial - typhoid fever, brucel-
losis, typhus, leprosy, pertussis tuberculous, pleurisy; fungal - South American
blastomycosis
live attenuated virus vaccinations against measles, mumps, polio, chicken pox
metabolic derangements - e.g. chronic renal failure
nutritional factors - e.g. severe protein depletion
diseases affecting lymphoid organs such as Hodgkin‘s disease, lymphoma,
chronic lymphocytic leukemia and sarcoidosis
corticosteroids and other immunosuppressive agents
age: newborns and elderly patients with ―waned‖ sensitivity
incubating or recent, far advanced or overwhelming infection with M. tuberculo-
sis
stresses such as surgery, burns, mental illness, and graft versus host reactions
complete anergy, giving negative skin test response to PPD as well or other skin
test antigens
Factors related to the tuberculin used
improper storage - exposure to light and heat
improper dilution
chemical denaturation
contamination
adsorption into the syringe - partially controlled by adding Tween 80
Factors related to the method of administration
injection of too little antigen
delayed administration after drawing into syringe
too deep injection
Factors related to error in reading and recording of results
inexperienced reader
conscious or unconscious bias
error in recording
89
month or those taking TNF-α antagonists).
The >10mm cut-off point is recommended for populations with high risks of having TB
infection and disease and for persons living in areas were TB is highly prevalent and
>15mm for populations with no risk factors.
National TB Prevalence Surveys
The national TB prevalence surveys done in 1981-1983 and in 1997 (using the 1TU-
PPD RT-23 and the 2-TU–PPD-RT-23 respectively) both showed the positive cut-off
point for tuberculin reactivity at 8mm induration30. In the 2007 nationwide TB preva-
lence survey, the positive cut–off was lowered from 8mm to 7mm due to the possibility
of under reading of the tuberculin reactions.
Rationale for Choosing the Positive Cut-off Value of the TST
Deliberations in the choice for the most appropriate definition of a positive cut point for
tuberculin reactivity should be weighed judiciously between over-diagnosis and under-
diagnosis of TB infection and disease. The positive cut-off value in tuberculin reactivity
should best discriminate TB infection and disease from normal subjects based on the
sensitivity, specificity, and positive predictive value of the test with clinical and epidemi-
ological considerations.
Sensitivity, Specificity and Positive Predictive Value (PPV)
The sensitivity of the diagnostic test refers to the probability that the test result will be
positive, while specificity refers to the probability that the test will be negative. 30 The
PPV, on the other hand, refers to the probability that a person with a positive result ac-
tually has the disease; it also serves as a crude measure of relative cost-efficiency, i.e.
it reflects the ratio of the screening program yield, i.e. number of true positives, to the
cost of misdiagnoses, i.e. no. of false positives and false negatives for a given number
of screens.30,31
Local studies using the 10mm induration size as the positive cut–off value for the TST
showed a sensitivity ranging from 64.7% to 99.7% and a specificity of 54% to 97.7%
26,27,,
False-positive tuberculin tests can occur in persons who have been infected with
nontuberculous mycobacteria and in persons who have received BCG vaccine. These
false-positive reactions result in lower specificity, low PPV and low probability of LTBI.
Epidemiologic and Clinical Considerations
The Mantoux PPD TST is the best screening test for patients with asymptomatic TB. In
places where the prevalence for TB infection is high, the specificity and PPV of the
PPD TST are remarkably increased, resulting to a superior tool for screening purpos-
es11. As the prevalence of the disease increases, it becomes more likely that the person
being tested actually has the disease with less false positive results. Hence, the more
prevalent the disease, the more sensitive the test must be.
A lower positive cut-off point e.g., from >10mm to >8mm or from >10mm to >5mm indu-
ration — results to less false negative cases, thus increasing the probability of diagnos-
ing more children with TB infection and disease. This is the rationale for a lower posi-
tive cut-off value (i.e. from 10 mm to 5 mm) in individuals with history of close contact
90
with a known or suspected infectious case of TB, in persons with clinical findings sug-
gestive of TB and in persons with chest x-ray readings suggestive of TB.
Since tuberculin reactivity provides a general measure of a person‘s cellular immune
responsiveness, a lower positive cut-off point, e.g. from 10mm to 5mm induration, is
recommended for immunocompromised individuals (e.g. HIV infection, and severely
malnourished children - those with clinical evidence of marasmus or kwashiorkor) who
may be unable to mount an immune response to the TST and thus have false negative
TST results7 or for persons taking immunosuppressive drugs that could decrease the
person‘s cellular immune responsiveness to the tuberculin.
Tuberculin skin test reactions using the Mantoux method should be interpreted in the
same manner for persons who have received BCG and for those who did not11,15. Ap-
proximately 10% of children with culture-proven TB are Mantoux test negative.38,39 Like-
wise, a negative reaction to the TST does not necessarily rule out TB.
Individuals with positive TST results is likely to have a positive reaction to the TST for
life; hence, an individual with a previous positive TST is not required to undergo subse-
quent testing11,14,39 The PPD tuberculin test reaction should always be correlated with
the recipient‘s history of exposure to an infectious TB source, presence of clinical signs
and symptoms suggestive of TB or with chest radiograph findings (if done), in order to
judiciously diagnose TB infection and disease and immediately manage such cases
appropriately.
91
infection (LTBI). They are surrogate markers of Mycobacterium tuberculosis infection
and indicate a cellular immune response to M. tuberculosis Blood assays for tuberculo-
sis, commonly referred to as interferon gamma release assays (IGRAs), are based on
the principle of interferon-gamma being critical to regulation of the cell-mediated im-
mune response to Mycobacterium tuberculosis (MTB) infection1.
The currently available and FDA-approved IGRA assays for MTB include QuantiFER-
ON-TB Gold in-Tube test (QFT-GIT) and the T-SPOT.TB. Both assays measure the
interferon-gamma response to MTB proteins, including early secretory antigenic target-
6 (ESAT-6) and culture filtrate protein-10 (CFP-10).1 Because these assays quantitate
a biologic response, testing of a fresh blood specimen having adequate viable white
blood cells is crucial to obtaining accurate results.3
Key Facts: TSTs and IGRAs testing for TB infection and TB disease1,2,3
TST and IGRA generally should not be tested on persons with low risk of TB infec-
tion and TB disease.
Neither IGRA nor TST can distinguish active TB from latent tuberculosis infection
IGRA can distinguish LTBI from previous BCG administration
Routine testing with both TST and an IGRA is not generally recommended. Excep-
tions include suspected active TB in immune compromised patients, or indetermi-
nate results from either test.
An IGRA is the test of choice in two instances; for people who have received BCG,
either as a vaccine or as chemotherapy and under circumstances where the tested
person is unlikely to return to have the TST read.
TST is preferred over IGRA for children less than 5 years of age.
Live vaccines may affect both TSTs and IGRA results. IGRA testing should be done
either on the same day of live vaccine administration or be postponed at least four-to-
six weeks from a live-vaccine administration
Advantages, Limitations and Disadvantages of IGRAs over TST1,2,3
IGRAs can be used as a diagnostic tool for TB infection similar to TST e.g. in contact
investigations, test during pregnancy, screening for health workers and for others un-
dergoing serial evaluation for M. tuberculosis infection. However its use is not recom-
mended in high-burden low-resource countries.4,7
IGRAs blood tests require only a single visit to a health care provider to draw the blood
and the results can be available in 24 hours. Results are not affected by prior BCG ad-
ministration through vaccination or chemotherapy. It also does not boost responses by
subsequent tests.
Apart from its availability and a much higher cost of the IGRA tests over the TST, some
disadvantages include immediate processing of blood samples within 8 – 30 hours after
collection while white blood cells are still viable, errors in collecting/transporting blood
specimens or in running and assay interpretation. False positive results also may occur
due to infection with other mycobacterial organisms, including M. kansasii, M. szulgai,
and M. marinum, due to their production of ESAT-6 and CFP-10
92
The Mantoux TST is preferred over IGRA for children less than 5 years of age. Blood
extraction for IGRA testing can pose as a challenge in young children requiring at least
1 – 2 mL. as blood test specimen. Limited studies show a reduced sensitivity in the
IGRA test over TST in young children and HIV-infected children.
93
REFERENCES:
1. National Institute for Health and Clinical Excellence. Clinical Diagnosis and Man-
agement of Tuberculosis, and Measures for its Prevention and Control. March
2011
2. World Health Organization. Tuberculosis IGRA TB Tests Policy Statement 2011
3. Centers for Disease Control and Prevention. Updated Guidelines for Using Interfer-
on Gamma Release Assays to Detect Mycobacterium tuberculosis Infection, United
States. MMWR 2010; 59 (No.RR-5)
4. Guidance for national tuberculosis programmes on the management of tuberculo-
sis in children. Geneva, World Health Organization, 2014. (WHO/HTM/TB/2014.03)
5. Manual of Procedures of the National Tuberculosis Control Program, 2014.
6. Philippine Pediatric Society. Evidence-based clinical practice guidelines for child-
hood tuberculosis. 2008.
7. TB Care I. International Standards for Tuberculosis Care, Edition 3, TB CARE I,
The Hague, 2014.
8. WHO Guidance on the Management of Latent TB Infection (LTBI), 2015
9. Consensus Statements on Child TB. Philippine Pediatric Society 1997
10. Menzies D. EDITORIALS: Tuberculin surveys - why?. Int J Tuberc Lung Dis.
1998;2 (4):263-264.
11. American Thoracic Society. Diagnostic Standards and Classification Tuberculosis
in Adults and Children. Am J Respir Crit Care Med. 2000;161:1387.
12. Huebner RE, Schein W, Bass, Jr JB. The tuberculin skin test. Clin Infect Dis.
1990;17:968-975.
13. Centers for Disease Control and Prevention. Targeted tuberculin testing and inter-
preting tuberculin skin test results -Fact sheet. 2009. Available from: http://
www.cdc.gov/tb/publications/factsheets/testing/skintestresults.htm.
14. Pediatric Tuberculosis Collaborative Group. Targeted Tuberculin Skin Testing and
Treatment of Latent Tuberculosis Infection in Children and Adolescents. PEDIAT-
RICS. 2004 Oct;114(4):1175-1201.
15. National Tuberculosis Controllers Association; CDC. Guidelines for the Investiga-
tion of Contacts of Persons with Infectious Tuberculosis. MMWR; 16 December
2005;54(RR15):1-37.
95
32. Sackett DL, Haynes RB, Tugwll P. Clinical Epidemiology: A Basic Science for Clini-
cal Medicine. Boston: Little Brown and Company; 1985.
33. Fletcher RH, Fletcher SW ,Wagner EH. Clinical Epidemiology- The Essentials. Bal-
timore: Williams & Wilkins; 1987.
34. Alon MJC, Gatchalian S, Tabora C. PPD tuberculin Test reactivity of Filipino infants
aged 6 months and 1 year vaccinated with BCG at birth. The Philippine Journal of
Pediatrics. 2001;50:32-35.
35. De la Cruz JM, Garcia I, PahatiL, de Leon MNA. PPD reactivity among high risk Fili-
pino children with or without BCG vaccination. The Philippine Journal of Pediatrics.
1999;48:70-74.
36. Johnson H, Lee B, Kelly E, McDonnell T. Tuberculin sensitivity and the BCG scar in
tuberculosis contacts. Tuber Lung Dis. 1995;35:113-7.
37. Smith MHD, Starke JR, Marquis JR. Tuberculosis and opportunistic mycobacterial
infections. Philadelphia: W.B. Saunders Co; 1992. p.1321-1362. (Feigin RD, Cherry
JD, editors. Textbook of Pediatric Infectious Diseases, 2 vols, 3rd ed.).
38. Steiner P, Rao M, Victoria MS, et al. Persistently negative tuberculin reactions. Am
J Dis Child. 1980;134:747-750.
39. Core Curriculum on Tuberculosis. What the Clinician Should Know: Centers for
40. Disease Control and Prevention National Center for HIV, STD, and TB Prevention
41. Division of Tuberculosis Elimination; http://www.cdc.gov/tb/education/corecurr/
index.htm 2010.
96
9
Radiologic Findings
Maricar Paguia, MD
Myra R Asuncion-Son, MD
OUTLINE
1 Overview of Chest Radiography in
Children and Other Ancillary Tests
97
OVERVIEW OF CHEST RADIOGRAPHY IN CHILDREN AND OTHER ANCIL-
LARY TESTS
The chest x-ray is the most basic and widely used radiological investigation used for
pulmonary tuberculosis.
Chest radiography is helpful in localizing the site of tuberculosis (TB) lesions. Although
not specific for the diagnosis of pulmonary TB, it becomes important when diagnosing
in places where TB is highly prevalent and where facilities for bacteriologic examination
are not easily available.1 Tuberculin skin test reaction lags behind radiological findings2.
Sputum almost always is unavailable for testing, and gastric washings are only positive
in 30-40% of cases.3 Because of all these limiting factors, Mycobacterium tuberculosis
is isolated in less than 50% of pediatric cases. Hence, the diagnosis of tuberculosis
(especially during the early stages), often relies on imaging findings. A study done by
Gwee et al showed that chest x-ray was able to demonstrate tuberculosis in a small but
noteworthy number of children in the absence of clinical symptoms.
Chest radiographic evidence parallel that of the pathologic changes of tuberculous in-
fection. These are divided into: 1) initial exposure or primary TB; and 2) post primary or
primary progressive TB. Reactivation of previous infection or progression of an initial
infection exemplifies the latter.
The primary complex is composed of the primary focus, lymphangitis, localized
pleural effusion and regional lymphadenitis. The hallmark of the initial disease is the
relatively large size of the adenitis compared with the relatively insignificant size of
the initial focus in the lungs.31
In radiological studies, it is important that we consider the following points in the inter-
pretation of chest film in children, particularly in those suspected with TB:
98
Lateral view of chest radiograph increases the sensitivity/yield of diagnosis in detecting
hilar adenopathy,16 but the use of high kilovolt (kv) view does not significantly improve
the accuracy of diagnosis.17 Partially calcified mediastinal nodes may be visible only in
lateral projections.
The common chest radiologic finding in primary TB is lymphadenopathy. Should an en-
larged lymph node be noted on chest x-ray, it should be interpreted as nonspecific
lymph node enlargement. Radiologists should refrain from using ―primary complex‖ un-
less the lymph node enlargement is associated with parenchymal disease and that per-
tinent clinical information strongly suggests such diagnosis. Providing a list of differen-
tial diagnosis is encouraged by clinicians. Lymphadenopathy can also be seen in viral
and bacterial infections. A radiological follow-up allows excluding of sequela or a preex-
isting pulmonary abnormality such as TB; however, follow-up x-rays are to be delayed
two to three weeks after the clinical recovery due to the late radiological recovery after
treatment.26 If lymphadenopathy persists, mycobacterial etiology can be considered 15.
The most common cause of calcification in children is TB. However, other diseases,
such as Histoplasmosis, Coccidioidomycosis and Aspergillosis, may also produce in-
trathoracic calcifications which a radiograph cannot differentiate from those due to TB.
Calcifications can also be demonstrated in the nodes regional to inoculations with BCG
vaccine; these, however, tend to disappear more quickly than those associated with
natural infections.
There are no pathognomonic radiographic findings. Perhaps the only finding that may
be highly suggestive of TB in infants and children is the uniform stippling of both lungs
found in miliary TB.
Historically, bronchography was useful in the definition of bronchial stenosis or bronchi-
ectasis. At present, special imaging techniques, such as computed tomography and
magnetic resonance imaging, may be of particular value in defining nodules, cavities
cysts, calcifications, contours of large bronchi and vascular details in lung parenchy-
ma.6
A cross-sectional study among Filipino children six months to 18 years old with a posi-
tive chest x-ray, positive tuberculin test and positive history of TB contact with symp-
toms suggestive of TB disease like cough, fever, weight loss, hemoptysis, anorexia,
dyspnea, weakness was done at the Philippine Heart Center. The over-all positive pre-
dictive value for diagnosing TB disease among these children using chest x-ray was
only 59% compared to chest computed tomography CT scan which was 98%. In Filipi-
no children, the specific lesions seen in chest CT scans are subcentimeter (lymph
nodes which are enlarged in the peri-hilar region of the lungs, but these are less than
one centimeter in greatest diameter), calcified nodes, peribronchial, axillary, bilateral
hilar nodes, pericardiac nodes with ring enhancement, and granuloma.18 This is similar
to the characteristic CT findings of childhood TB in other countries of low attenuation
with rim enhancement or calcification of mediastinal and hilar lymph nodes.19
In another local study done among diagnosed patients four months to 18 years old
(n=83), review of radiographs revealed the following findings: lymphadenopathies, in-
99
cluding lymph node enlargement as a solitary finding and as part of Ghon focus (64%),
parenchymal infiltrates (78%), air-space opacities (63%), cavitary lesions (21%), mili-
tary pattern (10%), bronchial and tracheal compressions (12%), and atelectasis (22%).
Only 7% of the subjects had normal radiographs.
Pleural effusion was present in one-fourth of the primary cases. Five had massive effu-
sion, one of which was proven to be empyema. Two patients presented with cardiac
shadow enlargement. One had pericardial effusion, while the other had constrictive per-
icarditis. Paraspinal masses associated with osseous destruction were appreciated in
10%; Pott‘s disease was observed in the old age bracket while a significantly higher
proportion of lymphadenopathy was noted among the younger group. Miliary TB was
linked with younger age bracket, positive culture results, and high mortality.40.
In the evaluation of children with known or suspected pulmonary TB, CT scans cannot
be routinely recommended because of its high cost, the need for IV sedation and the
risk involved in administering a contrast medium. However, when chest radiographs are
normal or when the presence of mediastinal lymphadenopathy cannot be seen by chest
x-ray but TB disease is clinically suspected, CT scan may be used in the diagnosis of
TB disease in pediatric age groups, especially when there is difficulty in establishing
definite diagnosis. 18,19,27,28
100
The most common chest radiograph findings in childhood TB are lymphadenopathy and
parenchymal abnormalities, which occurs in 83% to 92% in the first case19,20 and in
60.8% in the second case.20,21
As a rule, pleural exudates and lymphangitis are not clearly seen. Common radiological
findings are enlarged retrocardiac lymphadenopathy in 70% of the cases, hilar ade-
nopathy with pulmonary infiltrates in 20% and pleural effusion in 6-11%.5,20 Chest radio-
graphs may be normal in up to 10% of patients who have proven primary TB.8
Hilar adenopathy has a specificity of 36%. Because of its low specificity, this radiologi-
cal finding should not be used as a sole basis in initiating treatment, especially when
there are no other manifestations suggesting TB.5 The lymphatic drainage of the lungs
occurs predominantly from left to right, and therefore, the nodes in the right upper para-
tracheal area appear to be the ones most often affected.
Mediastinal lymphadenopathy, on the other hand, has been found to be a more likely
characteristic in children with both tuberculous meningitis and military TB than those
with miliary TB or TB meningitis alone.
Many primary lesions are subpleural and the lymphatic drainage of the apical pleura is
in the cervical nodes. The primary focus can look like any pneumonia and be situated
anywhere. All areas of the lung may be involved.
The principal components of the primary complex and their fate are discussed further
below.
1. Parenchymal Involvement
The parenchymal reaction is typically that of acinar consolidation, which is usually ho-
mogenous in density and with ill-defined margins, except where it abuts against a fis-
sure. It predominantly involves the upper lobes with preferential location between ante-
rior or posterior segments and between right and left. Quite frequently, it also involves
the apex of a lower lobe.
Atelectasis is important in the production of massive shadows. It frequently affects the
anterior segments of the upper and middle lobes of the right lung due to bronchial com-
pression by enlarged lymph nodes. There is surprisingly little shift in the heart and other
mediastinal structures; instead, the healthy lung on the same side expands and fills in
the space previously occupied by the collapsed segment. Lobar involvement is most
probably a combination of parenchymal consolidation and parenchymal atelectasis. An
entire lobe, often the right middle lobe, may be affected.
Large primary foci and their perifocal reactions may occupy all or most of a lobe and
obscure the lymphangitis and pleural exudate. Perifocal shadows may represent pneu-
monic consolidation, atelectasis or pleural exudate, singly or in combination. With the
gradual clearing of the perifocal shadows, the pulmonary lesion (primary focus) may
remain obscured or may become visible later. In other cases, large calcifying pulmo-
nary foci may dwarf the remainder of the tuberculous complex. Cavitation of the primary
focus with a perifocal exudate may occasionally be seen but is considered a rare mani-
101
festation of pulmonary TB in infants and children.
2. Lymph Node Involvement
Hilar or paratracheal lymph node enlargement is the radiologic finding that clearly differ-
entiates primary from postprimary TB. The lymph node enlargement is usually unilat-
eral. Highly suggestive of primary TB is the large size of the adenitis relative to the in-
significant size of the primary lung focus.
Progression from Ghon focus and lymph node disease tend to occur more frequently
among the younger age group (less than 5 years of age) and adolescents (>10
years).40
3. Airway Involvement
Tracheobronchial involvement is a common occurrence; it is usually the result of bron-
chi compression due to enlarged lymph nodes. It may also result from a long-standing
or untreated infection. Bronchial obstruction due to tuberculous lymph nodes may pre-
sent radiographically as:
102
disease. The pleural effusion may also be generalized, unilateral, or bilateral as seen
more commonly in progressive type of pulmonary TB. It may obscure the underlying
pulmonary changes, especially when it is large. The pleuritis may be small, localized
and asymptomatic or it may occur in the form of generalized effusion usually three to
six months after the infection occurs. Differentiating effusion from extensive pneumonic
lesion is difficult; thus, lateral decubitus radiographic views are helpful in confirming the
presence of pleural fluid.31
5. Resolution of Radiologic Changes
Regression of radiographic abnormalities in pulmonary TB is a slow process. In the first
three months of treatment, worsening of radiographic findings, which consists of exten-
sion of parenchymal involvement and enlargement of nodes, may be observed in al-
most one third of pediatric patients receiving appropriate therapy.20 Due to the great
variation in the velocity of healing in the different components of each complex, healing
may present a wide variety of findings in different cases. Resorption may begin in the
different stages of healing: during fibrosis, hyalinization, or calcification in different parts
of the complex.
In the small proportion of children with radiological evidence of the disease, clearing
usually occurs within six months to two years after institution of therapy. 9 In other re-
ports, the enlarged lymph node and parenchymal disease can persist for two to three
years or even longer.20 Chest x-ray findings should always be correlated with the clini-
cal manifestations.
Complete resolution, but without apparent residual, is seen in the majority of cases. In
some cases, atelectasis remains due to residual lymph node enlargement. A significant
percentage of children who have had atelectasis during the active stage of the disease
showed residual bronchiectasis in the affected areas.
In interpreting calcification on chest x-ray, calcification may be due to healed, healing,
or quiescent infection; thus, it should be correlated with the history of treatment.
Duration of follow up for patients with primary TB depends on the patient‘s clinical sta-
tus after therapy. Radiologists can recommend follow up study for as early as three to
six months or sooner if the patient is not responding to treatment. If patient has good
clinical response, a follow up x-ray would be to the clinician‘s discretion.15
Postprimary Tuberculosis
103
The following are radiologic features seen in chronic pulmonary tuberculosis:
Local fibroproductive TB: The relatively poor definition of the exudative lesion is re-
placed by shadow, which is more sharply circumscribed, homogenous, and usually
of somewhat irregular and angular contour. Cavitation may be seen. Healing occurs
by fibrosis and the resultant cicatrisation may result in loss of volume. If there is sig-
nificant volume loss, compensatory signs may become evident such as elevation of
the ipsilateral hilum, over inflation of the rest of the affected lung, and in some cas-
es, formation into bullae. This pattern is more commonly seen in chronic pulmonary
TB.
Cavitation: Cavities are the radiological hallmark of reactivation TB; they are evident
radiographically in 20% to 45% of patients.27 The wall of an untreated tuberculous
cavity is moderately thick, and its inner surface is fairly smooth. An air-fluid level is
seldom seen. With adequate therapy, a cavity may disappear or regress into a pa-
per-thin, air-filled cystic space. Cavitation is seen both in progressive and chronic
types of pulmonary TB.
Caseous material in a tuberculous focus may simulate cavitation because of its high
lipid content that produces a shadow slightly less dense than the wall. An aid to dif-
ferentiation is the contrast between the inner wall and the central radiolucency. Air
in apposition to a wall tends to show a sharper definition than when the interface is
liquid against solid.
104
2. Miliary Tuberculosis
In the event of an explosive and massive spread into the bloodstream, both lungs are
evenly seeded with innumerable foci of approximately the same sizes. At first, these
lesions are too small to be visualized. The pattern may not become apparent until sev-
eral weeks after the patient is seen .The lesions require at least 2 ½ weeks to become
perceptible and would first appear as a indistinguishable nodule of a uniform size of 2
mm.10
As the foci become larger and older, multiple small shadows appear which stipple both
lungs more or less uniformly (―millet-seed‖ densities). This stippling of the lungs is the
most diagnostic radiological change in pulmonary TB during infancy and childhood. Lat-
er, stippling in the lungs coalesce thus, producing a richly stippled pattern (―snowstorm
effect‖).
A local study was done correlating the different radiographic patterns of primary TB with
clinical presentation, microbiologic, as well as polymerase chain reaction (PCR) find-
ings. Results of said study showed that the yield of PCR was highest among the group
with disseminated military pattern compared to those who have lymph node disease,
progression from Ghon focus, and pleural effusion.
With adequate treatment, clearing is quite rapid and without residual. Considerable im-
provement may be observed within five weeks after initiating treatment, while clearing
is complete in seven to 22 months, with a mean of 16 weeks.
This miliary pattern may also be seen in sarcoidosis, pneumoconiosis, disseminated
carcinomatosis, and fungal and viral infections. Miliary calcification is extremely rare in
miliary TB. Other causes are to be considered such as histoplasmosis, coccidioidomy-
cosis, schistosomiasis and paragonimiasis.
3. Airway Tuberculosis
Tuberculous bronchiectasis: In endobronchial TB, the bronchi are affected in sever-
al different ways: a) exogenous lymph node compression; b) intrinsic granuloma
formation; and c) bronchiectasis.
Bronchographic evidence of distortion and dilatation of the bronchial tree
(bronchiectasis) may develop when the bronchial wall is infected. Healing by fibro-
sis and cicatrization leads to irreversible dilatation. Obstruction of a segmental bron-
chus from compression by enlarged lymph nodes or by bronchostenosis can lead to
destructive pneumonitis and subsequent bronchiectasis.
Bronchiectasis is twice more frequent in patients with hemoptysis. This may be
asymptomatic and suggested only by non-resolving radiographic shadows despite
adequate treatment. A definitive diagnosis of bronchiectasis is made through bron-
chography, CT scan, and bronchoscopy.
105
almost inevitable with its resultant obstructive atelectasis, pneumonitis, and bronchi-
ectasis. Persistent respiratory wheeze may suggest the diagnosis. The principal CT
scan findings in airway TB are circumferential wall thickening and luminal narrow-
ing, which involves a long segment of the bronchi. In active diseases, the airways
are narrowed irregularly in their lumen and have thick walls whereas, in fibrotic dis-
ease, the airways are narrowed smoothly and have thin walls. The left main bron-
chus is involved more frequently in fibrotic disease; whereas, both main bronchi are
equally involved in active disease.30
Extrapulmonary Tuberculosis
Diagnosis of extra-pulmonary TB is often difficult. Although positive chest radiographic
findings or a positive tuberculin skin test supports the diagnosis, negative results do not
exclude extra-pulmonary TB.4
Musculoskeletal Tuberculosis
107
The frontal and parietal lobes are commonly the most affected regions. At CT, tubercu-
lomas appear as rounded or ovulated masses with low or high attenuation and with var-
ying degree of edema. Calcification is uncommon. They demonstrate homogenous or
ring enhancement and have irregular walls of varying thickness.4 If the caseous core of
a tuberculoma liquefies, a TB abscess results. These patients with TB abscess tend to
be clinically worse overall than those with correspondingly sized tuberculoma. Clinical
sequelae of CNS tuberculomas and abscesses primarily include seizures and corre-
lates of increased intracranial pressure. Tuberculous abscess is a rare complication of
CNS TB in the general population, which is seen in four percent to eight percent of pa-
tients with CNS TB but without HIV infection. In a study done by Whiteman et al in
1995, the incidence of CNS TB increases in patients infected with HIV, reaching up to
20%. In contrast to the solid caseation observed in tuberculoma with few tubercle bacil-
li, the center of the abscess is semiliquid pus teeming with tubercle bacilli. The wall of a
tuberculous abscess lacks the giant cell epithelioid reaction of a TB granuloma. Further
distinguishing characteristics of abscesses include size and multiplicity; they tend to be
larger than tuberculomas and have a more accelerated clinical course. Tuberculomas
are often multiple, while abscesses are usually solitary.31
3. Spinal Tuberculous Meningitis
The MR imaging features of spinal TB meningitis consist of CSF loculation and oblitera-
tion of the spinal subarachnoid space, with loss of outline of the spinal cord in the cervi-
cothoracic spine, and matting of the nerve roots in the lumbar region. Contrast-
enhanced imaging reveals nodular, thick, linear, intradural enhancement, which can
completely fill the subarachnoid space.23 This could sometimes give a normal appear-
ance on an enhanced, non-contrast MR image; it emphasizes the importance of utiliz-
ing contrast when there is a clinical question of possible spinal infection.23
Head and Neck Tuberculosis
Tuberculosis in the head and neck represents about 15% of cases of extra-pulmonary
TB. The most common location is within neck nodes, often manifesting as bilateral,
painless cervical lymphadenitis, also known as scrofula. The nodes involved are initially
homogeneous but later undergo central necrosis. These nodes may be difficult to differ-
entiate from the necrotic nodes seen in metastatic head and neck squamous cell carci-
nomas. Nodal calcification often develops late in TB, which helps differentiate TB nodes
from malignancy.23
Tuberculous otomastoiditis can result from hematogenous spread or from direct exten-
sion from the upper respiratory tract. CT demonstrates soft-tissue attenuation in the
tympanic cavity in the early stages and destruction of middle ear structures in the later
stages. Associated retroauricular or epidural abscess may also be seen. Ocular TB re-
sults from hematogenous spread and can involve any part of the eye. Chorioretinitis
and uveitis are the most common manifestations.22
Abdominal Tuberculosis
Lymphadenopathy is the most common radiographic manifestation of abdominal TB,
which is seen in 55% to 66% of patients.23 Mesenteric, omental, and peripancreatic
108
lymph nodes are the most commonly involved. The nodes are usually large and multi-
ple and demonstrate peripheral enhancement with central areas of low attenuation at
contrast-enhanced CT.22
Peritonitis is the most common clinical manifestation of abdominal TB, affecting one-
third of all patients. The condition is subdivided into three main types: wet, fibrotic, and
dry. The wet type, which is characterized by a large amount of free or loculated viscous
fluid—which at CT is usually slightly hyperattenuating relative to water due to its high
protein and cellular content—is seen in most patients. The fibrotic-fixed type and the
dry or plastic type are less common. Fibrotic type is characterized by large omental and
mesenteric cake-like masses with matting of bowel loops. At CT, it manifests as mott-
led, low-attenuation masses with nodular soft-tissue thickening. Omental thickening and
caking can also be seen at ultrasound. Dry type peritonitis is seen in ten percent of cas-
es and is characterized by mesenteric thickening, fibrous adhesions, and caseous nod-
ules. Its imaging manifestations are highly suggestive of, but not specific for, TB. 23
Ileocecal involvement is seen in 80% to 90% of patients with abdominal
TB.1.Thickening of the valve lips or wide gaping of the valve with narrowing of the termi-
nal ileum (Fleischner sign) has been described as a characteristic of TB. At CT, one
half of patients with gastrointestinal TB shows circumferential thickening of the cecum
and terminal ileum, enlargement of the ileocecal valve and mesenteric lymphadenopa-
thy.4 The differential diagnosis for ileocecal TB includes amebiasis, Crohn‘s disease,
and primary cecal malignancy. Involvement of the esophagus, stomach, and proximal
small bowel is rare. Esophageal TB is usually due to extrinsic compression at the level
of the carina from lymphadenopathies.23
Hepatosplenic TB generally manifests in a micronodular (miliary) or macronodular
(tuberculoma) form. On CT scans, numerable tiny, low attenuation foci may be seen.
The macronodular form is rare. The spleen is probably always seeded during the initial
lymphohematogenous spread. Only rarely are the tubercles which are numerous
enough and large enough to undergo cassation and calcify.5
Adrenal involvement in TB is rare. It may manifest as unilateral or bilateral adrenal
masses with central areas of necrosis. Enlarged adrenal glands are also seen in pa-
tients with recent or concurrent renal tuberculous infection. Adrenal atrophy with calcifi-
cation may be seen in patients with healed, prior TB, and these patients may present
with Addison disease.22
Tuberculosis affecting the pancreas is rare. Its occurrence may pose a diagnostic prob-
lem when differentiating it from carcinoma of the pancreas. Most patients have constitu-
tional symptoms and vague abdominal pain. Fine needle aspiration should be consid-
ered in young patients presenting with a cystic pancreatic mass, especially, in those
with constitutional symptoms living in endemic regions.32
Renal Tuberculosis
Tuberculosis may involve the genitourinary tract as a secondary site following hematog-
enous dissemination from the lungs.22 The earliest radiographic abnormality is a ―moth-
109
eaten‖ calyx due to erosion. This finding is followed by papillary necrosis. Poor renal
function, dilatation of the pelvocalyceal system due to a stricture of the ureteopelvic
junction, or destructive dilatation or localized hydrocalycosis related to an infundibular
stricture may be seen. Cavitations within the renal parenchyma may be detected as
irregular pool of contrast material.4 Ureteral involvement is seen in 50% of patients with
genitourinary TB. CT may demonstrate thickening of the ureteral wall and inflammatory
changes of the periureteral. As the disease progresses, urography demonstrates short-
or long-segment ulceration of the ureter. Chronic fibrotic strictures result in a beaded or
corkscrew appearance. Chronic thickening of the ureteral wall may also cause fore-
shortening (pipe-stem ureter). The most common finding in TB cystitis is reduced ca-
pacity of the bladder.22
Genital Tuberculosis
Genital TB involves, almost always, the fallopian tubes in women (94% of cases), usu-
ally causing bilateral salpingitis. Findings at hysterosalpingography are always abnor-
mal, with obstruction and multiple constrictions of the fallopian tubes and endometrial
adhesions or deformity of the cavity. Male involvement is confined to the seminal vesi-
cles or prostate gland, with occasional calcification (10% of cases). Contrast-enhanced
CT shows hypoattenuating prostatic lesions, which likely represent foci of caseous ne-
crosis and inflammation. Nontuberculous pyogenic prostatic abscesses have a similar
CT appearance; its spread is hematogenous and self-limiting. The testes and epididym-
ides are rarely involved. US shows focal or diffuse areas of decreased echogenicity;
however, these findings are very nonspecific.22,23
Cardiac Tuberculosis
Tuberculosis involving the heart is rare, accounting for only 0.5% of cases of extrap-
ulmonary TB. The main presenting finding is pericardial involvement, particularly in im-
munocompromised patients. Myocardial involvement is seen less often. The primary
sign of tuberculous pericarditis is pericardial thickening of more than 3 mm as seen on
CT. CT demonstrates a thickened, irregular pericardium, frequently, with associated
mediastinal lymphadenopathy. Most patients have distention of the inferior vena cava
to a diameter greater than 3 cm; pleural effusions, typically bilateral; and deformities of
the intraventricular septum. Less than 20% have pericardial effusions or develop local-
ized pericardial calcifications.23 A historical cohort study (2000 to 2009) of 179 patients
(0 to 19 years old) with an initial presentation of either pleural effusion or pericardial
effusion was done at the Philippine Heart Center. Ninety-one patients (51%) presented
initially with pleural effusion while 88 patients (49%), with pericardial effusion. Those
patients who initially presented with pericardial effusion are highly associated with ac-
quired heart disease (74%) then TB (45%).30
110
REFERENCE:
1. Balthazar EJ, Gordon R, Hulrick D. Ileocecal Tuberculosis: CT and Radiologic Eval-
uation. Am J Roentgenol. 1989;21:449-503.
2. Starke JR, Taylor-Watts KT. Tuberculosis in the pediatric population of Houston,
Texas. Pediatrics 1989;84:28-35.
3. Leung AN, Muller NL, Pineda PR, Fitzgerald JM. Primary tuberculosis in childhood:
Radiographic manifestations. Radiology 1992;182:87-91.
4. Bass JB, Fares LS, Hopewell PC, Jacobs RF, Snider JR DE. Diagnostic Standards
and Classification of Tuberculosis. Am Rev Respir Dis. 1990;142: 725.
5. Davidson PT. Drug Resistance and Selection. Tuberculosis. Am Rev Respir Dis.
1987;133:255-256.
6. Engin G, Acunas B, Acunas G, Tucani M. Imaging of Extrapulmonary Tuberculosis.
Radiographics. 2000;20:471-488.
7. Feign RD, Cherry JD. Textbook of Pediatrics Infection Disease. 6th ed. Philadelph-
ia:Saunders;2009.
8. Jacobs R, Srtarke J. Tuberculosis in Children. Medical Clinical of North America.
1993;77:1335-1405.
9. Kendig EL. Pulmonary and Pleural TB. Seminars in Pediatric Infectious Disease.
1993;4:214-249.
10. Moon MS. Tuberculosis of the Spine. Spine. 1997;22:1791-1797.
11. Resnick D. Bone and Joint Imaging. 2nd ed. Philadelphia, Pa: Saunders; 1996. 684-
716 p.
12. Sahn SA, Reff TA. Miliary Tuberculosis: The American Journal of Medicine.
1974;58:495.
13. Singson-Perlas P. Phil. Surgical Journal. 1995 July-December;28:47-52.
14. Yao DC, Sartoooris DJ. Musculoskeletal Tuberculosis. Radiol Clin North Am.
1995;33:679-689.
15. Summary Consensus in the Clinico-Radiologic forum on TB. SLMC. September 14,
2005
16. Woodring JH, Vandiviere HM, Fried AM, Dillon ML, Williams TD, Melvin IG. Update:
The radiographic features of pulmonary tuberculosis. AJR Am J Roentgenol. 1986
mar;146(3):497-506.
17. Smuts NA, Beyers N, Gie RP, Schaf HS, Talent JM, et al. Value of the lateral chest
radiograph in tuberculosis in children. Pediatr Radiol. 1994;24(7):478-80.
111
18. Dela Cruz BD (MD), De Leon NA (MD), Policarpio MT (MD), Reyes A (MD), De
Leon A (MD), Bautista MS (MD), De Guia TS (MD). The Agreement between the
Chest CT scan and Chest x-ray Findings Among Tuberculin Positive Filipino Chil-
dren- A Prospective Study. Chest. 2009 suppl vol136 pp 405-a
19. Kim WS, Moon WK, Kim IO, et al. Pulmonary tuberculosis in children: Evaluation
with CT. AJR. 1997 168: 105-109.
20. Leung, AN, Muller NL, Pineda PR, FitzGerald JM. Primary Tuberculosis in Child-
hood: Radiographic Manifestations. Radiology. 1992;182(1)87-91.
21. Donald PR, Ball JB, Burger PJ. Bacteriologically confirmed pulmonary tuberculosis
in childhood. Clinical and radiological features. Afr Med J. 1985 Apr;67(15):588-90.
22. Philippine Pediatric Society. Clinical Practice Guidelines for Childhood Tuberculo-
sis. 2008.
23. Paguia MR, Laya BF, de Jesus JM, Piedad RO. Accuracy of Established CT Scan
Findings in the Diagnosis of Cranial Tuberculosis. SLMC. 2008.
24. Harisinghani MG, McLoud TC, Shepard JO. Tuberculosis from Head to Toe. Radi-
ographics. 2000 March;20:449-470.
25. Burrill J, Williams CJ, Bain G. Tuberculosis: A Radiologic Review. Radiographics.
2007 September;27:1255-1273.
26. Monroc M, Marguet C, Dacher JN, Le Guillou A, Eurin D, Le Dosseur P. Evolution
and radiologic surveillance on community-acquired pneumonia in children. Arch Pe-
diatr. 1998;5 Suppl 1:37s-44s.
27. Taussig LM, Landau LI. Pediatric Respiratory Medicine. Mosby. c1999. 713 p.
28. Dimaculangan ME, Beltran GL. Computed Tomography Patterns of Pulmonary Tu-
berculosis in Pediatric Population. Makati Medical Center Proceedings. 2003. 40-43
p.
29. Krysl J, Korzeniewska-Kosela M, Muller NL, FitzGerald JM. Radiologic features of
pulmonary tuberculosis: an assessment of 188 cases. Can Assoc Radiol J.
1994;45:101–107.
30. Yeon JJ, Kyung SL. Pulmonary Tuberculosis: Up-to- Date Imaging and Manage-
ment. AJR. 12008 September; 91.
31. Behrman RE, Kliegman RM, Arvin AM. Textbook of Pediatrics. 18th ed. Philadelph-
ia: Saunders; 2007.
32.
33. Damian MS (MD), De leon NA (MD), Bautista MS (MD), De Guia TS (MD), Ayuyao
FG (MD). Risk Factors of Pleuro-pericardial Effusion among Pediatric Patients
112
initially Presenting with Pleural effusion and Pericardial Effusion alone at
Philippine Heart Center. 2009. (Presented at the Asia Pacific Society of Respirology
Convention, Seoul, South Korea, November 2009)
34. Whiteman M, Espinoza L, Post MJ , Bell MD, and Falcone S. Central nervous sys-
tem tuberculosis in HIV-infected patients: clinical and radiographic findings. AJNR
Am. J. Neuroradiol. 1995 Jun;16:1319-1327.
35. Lo SF, Ahchong AK, Tang And CN, Yip AWC. Pancreatic tuberculosis: case reports
and review of the literature. J.R.Coll. Surg. Edinb. 1998 February;43:65-69.
36. Gwee A, Pantazidou A, Ritz N, et al. To x-ray or not to x-ray? Screening asympto-
matic children for pulmonary TB: a retrospective audit. Arch Dis Child 2013; 98:401
37. van den Bos F, Terken M, Ypma et al. Tuberculous meningitis and military tubercu-
losis in young children. Trop Med Intl Health 2004; 9:309
38. Ip MS, So SY, Lam WK, Mok CK. Endobronchial tuberculosis revisited. Chest 1986;
89: 727-730
39. Smith LS, Schillaci RF, Sarlin RF. Endobronchial tuberculosis: Serial fiberoptic
bronchoscopy and natural history. Chest 1987; 91: 649-647.
40. Laya BF, Concepcion NDP, dela Eva RC, Mendoza AR, Sanchez MO, Goco NB,
Buenaflor MTS. Computed tomography with Multiplanar Reformation and 3D-
Volume Rendering Technique in Correlation with Fiberoptic Tracheobronchoscopy
for Thoracic Evaluation of Children with Primary Progressive Tuberculosis and Tra-
cheobronchial Involvement. St. Luke‘s J Med 2011; 07: 17-19.
41. Sanchez MO, Mendoza AR, Laya BF. Primary Progressive Tuberculosis in Chil-
dren: Radiographic Patterns in Correlation with Clinical Presentation and Microbio-
logic Studies. St. Luke‘s J Med 2011; 07: 59-65
113
10
Diagnostic Tests/
Laboratory Findings
Ma. Liza Antoinette M Gonzales, MD
Maria Rosario Z Capeding, MD
Carmina A Delos Reyes, MD
OUTLINE
1 Diagnostic Mycobacteriology
3 Histologic Examination
4 Serological Tests
114
DIAGNOSTIC MYCOBACTERIOLOGY
Detection of acid fast bacilli (AFB) in microscopically examined stained smears is the
first bacteriologic evidence of the presence of mycobacteria in clinical specimens using
either Ziehl-Neelsen or Kinyoun carbol fuchsin stains. AFB smear microscopy is the
easiest, least expensive and most rapid procedure where the results can be obtained
within an hour from obtaining preliminary information. While it provides the physician
with a presumptive diagnosis of active tuberculosis (TB), it also gives a quantitative es-
timation of the number of bacilli on the smear and implies the degree of infectiousness
of the patient.1,2 A positive AFB smear may represent either M. tuberculosis (MTB) or
some non-tuberculous mycobacterium; therefore, it is not sufficient evidence for bacte-
riologic diagnosis.
The overall sensitivity of AFB smear is variable, ranging from 20 to 80%; however, its
specificity is high (≥97%).3 Sensitivity of smear microscopy is generally higher in older
children (more than 5 years of age) and adolescents, and in children of all ages with
severe disease4, however, is significantly reduced in patients with extrapulmonary TB
and in HIV-infected TB patients. Several factors can influence the sensitivity of AFB
smear: specimen type, number of obtained specimens, and staining method. It is esti-
mated that the lowest concentration of organisms that can be detected by microscopic
examination is 104 AFB per ml of sputum.2,3
Fluorescence microscopy (FM), in which auramine-based staining causes the acid-fast
bacilli to fluoresce against a dark background, are increasingly used in many parts of
the country. A comprehensive systematic review of 45 studies using culture as the gold
standard indicates that FM is 10% more sensitive than conventional microscopy and
examination of fluorochrome-stained smears takes less time.5 It allows a much larger
area of the smear to be seen resulting to a more rapid examination of the specimen up
to four times faster and easier counting of the bacilli. Its specificity is comparable to
conventional microscopy.6
Fluorescent light-emitting diode (LED) microscopy for diagnosis of tuberculosis provide
a relatively inexpensive light source for fluorescence microscopy. Evidence show that
the diagnostic accuracy of LED microscopy is comparable to that of conventional fluo-
rescence microscopy and it surpasses that of conventional Ziehl-Neelsen microscopy
by an average of 10%. It showed 84% sensitivity and 98% specificity against culture as
the reference standard. This is recommended for use in high-volume laboratories.7
115
Mycobacterial culture has a low sensitivity in children, where only about 10 to 50% of
tuberculous cases are culture-proven10,11 owing to the paucibacillary nature of the dis-
ease and difficulty of specimen collection.12 Fortunately, for most children with pulmo-
nary TB, culture confirmation is not necessary if the epidemiologic, tuberculin skin test
and roentgenographic information are compatible with the disease.13 However, cultures
should be obtained from the child if the source case is unknown or has a drug-resistant
organism, or if the child is immunocompromised or has extrapulmonaryTB.
Traditional culture methods utilize solid culture media that include the egg-potato-based
media (e.g. Lowenstein-Jensen, Ogawa) and the agar-based media (e.g. Middlebrook
7H10, Middlebrook 7H11 and Mitchison‘s selective 7H11). Incubation of inoculated me-
dia in 5% to 10% carbon dioxide atmosphere enhances the number of positive isolates
and the number of culturable colonies. It is limited by suboptimal sensitivity, slow turna-
round time and the low bacteriologic yields in children with active TB. The isolation of
organisms often require four-to-six weeks and another two-to-four weeks for suscepti-
bility testing.1,2,3,4,14
Mycobacterial culture with the use of commercial broth-based liquid culture media are
more rapid and sensitive than solid culture techniques in detecting TB bacilli. It has re-
duced the average detection time of broth smear-positive and smear-negative samples
by nearly 50%. Liquid culture systems provide results in 7-14 days (up to 42 days for a
negative result) significantly faster than solid culture techniques. 15 Liquid systems are
more prone to contamination and its manipulation of large volumes of infectious materi-
al mandates appropriate biosafety measures. Both culture systems have specificities of
nearly 100% but have lower sensitivity.1,2,3,11,13,15
The growth of mycobacteria in liquid media, regardless of its type, requires 5% to 10%
carbon dioxide (CO2), which is already provided in the culture vials or is added accord-
ing to manufacturer‘s instructions. The most commonly used are Septi-Check AFB and
Mycobacteria Growth Indicator Tube (MGIT). The liquid culture system, such as BacT/
Alert, also has a feature that monitors the continuous growth of mycobacteria. In this
system, growth is detected by CO2 production through the use of colorimetric CO2 sen-
sor in each bottle and through BACTEC 9000 MB by monitoring O2consumption with
the use of a fluorescent sensor. Both manual and automated systems are availa-
ble.1,2,3,4,13,16,17
Drug-susceptibility testing (DST) is performed in two methods: phenotypic and genotyp-
ic. Phenotypic methods involve culturing M. tuberculosis in the presence of anti-TB
agents to detect growth (indicates resistance) or inhibition of growth (indicates suscepti-
bility). Phenotypic DST methods are performed as direct or indirect in solid or liquid me-
dia. Liquid culture systems for DST reduce the turn-around time to as short as 10 days,
compared with the 28-42 days needed for conventional solid media. Genotypic meth-
ods target specific molecular mutations associated with resistance against anti-TB
agents.18
116
Collection and Transport of Specimens for Demonstration of Tubercle
Bacilli
Mycobacterial infections occur in almost any site in the body; therefore, a variety of clin-
ical samples are appropriate specimens for laboratory analysis. In children, specimens
include gastric aspirate, sputum, cerebrospinal fluid, pleural fluid, bronchial washings,
urine, pus, bone marrow, and biopsy or resected tissue. Proper collection of specimens
is essential for the diagnosis in children because of the fewer tubercle bacilli present.
Specimens must be collected in sterile, leak proof, disposable and appropriately la-
belled containers and must be stored under conditions that inhibit the growth of con-
taminants, since most specimens will contain bacteria other than mycobacteria. Certain
specimen collection procedures are optimal for MTB. All specimens from non-sterile
sites must be decontaminated before culture.1,2,3,4
Tissue/Lymph Node
Tissue or lymph node specimen can be collected aseptically during surgery or biopsy and
placed in a sterile, leak-proof container such as a 50mL conical tube. Two to 3mL of sterile sa-
line must be added to the tissue for transport. The tissue specimen must be transported as soon
as possible at room temperature. If transport is delayed for more than one hour, the tissue must
be stored and refrigerated. Always submit as much tissue as possible.
HISTOLOGIC EXAMINATION
Biopsy of a node, skin lesion, bone or pleura may permit a quick presumptive diagno-
sis, as well as yield specimens for bacteriologic examination. The classic pathologic
lesion in TB consists of caseating granulomas, which may demonstrate AFB. The path-
ologic events in the initial tuberculous infection will depend upon the balance among
the mycobacterial antigen load, cell-mediated immunity which enhances intracellular
killing and tissue hypersensitivity which promotes extracellular killing. 3,19 When the anti-
gen load is small and the degree of tissue sensitivity is high, granuloma formation re-
sults from the organization of lymphocytes, macrophages, Langerhans giant cells, and
fibroblasts. When both antigen load and the degree of hypersensitivity are high, granu-
loma formation is less organized.
SEROLOGICAL TESTS
Immunodiagnostic methods based on the detection of antigen or antibodies to TB bacilli
have not come into widespread clinical use unlike with other infectious diseases. De-
spite hundreds of studies published, serology has found little place in the routine diag-
nosis of TB in adults and children because both sensitivity and specificity of the various
tests were highly variable.20,21 Results have shown that for all commercial serological
tests, sensitivity ranged from 0-100% for diagnosing both pulmonary and extrapulmo-
119
nary TB while specificity ranged from 31-100% for PTB and from 59-100% for extrap-
ulmonary TB.20 None of these studies were conducted in children. Due to the incon-
sistent and imprecise findings for currently available serological tests, WHO strongly
recommends that commercial serodiagnostic or serological tests should not be used for
the diagnosis of pulmonary and extrapulmonary TB.21
125
collection while white blood cells are still viable. The test is expensive, and there is pau-
city of data on the use of IGRAs particularly in children younger than 5 years, persons
with recent exposure to MTB, and immunocompromised patients. 88-90
One systematic review showed that IGRAs based on RD1-specific antigens, ESAT-6 or
CFP-10, correlate better with intensity of exposure and are more likely to detect LTBI
accurately compared with TST/PPD-based assays. IGRAs are also more likely to be
independent from BCG vaccination and HIV status.15 In recent years, IGRAs have been
widely endorsed in high-income countries for diagnosing LTBI.78
However, due to the lower sensitivity of IGRAs in low and middle-income countries,
there are no current guidelines for their use in these high-burden settings to screen in-
dividuals with repeated TB exposure such as healthcare workers and contacts of active
TB cases and for identifying individuals at risk of developing active TB. 89
Use of IGRAs in health care worker (HCW) screening
In the systematic review conducted by WHO, evidence for use of IGRAs for LTBI
screening of HCW in low and middle-income countries was limited and based on low
quality data from two cross-sectional studies. Results showed that positivity of both
IGRA and TST were high at 40-66%, although IGRA positivity was slightly lower than
the TST. Data are lacking on serial IGRA testing and on the predictive value of IGRAs
in HCWs for high-burden TB and/or HIV settings. 89
Use of IGRAs in contact screening and outbreak investigations
IGRAs have not shown additional benefit over the TST in screening for LTBI in adult
and pediatric contacts, or in outbreak investigations. In the systematic review, 16 low
quality studies evaluated IGRAs in contact screening and outbreak investigations, 75 %
of contact studies included children. Majority of studies were cross-sectional and the
heterogeneity in study designs and outcomes assessed made it impossible to pool the
data. Despite the limitations, majority of the studies showed comparable LTBI preva-
lence by TST or IGRAs in contacts. The most commonly observed discordance was of
the TST-positive/IGRA negative type. Both IGRAs and TST showed positive associa-
tions with higher levels of exposure in cross-sectional studies but the strength of asso-
ciation varied across studies. Concordance between TST and IGRAs was variable.
Predictive value of IGRAs
Neither IGRAs nor the TST should be used for the identification of individuals at risk of
developing active TB. Results are based on three very low quality studies that provided
the incidence rate ratios (IRR) of TB stratified by IGRA as well as TST status at base-
line. Association with subsequent incident TB in those who tested positive compared to
test negatives appeared higher for IGRA than for TST but this was not statistically sig-
nificant. Both the TST and IGRA show positive associations between exposure gradient
and test results but there was variability in the strength of the associations across popu-
lations, irrespective of BCG vaccination. There was no statistically significant increase
in the incidence of TB in IGRA-positives compared to IGRA-negatives and majority of
individuals (95%) with a positive IGRA result did not progress to active TB on follow-up.
Both IGRAs and the TST appeared to have a modest predictive value and did not help
identify those who are highest risk of progression to TB disease.
126
Use of IGRAS for Diagnosis of Active TB
IGRAs were developed to replace the TST in the diagnosis of latent TB infection. It was
not intended for detecting cases of active TB. IGRAs, just like the TST, cannot distin-
guish LTBI from active TB. Because of this, they are expected to have poor specificity
for active TB in high-burden settings due to a high background prevalence of LTBI. The
low performance of IGRAs in these settings was partly attributed to anergy due to ad-
vanced disease, malnutrition, and HIV-associated immune suppression. Laboratory
procedures and infrastructure may have also contributed to the low performance of
IGRAs.
The systematic review89 showed that for T-SPOT, the pooled sensitivity was 83% (95%
CI 70%-91%) and the pooled specificity was 58% (95% CI 42%-73%). For QFT-GIT
the pooled sensitivity was 73% (95% CI 61%-82%) and pooled specificity was 49%
(95% CI 40%-58%). On the average about 25% of patients with culture-confirmed ac-
tive TB could be expected to be IGRA negative in low and middle-income countries,
and this could lead to serious patient consequences. However, it is important to note
that the quality of evidence for the use of IGRAs in the diagnosis of active TB was low.
There was no evidence to show that IGRAs were more sensitive than the TST for diag-
nosing active TB, and IGRAs have no added value beyond conventional microbiologic
tests for the diagnosis of active TB.
Use of IGRAs in children
Results from two small studies with low methodological quality showed that there was
no difference in the performance of TST and IGRAs for diagnosing LTBI and for deter-
mining incidence of active TB in children. 89
Use of IGRAs in HIV-infected individuals
Studies on use of IGRAs in HIV-infected individuals had small sample size and were of
very low quality, hence results should be interpreted cautiously. In persons with active
TB (used as a surrogate reference for LTBI), pooled sensitivity estimates were higher
for T-SPOT (72%, 95% CI 62%-81%) than for QFT-GIT (61%, 95% CI 41%-75%). Oth-
er studies showed that IGRAs have a poor positive predictive value but a high negative
predictive value for active TB, implying that that no HIV-infected person with a negative
IGRA result developed culture positive TB. Generally, the sensitivity of IGRAs was low-
er at 60-70% among HIV-positive individuals, suggesting that nearly 33% of HIV-
positive patients with active TB would be IGRA-negative. The IGRA was no more con-
sistently sensitive that the TST and the impact of advanced immunosuppression on
IGRA validity remains unclear.
Overall, the studies show that there is insufficient data and low-quality evidence on the
performance of IGRAs in low and middle-income countries, typically those with a high
TB and/or HIV burden. 89 Both the IGRAs and the TST cannot accurately diagnose
active TB nor can they predict the risk of infected individuals developing active TB.
Moreover, IGRAs are more costly and technically complex to do than the TST and the
increased cost of the former given the comparable performance of the TST, does not
justify the replacement of the TST by IGRAs as a public health intervention in resource-
limited settings.
127
Based on existing evidence, the WHO issued policy statements in 2011 that IGRAs
should not be used for the diagnosis of pulmonary or extra-pulmonary TB in low and
middle-income countries, nor for the diagnostic work-up of adults and children suspect-
ed of active TB, irrespective of HIV status. 89 IGRAs should not replace the TST for the
diagnosis of latent TB infection in children and should not be used for the screening of
latent TB infection in adult and pediatric contacts (including HIV positive individuals),
nor in outbreak investigations. IGRAs should not be used in HCW screening programs.
Neither IGRAs nor the TST should be used for the identification of individuals at risk of
developing active TB. In children, there may be additional harm associated with blood
collection and issues such as acceptability and cost have not been adequately ad-
dressed in previous studies.
128
REFERENCES:
1. Stop TB Partnership and World Health Organization (WHO). New Laboratory Diag-
nostic Tools for TB control. Geneva: World Health Organization; 2008.
2. Gooring RV, Rocknell HM, Zuckerman M, et al. Medical Microbiology. 4th ed. Else-
vier; 2008.
3. Forbes BA, Sahm DF, Weisfeild AS. Bailey & Scott‘s Diagnostic Microbiology. 12th
Ed. Mosby Elsevier; 2007.
4. World Health Organization. Guidance on national tuberculosis programmes on the
management of childhood tuberculosis 2nd ed. Geneva: World Health Organization
2014. WHO/HTM/TB/2014.03
5. Steingart KR. Fluorescence versus conventional sputum smear microscopy for tu-
berculosis: a systematic review. Lancet Infect Dis. 2006; 6(9): 570-81.
6. TB CARE I. International Standards for Tuberculosis Care, Edition 3.TB CARE I,
The Hague, 2014.
7. World Health Organization. Fluorescent light-emitting diode (LED) microscopy for
diagnosis of tuberculosis 2011. WHO/HTM/TB/2011.8
8. Heifets LB, Good RC. Current Laboratory Methods for the Diagnosis of Tuberculo-
sis. In Tuberculosis: Pathogenesis, Protection and Control. Bloom BR, editor.
Washington DC: American Society for Microbiology; 1994.
9. Marais BJ, Pai M. New Approaches and Emerging Technologies in the Diagnosis
of Childhood Tuberculosis. Paediatric Respiratory Reviews. 2007;(8): 124-133.
10. Starke JR, Taylor-Watts KT. Tuberculosis in the Pediatric Population of Houston,
Texas. Pediatrics. 1989: 84(1): 28-35.
11. Perez-Velez CM, Marais BJ. Tuberculosis in children. New England Journal of Med-
icine 2012; 367(4):348-361.
12. Marais BJ, Schaaf HS, Donald PR. Pediatric TB: issues related to current and fu-
ture treatment options. Future Microbiol. 2009;4(6): 661-675
13. Starke JR. Modern Approach to the Diagnosis and Treatment of Tuberculosis in
Children. Pediatr Clin N Am. 1988; 53:441.
14. Clinical and Laboratory Standards Institute (CLSI). Laboratory Detection and Identi-
fication of Mycobacteria: Approved Guideline. Wayne PA, USA; 2009. M48-A; 28
(17).
15. Dinnes J, Deeks J, Kunst H, et al. A Systemic Review of Rapid Diagnostic Tests for
the Detection of Tuberculosis Infection. Health Technology Assessment. 2007;11
(3): 1-178.
129
16. Yanina Balabanova, Francis Drobniewski, Vladyslav Nikolayevsky et al. An Inte-
grated Approach to Rapid Diagnosis of Tuberculosis and Multidrug Resistance Us-
ing Liquid Culture and Molecular Methods in Russia. PLoS ONE 2009; 4 (9):
e7129. doi: 10.1371/journal.pone.0007129
17. Claudio Piersimon, Claudio Scarparo, Annapaola Callegaro et al. Comparison of
MB/BacT Alert 3D System with Radiometric BACTEC System and Lowenstein-
Jensen Medium for Recovery and Identification of Mycobacterium from Clinical
Specimens: a Multicenter Study. J Clin Microbiol. 2001 Feb; 39(2): 651–657
131
41. Da Costa Lima D, Lapa Montenegro LM, de Albuquerque Montenegro R, Cabral
MML, Santos Lima A, et al. Performance of nested PCR in the specific detection of
Mycobacterium tuberculosis complex in blood samples of pediatric patients. J
BrasPneumol.2009;35(7):690-697.
42. Gomez-Pastrana D, Torronteras R, Caro P, Anguita ML, Lopez Barrio AM, Andres
A, et al. Diagnosis of tuberculosis in children using a polymerase chain reaction.
Pediatr Pulmonol. 1999;28: 344-351.
43. Gomez-Pastrana D. Tuberculosis in children - is PCR the diagnostic solution? Clin
Microbiol Infect. 2002;8:541-544.
44. Montenegro SH, Gilman RH, Sheen P, Cama R, Caviedes L, et al. Improved detec-
tion of Mycobacterium tuberculosis in Peruvian children by use of a hemi-nested
IS6110 polymerase chain reaction assay. Clin Infect Dis. 2003; 36(1):16-23.
45. Khan EA, Starke JR. Diagnosis of tuberculosis in children: Increased need for bet-
ter methods. Emerging Inf Dis. 1995;1(4):115-123.
46. Flores LL, Pai M, Colford JM, Riley LW. In-house nucleic acid amplification tests for
the detection of Mycobacterium tuberculosis in sputum specimens: met-analysis
and meta-regression. BMC Microbiology. 2005;5:55. Available from: http://
www.biomedcentral.com/1471-2180/5/55.
47. Greco S, Girardi E, Navarra A, Saltini C. Current evidence on diagnostic accuracy
of commercially based nucleic acid amplification tests for the diagnosis of
pulmonary tuberculosis. Thorax 2006; 61: 783-790.
48. Greco S, Rulli M, Girardi E, Piersimoni C, Saltini C. Diagnostic accuracy of in-
house PCR for pulmonary tuberculosis in smear-positive patients: meta-analysis
and metaregression. J ClinMicrobiol. 2009;47(3):569-576.
49. Ling DI, Flores LL, Riley LW, Pai M. Commercial nucleic acid amplification tests for
diagnosis of pulmonary tuberculosis in respiratory specimens: meta-analysis and
meta-regression. PloS ONE. 2008;3(2):e1536.doi: 10.1371/ journal.pone.0001536.
50. Sarmiento OL, Weigle K, Alexander J, Weber DJ, Miller WC. Assessment by meta-
analysis of PCR for diagnosis of smear-negative pulmonary tuberculosis. J ClinMi-
crobiol. 2003;41(7):3233-3240.
51. Pai M, Flores L, Hubbard A, Riley L, Colford JM. Nucleic acid amplification tests in
the diagnosis of tuberculous pleuritis: a systematic review and meta-analysis. BMC
Infect Dis. 2004;4:6. available from: http://www.biomedcentral.com/1471-2334/4/6.
52. Pai M, Flores L, Pai M, Hubbard A, Riley L, Colford JM. Diagnostic accuracy of nu-
cleic acid amplification tests for tuberculous meningitis: a systematic review and
meta-analysis. Lancet Infect Dis. 2003;3:633-643.
53. Daley P, Thomas S, Pai M. Nucleic acid amplification tests for the diagnosis of tu-
132
berculous lymphadenitis: a systematic review. Int J Tuberc Lung Dis. 2007;11:1166-
1176.
54. Drobniewski F, Nikolayevskyy V, Maxeiner H, Balabanova Y, Casali N, Kontsevaya
I, Ignatyeva O. Rapid diagnostics of tuberculosis and drug resistance in the indus-
trialized world: clinical and public health benefits and barriers to implementation.
BMC Medicine 2013, 11:190. http://www.biomedcentral.com/1741-7015/11/190
55. Morgan M, Kalantri S, Flores L, Pai M: A commercial line probe assay for the rapid
detection of rifampicin resistance in Mycobacterium tuberculosis: a systematic re-
view and meta-analysis. BMC Infect Dis 2005, 5:62;
56. World Health Organization. Molecular line probe assays for rapid screening of pa-
tients at risk of multidrug-resistant tuberculosis (MDRTB).Policy statement. Availa-
ble at: http://www.who.int/tb/laboratory/lpa_policy.pdf. 27 June 2008.
57. Seoudi N, Mitchell SL, Brown TJ, Dashti F, Amin AK, Drobniewski FA: Rapid molec-
ular detection of tuberculosis and rifampicin drug resistance: retrospective analysis
of a national UK molecular service over the last decade. Thorax 2012; 67:361–367.
58. Ling DI, Zwerling AA, Pai M. GenoType MTBDR assays for the diagnosis of multi-
drug resistant tuberculosis: a meta-analysis. Eur Respir J 2008; 32:1165–74.
59. Lin Shou-Yean G, Desmond EP. Molecular diagnosis of tuberculosis and drug re-
sistance. Clin Lab Med 2014; 34: 297–314.
60. Luetkemeyer AF, Kendall MA, Wu X, Lourenço MC, Jentsch U, Swindells S, Qasba
SS, Sanchez J, Havlir DV, Grinsztejn B, Sanne IM, Firnhaber C; Adult AIDS Clinical
Trials Group A5255 Study Team.
61. World Health Organization. Evaluation of two line probe assays for rapid detection
of Mycobacterium tuberculosis, tuberculosis (TB) drug resistance, and non-TB My-
cobacteria in HIV-infected individuals with suspected TB.J Clin Microbiol. 2014
Apr;52(4):1052-9. doi: 10.1128/JCM.02639-13. Epub 2014 Jan 15.
62. Connell TG, Zar HG, Nicol MP. Advances in the Diagnosis of Pulmonary Tuberculo-
sis in HIV-Infected and HIV Uninfected Children. Journal of Infectious Diseases
2011: 204 (Suppl 4): S1151–S1158.
63. Boehme CC, Nabeta P, Hillemann D, Nicol MP, Shenai S, et al. Rapid molecular
detection of tuberculosis and rifampin resistance. N Engl J Med 2010: 363(11):
1005–1015
64. Evans CA. GeneXpert—A Game-Changer for Tuberculosis Control? PLoS Med
2011; 8(7): e1001064. doi:10.1371/journal.pmed.1001064.
65.Lawn SD, Nicol MP. Xpert(R) MTB/RIF assay: development, evaluation and imple-
mentation of a new rapid molecular diagnostic for tuberculosis and rifampicin re-
sistance. Future Microbiol 2011; 6 (9):1067–1082..
133
66. Helb D, Jones M, Story E, Boehme C, Wallace E, et al. (2010) Rapid detection of
Mycobacterium tuberculosis and rifampin resistance by use of on demand, near-
patient technology. J ClinMicrobiol 48(1): 229–237
67. Blakemore R, Story E, Helb D, Kop J, Banada P, et al. (2010) Evaluation of the an-
alytical performance of the Xpert MTB/RIF assay. J ClinMicrobiol 48(7): 2495–2501
68. WHO. Automated real-time nucleic acid amplification technology for rapid and sim-
ultaneous detection of tuberculosis and rifampicin resistance: Xpert MTB/RIF sys-
tem for the diagnosis of pulmonary and extrapulmonary TB in adults and children.
Policy Update. Geneva, World Health Organization, 2013.
69. Detjen AK, DiNardo AR, Leyden J, Steingart KR, Menzies D, Schiller I, N Denduku-
ri, Mandalakas AM. Xpert MTB/RIF assay for the diagnosis of pulmonary tuberculo-
sis in children: a systematic review and meta-analysis. The Lancet Respiratory
Medicine 2015;3 (6) : 451–461.
70. World Health Organization. Rapid implementation of the Xpert MTB/RIF diagnostic
test. Technical and operational ‗how-to‘: practical considerations. Geneva, World
Health Organization 2011 (http://whqlibdoc.who.int/publications/2011/
9789241501569_eng.pdf)
71. World Health Organization. Automated real-time nucleic acid amplification technolo-
gy for rapid and simultaneous detection of tuberculosis and rifampicin resistance:
Xpert MTB/RIF system. Policy statement. Geneva, World Health Organization,
2011 (http://whqlibdoc.who.int/publications/2011/9789241501545_eng.pdf)
72. World Health Organization. The use of the Xpert MTB/RIF® assay for the detection
of pulmonary, extrapulmonary tuberculosis and rifampicin resistance in adults and
children. Geneva; World health Organization, 2013, WHO/HTM/TB/2013.14.
73. Nicol MP, Workman L, Isaacs W, et al. Accuracy of the Xpert MTB/ RIF test for the
diagnosis of pulmonary tuberculosis in children admitted to hospital in Cape Town,
South Africa: a descriptive study. Lancet Infect Dis 2011; 11: 819–24.
74. Zar HJ, Workman L, Isaacs W, et al. Rapid molecular diagnosis of pulmonary tuber-
culosis in children using nasopharyngeal specimens. Clin Infect Dis 2012; 55: 1088
–1095.
75. Sekadde MP, Wobudeya E, Joloba ML, Ssengooba W, KisemboH,Bakeera-Kitaka
S ,Musoke P. Evaluation of the Xpert MTB/RIF test for the diagnosis of childhood
pulmonary tuberculosis in Uganda: a cross-sectional diagnostic study . BMC Infec-
tious Diseases 2013, 13:133
76. Nhu NTQ, Ha DTM, Anh ND, Thu DD, Duong TN, Quang ND, Lan NTN et al. Evalu-
ation of Xpert MTB/RIF and MODS assay for the diagnosis of pediatric tuberculosis.
BMC Infectious Diseases 2013, 13:31.
77. Tortoli E, Russo C, Piersimoni C et al. Clinical validation of Xpert MTB/RIF for the
134
diagnosis of extrapulmonary tuberculosis. Eur. Respir. J. 2012.
doi:10.1183/09031936.00176311
78. Centers for Disease Control and Prevention. Updated Guidelines for Using Interfer-
on Gamma Release Assays to Detect Mycobacterium tuberculosis Infection, United
States. MMWR 2010; 59.
79. Somoskovi, A, Song Q, Mester J, Tanner C, Hale YM, Parsons LM, Salfinger M.
Use of molecular methods to identify the Mycobacterium tuberculosis complex
(MTBC) and other mycobacterial species and to detect rifampicin resistance in
MTBC isolates following growth detection with the BACTEC MGIT 960 system. J
Clin Microbiol. 2003 Jul;41(7):2822-2826.
80. Cellestis. Clinicians Guide to Quantiferon-TB Gold. Available from: http://
www.cellestis.com/IRM/contentAU/gold/clinicians guide.pdf. 2004.
81. Forbes BA, Sahm DF, Weisfeild AS. Bailey & Scott‘s Diagnostic Microbiology. 12th
Ed. Mosby Elsevier; 2007.
82. Pottumarthy S, Morris AJ, Harrison AC, Wells VC. Evaluation of the tuberculin gam-
ma interferon assay: potential to replace the Mantoux skin test. J Clin Microbiol.
1999; 37:3299-32.
83. Fietta A, Meloni F, Cascina A, Morosini M, Marena C, Troupioti P, et al. Compari-
son of a whole-blood interferon-gamma assay and tuberculin skin testing in patients
with active tuberculosis and individuals at high or low risk of Mycobacterium tuber-
culosis infection. Am J Infect Control. 2003; 31:347-353.
84. Oxford Immunotech Limited. T Spot TB. Available from: http://www/
oxfordimmunotec.com/products.htm.2004. Mabtech AB. EliSpot-assay procedure.
URL:http:///.mabtech.com/elispot.asp.2004.
85. Streeton JA, Desem N, Jones SL. Sensitivity and specificity of a gamma interferon
blood test for tuberculosis infection. Int J Tuberc Lung Dis. 1998;2:443-450.
86. Mazurek GH, LoBue PA, Daley CL, Bernardo J, Lardizabal AA, Bishai WR et al.
Comparison of a whole-blood interferon gamma assay with tuberculin skin testing
for detecting latent Mycobacterium tuberculosis infection. Centers for Disease Con-
trol and Prevention. MMWR Recomm Rep. 2003; 52 (RR-w): 15-18..
87. Chapman AL, Munkata M, Wilkinson KA, Pathan AA, Ewer K, Ayles H et al. Rapid
detection of active and latent tuberculosis infection in HIV-positive individuals by
enumeration of Mycobacterium tuberculosis-specific T cells. AIDS 2002; 16:2285-
2293.
88. Barnes PF. Diagnosing latent tuberculosis infection: the 100 year upgrade. Am J
Resp Crit Care Med 2001; 163:807-8.
89. World Health Organization. Use of tuberculosis interferon-gamma release assays
135
(IGRAs) in low and middle-income countries: policy statement. WHO 2011
90. Morris K. WHO recommends against inaccurate tuberculosis tests. Lancet 2011;
377:113-114
136
11
Management
Anna Lisa T Ong-Lim, MD
OUTLINE
1 Aims and Basic Principles of Ther-
apy
2 Anti-Tuberculosis Drugs
3 Recommended Regimens
4 Preventive Therapy
137
AIMS AND BASIC PRINCIPLES OF THERAPY
The drug therapy of tuberculosis aims to achieve several objectives. These include
cure of the patient by rapid elimination of most of the metabolically active and rapidly
replicating bacilli; prevention of death from active TB or its late effects; prevention of
relapse by eliminating slowly and intermittently multiplying bacilli; prevention of drug
resistance by using a combination of drugs; and finally, decrease of TB transmission to
others.1
A thorough knowledge of the microbiology of tuberculosis is necessary to understand
the basis for the treatment strategies currently being utilized. The need for multiple
drugs and the prolonged duration of therapy is explained by the following characteris-
tics of the causative organism 2:
1. bacilli live in several sites within the host, and each site (ex. open cavities, closed
lesions, macrophages) contains organisms with different population sizes, metabol-
ic activities and replication rates;
2. mycobacteria replicate slowly, can remain dormant for prolonged periods, and can
be eradicated only during replication;
3. naturally occurring drug resistant mutants are present within large bacterial popula-
tions even before chemotherapy is started.
In addition, it is thought that within each patient, there are TB populations consisting of
four groups, defined by drug efficacy 3:
1. actively growing bacilli found in open cavities, which are killed by bactericidal drugs;
2. slowly multiplying or non-replicating TB bacilli (located in caseous lesions) that un-
dergo spurts of metabolism and are killed by sterilizing drugs;
3. intracellular bacilli present in the acidic compartments of macrophages or in acidic
lung lesions, for which sterilizing drugs are also effective;
4. TB persisters found in hypoxic microenvironments, which are non-responsive to
most anti-TB drugs.
Controlled clinical trials have thus demonstrated the principles on which treatment rec-
ommendations are based: treatment of disease must contain multiple drugs to which
the organisms are susceptible; drugs must be taken regularly, and drug therapy must
continue for a sufficient length of time.4
Although the therapeutic principles stated above are valid regardless of the age of the
patient, several pharmacologic considerations unique to children need to be considered
in their treatment 5:
1. the pharmacokinetics and pharmacodynamics of various anti-tuberculosis drugs are
different in children;
2. children generally have fewer mycobacterial organisms and are thus less likely to
develop secondary drug resistance (see Chapter 12 Treatment of Drug-Resistant
Tuberculosis);
3. extrapulmonary disease is more common in children; medications used must there-
138
fore penetrate specific body sites and tissues;
4. children tolerate higher doses of anti-tuberculous medications per kilogram of body
weight with fewer side effects;
5. there may be a need to modify medications to a form that children can tolerate,
which can cause problems with stability, bioavailability and compliance.
Present treatment regimens consist of multiple drugs given simultaneously. Since mu-
tant organisms naturally resistant to multiple drugs are extremely rare, this strategy de-
creases the likelihood of selecting out drug-resistant organisms and prevents the emer-
gence of resistance. An initial intensive phase (consisting of three or four drugs to
which the TB bacilli are susceptible) promotes efficient killing of actively dividing organ-
isms and leads to the rapid reduction of large bacillary populations; this provides relief
of symptoms, terminates transmission and prevents the emergence of drug resistance.
A longer continuation phase (using fewer drugs) then follows, which kills slowly or inter-
mittently dividing bacilli, sterilizes lesions and prevents relapse.
In both adults and children, numerous trials have proven the efficacy of short-course, 6-
month chemotherapy for drug-sensitive pulmonary tuberculosis. Efforts to shorten the
duration of therapy to even fewer than 6 months, however, have been associated with
significant relapse rates.6
Because of the need for prolonged treatment, adherence to therapy is still the major
issue determining its effectiveness. Several strategies have thus been explored to de-
crease treatment costs and promote patient compliance. The World Health Organiza-
tion endorses directly observed treatment (DOT), which involves direct observation of
medication intake. This ensures that medication is taken in the right combination and
appropriate dosage, maximizes the likelihood of completion of therapy, and prevents
the development of drug resistance.
The optimal dosing frequency for new patients with pulmonary TB is once daily
throughout the course of therapy, however adherence to such regimens remains a
challenge. There are differing recommendations on the use of intermittent regimens to
treat TB in children; a recent systematic review comparing intermittent twice-weekly
versus daily treatment among children 15 years and below concluded that ―trials con-
ducted to date are insufficient to support or refute the use of intermittent twice- or thrice
-weekly, short-course treatment regimens‖. 7
The WHO does not recommend twice-weekly regimens, since any missed dose leads
to a significant reduction of the total quantity of drugs received by the patient and can
lead to treatment failure.8 In addition, the 2009 WHO Treatment of Tuberculosis states
that the rates of acquired drug resistance are higher among patients receiving intermit-
tent dosing throughout therapy; in patients with pre-treatment isoniazid resistance,
three times weekly dosing during the intensive phase was associated with significantly
higher risks of failure and acquired drug resistance.9 Thus, the WHO‘s Rapid Advice
2010 specifies that thrice-weekly regimens may be considered only during the continu-
ation phase of treatment, for children known to be HIV-uninfected and living in settings
with well-established DOT.
139
Contact tracing remains a key step for effective tuberculosis therapy and control. The
treatment of tuberculosis ―can only be successful within the framework of overall medi-
cal and social management of patients and their contacts. The ultimate elimination of
tuberculosis requires an organized and smoothly functioning network of primary and
referral services based on cooperation between the private and public sectors of medi-
cal care.‖ 4
140
culture conversion in adults with pulmonary MDR-TB. Likewise delamanid has been
shown to be effective in increasing the proportion of patients achieving sputum culture
conversion after 8 weeks of treatment. 12
Table 11.1 lists the first-line anti-tuberculosis drugs, including details on the dosing,
mechanisms of action and adverse effects associated with these agents. The recom-
mended pediatric drug doses are based on the World Health Organization‘s 2010 Rap-
id Advice: Treatment of TB in Children. The Department of Health‘s 2014 Manual of
Procedures has adopted these recommended doses for the country‘s National Tuber-
culosis Control Program, with the exception of pyrazinamide; the local recommendation
for this drug specifies a slightly lower dose and is used in the table below.
Since adolescents show greater similarity to adults than to young children with respect
to dosage requirements, risk of MDR-TB, and adverse drug reactions, it is recommend-
ed that older children and adolescents be treated at adult dosages once they reach a
body weight of 25 kg. 1
Table 11.1. First-line anti-tuberculosis drugs (continued on next page)
Drug Drug Dose per Kg Mechanism Adverse Reac- Comments
Body Weight of Action tions / Interac-
Children Adults tions
Pyra- 30 (20- 25 (20- Disruption of Nausea, vomit- Discontinue
zinamide 40) mg/ 30) mg/ membrane ing; most com- if AST >3-5x
(Z) kg/day kg/day energy me- mon cause of normal val-
(max 2 g/ (max 2 g/ tabolism hepatotoxicity uesa
day) day) in regimens
also containing Requires
H and R; dose modifi-
hypersensitivity cation in
reactions and renal failure
polyarthralgia
Ethambutol 20 (15- 15 (15- Inhibits Peripheral neu- Previously
(E) 25) mg/ 20) mg/ transferase ropathy and omitted from
kg/day kg/day enzymes retrobulbar op- regimens for
(max 1.2 (max 1.2 involved in tic neuritis children be-
g/day) g/day) cell wall (impairment of cause of
synthesis visual acuity difficulty of
and red-green monitoring
color vision) optic neuri-
tis; new evi-
dence
shows safe-
ty in children
at recom-
mended
doses in the
absence of
renal impair-
ment
141
Isoniazid (H) 10 (10-15) 5 (4-6) mg/ Bactericidal Hepatitis, periph- Plasma half-
mg/kg/day kg/day against ac- eral neuropathy, life of H var-
(max 300 (max 300 tively growing allergic skin re- ies from < 1
mg/day) mg/day) M. tuberculo- actions; possible hour in fast
sis; bacterio- hemolysis acetylators to
static against among G6PD- > 3 hours in
non- deficient patients slow acetyla-
replicating tors
organisms; Inhibits drug-
inhibits my- metabolizing For H and R,
colic acid enzymes (DME), discontinue if
synthesis; leading to in- AST >3-5x
also likely creased risk of normal val-
inhibits the phenytoin, uesa
catalase- ethosuximide
peroxidase and carbamaze- For H and R,
enzyme pine toxicity no dose ad-
justment for
renal dys-
Rifampicin 15 (10-20) 10 (8-12) Inhibits DNA- Hepatitis, hyper- function.
(R) mg/kg/day mg/kg/day dependent sensitivity reac-
(max 600 (max 600 RNA poly- tions (including a
mg/day) mg/day) merase systemic flulike H and R are
syndrome + best ab-
thrombocytope- sorbed on an
nia in patients empty stom-
given intermittent ach
high-dose thera-
py), orange dis- When co-
coloration of administering
body fluids anti-TB and
antiretroviral
Induces DME, drugs, seek
resulting in de- guidance
creased plasma from clini-
levels of some cians familiar
drugs such as with potential
AEDs (including interactions
phenytoin), anti -
infectives (ex.
clarithromycin,
erythromycin,
chlorampheni-
col), hormonal
therapy agents
(ethinylestradiol,
norethindrone,
tamoxifen, levo-
thyroxine), corti-
costeroids, etc.
142
a
Drug-induced liver injury is defined as AST level ≥ 3 times the upper limit of normal in the pres-
ence of symptoms, or 5 times more than the upper limit in the absence of any symptoms
References:
1. American Thoracic Society.CDC,MMWR. Treatment of tuberculosis infection in
adults and children. 20 June 2003;52(RR-11).
2. CDC. TB Guideline. Accessed from: http://www.CDC.gov/tb/pubs/PDA
3. CDC. Core Curriculum on Tuberculosis. What the Clinician Should Know. 6th ed.
2013. Accessed from: https://www.cdc.gov/tb/education/corecurr/pdf/corecurr_all.pdf
4. Curry InternationalTuberculosis Center and California Department of Public Health,
2012: Tuberculosis Drug Information Guide, 2nd edition. Accessed at http://
www.currytbcenter.ucsf.edu/tbdruginfo
5. Davis PDO,(ed).Clinical Tuberculosis.London:Chapman and Hall;1994:151-152.
6. Diagnosis, Treatment, Prevention and Control of Tuberculosis in Adult Filipinos: Up-
date.2006
7. Stop TB Partnership Childhood TB Subgroup. Chapter 2: Anti-tuberculosis treatment
in children [Official Statement. Guidance for National Tuberculosis Programmes on the
management of tuberculosis in children. Chapter 2 in the series]. The International
Journal of Tuberculosis and Lung Disease.Nov 2006. 10(11);1205-1211(7).
143
Table 11.3 Symptom-based approach to management of minor side effects
MINOR SIDE EFFECTS DRUG(S) IMPLI- MANAGEMENT
CATED
Anorexia, nausea, ab- INH Give drugs with small meals or sched-
dominal pain Rifampicin ule before bedtime; advise to swallow
PZA medications slowly with small sips of
water
If symptoms persist or worsen, or
there is prolonged vomiting or any
signs of bleeding, consider as MAJOR
side effect (stop TB drugs and refer)
Joint pains PZA Aspirin, NSAID or paracetamol
Burning, numbness or tin- INH Add pyridoxine (5-10 mg/day) to treat-
gling sensation in hands / ment regimen for exclusively-
feet breastfed infants, infants of breast-
feeding mothers on isoniazid, pa-
tients on meat- and milk-deficient di-
ets, pregnant adolescents, patients
with conditions predisposing to neu-
ropathy (including diabetes, uremia,
malnutrition, and HIV infection).
Since it is often difficult to determine which of the multiple drugs being taken is the rea-
son for an adverse reaction, the following table may provide guidance as to the likeliest
cause.
144
Table 11.4 Likeliest causes of adverse drug reactions
Most likely à least likely cause
Hepatitis- ALT/ Pyrazinamide Isoniazid Rifampicin Ethambutol
AST predomi- (very rare)
nant
Cholestatic Rifampicin
jaundice
Upper GIT Rifampicin Pyrazinamide Isoniazid Ethambutol
symptoms
Arthralgia Pyrazinamide Rifampicin (flu- Isoniazid (drug- Ethambutol
like syndrome) induced lupus)
Hypersensitivity Isoniazid Rifampicin Pyrazinamide Ethambutol
(fever, rash,
other symp-
toms)
145
Table 11.5 Dosing table for FDCs
*Ethambutol (15-25 mg/kg) should be added in the intensive phase for children with
extensive disease or living in settings where the prevalence of HIV or of isoniazid re-
sistance is high
Though these FDCs will provide isoniazid at a dose below 10-15 mg/kg/day in some
weight bands, the WHO has noted that an isoniazid dosage of 7 mg/kg will provide ade-
quate levels in almost all children; thus the isoniazid dosing range may be extended to
7-15 mg/kg, with the mid-range placed at 10 mg/kg. 1
RECOMMENDED REGIMENS
Drug therapy regimens employed in tuberculosis are designed to achieve varying
goals. For the patient exposed to Mycobacterium tuberculosis without evidence of in-
fection or active disease, the objective is to prevent the onset of infection (―primary
prophylaxis‖). For the patient with latent TB infection, the drug regimen aims at prevent-
ing progression of infection to active disease (―secondary prophylaxis‖). Finally, for the
patient classified as having disease, the goals include not only cure for the individual
patient, but also decrease in transmission to the community.
There is a standard code for anti-TB treatment regimens, which uses an abbreviation
for each anti-TB drug, e.g. isoniazid (H), rifampicin (R), pyrazinamide (Z) and ethambu-
tol (E). A regimen consists of two phases: the initial and continuation phases. The num-
ber at the front of each phase represents the duration of that phase in months. A sub-
script number (e.g. 3) following a drug abbreviation is the number of doses per week of
that drug. If there is no subscript number following a drug abbreviation, treatment with
that drug is daily. An alternative drug (or drugs) appears as an abbreviation (or abbrevi-
ations) in parentheses.
Example: 2HRZE/4HR
The initial phase is 2HRZE. Duration of this phase is 2 months. Drug treatment is daily
(no subscript numbers after the abbreviations) with isoniazid, rifampicin, pyrazinamide
and ethambutol. The continuation phase is 4HR. Duration of this phase is 4 months,
146
with isoniazid and rifampicin once daily on empty stomach (one hour before, or two
hours after, a meal) 15.
Treatment regimens are assigned based on the anatomical site of involvement, bacteri-
ologic confirmation status as well as history of previous treatment. The World Health
Organization‘s most recent guideline (Guidelines for Treatment of Tuberculosis, 4th edi-
tion, 2010) as well as the 2nd edition of the Guidance for National Tuberculosis Pro-
grammes on the Management of Tuberculosis in Children (2014) outline a treatment
strategy that has become the basis of the Department of Health‘s recommendations,
listed below.
The following table describes the different registration groups based on treatment histo-
ry.
Table 11.6 TB disease registration groups
Registration Group Definition of Terms
New A patient who has never had treatment for
TB* or who has taken anti-TB drugs for <1
month
Relapse A patient previously treated for TB who has
been declared cured, or completed treatment
in their most recent treatment episode, and is
presently diagnosed with bacteriologically-
confirmed or clinically-diagnosed TB.
Treatment after failure A patient who has been previously treated
for TB and whose treatment failed at the end
of their most recent course. This includes:
A patient whose sputum smear or culture
Re-treatment
148
The issues of drug resistance and HIV prevalence were also taken into consideration in
the 2003 joint recommendation for TB treatment issued by the American Thoracic Soci-
ety, CDC and the Infectious Disease Society of America; this recommendation states
that a four-drug intensive phase including ethambutol can be safely used in children
when warranted by epidemiologic circumstances.17
PREVENTIVE THERAPY
Prophylaxis aims to prevent the following conditions:
1. Development of infection among contacts exposed to active disease: primary
prophylaxis
2. Progression of latent TB infection to disease: secondary prophylaxis
Prophylaxis is recommended for children under 5 years or among those with other risk
factors for rapid development of disease, particularly immunosuppressed individuals.
This is because disease may set in even before conversion of the tuberculin skin test.
Several well-controlled studies have demonstrated the favorable effect of isoniazid on
reduction of complications due to lymphohematogenous and pulmonary spread after
infection. The protective effect of isoniazid in the latter situation has been shown to last
from 19 to 30 years.18
One approach to prophylaxis is proposed by the Department of Health in its National
Tuberculosis Control Program. Once TB disease has been excluded, isoniazid preven-
tive therapy (IPT) is recommended for (1) all HIV-positive individuals, (2) children less
than 5 years old who are household contacts of a bacteriologically confirmed TB case,
regardless of the TST results or (3) children less than 5 years old who are household
contacts of a clinically diagnosed TB case, if the TST result is positive. The regimen
consists of a 6-month course of isoniazid, given at a dose of 10mg/kg once daily. 19
The following algorithm illustrates a more comprehensive approach to prophylaxis. This
was proposed in the National Consensus on Childhood Tuberculosis 1997 and contin-
ues to be recommended by the Philippine Pediatric Society:
149
TB Exposure
< 5 years
Yes
Start INH for 3
old months
No
Radiologic
Repeat findings
Positive and / or, Yes TB Disease
Mantoux
test af- signs /
symptoms Multiple Drug Ther-
ter 3
suggestive apy
months
of TB
Negative
No
Discontinue H LTBI
If no BCG scar, Continue > 6H
give BCG
150
REFERENCES
1. World Health Organization. Guidance for national tuberculosis programmes on the
management of tuberculosis in children. 2nd ed. 2014.
2. American Academy of Pediatrics Committee on Infectious Diseases. Chemotherapy
for tuberculosis in infants and children. Pediatrics 1992; 89(1): 161-165.
3. Evangelopolous D et al. Understanding anti-tuberculosis drug efficacy: rethinking
bacterial populations and how we model them. International Journal of Infectious
Diseases 32 (2015) 76–80.
4. American Thoracic Society and the Centers for Disease Control. Treatment of tu-
berculosis and tuberculosis infection in adults and children. Am J Respir Critic Care
Med 1994 May; 149 (5): 1359-74.
5. Rom WN and Stuart Garay, eds. Tuberculosis. 1996. Boston: Little, Brown and Co.
p 683.
6. Rom WN and Stuart Garay, eds. Tuberculosis. 1996. Boston: Little, Brown and Co.
p 758.
7. Bose A, Kalita S, Rose W, Tharyan P. Intermittent versus daily therapy for treating
tuberculosis in children. Cochrane Database Syst Rev. 2014 Jan 28;1:CD007953.
8. World Health Organization. Treatment of tuberculosis: guidelines for national pro-
grams, 3rd edition. 2003.
9. World Health Organization. Treatment of tuberculosis: guidelines. Fourth edition,
2010.
10. Gumbo T et al. Pharmacokinetics-pharmacodynamics of pyrazinamide in a novel in
vitro model of tuberculosis for sterilizing effect: a paradigm for faster assessment of
new antituberculosis drugs. Antimicrob. Agents Chemother. August 2009; 53(8):
3197-3204).
11. CDC. Core Curriculum on Tuberculosis. What the Clinician Should Know. 6th ed.
2013. Accessed from: https://www.cdc.gov/tb/education/corecurr/pdf/
corecurr_all.pdf.
12. NHS England. Clinical Commissioning Policy: Bedaquiline and Delamanid for de-
fined patients with MDR-XDR-TB. Accessed from https://
www.engage.england.nhs.uk/consultation/specialised-services-consultation/
user_uploads/bedaquiline-delamanid-policy-upd.pdf.
13. Wallace Jr. RJ et al. 2015. Chap 38. Antimycobacterial agents. In Bennett JE et al.
eds. Mandell, Douglas, and Bennett‘s Principles and Practice of Infectious Diseas-
es, 8th ed. Phladelphia, Elsevier Saunders.
14. World Health Organization. Factsheet: New fixed-dose combinations for the treat-
151
ment of TB in children. Accessed from: http://www.who.int/tb/FDC_Factsheet.pdf?
ua=1.
15. World Health Organization. Guidance for national tuberculosis programmes on the
management of tuberculosis in children. 1st ed. 2006.
16. USAID/Philippines: External Evaluation of the Tuberculosis Portfolio (2006–2011).
Accessed from http://pdf.usaid.gov/pdf_docs/pdact786.pdf.
17. Centers for Disease Control and Prevention. Treatment of Tuberculosis, American
Thoracic Society, CDC, and Infectious Diseases Society of America. MMWR
2003;52(No. RR-11):1-77.
18. Starke JR and Margaret HD Smith. 1998. Tuberculosis. In Feigin RD and JD Cher-
ry, eds. Textbook of Pediatric Infectious Diseases 4th ed. Philadelphia: WB Saun-
ders Co. p 1225.
19. Department of Health. (2014). Manual of Procedures of the National Tuberculosis
Control Program. 5th edition.
152
12
Drug-Resistant TB
Anjanette de Leon,MD
Heidilita Espinoza,MD
OUTLINE
1 Overview
2 Definitions
3 Pathophysiology
4 Diagnosis
5 Risk Factors
6 Treatment
7 Prevention
153
OVERVIEW
The management of drug-resistant tuberculosis remains to be a challenge among
health care professionals. Thus, it is vital to identify such patients with the help of ex-
pertise in this area. The WHO has a revised definition of cases of drug-resistant tuber-
culosis based on drug susceptibility testing (DST) of clinical isolates confirmed to be M.
tuberculosis (MTB) namely: monoresistance, multidrug-resistance, polydrug re-
sistance, extensive drug resistance and rifampicin resistance.1 Establishing a diagnosis
is quite difficult so a detailed and careful history should be obtained. Likewise, radiolog-
ic findings are comparable with that of drug-susceptible tuberculosis thus making the
diagnosis more difficult. Other diagnostic procedures include baseline sputum culture
and sensitivity prior to treatment and nucleic acid tests such as GeneXpert MTB/RIF,
MTBDRplus and MTBDRs which are all helpful but expensive. The approach to man-
agement can be medical and surgical. With medical treatment, this is based on three
strategies which includes standardized, empirical and individualized. Surgery is an op-
tion in cases where patient remains to be smear positive, with resistance to a large
number of drugs and has localized pulmonary disease.
DEFINITIONS
Uniform definitions among health care professionals are vital to have standardized pa-
tient registration, correct assignment to treatment regimens and in comparing research
outcomes. Drug-Resistant Tuberculosis (DRTB) is confirmed through laboratory tests
that show that the infecting isolates of MTB grow in vitro in the presence of one or more
anti-tuberculosis drugs.
Primary resistance refers to the bacterial resistance present in patients who have
not received prior treatment with anti-tuberculosis drugs.
Acquired resistance refers to the bacterial resistance in patients with some record
of previous treatment.1
The World Health Organization defines five categories:2
Monoresistance: resistance to one anti-tuberculosis drug
Multidrug-resistance (MDR): resistance to at least both isoniazid and rifampicin
Polydrug-resistance: resistance to more than one first line anti-tuberculosis drug,
other than both isoniazid and rifampicin
Extensive drug-resistance: resistance to isoniazid and rifampicin, any flouroquin-
olone, and at least one of three second line injectable drugs: kanamycin, capreomy-
cin, or amikacin.
Rifampicin resistance: resistance to rifampicin detected using phenotypic or geno-
typic methods, with or without resistance to other anti-TB drugs. It includes any re-
sistance to rifampicin, whether mono-resistance, multidrug resistance, polydrug re-
sistance or extensive drug resistance.28
154
The National Consensus on Childhood Tuberculosis proposed the following criteria for
defining DRTB:5,24,28
Confirmed MDR-TB disease is defined as clinical evidence of TB disease together
with the detection of MTB (either by culture or molecular probe) from a specimen col-
lected from the subject with genotypic (e.g., GeneXpert) or phenotypic resistance to at
least Rifampicin.3,5,6,7,8,9
Clinical evidence of TB (TB Disease) would mean the presence of at least one
of the following signs or symptoms: persistent cough, weight loss, failure to
thrive, persistent unexplained fever, persistent unexplained lethargy or reduced
playfulness, or the presence of any of the following in the neonate like pneumo
nia, unexplained hepatosplenomegaly, or sepsis-like illness
Probable MDR-TB disease is defined as the clinical evidence of TB disease AND pres-
ence of one of the following: (a) positive clinical response to MDR-TB treatment, or (b)
immunological evidence of TB infection (i.e., a positive TST or IGRA test result at
screening), AND documented exposure to a source case of MDR-TB based on a stand-
ardized questionnaire or an adult who died of TB.
Clinical evidence of TB (TB Disease) would mean the presence of at least one
of the following signs or symptoms
(1) Lack of clinical improvement despite adequate therapy for at least 2
months. This would mean any of the following: persistent fever with no
apparent cause; worsening of radiologic findings; new complications
arise; and poor weight gain(less than 10%) and well-being;
(2) Previous inadequate treatment and the presence of other risk factors like
cavitary lesions and HIV/AIDS infection.
PATHOPHYSIOLOGY
Drug resistance can be attributed to two pathways: primary drug resistance and sec-
ondary/acquired drug resistance.
Primary drug resistance would mean that the patient was exposed to a drug resistant
TB strain and would have the same way of transmission as that of regular TB. On the
other hand, secondary drug resistance is a result of inadequate, inappropriate and in-
complete treatment resulting to development of resistant strains.
Treatment Outcome Definition
Cured Treatment completed as recommended by the national policy without
evidence of failure AND three or more consecutive cultures taken at
least 30 days apart are negative after the intensive phase
DIAGNOSIS
In pediatric cases, diagnosis of DRTB can be a problem. For all cases of childhood TB,
whether suspected to be of a drug-resistant strain or not, the adult source case should
be sought, and if resources are available, the drug susceptibility of the MTB isolate of
either adult source case or child be determined. Risk factors for resistance should al-
ways be elicited. For most children, drug susceptibility information come from the MTB
isolate that is obtained from the adult who transmitted the infection 3 since no isolate is
usually obtained from the child. Thus, studies have shown that patterns of drug re-
sistance in children tend to mirror those found in adult patients.4 In most communities, a
good clinical response to treatment may be a reasonable basis to assume that the or-
ganism is susceptible. If the clinical response is slow or when new complications arise,
the possibility of DRTB should be considered,5 thus requiring revision of treatment regi-
men.
In May 2013, WHO convened an Expert Group to evaluate the data and formulate rec-
ommendations on the use of Xpert MTB/RIF in children. This was later approved upon
by the WHO Guideline Review Committee in October 2013.
This would mean that using the Xpert MTB/RIF can: (i) help in the diagnosis of pulmo-
nary TB and rifampicin resistance in children; and (ii) Xpert MTB/RIF for the diagnosis
of extrapulmonary TB in children. The technology is recommended, especially in se-
verely ill children where rapid diagnosis is important although this does not negate or
exclude TB if a negative Xpert MTB/RIF result would come out, needing for a clinical
decision on these cases be made. 26
RISK FACTORS
The following are important risk factors for the development of drug resistance:12
TREATMENT
The treatment regimens for MDR-TB is highly complicated. Most of the time, treatment
is empiric on initial intensive phase of treatment until drug susceptibility testing is avail-
able on the MTB isolate to address the treatment to be given, the patient‘s previous
156
treatment history for TB, information on drug resistance patterns of known MDR-TB pa-
tients with whom a given patient has been in close contact, and available information
on HIV status.
Common treatment strategies for DRTB are as follows:
STANDARDIZED, where drug resistance and susceptibility (DRS) data from repre-
sentative patient populations are used to base regimen design in the absence of
individual DST, and all patients in a defined group or category receive the same
regimen.
157
(i.e. Dr. Jose N. Rodriguez Memorial Hospital in Tala Caloocan, Lung Center of the
Philippines and KASAKA DOTS Treatment Center, Quezon Institute both in Quezon
City, and PTSI in Tayuman, Manila) and provinces as well.
Drug dosages are determined by body weight. When adverse effects are noted, they
should be treated immediately and adequately in order to minimize the risk of treatment
interruptions and prevent increased morbidity and mortality due to serious adverse ef-
fects. Most importantly, each dose is given as directly observed therapy (DOT)
throughout the treatment and a treatment card is marked for each observed dose.13,14
The table below summarizes the drugs used in the treatment of MDRTB.
Table 12.1. Drugs used in the treatment of MDRTB
Grouping Drugs
Group 1 Isoniazid, Rifampicin, Ethambutol, Pyra-
First-line oral agents zinamide
Group 2 Injectable agents Kanamycin, Amikacin, Capreomycin,
Streptomycin
Group 3 Fluoroquinolones Moxifloxacin, Levofloxacin, Ofloxacin
Group 4 Oral bacteriostatic Ethionamide, Prothionamide, Cyclo-
second-line agents serine, Terizidone, p-aminosalicylic acid
(PAS)
Group 5 Clofazimine, Linezolid, Amoxicillin-
Agents with unclear efficacy clavulanic acid, thioacetazone,
(not recommended by WHO Imipenem/cilastatin, clarithromycin, high
for routine use in MDR-TB -dose isoniazid (16-20 mkd)
patients)
158
5. Use two or more agents from Group 5.
6. Consider high-dose isoniazid treatment if low-level resistance is documented.
7. Consider adjuvant surgery if there is localized disease.
8. Ensure strong infection control measures.
9. Treat HIV.
10. Provide comprehensive monitoring and full adherence support.
159
Table 12.2 Different Characteristics of Second-line Anti-tuberculosis Agents15,16
Daily
Mecha-
dose Max daily Adverse ef- Other com-
Grouping Drugs nism of
in mg/ dose fects ments
Action
kg,
Kanamy- 15-30 1g Nephrotoxici- Avoid in
cin ty pregnancy
Group 2 inhibits 15- Ototoxicity
Injectable Amikacin protein 22.5
agents synthesis Neuromuscu- Dose adjust-
Capreo- 15-30 lar blockade ment in renal
mycin insufficiency
15-20 800 mg Gastric irrita- avoid in
Ofloxacin div q tion. pregnancy
12 h
Group 3
Inhibits 7.5-10 750 mg
Fluoro- Levofloxa- DNA gy- Not approved dose adjust-
quinolon cin rase for children ment in renal
es
7.5-10 400 mg due to con- insufficiency
Moxifloxa-
cern with
cin
arthopathy
Ethiona- 15-20 1g vomiting, gas- teratogenic;
mide Inhibits tric irritation, contraindi-
mycolic hypothyroid- cated in
acid syn- ism, malab- pregnancy
Prothiona- thesis sorption
mide
160
PREVENTION OF MDR-TB
Many new cases of MDR-TB are created each year by a combination of physician error
(prescribing inadequate regimens), genetic predisposition and poor patient compliance
with treatment, which turn fully susceptible organisms, or those with less complex re-
sistance pattern develop to MDR-TB.
Programmatic failures may result in an insufficient or erratic supply of antituberculosis
drugs or poor drug quality which may affect bioavailability and lead to a reduction in
drug efficacy. Poor case management, such as permitting patients to self-administer or
tailor their own regimens may also induce drug resistance.17
MDR-TB can be controlled by the universal implementation of the WHO TB control
strategy (DOTS), which emphasizes use of standardized treatment regimens, an unin-
terrupted supply of high-quality drugs, and directly-observed treatment. Given the high
morbidity and mortality, as well as, the high cost of treating MDR-TB patients, it is es-
sential that TB control programs employ these measures to prevent MDR TB.18
161
REFERENCES:
1. Crofton J, Chaulet P, Maher D. Guidelines for the management of drug-resistant
tuberculosis. Geneva: World Health Organization; 1997.
2. Rich M, et al. Guidelines for the programmatic management of drug-resistant tuber-
culosis, Emergency update. Geneva: World Health Organization; 2008.
3. Swanson D, Starke J. Drug Resistant Tuberculosis in Pediatrics. Pediatric Clinics of
North America. 1995;42:553-575.
4. Steiner P, Rao M, Mitchell M, et al. Primary drug-resistant tuberculosis in children.
Correlation of drug-susceptibility patterns of matched patient and source case
strains of Mycobacterium tuberculosis. Am J Dis Child. 1985;139:780-782.
5. Jacobs R, Starke J. Tuberculosis in Children. Medical Clinics of North America.
1993;77:1335-1405.
6. Starke J. Modern approach to the diagnosis of tuberculosis in children. Pediatric
Clinics of North America. 1988; 35:441-459.
7. Davidson P. Drug Resistance and Selection of Therapy for Tuberculosis. Am. Rev.
Respiratory Diseases. 1987;136:255-256.
8. Jacobs, Richard F.M.D. Multiple Drug Resistance Tuberculosis. Clinical Infectious
Diseases. 1994;19:1-10.
9. Neville K, et.al. The Third Epidemic MultiDrug Resistant tuberculosis. Chest.
1994;105:45-47.
10. Global tuberculosis control: epidemiology, strategy, financing: WHO report 2009.
11. Tuberculosis in the Philippines. Philippine Tuberculosis Society, Inc. 2009.
12. Data on the Tuberculosis Epidemic. TB: A Crossroads. WHO Report on the Global
Tuberculosis Epidemic; 1998:42-51.
13. Guidance for the national tuberculosis programme on the management of tubercu-
losis in children. Geneva. World Health Organization. 2006 (WHO/HTM/
TB/2006.371).
14. Centers for Disease Control and Prevention. Treatment of Tuberculosis. American
Thoracic Society, CDC, and Infectious Disease Society of America. MMWR.
2003;52 (No. RR-11).
15. Zhang Y. The Magic Bullets and Tuberculosis Drug Targets. Annu Rev Pharmacol
Toxicol. 2005;45:529-64.
16. Brunton L, editor. Goodman & Gilman‘s the pharmacologic basis of therapeutics.
11th edition. The McGraw-Hill Companies. 2006.
162
17. Ormerod L. Multidrug-resistant tuberculosis (MDR-TB): epidemiology, prevention
and treatment. British Medical Bulletin. 2005;73-74(1):17-24.
18. Spradling P. Prevention of Multidrug-Resistant Tuberculosis. Division of TB Elimi-
nation, NCHSTP, Centers for Disease Control and Prevention, Atlanta, GA.
19. Mendoza MT, Ang CF, Lazo S, Isaac C. In vitro susceptibility of multi-drug resistant
Mycobacterium tuberculosis isolated from Filipino patients. Phil J Microbiol Infect
Dis 2004; 33(3):95-97.
20. The Global MDR-TB & XDR-TB Response Plan 2007-2008.
21. www.stoptb.org/wg/mdrtb.
22. The use of delamanid in the treatment of multidrug-resistant tuberculosis Interim
policy guidance. WHO 2014.
23. Robert Roos .European Group Offers Treatment for MDRTB, XDRTB. CIDRAP
News | Mar 24, 2014
24. Companion handbook to the WHO guidelines for the programmatic management of
drug-resistant tuberculosis. WHO 2014.
25. Management of MDR-TB: A field guide A companion document to Guidelines for
the programmatic management of drug-resistant tuberculosis. WHO 2009.
26. Guidance for national tuberculosis programmes on the management of tuberculosis
in children Second edition. WHO 2014.
27. 2013 Manual of Procedures of the National TB Control Program, 5th edition. De-
partment of Health, Philippines 2013.
28. Definitions and reporting framework for tuberculosis – 2013 revision (updated De-
cember 2014) . WHO 2013.
29. Seddon JA, Perez-Velez CM, Schaaf HS, et al. Consensus Statement on Research
Definitions for DrugResistant Tuberculosis in Children. J Pediatric Infect Dis Soc.
2013; 2(2):100-109.
163
13
TB in Special Situations
Clara R Rivera, MD
Maria Theresa T Policarpio, MD
Grace Devota G Go, MD
OUTLINE
1 Overview
6 Tuberculosis in Immunocompro-
mised Children
164
OVERVIEW
This chapter presents the treatment of tuberculosis (TB) in special situations like preg-
nant and breastfeeding mothers with TB, the newborns of tuberculous mothers in differ-
ent circumstances like latent tuberculosis infection (LTBI) and active TB. Treatment
difficulties in the management of TB in children with drug-induced liver disease, liver
and renal impairment and specific dosing guidelines for patients with renal insufficiency
and end-stage renal disease are provided.
Immunodeficiency, especially in people living with Human Immunodeficiency Virus
(PLHIV) and Human Immunodeficiency Virus (HIV) infected children, and other special
situations where patients are immunosuppressed have increased susceptibility to tu-
berculosis infection and disease are discussed.
Combination chemotherapy for tuberculosis is associated with a predictable incidence
of adverse effects, some mild, some serious. Mild adverse effects can generally be
managed with symptomatic therapy, whereas with more severe effects, the offending
drug or drugs must be discontinued. Although it is important to be attuned to the poten-
tial for adverse effects, it is at least equally important that first-line drugs not be stopped
without adequate justification. A summary of the approaches that should be taken in
managing the common adverse effects of tuberculosis treatment will be discussed but
proper management of more serious adverse reactions often requires expert consulta-
tion.1
165
Management of pregnant mothers with latent TB infection (LTBI)
Pregnant women diagnosed with LTBI are at high risk of developing active TB, espe-
cially if they are HIV-positive, have had contact with active TB cases, or reveal new tu-
berculin skin test (TST) positivity within the past 2 years.13
Asymptomatic pregnant women with positive TST results, normal chest radiographic
findings, and recent contact with a contagious person should be considered for isonia-
zid preventive therapy (IPT). The recommended duration is 9 months. Therapy in these
circumstances should begin after the first trimester. However, under WHO/DOH-NTP,
the mother should be given isoniazid for at least 6 months immediately without delay
with pyridoxine supplementation to diminish the risk of peripheral neuropathy.14
Management of pregnant mothers with TB disease
Most anti-tuberculosis drugs are safe for pregnant women. Streptomycin (S) and other
aminoglycosides should be avoided because of their ototoxicity. The use of capreomy-
cin. ethionamide, fluoroquinolones or cyloserine is not recommended.1,4 Women who
become pregnant while on treatment should continue therapy, except for streptomycin
and fluoroquinolone. Treatment includes 2HRZE4HR.1,2,3,4 Advise pregnant women that
successful treatment of TB with the recommended standardized treatment regimen (i.e.
2HRZE/4HR) is important for a successful outcome of pregnancy. Pregnant women
taking isoniazid should be given pyridoxine at 25mg/day.12
166
for granulomata. The baby should be evaluated for congenital TB. Once the infant has
received isoniazid or rifampicin and the mother has undergone appropriate therapy,
separation is no longer required. If initial TST turns out negative, the test should be re-
peated after three months. If TST turns out positive but chest X-ray is negative, isonia-
zid or rifampicin should be continued to complete six months. If the TST and chest X-
ray of the mother are negative and she has completed her treatment, BCG should be
administered to the infant while isoniazid or rifampicin should be discontinued.
If the mother has extrapulmonary disease, such as tuberculous meningitis, miliary,
bone or joint TB, the infant must be monitored closely for possible congenital tuberculo-
sis. If a newborn infant is suspected of having congenital tuberculosis, a TST, chest
radiography, lumbar puncture, and appropriate cultures and radiography should be per-
formed promptly. The TST result is usually negative in newborn infants with congenital
or perinatally acquired infection. Hence, regardless of the TST results, treatment of the
infant should be initiated promptly with isoniazid, rifampicin, pyrazinamide and an ami-
noglycoside (eg. streptomycin).14
Currently, the accepted mode of treatment is isoniazid, rifampicin, pyrazinamide and
either ethambutol or streptomycin for first 2 months followed by isoniazid and rifampicin
for 4 to 10 months.15,16 Depending on the disease category, recovery has been ob-
tained by the combination of isoniazid, rifampicin, streptomycin and pyrazinamide for
initial 2 months. If congenital TB is ruled out, preventive therapy should be given similar
to cases of newborns whose mother has TB disease.
167
substituted with a fluoroquinolone or an aminoglycoside as per the clinician‘s prefer-
ence. The proposed regimen is a combination of rifampicin, isoniazid, fluoroquinolone/
aminoglycoside and ethambutol for 2 months followed by 4 months of rifampicin and
isoniazid.18
Hepatotoxic drugs are completely avoided in decompensated liver cirrhosis. The vari-
ous drugs which can be used in a combination regimen are ethambutol, aminoglyco-
sides, fluoroquinolones, cycloserine and other newer non-hepatotoxic drugs with at
least 3 anti-TB drugs to be used in any combination regimen for around 18 to 24
months. One such possible regimen is the combination of streptomycin, ethambutol
and a fluoroquinolone for 18 to 24 months.20 However, there are no clinical trials to
prove the efficacy of such combinations.
Tuberculosis in Children with Drug-Induced Hepatitis
Hepatotoxicity may occur anytime during treatment but usually manifest in the first 2-4
weeks of therapy. Other causes of hepatitis must be ruled out. Children have a low risk
of developing drug-induced hepatitis. It has been noted that there is an increasing risk
of hepatitis with increasing age. The main principle is to reduce the number of hepato-
toxic drugs from the treatment regimen beyond 20 years of age and to increase the du-
ration of treatment.
Increases in transaminase values are frequently encountered during the first two
months of treatment. Elevation of serum liver enzymes up to two to four times occurs
rarely in children. In children less than 5 years old with elevated transaminases less
than 5 times normal and who are asymptomatic, stopping of drugs is not warranted.
However, if gastrointestinal symptoms such as nausea, vomiting and abdominal pain or
jaundice is noted, hepatotoxic first-line drugs should be discontinued or modified de-
pending on the level of serum liver enzyme aspartate aminotransferase (AST). Drug-
induced liver injury is defined as AST level three or more times than the upper limit of
normal in the presence of symptoms or five times more than the upper limit in the ab-
sence of any symptoms. AST of less than five times more than normal is mild toxicity;
AST of five to ten times more than normal is moderate; and a level of ten times more
than normal is severe.1,2,4,8
Restarting of the anti-TB drugs should be done slowly. The suspect anti-TB medica-
tions should be reintroduced one at a time after the AST level returns to less than two
times more than the upper limit of the normal. Rifampicin should be restarted first be-
cause it is less likely to cause hepatotoxicity compared to isoniazid and pyrazinamide.
There is no risk for hepatitis using rifampicin alone. However, risk increases when isoni-
azid and rifampicin are given together.2 If there is no increase in AST after one week,
isoniazid may be restarted. PZA will be restarted after INH if AST is not increasing. If
symptoms recur or AST increases, the last drug added must be stopped.
If hepatitis is severe, PZA must be discontinued and replaced by ethambutol or INH
and rifampicin be continued for nine months. Among the first-line anti-TB drugs the inci-
dence of pyrazinamide-induced hepatotoxicity during treatment is the highest and may
be severe and prolonged.1,3
168
TUBERCULOSIS IN CHILDREN WITH RENAL IMPAIRMENT
The diagnosis of TB in patients with renal failure is often delayed and may be masked
by either the underlying disease or by the symptoms of renal failure. Extrapulmonary
presentations are common. 51.6% incidence of extrapulmonary TB, with the peritoneal
and pleural involvement being the most common sites.57 Tuberculosis should be sus-
pected in end-stage renal failure patients with fever of unknown origin (T > 38.3C on
several occasions) and unexplained weight loss, especially when attempts to obtain a
clinical diagnosis fail despite an extensive laboratory and diagnostic work-up. All lymph
node groups should be carefully palpated as axillary and inguinal adenopathy may be
overlooked. Cardiovascular examination should identify clinical signs of pericardial effu-
sion. Respiratory examination may identify the presence of pleural effusion. The pres-
ence of ascites should be noted and the macroscopic appearance of peritoneal dialy-
sate should be checked for a cloudy appearance. Abdominal adenopathy and renal en-
largement associated with obstructive uropathy as well as bladder enlargement related
to urinary obstruction should be identified.
Sputum examination and chest radiography are needed. Radiography of the chest pat-
tern may be atypical (lower zone disease, miliary involvement, adenopathy and/or pleu-
ral effusion). An enlarged cardiac shadow may indicate the presence of pericardial effu-
sion. Hypercalcemia has been described as a feature of TB in patients with end-stage
renal failure .Ultrasound examination of the abdomen is useful for confirming abdominal
sites of TB including the presence of ascites, adenopathy, and splenic and renal TB.
Biopsy and culture of needle aspirates from extrapulmonary and pulmonary sites of pa-
thology are often required: the pleura (preferably biopsy), bronchial lavage and trans-
bronchial biopsies, peritoneal aspirates, lymph nodes (aspirate or biopsy), liver (biopsy)
and joints (aspirate or biopsy). Dialysate from peritoneal lavage should always be sent
for culture as acid-fast bacilli (AFB) staining alone on lavage fluid has a low diagnostic
yield.
Management
The recommended initial TB treatment regimen for patients with renal failure or severe
renal insufficiency is 2 months of isoniazid, rifampicin, pyrazinamide and ethambutol,
followed by 4 months of isoniazid and rifampicin. Although the first-line drugs, isoniazid,
rifampicin and pyrazinamide, are eliminated by the liver, management of patients with
renal insufficiency could be complicated. There are some alterations in dosages and
dosing interval of the anti-TB drugs in order to maintain peak plasma concentration.
Isoniazid and rifampicin are eliminated almost entirely by liver metabolism and biliary
excretion; these drugs could be given at the same doses in case of renal impairment.
Although pyrazinamide is metabolized in the liver, its metabolites may accumulate in
patients with renal disease. Ethambutol is 80% cleared by the kidney; hence it may also
accumulate in patients with renal disease. A longer dosing interval and the administra-
tion of PZA and EMB three times a week is recommended. Because of an increased
risk of nephrotoxicity and ototoxicity, streptomycin should be avoided in patients with
renal failure. If streptomycin must be used, the dosage is 15 mg/kg, two or three times
per week to a maximum of 1 gram per dose, and serum levels of the drug should be
monitored.
169
A thorough drug chart review should be performed prior to prescribing TB medication to
identify potential drug interactions especially in transplant patients. The selection and
doses of anti-TB drugs should preferably be decided in consultation with a nephrologist.
Anti-TB drugs that are significantly dependent on renal clearance are ethambutol,
levofloxacin, cycloserine, streptomycin, kanamycin, capreomycin, and amikacin.
Drug absorption may be unpredictable in patients with renal failure due to nausea and
vomiting resulting from uremia, and patients should be questioned regularly about the-
se symptoms.
Creatinine clearance should be measured in all patients with renal disease prior to
treatment. The timing of drug administration is important. Drugs should generally be
administered after hemodialysis to prevent loss of drug during dialysis. This is especial-
ly relevant for PZA, which is efficiently removed by hemodialysis. The injectable agents
– streptomycin, kanamycin, capreomycin and amikacin – are also partially removed and
should be given after hemodialysis. Of the standard TB drugs, rifampicin is not re-
moved by hemodialysis due to its wide volume of distribution and high degree of protein
binding. Isoniazid and ethambutol are removed by hemodialysis but to a lesser extent
than PZA.
End-stage renal failure (ESRF) patients with TB may pose an infective risk to immuno-
compromised renal transplant cases and other patients in dialysis units. Infection con-
trol measures and contact tracing should be applied. In patients with ESRF on TB treat-
ment, the same general principles for management of patients with renal failure apply.
A thorough drug chart review should be performed prior to prescribing TB medication to
identify potential drug interactions especially in transplant patients on cyclosporine and
mycophenolate mofetil. The selection and doses of TB drugs should preferably be de-
cided in consultation with a nephrologist.
Box 13.1 Anti-tuberculous drugs that significantly dependent on renal clearance
Ethambutol
Levofloxacin
Cycloserine
Kanamycin
Capreomycin
Amikacin
(Metabolites of pyrazinamide may accumulate.)
The recommended initial TB treatment regimen for patients with renal failure or severe
renal insufficiency is 2 months of isoniazid, rifampicin, pyrazinamide and ethambutol,
followed by 4 months of isoniazid and rifampicin.13 Although the first-line drugs, isonia-
zid, rifampicin and pyrazinamide, are eliminated by the liver, management of patients
with renal insufficiency could be complicated. There are some alterations in dosages
and dosing interval of the anti-TB drugs in order to maintain peak plasma concentration
Isoniazid and rifampicin are eliminated almost entirely by liver metabolism and biliary
excretion; these drugs could be given at the same doses in case of renal impairment.
Although pyrazinamide is metabolized in the liver, its metabolites may accumulate in
170
patients with renal disease. Ethambutol is 80% cleared by the kidney; hence it may also
accumulate in patients with renal disease. A longer dosing interval and the administra-
tion of PZA and EMB three times a week is recommended. Because of an increased
risk of nephrotoxicity and ototoxicity, streptomycin should be avoided in patients with
renal failure. If streptomycin must be used, the dosage is 15 mg/kg, two or three times
per week to a maximum of 1 gram per dose, and serum levels of the drug should be
monitored.
In severe renal impairment with creatinine clearance less than 10 mL/min, reduction of
isoniazid dose to 200mg is recommended.1,6,10 Serum concentration of streptomycin is
allowed at 5 μg/mL and should be given eight hours before dialysis. Ethambutol should
be given eight hours before dialysis and at 25 mg/kg, three time a week when creati-
nine level is between 50 to 100 mL/min; and two times a week when creatinine level is
between 30 to 50 mL/min. Pyrazinamide is recommended at the low end of 15 to 20
mg/kg and requires supplemental dosing if given before dialysis. Generally, anti-
tuberculous drugs should be given after hemodialysis to avoid filtration of the drugs dur-
ing the procedure.2,4 Directly-observed therapy (DOT) should be facilitated in patients
undergoing hemodialysis for better outcome.1,2
171
in HIV-infected children, may depend on degree of immunocompromise, with severe,
disseminated forms more frequently found in patients with advanced HIV infection.
Approach to TB Diagnosis in Pediatric HIV
The clinical approach to TB diagnosis is similar to adults with sputum smear-negative
disease, taking into account the likelihood of TB exposure, proof of Mycobacterium tu-
berculosis (MTB) infection (tuberculin skin test induration of ≥5 mm, or a positive MTB
interferon-gamma-release assay), clinical features, and chest X-ray signs suggestive of
TB, as well as appropriate microbiological evaluation.27,28
The cornerstone of diagnostic methods for LTBI is the TST, administered by Mantoux
test. Because children with HIV infection are at high risk for TB, annual skin testing is
recommended to diagnose LTBI.
TST is less sensitive in children living with HIV than in HIV-negative children; induration
of > 5 mm is considered positive if the child is living with HIV.
Interferon-gamma release assays (IGRAs) should not replace the TSTin low- and mid-
dle-income countries for the diagnosis of latent TB infection in children or for the diag-
nostic work-up of children (irrespective of HIV status) suspected of TB disease in these
settings.29
The WHO has endorsed Xpert MTB/RIF as a primary TB diagnostic test in symptomatic
people living with HIV.30,31 However, Xpert MTB/RIF cannot be used to rule out TB, and
MTB culture remains a necessary diagnostic tool.
172
vices if living in settings with a medium or low TB prevalence ((Conditional recommen-
dation acknowledging resource implications, low quality of evidence)
The WHO recommended that children over 12 months of age and living with HIV
should be screened and be given IPT for tuberculosis.36 Although not a requirement for
starting IPT, TST may be done as part of eligibility screening in some settings.
If a child presents with any of the following symptoms below, an investigation for tuber-
culosis in accordance with existing national guidelines and other diseases should be
done. 36
1. poor weight gain defined as reported weight loss, very low weight ( weight-for-age
less than -3 z-score), underweight (weight-for-age less than -2 z-score), confirmed
weight loss (>5%) since last visit, or growth curve flattening;
2. fever;
3. current cough; and
4. contact history with a TB case
If the case is not TB or there are other diagnoses, follow up patient and give appropri-
ate treatment respectively and consider giving IPT to both cases. If the diagnosis is
consistent with tuberculosis, treat patient accordingly.
If the child does not manifest any of the symptoms, the child may receive IPT if there
are no contraindications like active hepatitis (acute or chronic), and symptoms of pe-
ripheral neuropathy. Past history of TB should not be a contraindication for starting IPT.
All children and infants less than one year old should be provided with IPT if they have
household contact history with an infectious TB case.
2. BCG vaccination in HIV infected children
In children who are known to be HIV-infected, BCG vaccine should not be given be-
cause of the risk of disseminated BCG disease.37 (Recommendation strength and evi-
dence quality have not been graded)
In infants whose HIV status is unknown, are born to HIV-positive mothers, and lack
symptoms suggestive of HIV, BCG vaccine should be given after considering local fac-
tors.37
174
start of ART and pyridoxine supplementation along with nutritional support.
2. Co-trimoxazole Preventive Therapy
Co-trimoxazole (CTX) is a broad-spectrum antimicrobial agent that prevents a range of
secondary bacterial and parasitic infections in eligible adults and children living with
HIV. Daily prophylaxis co-trimoxazole preventive therapy (CPT) prolongs survival in
children living with HIV and reduces the incidence of co-morbidities. It also reduces the
risk of co-infections such as pneumocystis pneumonia in HIV-exposed infants. CPT is
therefore recommended for all HIV-exposed infants and children living with HIV, includ-
ing those with TB41 and should be implemented as an integral component of a package
of HIV- related services.42
CTX prophylaxis is indicated in all HIV exposed infants starting at 4-6 weeks and all
infants with presumptive clinical diagnosis of severe symptomatic HIV until no longer
breastfeeding and HIV has been excluded.43 This strategy may be considered in set-
tings such as TB programs with a high prevalence of HIV and with limited health infra-
structure.
The following confirmed HIV-infected infants and children should receive CTX prophy-
laxis:43
For children aged 1-5 years diagnosed with WHO stages 2,3, and 4 regardless of
CD4% OR any WHO stage and CD4<25% (in resource –limited settings, CTX may
be started when the CD4 count has dropped to <25% at age <5 years or is <350
cells/mm3 at <6 years.)
The aim is to reduce the morbidity and mortality associated with malaria, bacte
rial diarrheal diseases and pneumonia, in addition to the prevention of PCP and
toxoplasmosis. In other settings where the use of CTX is limited to preventing
PCP, CTX may be started when the CD4 count has dropped to <20% at age 5
or is <200 cell/mm3 at >6 years.
For less than 6 years diagnosed with any WHO clinical stage and CD4<350 (same
as in re source-limited settings as above) or with WHO stage 3 or 4 and any CD4
level (asymptomatic children in WHO clinical stage 1 do not require CTX prophylax-
is. However, it is strongly recommended to measure the CD4 count as asymptomat-
ic children may also have laboratory signs of immunodeficiency)
The indications for initiating, discontinuing and monitoring CPT are included in
the 2006 WHO guidelines on co-trimoxazole prophylaxis for HIV-related infec-
tions among children, adolescents and adults: recommendations for a public
health approach.44 The recommended dose of CTX is 150 mg TMP/m2 SMX 750
mg/m2 .45
3. Antiretroviral Therapy
Antiretroviral therapy (ART) in children living with HIV aims to improve the length and
quality of life, reduce HIV-related morbidity and mortality by reducing the incidence of
175
opportunistic infections (including TB), reduce the viral load, restore and preserve im-
mune function, and restore and preserve normal growth and development. ART im-
proves TB treatment outcomes for children living with HIV.
WHO 46 recommends that ART be provided to all people with a confirmed HIV diagno-
sis and a CD4 count of 500 cells/mm3 or less, giving priority to those with severe/
advanced HIV disease or a CD4 count of 350 cells/mm3 or less.
WHO also recommends that ART be initiated in people with active TB and HBV co-
infection with severe liver disease, all pregnant and breastfeeding women with HIV, all
children younger than 5 years living with HIV and all individuals with HIV in serodiscord-
ant relationships, regardless of CD4 cell count.
TB treatment should be started first, followed by ART within 8 weeks of the start of TB
treatment. For those with a CD4 count below 50 cells/mm3, ART should be provided
within 2 weeks of the start of TB treatment.46
For first-line ART, use of simplified and less toxic regimens – as fixed-dose combina-
tions whenever possible – is recommended as the most effective and convenient ap-
proach. Regimens comprising of a non-thymidine nucleoside reverse-transcriptase in-
hibitor (NRTI) backbone (tenofovir disoproxil fumarate (TDF) or abacavir (ABC) + lamiv-
udine (3TC)) and one non-nucleoside reverse-transcriptase inhibitor (NNRTI) efavirenz
(EFV) are maintained as the preferred choices in adolescents and children older than 3
years. For children younger than 3 years, a protease inhibitor (PI)-based regimen is the
preferred approach in combination with ABC or zidovudine (AZT).46
The following is the summary of recommendations on when to start ART in children.46
1. For all infants less than 1 year regardless of CD4 count
2. For one to less than 5 years ( priority are children ≤ 2 years, or with WHO stage 3
or 4, or CD4 count ≤750 cells/mm3 OR <25% )
3. For 5 years and above diagnosed with WHO stage 3 or 4 or CD4≤500 cells/mm3
(priority case if CD4≤350 cells/mm3)
Selecting ARV regimens that are compatible with TB therapy is essential. Interactions
between rifampicin (rifabutin preferred because of less enzyme induction property) and
lopinavir/ritonavir (LPV/r) or nevirapine (NVP) mean that co-treatment in children under
three years is challenging, but a recent large randomized controlled trial of ART in chil-
dren has generated preliminary evidence on the efficacy of triple nucleoside therapy
which, despite limited data in the context of TB co-treatment, offers a suitable option for
children who require TB treatment while already receiving ART. 46, 47
WHO also recommends ART for all patients with HIV and drug-resistant TB requiring
second-line anti-TB drugs, irrespective of CD4 count, as early as possible (within the
first 8 weeks) following the start of anti-TB treatment.46
The recommended regimen for children and infants starting ART while on TB treatment
is as follows:46
176
1. For younger than 3 years, start 2 NRTIs NVP ensuring that dose is 200mg/m2 OR
Triple NRTI (AZT + 3TC + ABC)a
2. For 3 years and above, start 2 NRTIs + EFV OR Triple NRTI (AZT + 3TC + ABC)a
The following is the recommended regimen for children and infants starting TB treat-
ment while on ART.46
1. For children on standard NNRTI-based regimen (2 NRTIs +EFV or NVP) younger
than 3 years, continue NVP ensuring that dose is 200 mg/mm3 or Triple NRTI
(AZT+3TC +ABC)a
2. For children on standard NNRTI-based regimen (2 NRTIs +EFV or NVP) 3 years
and older, may continue regimen if on EFV or If on NVP, change to EFV or Triple
NRTI (AZT+3TC+ABC)a
3. For children on standard PI-based regimen(2 NRTIs +LPV/r) younger than 3 years,
may receive Triple AZT (AZT+3TC+ABC)a or change to NVP ensuring that dose is
200mg/m2 or continue LPV/r and consider adding RTV to achieve full therapeutic
dose (increase RTV until same dose as LPV in mg, in a ratio of 1:1)
4. For children on standard PI-based regimen (2 NRTIs +LPV/r) 3 years and above
and with no history of NNRTI- based regimen failure, change to EFV (as the pre-
ferred option and could be maintained after termination of TB treatment to allow
simplification and harmonization with ARVs regimen in older children) or Triple
NRTI (AZT+3TC+ABC)a or continue LPV/r consider adding RTV to achieve full ther-
apeutic dose (RTV;LPV/r in 1:1 ratio in mg)
5. For children on standard PI-based regimen (2 NRTIs +LPV/r) 3 years and above
and with history of NNRTI- based regimen failure, start on Triple NRTI
(AZT+3TC+ABC)a or continue LPV/r and consider adding RTV to achieve full thera-
peutic dose (RTV:LPV/r in 1:1 ratio in mg). Consultation with experts for construc-
tion of second-line regimen should be considered.
Notes:
Abbreviations used: ABC abacavir; AZT zidovudine; EFV efavirenz; LPV/r lop-
inavir/ritonavir; NNRT non-nucleoside reverse-transcriptase inhibitor; RTV ritonavir;
3TC lamivudine
a
Triple NRTI is recommended only for the duration of TB treatment; an age –
appropriate PI-or NNRTI-based regimen should be restarted at termination of rifam-
picin-based therapy. This regimen should be considered as the preferred option for
children less than 3 years on LPV/r-based regimen when starting TB treatment and
also as the preferred regimen for children older than 3 years with history of NNRTI
failure.
4. Immune Reconstitution Inflammatory Syndrome (IRIS)36
Sometimes known as a paradoxical reaction, immune reconstitution inflammatory syn-
drome (IRIS) is a temporary clinical deterioration that may occur within 3 months of
177
starting ART and most commonly within the first month. It is the result of reconstitution
of cell-mediated immunity in response to mycobacterial antigens and can give rise to
diagnostic and management challenges. Immune reconstitution can also occur with im-
proved nutritional status during anti-TB treatment. It can simulate worsening of TB dis-
ease, with fever and increased size of lymph nodes or tuberculomas. Risk factors for
IRIS include low baseline CD4 count, extensive TB, early initiation of ART, and rapid
immunological and virological responses to ART. Though a cause of significant morbid-
ity, TB-IRIS and BCG-IRIS are not associated with an increased mortality risk.
With respect to TB, there are two main presentations: exacerbation of known TB dis-
ease in a child living with HIV who started anti-TB treatment and ART; or development
of TB disease in a child starting ART.
In all cases, anti-TB treatment should be continued; the addition of corticosteroids may
sometimes be useful. If there is any doubt, the child should be referred to the next level
of care.
Tuberculosis in non-HIV immunosuppressed patients
Patients who are immunosuppressed have increased susceptibility to tuberculosis in-
fection and disease. The risk associated with each cause of immunosuppression var-
ies, but is most profound with drug-induced immunosuppression in the management of
malignancy and organ transplantation, and is milder in diabetes mellitus. In some dis-
eases, the risk represents the dual effects of susceptibility conferred by the disease
state and its treatment. The risk, presentation, investigation and approach to manage-
ment varies according to the cause.
Diabetes Mellitus
The increased risk of developing TB in diabetes mellitus (DM) varies according to the
background prevalence and various host factors, including patient's age and sex, body
mass, duration of diabetes and most importantly, adequacy of glycemic control. TB may
result in impaired glucose tolerance that improves/resolves after successful treatment.
In pediatrics, this host factor of concern is the a) duration of diabetic disease; b) male
sex considered to be greater risk of TB than females; c) glycemic control; and d) body
mass. Most important determinant of developing TB is the level of diabetic control that
patients maintain. Increased risk of smear-positive disease has been demonstrated at
HbA1c levels of 9% or more. Low body mass in younger ones has been demonstrated
to be an independent risk factor for TB. Active TB may also be associated with impaired
glucose tolerance that improves/resolves after successful TB chemotherapy.50 This im-
pairment maybe transient; thus, patients diagnosed with diabetes while on TB treat-
ment should be reviewed once treatment has been completed.
1. Clinical Features of TB in Diabetes Patients
Clinical features and presentations are similar to those without diabetes. Unusual
presentation is the higher incidence of extra-pulmonary disease (i.e. laryngeal TB).
Some of the symptoms of TB are similar to poorly controlled diabetes and its co-
morbidities, hence the diagnosis of TB maybe overlooked or delayed.
178
BOX 13.2. Comprehensive history and evaluation of the tuberculosis-diabetes
mellitus patient49
Diabetes History
Type and onset of diabetes.
Diabetic treatment history and drug administration and supply.
Level of prior diabetes education.
Dietary history and eating plan.
Monitoring and Glycemic control.
Episodes and symptoms of hypoglycemia
Evidence of target organ damage.
Micro vascular: renal, ocular, neuropathy.
Macro vascular: cardiac, arterial disease, stroke.
Other: gastroparesis, impotence
Tuberculosis history
Duration and symptoms.
Presence of Extra pulmonary disease
Presence of complication of the disease
Smoking and prior lung disease
179
these reports, others have described better patient compliance and treatment success
rates in diabetics.13 However, relapse rates might also be higher.56
180
BOX 13.3. History and clinical signs suggesting bronchial malignancy in patients
with tuberculosis49
A history of smoking and previous bronchial or upper respiratory tract malignancy.
Failure to gain weight on therapy especially in patients with drug- susceptible M.
tuberculosis on initial cultures.
The presence of persistent chest pain.
Ongoing or new hemoptysis while on TB treatment.
Hoarseness of new onset.
Persistence of radiological infiltrates or appearance of new ones.
In patients with TB and suspected malignancy, repeated sputum cytology has been
shown to improve the diagnostic yield but this is not feasible when hemoptysis is pre-
sent and or when patients are unable to expectorate.60 The latter can be overcome in
some by use of sputum-inducing methods, but fine needle aspiration under radiograph-
ic guidance (preferably using a CT scan) or bronchoscopy has the highest success rate
in confirming the diagnosis.
2. Treatment and Prognosis
Tuberculosis occurring in malignancy is treated along standard lines, but attention must
be paid to potential drug interactions and side effects in patients who are debilitated by
cancer or on aggressive chemotherapy or radiotherapy. Where surgery for cancer is
planned, it is advisable to delay this for at least some weeks until TB chemotherapy has
been started. Where possible, a longer delay and even completion of TB treatment is
advised in patients scheduled to receive chemotherapy and bone marrow transplanta-
tion. However, this must be weighed against the impact of such delays upon prognosis
of the malignancy.
3. Complications
Clinicians prescribing TB medication and following up patients with co-morbid disease
should be attentive to potential drug interactions between TB drugs and multiple
chemotherapeutic agents; the effects of nausea and vomiting on drug absorption; the
effects of general debility and impaired mobility on drug collection and treatment com-
pliance; the risk of transmission of TB from these to other susceptible patients in clinic
and hospital facilities serving patients with cancer and hematological diseases; and the
development of other cancer and treatment-related lung disease including: opportunis-
tic lung infections such as fungal pneumonia, drug-induced lung disease, radiation lung
injury, and pulmonary thromboembolic disease.
181
volvement.61-65 Corticosteroids can also be used for treating the paradoxical response
and IRIS associated with TB treatment, especially in patients on antiretroviral therapy.66
SLE and collagen vascular diseases comprise a major category of disease in which
corticosteroids are used – either as a long-term remittive agent or for the treatment of
flare-ups of acute arthritis in rheumatoid arthritis and SLE. The risk of TB appears to
vary by disease, indication and dosing schedule. It is greater in patients with lupus than
those with rheumatoid arthritis,67 and is more common in patients with organic brain
syndrome, vasculitis and nephritis, and in patients who receive intravenous ‗pulse‘
methylprednisolone or high cumulative doses of prednisolone.68 The use of prophylactic
isoniazid (INH) does not appear to effectively prevent recurrence of TB in lupus patients
in highly endemic areas, and may be associated with an increased risk of lupus flare.
In asthma and chronic obstructive pulmonary disease (COPD), inhaled corticosteroids
are used in patients and severe COPD. In addition, short courses of systemic cortico-
steroids are used for exacerbations of both diseases. Inhaled corticosteroids are also
considered to be safe in children and adults with asthma who are tuberculin-positive
and there is no evidence of increased risk of tuberculous infection or disease.69
In diffuse parenchymal lung disease, systemic corticosteroids are the most widely used
treatment like sarcoidosis, idiopathic pulmonary fibrosis, lymphocytic interstitial pneu-
monitis, cryptogenic organizing pneumonia and non-specific interstitial pneumonia.
Studies of the potential risk associated with their use in these diseases are confounded
by several factors. Firstly, studies from places with both high and low prevalence of TB
have confirmed an increased incidence of TB in patients with diffuse parenchymal lung
disease unrelated to treatment.70 Secondly, the presence of diffuse radiographic chang-
es and frequent follow-up provided to these patients have an impact, both positive and
negative upon the detection and diagnosis of intercurrent TB. Thirdly, for sarcoidosis,
there is a long-standing debate concerning a possible etiological role for MTB, which, if
accepted, would alter the interpretation of TB as a complication of treatment.71,72 How-
ever, cases of TB developing in patients with sarcoidosis receiving corticosteroids have
been reported.73,74
1. Clinical features and Investigations
Tuberculosis associated with corticosteroid treatment frequently presents in unusual
forms, or as extra-pulmonary or disseminated disease. A high index of suspicion is re-
quired, particularly in high-prevalence countries, and the general approach and selec-
tion of investigations is similar to that for other immunocompromised patients. In those
with interstitial disease, detection of new pulmonary lesions, particularly if these are
nodular or miliary, is a particular problem. Computed Tomography (CT) scanning is of
value, not only for defining new lesions but also for seeking patterns of specific diag-
nostic value. These include cavitation and new pleural involvement as features of TB,
aspergillomas in pre-formed cavities and sequestra suggesting locally invasive asper-
gillosis.
2. Treatment and Prognosis
The treatment and outcome of TB patients occurring in association with corticosteroid
use is similar to those without such treatment. However, in those with pre-existing lung
182
disease, the combined effects with TB may result in significant long-term loss of pulmo-
nary reserve and/or development of bronchiectasis and susceptibility to infections. Sup-
portive care and even domiciliary oxygen might be necessary.
3. Prevention
Recommendations concerning INH prophylaxis in patients on systemic corticosteroids
vary and are tentative because of lack of firm evidence on the doses of corticosteroids
at which the risk increases, and a similar absence of cost/benefit analyses of chemo-
prophylaxis in this setting. Any benefits of treatment must be weighed against the risk
of side effects, especially hepatotoxicity. The American Thoracic Society Statement
notes these facts and suggests that chemoprophylaxis is unnecessary for patients re-
ceiving moderate doses of oral prednisolone up to 10 mg per day, as the risk of devel-
oping TB is uncertain, even in high-prevalence communities.70,75 According to these
guidelines, chemoprophylaxis should be considered for patients receiving doses higher
than this. Chemoprophylaxis is not necessary for patients receiving inhaled corticoster-
oids.
183
several affected segments of lung and collection of a post-bronchoscopy sputum sam-
ple provides the highest yield. Transbronchial and/or endobronchial biopsies are also
advised, especially if alternative diagnoses are being considered, but these increase
the risk of hemoptysis, pneumothorax and other complications of the procedure.
A less invasive option, suitable for patients with accessible discrete lesions that can be
accurately located on CT scanning, is transthoracic fine needle aspiration. Transbron-
chial needle aspiration is also useful where hilar or mediastinal nodes are observed. In
view of the risks of bleeding, particularly in renal transplant recipients, measurement of
blood counts including hemoglobin, platelet count and international normalized ratio
(INR) as well as bleeding time are necessary.
Biopsies and needle aspiration should not be attempted on patients with abnormal INR,
low platelet count or prolonged bleeding times, and if other attempts to make the diag-
nosis fail and, upon review of risks and benefits, these procedures are still considered
essential, the abnormality should be corrected using standard measures before they
are attempted.77
Depending on the suspected organ involvement of TB, other tests might be required.
These include liver function tests, ultrasound examination of the heart, and CT scans of
abdomen and chest. Examination of urine for sterile pyuria, a feature in renal TB, ex-
amination of cerebrospinal fluid for suspected meningitis and biopsies of liver, skin le-
sion, lymph node and less obvious sites like the synovium and vertebra may be re-
quired. An aggressive approach is often necessary since the differential diagnoses in-
clude other opportunistic infections such as disseminated aspergillosis that requires
specific therapy, and malignancies (lymphomas, Kaposi‘s sarcoma and both solid or-
gan and hemopoeitic malignancies) as these are more common in transplant recipients.
3. Isoniazid prophylaxis in solid organ transplantation
All transplant patients should receive a tuberculin skin test prior to transplantation. Ap-
proximately 70% have negative reactions but those that are positive are at greater risk.
The recommendations for INH chemoprophylaxis in renal and heart transplant recipi-
ents are described below. These recommendations are based on the fact that these
patients do not appear to be at a higher risk of developing isoniazid hepatotoxicity than
the general population, that the case fatality rate for transplant recipients is almost
30%, and that in a further similar proportion the allograft is rejected. These dire conse-
quences of untreated dormant disease argue for chemoprophylaxis, at least in low-
prevalence areas.
184
BOX 13.4. Indications of isoniazid prophylaxis for solid organ transplant recipi-
ents49
1. Tuberculin reactivity > 5mm before transplantation.
2. Patients with the following characteristics regardless of reactivity:
Radiographic evidence of old TB and no prior prophylaxis;
History of inadequately treated TB;
Close contact with an infectious patient; and
Recipient of an allograft from a donor with a history of untreated TB
or tuberculin reactivity without adequate prophylaxis.
3. Newly infected persons (recent conversion of tuberculin test to positive).
185
and 0.4% of patients who have undergone this form of surgery (with most of whom re-
siding in countries with a low incidence of TB) develop TB. Tuberculosis following jeju-
noileostomy is usually extrapulmonary: 82% in one series.81 In a small case series of
seven cases, the mean interval between surgery and the diagnosis of TB was 16
months.82 Initial symptoms included fever and rapid weight loss. Weight loss is often
observed as a second period of decline after stabilization of the initial phase of weight
loss resulting from the bypass surgery.81
1. Management
Chest radiography and TST should be performed on all patients being considered for
bypass surgery.82 Tuberculous chemoprophylaxis is advised for patients with a positive
TST response measuring 10 mm or more prior to surgery, which should be delayed for
at least 6 months. As for patients with TB following gastrectomy, therapeutic drug moni-
toring should be performed because drug absorption in patients following jejunoileal
bypass surgery is often unpredictable.
Tumor necrosis factor-a (TNF-A)49 is a pro-inflammatory cytokine that exists in trans-
membrane and soluble forms and acts as a ligand that stimulates apoptosis. Through
its receptor binding, particularly to receptor TNFR1, it is involved in host defenses
against intracellular organisms like MTB, by enhancing intracellular killing and stimulat-
ing granuloma formation, thereby limiting dissemination. However, excessive produc-
tion of TNF-a can have detrimental effects, including the development of caseous ne-
crosis and excessive wasting in patients with TB. The most extensively studied anti-
TNF-a agents are infliximab, etanercept and adalimumab.
Infliximab is a humanized murine chimeric monoclonal antibody with high bind
ing affinity and specificity for TNF-a that forms stable complexes with monomer
ic and trimeric forms of TNF-a, crosslinking between molecules. It also binds to
transmembrane TNF-a. Infliximab is administered as an intravenous infusion at
weeks 0 and 2 of therapy followed by an infusion every 8 weeks. Its half-life is
10.5 days. It can be used for treatment of rheumatoid arthritis, Crohn‘s disease,
ankylosing spondylitis and psoriatic arthritis.
Etanercept is a dimeric fusion protein that forms less stable complexes with
monomeric and membrane bound TNF, but binds well to trimeric TNF-a and
forms stable complexes with TNF-b. It is given twice weekly as a subcutaneous
injection and has a shorter half-life of approximately 3 days.
a. Epidemiology of anti-TNF therapy and tuberculosis
The association between anti-TNF treatment and increased risk of TB was first ob-
served in post-approval pharmacovigilance data from patients with rheumatoid arthritis
and other autoimmune conditions treated with these agents. This was later confirmed
by information gathered by the Spanish Society of Rheumatology. It appears that when
used in countries other than the USA, the risk is positively associated with the preva-
lence of TB in the general population. The onset of TB is earlier with infliximab than
with etanercept, and dissemination is common. Reactivation of latent TB infection
(LTBI) rather than reinfection is often the suspected cause of disease.86
186
The Food and Drug Administration has estimated that in the USA, the age-adjusted in-
cidence of TB in all patients exposed to anti-TNF-a agents is 8.2 cases per 100,000
patient years of exposure. The British Thoracic Society estimates that the risk of TB in
patients with rheumatoid arthritis treated with infliximab is increased five-fold.80 Com-
parisons of the rates of developing TB with these two agents should be interpreted with
caution as these are influenced by the medical indication for which the agent was giv-
en, duration of treatment, concurrent therapy and other risk factors for TB. However, it
appears that the risk is higher with infliximab than with etanercept given these limita-
tions of interpretation.
b. Assessment of patients being considered for Anti-TNF-a treatment
Risk assessment for TB infection should be performed in a patient eligible for anti-TNF-
a therapy comprising a history, physical examination, chest radiology and tuberculin
skin test.
Patients should be questioned on their TB history including BCG vaccination, results of
previous TST, previous treatment for TB and outcome of treatment. When anti-TNF-a
treatment is commenced, inquiry into symptoms of TB should be made at each visit,
and features of TB, including extrathoracic manifestations – nodes, organomegaly and
abdominal ascites – which occur in over 50% of cases, should be sought.
Chest radiography
The chest radiograph is part of the minimum requirement in the work-up of patients for
therapy as there is evidence that TST alone, even when steroid therapy is stopped a
week prior to skin testing, may not be sufficiently sensitive to identify latent TB in the
target group of patients (rheumatoid arthritis, spondyloarthropathies and patients with
inflammatory bowel disease).85 However, as the chest radiograph will be normal in
most patients with LTBI, it has limited usefulness as a screening test. Three outcomes
with the radiographic investigation can potentially be expected.
rifampicin plus isoniazid for 3 months (3RH). This approach is thought to be associ-
ated with better adherence, but a disadvantage is the higher rate of drug-induced
hepatitis in the face of co-administration of other potentially hepatotoxic immuno-
suppressive drugs.
Liver function testing (aminotransferases) is recommended at the start of chemopro-
phylaxis and should be monitored at least every 3 months. If the regimen contains ri-
fampicin, the dose of prednisone (if in use) will need to be doubled. In patients with a
normal chest radiograph and who are not tuberculin test-assessable (because of con-
current immunosuppressive therapy), it may be started at the same time as chemopro-
phylaxis. However, a pragmatic approach is, when possible, to defer anti-TNF-a treat-
ment until chemoprophylaxis is complete.
d. Management of TB in patients on anti-TNF-a therapy
Current evidence suggests that patients who are currently receiving anti-TNF-a therapy
should also receive full standard anti-TB chemotherapy. In this situation, where a pa-
tient is already on anti-TNF-a therapy and withdrawal might place them at risk of dis-
ease deterioration, the patient may continue therapy provided that there is satisfactory
evidence of response to TB treatment.
188
ment; in those with major side effects the relevant drug should be stopped and usually
the patient needs hospitalization.
Factors influencing side effects of anti-TB treatment: age, HIV infection, alcohol and
substance abuse, nutritional status and diabetes. Old age is a risk factor for hepatotoxic
side effects of anti-TB treatment. The toxic effects of aminoglycosides on kidney func-
tion, balance and hearing are more frequent in old age. A serious concern in young
children is that they may be unable to communicate with caregivers regarding sensory
side effects such as reduced hearing (aminoglycosides), vision (ethambutol, isoniazid)
or peripheral neuropathy (isoniazid). Similar logic applies to a certain extent in the treat-
ment of pregnant women, since one will be unable to assess the occurrence of side
effects in the fetus. The main problem with alcoholic TB patients is ensuring treatment
compliance. Patients consuming significant quantities of alcohol may be more prone to
side effects, such as hepatitis. In the event of worsening liver function on anti-TB treat-
ment, it may be challenging to diagnose whether the liver damage is due to alcohol or a
result of anti-TB drug-induced hepatotoxicity. Malnutrition and particularly vitamin defi-
ciencies may increase the risk of side effects of anti-TB treatment. Most notably, lack of
B vitamins, such as pyridoxine, increases the risk of peripheral neuropathy, and proba-
bly also other neurological side effects. Visual impairment, renal failure and peripheral
neuropathy are common complications in patients with diabetes. Diabetic renal failure
may increase the risk of renal failure as a side effect of anti-TB therapy. Ethambutol-
induced optic neuritis may be more difficult to detect in a patient with diabetic retinopa-
thy. The consequences of even modest visual reduction due to ethambutol have great-
er significance if vision is already impaired by diabetic complications.
Symptom-based approach to managing minor and major side effects of anti-TB
drugs49
Table 13.1 Minor side effects
Symptom Drugs Management
Minor gastrointestinal up- Pyrazinamide, rifampicin Continue anti-TB drugs,
set, anorexia, nausea, check doses.
abdominal pain
Give drugs with small
meal or just before going
Joint pains Pyrazinamide Aspirin
Non-steroidal anti-
inflammatory drugs
Burning sensation in the Isoniazid Pyridoxine 100 mg daily
Orange/red urine Rifampicin Harmless
Reassurance
Inform patient at start of
treatment
189
Table 13.2 Major side effects
Symptom Drugs Management
Itching, skin rash Thioacetazone Stop anti-TB drugs
(streptomycin, isoniazid,
rifampicin, pyrazinamide)
Deafness (no earwax on Streptomycin Stop streptomycin and
otoscopy) give ethambutol instead.
Dizziness, vertigo, nystag- Drug: Streptomycin Stop streptomycin, use
mus ethambutol.
Jaundice (other causes Isoniazid, pyrazinamide, Stop anti-TB drugs
excluded), hepatitis Strep- rifampicin
tomycin
Confusion Most anti-TB drugs Stop anti-TB drugs.
Suspect acute liver failure
if jaundice is present.
Urgent liver function test
and prothrombin time
Visual impairment (other Ethambutol Stop ethambutol
causes excluded)
Shock, purpura, acute re- Rifampicin Stop rifampicin
nal failure
190
Generalized erythematous rash
a) If there is a generalized erythematous rash, especially if it is associated with fever
and/or mucous membrane involvement, all drugs should be stopped immediately. If the
patient has severe tuberculosis, three new drugs (e.g., an aminoglycoside and two oral
agents) should be started. When the rash is substantially improved the medications can
be restarted one by one, at intervals of 2–3 days. RIF should be restarted first (because
it is the least likely to cause rash, and it is the most important agent), followed by INH,
and then EMB or PZA. If the rash recurs the last drug added should be stopped. If no
rash appears after the first three drugs have been restarted, the fourth drug should not
be restarted unless the rash was relatively mild and the fourth drug is considered es-
sential for therapy. 1
If a reaction occurs during drug rechallenge (see rechallenge dose table under Hepati-
tis) and the causative drug cannot be discontinued, refer to specialist for drug desensiti-
zation. Drug desensitization should not be attempted with severe skin reactions or
those involving the mouth or mucous membranes (e.g. exfoliative dermatitis and Ste-
vens- JohnsonSyndrome).48
*Although nausea and vomiting are uncommon adverse effects of the aminoglycosides
(streptomycin, amikacin, kanamycin) and capreomycin, it may be an indication of vestibular tox-
icity (inner-ear toxicity). If this occurs, contact a TB Control physician.
191
Does the child take the medicine on an empty stomach? If so, give medications af-
ter meals (e.g. ask school nurse to administer medications after lunch).
Management
Food and drug administration
Isoniazid and rifampin should be administered 1 hour before or 2 hours after food in-
gestion for maximum drug absorption. If nausea and/or vomiting occurs, administer iso-
niazid and rifampin with food (better to give the drug with food and have some de-
creased absorption than to not have the patient ingest the drug at all because of the
side effect). All other TB medications can be administered without regard to food. Op-
tions for patients who cannot swallow tablets and capsules (some adults and infants/
children)
Liquid preparations
Availability: Isoniazid is the only commercially available liquid product. Ri-
fampin and pyrazinamide suspensions can be prepared from the tablets/
capsules. Ethambutol suspensions cannot be prepared because of drug
stability problems
Limitations of liquid preparations: The volume of the liquid required for each
dose may be too large for the patient to tolerate (especially in infants and
children). Diarrhea may occur due to the lactose and sucrose content in liq-
uid preparations. Prepared suspensions have limited stability and some sus-
pension are not palatable (bitter tasting)
192
The initial approach to gastrointestinal intolerance, not associated with hepatic toxicity,
is to change the hour of drug administration and/or to administer the drugs with food. If
patients are taking daily DOT, the timing of the drug administration should be altered,
preferably to be closer to meal time. Alternatively, food can be taken at the time of DOT
administration. (In many programs food is offered as an incentive with DOT.) Patients
receiving self-administered therapy can take the medications at bedtime. If gastrointes-
tinal intolerance persists, it may be best for all medications to be taken with meals.1
Diarrhea
Table 13.4 Causative Agents: Ranked by frequency
+++++ Clofazimine, ethionamide, para-aminosalicylic acid (PAS)
+++ Rifampin
++ rifabutin, ofloxacin, levofloxacin
+ isoniazid, ethambutol, pyrazinamide, rifapentine, cycloserine, amino-
glycosides, capreomycin
1. Management
Withhold drugs until diarrhea resolves. Then, restart drugs one at a time every 4 days
beginning with drugs that are least likely to cause diarrhea. If the patient was receiving
a twice or thrice weekly regimen when the diarrhea began, consider switching to a 5x/
week regimen. If diarrhea recurs when one particular drug is added to the regimen,
consider discontinuing the causative agent and adding other TB drugs and/or extend-
ing the duration of treatment. If diarrhea occurs with multiple drugs, consider separat-
ing medication administration times. Different drugs in the regimen should be adminis-
tered several hours apart. Do not split doses for individual drugs (possible exceptions:
ethionamide, ofloxacin). For example: administer INH 200mg in the morning and rifam-
pin 300mg in the evening. If diarrhea continues and an alternate regimen cannot be
utilized consider the addition of an anti-motility agent (loperamide). Adsorbents (kaolin-
pectin, polycarbophil) should not be prescribed because decreased absorption of the
TB drugs may occur.
Hepatitis
Hepatitis is not a common adverse effect in children on anti-TB treatment, but is proba-
bly the most serious. Three of the first-line anti-tuberculosis drugs, INH, RIF, and PZA,
can cause drug-induced liver injury (AST level three or more times the upper limit of
normal in the presence of symptoms, or five or more times the upper limit of normal in
the absence of symptoms).
If the AST level is less than 5 times the upper limit of normal, toxicity can be considered
mild; an AST level 5--10 times normal defines moderate toxicity; and an AST level
greater than 10 times normal (i.e., greater than 500 IU) is severe. In addition to AST
elevation, occasionally there are disproportionate increases in bilirubin and alkaline
phosphatase. This pattern is more consistent with rifampin hepatotoxicity.
193
Clinical Presentation
Symptoms: nausea, vomiting, abdominal tenderness, discomfort near the ribs on
the right upper abdomen, jaundice (yellowing of skin and whites of the eyes)
Signs: hepatic enlargement, increased liver function tests (LFTs)
2. Causative Agent
INH + Rifampin > INH alone >> pyrazinamide* alone > rifampin alone > ethiona-
mide
*Early reports of pyrazinamide hepatotoxicity occurred in patients who received 40-50mg/kg/d
for prolonged periods. Hepatotoxicity has not been reported with extensive use of lower doses
(15-30mg/kg/d) in short course regimens.
3. Management
a. Asymptomatic patients with an increase in LFTs from baselines
It is important to note that an asymptomatic increase in AST concentration occurs in
nearly 20% of patients treated with the standard four drug regimen. In the absence of
symptoms therapy should not be altered because of modest asymptomatic elevations
of AST, but the frequency of clinical and laboratory monitoring should be increased. In
most patients, asymptomatic aminotransferase elevations resolve spontaneously.
However, if AST levels are more than five times the upper limit of normal (with or with-
out symptoms) or more than three times normal in the presence of symptoms, hepato-
toxic drugs should be stopped immediately and the patient evaluated carefully. Similar-
ly, a significant increase in bilirubin and/or alkaline phosphatase is cause for a prompt
evaluation. Serologic testing for hepatitis A, B, and C should be performed and the pa-
tient questioned carefully regarding symptoms suggestive of biliary tract disease and
exposures to other potential hepatotoxins, particularly alcohol and hepatotoxic medica-
tions. Drug-induced hepatitis is usually a diagnosis of exclusion but in view of the fre-
quency with which other possible causes are present in any given patient, determining
the cause may be difficult.
Because the schedule for restarting anti-tuberculosis medications is slower with hepati-
tis than for rash or drug fever it is generally prudent to give at least three non-
hepatotoxic anti-tuberculosis drugs until the specific cause of hepatotoxicity can be de-
termined and an appropriate longer term regimen begun.
The suspect anti-tuberculosis medications should be restarted one at a time after the
AST concentration returns to less than two times the upper limit of normal. (In patients
with elevated baseline AST from preexisting liver disease, drugs should be restarted
when the AST returns to near baseline levels.) Because RIF is much less likely to
cause hepatotoxicity than is INH or PZA, and is the most effective agent, it should be
restarted first. If there is no increase in AST after about 1 week, INH may be restarted.
PZA can be started 1 week after INH if AST does not increase. If symptoms recur or
AST increases, the last drug added should be stopped. If RIF and INH are tolerated,
and hepatitis was severe, PZA should be assumed to be responsible and should be
discontinued. In this last circumstance, depending on the number of doses of PZA tak-
en, severity of disease, and bacteriological status, therapy might be extended to 9
months.
194
b. Symptomatic patients
Hold all drugs and obtain Liver Function Tests (LFTs). If LFTs are within normal ranges,
refer to the Management of Nausea/Vomiting section. If LFTs are elevated, hold drugs
until symptoms resolve and the transaminases decreases to < 2x normal. Ethambutol
and pyrazinamide should be started if drug therapy cannot be held secondary to the
patient‘s clinical condition. Use streptomycin if pyrazinamide is suspected to be the
cause of hepatotoxicity.
Rechallenge the patient after resolution of signs and symptoms by adding drugs to the
regimen every 4 days: (See Drug Rechallenge Initial Drug Dose below). Start Rifampin
for 3 days, if patient remains asymptomatic, then add INH for 3 days; if patient remains
asymptomatic, then add pyrazinamide (Initial dose 15-20mg/kg/d) for 3 days.
c. Timing of reintroduction of anti-tuberculosis drugs
If blood tests for assessing the liver function are available, reintroduction of anti-TB
drugs can be started as soon as the liver tests have normalized. However, in many high
-burden settings, laboratory testing facilities are scarce and liver function testing may
not be possible. In these situations, treatment will be guided by the symptoms. Usually,
jaundice and other symptoms will subside in 1 to 2 weeks. The WHO recommends that
the regular anti-TB drugs be reintroduced 2 weeks after jaundice has disappeared.
If signs and symptoms recur with rechallenge, discontinue the responsible drug and
modify the regimen and/or duration of therapy as required.
BOX 13.5 Drug Rechallenge Protocol: Sequence of reintroduction of antitubercu-
losis drugs
Drug sequence (Initial Dose Day 1): Likelihood of drug causing the reaction
INH (50mg): +
RIF (75mg): ++
PZA (250): +++
EMB(100mg): +
SMZ(125mg): ++
Drug fever
Recurrence of fever in a patient who has been receiving therapy for several weeks
should suggest drug fever, especially if the patient is showing microbiological and radi-
ographic improvement. It should be noted, however, that fever from tuberculosis may
persist for as long as 2 months after therapy has been initiated. Fever may also be a
manifestation of a paradoxical reaction, especially in patients with HIV infection The
clinical hallmark of drug fever is that the patient looks and feels well despite having a
high fever (often greater than 39°C). There is no specific pattern to the fever. Eosino-
philia may or may not be present.
The first step in management of a possible drug fever is to ensure that there is no su-
perinfection or worsening of tuberculosis. If these potential causes are excluded all
drugs should be stopped. Drug-related fever usually will resolve within 24 hours. Pa-
tients with severe tuberculosis should be given at least three new drugs in the interim.
195
Once the fever has resolved, the same protocol as described above for restarting drugs
in the presence of a rash should be followed.
Miscellaneous Adverse Effects
1. Arthalgias (joint pain)
Symptoms: Pain and tenderness of joints, fingers, shoulders, knees, etc. (usually
mild)
Causative Agents: pyrazinamide>>ethambutol>isoniazid
Management: TB medications do not require discontinuation. Low dose non-
steroidal anti-inflammatory agents (NSAIDS) can be used for pain relief as needed.
If symptoms persist, consider referral for rheumatologic evaluation.
2. Arthalgias Type 2 (Gouty Arthritis)
Causative Agents: pyrazinamide>>ethambutol
Symptoms: pain, tenderness and swelling of joints: fingers, shoulders, knees, etc.
(usually severe)
Signs: elevated serum uric acid concentrations
Management:
TB medications usually do not require discontinuation. If acute swelling is
present, the affected joint should be aspirated and examined for urate crys-
tals to confirm the diagnosis of acute gouty arthritis.
Low-dose non-steroidal anti-inflammatory agents (NSAIDS) can be used for
pain relief, which includes: indomethacin, ibuprofen, and naproxen. Colchi-
cine is an alternative to NSAIDS; the dose is 0.5-1.2 mg x1, then 0.5-0.6 mg
q 1-2 hours until joint pain is relieved or nausea, vomiting or diarrhea oc-
curs. Pain usually resolves after 4-8 mg cumulative dose. Maximum dose is
8mg. A steroid may be required for severe attacks. Recurrent episodes may
occur while the patient remains on pyrazinamide or ethambutol. Consider
using prophylactic colchicine at 0.6mg one to two times daily and continue
until causative agent is discontinued. Consider referral for rheumatologic
evaluation for acute gouty arthritis attacks.
3. Influenza Syndrome
Symptoms: fever, headache, bone pain usually occurs 1-2 hours after drug admin-
istration (usually resolves within 12 hours of drug administration)
Causative Agents: rifampin > rifabutin (intermittent regimens > daily regimens)
Management:
More common with high dose (>1200mg), intermittent therapy than with dai-
ly dosing. Ιt occurs in 10% of patients receiving 600mg twice weekly and
may also occur with daily therapy when administered irregularly (e.g. in
noncompliant patients).
Switch from intermittent therapy to daily dosing. Symptomatic therapy may
be required when switching from intermittent to daily therapy to prevent the
reaction with initial doses. Usually presents after 3-6 months of inter-
mittent therapy and 1 to 2 hours after rifampin administration. Resolution of
symptom usually occurs within 12 hours
196
4. Neurotoxicity
a. Peripheral Neuropathy
Causative Agents: INH>>>ethambutol
Clinical Presentation:
Prickling, tingling or burning sensation of the fingers and/or toes
usually occurs in a glove-and-stocking distribution.
Management
peripheral neuropathy rarely occurs in children unless severe malnutrition is
present
peripheral neuropathy is uncommon if the patient is receiving pyridoxine
(vitamin B6)
if peripheral neuropathy occurs, it can be treated with pyridoxine 100-200mg
po q daily while the patient is receiving INH.
b. Nervous System Effects in Children
Causative Agent: INH
Clinical Presentation: Drowsiness or hyperactivity, dizziness, tonic-clonic seizure
(rare)
Management
Drowsiness: Make sure the dose does not exceed 10mg/kg/day and add
pyridoxine 50mg to the regimen. Administer medications around afternoon
naps or at bedtime (e.g. administer school DOT at the end of the day so the
child can take a nap after school)
Hyperactivity: Make sure the dose does not exceed 10mg/kg/day. Switch to
twice weekly dosing as soon as possible. If the child becomes hyperactive
only on the days of medication administration, then the medication is the
cause. Add pyridoxine 50mg daily for 6 weeks, then twice weekly for the re-
mainder of therapy.
Dizziness: Make sure the dose does not exceed 10mg/kg/d
Tonic/clonic seizures: Hospitalize the child and administer isoniazid to docu-
ment the reaction. If a seizure occurs, discontinue isoniazid and add an al-
ternative agent to the regimen
c. Optic Neuritis
Causative Agents: ethambutol >>INH
Clinical Presentation
Blurred vision (decrease in the ―sharpness‖ of objects)
―spots‖ present in patient‘s field of vision
red/green color blindness
Management: Children with complaints of vision changes should be referred to spe-
cialist for evaluation and discontinue the drug.
197
REFERENCES:
1. American Thoracic Society.CDC,MMWR. Treatment of tuberculosis infection in
adults and children.20 June 20 2003;52(RR-11).
2. Diagnosis, Treatment, Prevention and Control of Tuberculosis in Adult Filipinos: Up-
to-date.2006.
3. Red Book,2009 Report of the Committee of Infectious Diseases,28th.ed.
4. TB Guideline.Available from : http://www.CDC.gov/tb/pubs/PDA
5. American Family Physician. Identification and Management of Tuberculosis.1 May
2000.
6. Philippine Pediatric Society. Philippine Coalition Against Tuberculosis,Pediatric In-
fectious Disease Society of the Philippines,National Consensus on Childhood Tu-
berculosis,1997.
7. Philippine Pediatric Society,Committee on Handbook on childhood Tubercul-
sis.Tuberculosis in Infancy and Childhood.2003.
8. Davis PDO,(ed).Clinical Tuberculosis.London:Chapman and Hall;1994:151-152.
9. Nolan CM,Goldberg SV.Hepatotoxicity associated with Isoniazid preventive thera-
py.JAMA.1999;281:1014-1018.
10. Rossman MD,McGregor RR.Tuberculosis: Clinical Management and New Chal-
lenges.McGraw Hill Inc; 1995.
11. Snider DE,Powell KE.Should women taking antituberculosis drugs breastfeed?Arch
Intern Med 1984;144:589-90.
12. National Tuberculosis Control Program Manual of Procedures, 5th edition
13. Bergeron KG, Bonebrake RG, Gray CJ. Tuberculosis in pregnancy: current recom-
mendations for screening and treatment in the USA. Expert Rev Anti Infect Ther
2004;2:589–98.
14. Redbook 2015 Report of the Committees on Infectious Diseases 30th ed, p.821.
15. Khilnani GC. Tuberculosis and pregnancy.Ind J Chest Dis Allied Sci 2004; 46(2):
105-11.
16. Cantwell MF,Sehab ZM,Costello AM et al.Brief report: congenital tuberculosis. New
Eng J Med 1994;330:1051-54.
17. WHO treatment of tuberculosis guidelines.2009.3rd edition.
198
18. Kumar N, Kedarisetty CK,Kumar S, Khillan V, Sarin SK. Antitubercular therapy in
patients with cirrhosis: Challenges and Options.World J Gasroenterol 2014 May
21;20(190: 5760-5772.
19. American Thoracic Society,CDC, Infectious Disease Society of America.Treatment
of tuberculous.Morbidity and mortality weekly report:recommendations and reports.
2003;52(rr-11):1-77.
20. WHO treatment of tuberculosis guidelines.4th edition.2010.p96-7.
21. American Thoracic Society and Center for Disease Control and Prevention. Target-
ed tuberculin testing and treatment of latent tuberculosis infection.AM J Respir Crit
Care Med.2000;161:S221-47.
22. Kimmoun E, Samuel D.Antituberculous drugs in patients with chronic liver disease,
Journal of Gastroenterology and Hepatology(2002) 17, S408-S412.
23. Luo C, Chintu C, Bhat G, Raviglione M, Diwan V, DuPont H, et al. Human immuno-
deficiency virus type-1 infection in Zambian children with tuberculosis: changing
seroprevalence and evaluation of a thioacetazone-free regimen. Tuberc Lung Dis
1994;75:110–5.
24. Marais B, Rabie H, Cotton M. TB and HIV in children—advances in prevention and
management. Paediatr Respir Rev 2011;12:39–45.
25. Marais BJ, Graham SM, Cotton MF, Beyers N. Diagnostic and management chal-
lenges for childhood tuberculosis in the era of HIV. J Infect Dis 2007; 196(Suppl
1):S76–85.
26. Swaminathan S, Padmapriyadarsini C, Narendran G. HIV-associated tuberculosis:
clinical update. Clin Infect Dis 2010;50:1377–86.
27. Marais BJ, Graham SM, Cotton MF, Beyers N. Diagnostic and management chal-
lenges for childhood tuberculosis in the era of HIV. J Infect Dis 2007; 196(Suppl
1):S76–85.
28. Gandhi NR, Moll A, Sturm AW, Pawinski R, Govender T, Lalloo U, et al. Extensively
drug-resistant tuberculosis as a cause of death in patients co- infected with tubercu-
losis and HIV in a rural area of South Africa. Lancet 2006;368:1575–80.
29. Use of tuberculosis interferon-gamma release assays (IGRAs) in low- and middle-
income countries: policy statement. Geneva, World Health Organization, 2011
(WHO/HTM/TB/2011.18)
30. World Health Organization. WHO policy on collaborative TB/HIV activities—
guidelines for national programmes and other stakeholders. Geneva: WHO; 2012.
31. World Health Organization. Automated real-time nucleic acid amplification technolo-
gy for rapid and simultaneous detection of tuberculosis and rifampicin resistance:
199
Xpert MTB/RIF system for the diagnosis of pulmonary and extrapulmonary TB in
adults and children. Policy Update. Geneva: WHO; 2013.
32. Guidelines for intensified tuberculosis case-finding and isoniazid preventive therapy
for people living with HIV in resource-constrained settings. Geneva, World Health
Organization, 2011.
33. Frigati LJ et al. The impact of isoniazid preventive therapy and antiretroviral therapy
on tuberculosis in children infected with HIV in a high tuberculosis incidence setting.
Thorax, 2011, 66(6):496-501.
34. Madhi SA et al. Primary isoniazid prophylaxis against tuberculosis in HIV-exposed
children. New England Journal of Medicine, 2011, 365(1):21-31.
35. Zar HJ et al. Effect of isoniazid prophylaxis on mortality and incidence of tuberculo-
sis in children with HIV: randomised controlled trial. British Medical Journal, 2006,
334:136.
36. Guideline for national tuberculosis programmes on the management of tuberculosis
in children, Second edition,pp.55-64.
37. Revised BCG vaccination guidelines for infants at risk for HIV infection. Weekly Epi-
demiological Record, 2007, 82:193-196.
38. Recommendations for investigating contacts of persons with infectious tuberculosis
in low- and middle-income countries. Geneva, World Health Organization, 2012
(WHO/HTM/TB/2012.9).
39. Rapid advice: treatment of tuberculosis in children. Geneva, World Health Organi-
zation, 2010 (WHO/HTM/TB/2010.13).
40. Guidelines for intensified tuberculosis case-finding and isoniazid preventive therapy
for people living with HIV in resource-constrained settings. Geneva, World Health
Organization, 2011.
41. WHO policy on collaborative TB/HIV activities: guidelines for national programmes
and other stakeholders. Geneva, World Health Organization, 2012 (WHO/HTM/
TB/2012.1).
42. Consolidated guidelines on the use of antiretroviral drugs for treating and prevent-
ing HIV infection: recommendations for a public health approach. Geneva, World
Health Organization, 2013
43. Guideline on the Integrated Management of Pediatric HIV and AIDS by the Cross-
ing Border Project,p.26.
44. Guidelines on co-trimoxazole prophylaxis for HIV-related infections among children,
adolescents and adults: recommendations for a public health approach. Geneva,
World Health Organization, 2006.
200
45. Provisional WHO/ UNAIDS Recommendations on the Use of Cotrimoxazole
Prophylaxis in Adults and Children Living with HIV/AIDS in Africa. WHO/UNAIDS
Geneva, 2000.African Health Sciences Vol 1.No1.August 2001 pp.30-31.
46. Consolidated guidelines on the use of antiretroviral drugs for treating and prevent-
ing HIV infection: recommendations for a public health approach. Geneva, World
Health Organization, 2013.
47. ARROW Trial Team. Routine versus clinically driven laboratory monitoring and first-
line antiretroviral therapy strategies in African children with HIV (ARROW): a 5-year
open-label randomized factorial trial. Lancet, 2013, 381:1391-1403.
48. Centers for Disease Control and Prevention.National Center for HIV/AIDS,Viral
Hepatitis,STD, and TB Prevention in partnership with New Jersey Medical School
Global Tuberculosis Institute 2013.Latent Tuberculosis Infection.A Guide for Prima-
ry Health Care Providers.
49. Schaaf, H. Simon MBChB, MMed Paed, DCM MD Paed, Zumla, Allimuddin BSc,
MBChB, MSc, PhD,FRCP .Tuberculosis: A Comprehensive Clinical Reference.
First edition. Saunders Elsevier 2009
50. Jawad F, Shera AS, Memon R, et al. Glucose intolerance in pulmonary tuberculo-
sis. J Pak Med Assoc 1995;45(9):237–238.Alisjahbana B, van Crevel R, Sa-
hiratmadja E, et al. Diabetes mellitus is strongly associated with tuberculosis in In-
donesia. Int J Tuberc Lung Dis 2006;10(6):696–700.
51. Singla R, Khan N, Al-Sharif N, et al. Influence of diabetes on manifestations and
treatment outcome of pulmonary TB patients. Int J Tuberc Lung Dis 2006; 10(1):74
–5.
52. Perez-Guzma ́n C, Torres-Cruz A, Villarreal- Velarde H, et al. Atypical radiological
images of pulmonary tuberculosis in 192 diabetic patients: a comparative study. Int
J Tuberc Lung Dis 2001;5(5): 455–461
53. Yip C, Lee AJ. Gatifloxacin-induced hyperglycemia: a case report and summary of
the current literature. Clin Ther 2006;28(11):1857–1866.
54. Guler M, Unsal E, Dursun B, et al. Factors influencing sputum smear and culture
conversion time among patients with new case pulmonary tuberculosis. Int J Clin
Pract 2007;61(2):231–236.
55. Restrepo BI, Fisher-Hoch SP, Crespo JG, et al. Type 2 diabetes and tuberculosis in
a dynamic bi-national border population. Epidemiol Infect 2007;13(3): 483–491.
56. Wada M, Yoshiyama T, Ogata H, et al. [Six-months chemotherapy (2HRZS or
E/4HRE) of new cases of pulmonary tuberculosis – six year experiences on its ef-
fectiveness, toxicity, and acceptability]. Kekkaku 1999;74(4):353–360.
201
57. Fang HC, Lee PT, Chen CL, et al. Tuberculosis in patients with end-stage renal dis-
ease. Int J Tuberc Lung Dis 2004;8(1):92–97.
58. Aoki K. Excess incidence of lung cancer among pulmonary tuberculosis patients.
Jpn J Clin Oncol 1993;23(4):205–220.
59. Silva FA, Matos JO, de Q Mello FC, et al. Risk factors for and attributable mortality
from tuberculosis in patients with hematologic malignances. Haematologica
2005;90(8):1110–1115.
60. Mok CK, Nandi P, Ong GB. Coexistent bronchogenic carcinoma and active pulmo-
nary tuberculosis. J Thorac Cardiovasc Surg 1978;76(4):469–472.
61. Gie RP, Beyers N, Schaaf HS, et al. The challenge of diagnosing tuberculosis in
children: a perspective from a high incidence area. Paediatr Respir Rev 2004; 5
(Suppl A):S147–149.
62. Strang JI, Nunn AJ, Johnson DA, et al. Management of tuberculous constrictive
pericarditis and tuberculous pericardial effusion in Transkei: results at 10 years fol-
low-up. QJM 2004;97(8):525–535.
63. Wiysonge CS, Ntsekhe M, Gumedze F, et al. Contemporary use of adjunctive corti-
costeroids in tuberculous pericarditis. Int J Cardiol 2008;124(3): 388–390.
64. Thwaites GE, Nguyen DB, Nguyen HD, et al. Dexamethasone for the treatment of
tuberculous meningitis in adolescents and adults. N Engl J Med 2004;351(17):1741
–1751.
65. Simmons CP, Thwaites GE, Quyen NT, et al. The clinical benefit of adjunctive dex-
amethasone in tuberculous meningitis is not associated with measurable attenua-
tion of peripheral or local immune responses. J Immunol 2005;175(1): 579–590.
66. Hammer SM, Saaq MS, Schechter M, et al. Treatment for adult HIV infection: 2006
recommendations of the International AIDS Society – USA panel. Top HIV Med
2006;14(3):
827–843
67. Yun JE, et al. The incidence and clinical characteristics of Mycobacterium tubercu-
losis infection among systemic lupus erythematosus and rheumatoid arthritis pa-
tients in Korea. Clin Exp Rheumatol 2002;20(2):127–132.
68. Mok MY, Lo Y, Chan TM, et al. Tuberculosis in systemic lupus erythematosus in an
endemic area and the role of isoniazid prophylaxis during corticosteroid therapy. J
Rheumatol 2005;32(4):609–615.
69. Bahceciler NN, Nuhoglu Y, Nursoy MA, et al. Inhaled corticosteroid therapy is safe
in tuberculin- positive asthmatic children. Pediatr Infect Dis J 2000; 19(3):215–218.
202
70. Bateman ED. Is tuberculosis chemoprophylaxis necessary for patients receiving
corticosteroids for respiratory disease? Respir Med 1993;87(7): 485–487.
71. Fidler H, Rook GA, Johnson NM, et al. Search for mycobacterial DNA in granulom-
atous tissues from patients with sarcoidosis using the polymerase
chain reaction. Am Rev Respir Dis 1993;147(3): 777–778.
72. Wong CF, Yew WW, Wong PC, et al. A case of concomitant tuberculosis and sar-
coidosis with mycobacterial DNA present in the sarcoid lesion. Chest 1998;114
(2):626–629.
73. Knox AJ, Wardman AG, Page RL. Tuberculous pleural effusion occurring during
corticosteroid treatment of sarcoidosis. Thorax 1986;41(8):651.
74. 74. Baughman RP, Lower EE. Fungal infections as a complication of therapy for
sarcoidosis. QJM 2005; 98(6):451–456.
75. Targeted tuberculin testing and treatment of latent tuberculosis infection. American
Thoracic Society. MMWR Recomm Rep 2000;49(RR-6):1–51.
76. Singh N, Paterson DL. Mycobacterium tuberculosis infection in solid-organ trans-
plant recipients: impact and implications for management. Clin Infect Dis 1998;27
(5):1266–1277.
77. British Thoracic Society guidelines on diagnostic flexible bronchoscopy. Thorax
2001;56(Suppl 1): i1–21.
78. Hanngren A, Hedenstedt S, Reizenstein P. Nutritional studies in patients with
dumping syndrome. I. Subjects with postcibal symptoms. Am J Dig Dis 1967;12
(1):71–80.
79. Yokoyama T, Sato R, Rikimaru T, et al. Tuberculosis associated with gastrectomy.
J Infect Chemother 2004;10(5):299–302.
80. Snider DE Jr. Tuberculosis and gastrectomy. Chest 1985;87(4):414–415.
81. Snider DE Jr. Jejunoileal bypass for obesity: a risk factor for tuberculosis. Chest
1982;81(5):531.
82. Yu VL. Onset of tuberculosis after intestinal bypass surgery for obesity. Guidelines
for evaluation, drug prophylaxis, and treatment. Arch Surg 1977;112(10): 1235–
1237.
83. Mow WS, Abreu-Martin MT, Papadakis KA, et al. High incidence of anergy in in-
flammatory bowel disease patients limits the usefulness of PPD screening before
infliximab therapy. Clin Gastroenterol Hepatol 2004;2(4):309–313.
84. Provenzano G, Ferrante MC, Simon G. TB screening and anti-TNFa treatment.
Thorax 2005;60(7):613.
203
85. Sichletidis L, Settas L, Spyratos D, et al. Tuberculosis in patients receiving anti-
TNF agents despite chemoprophylaxis. Int J Tuberc Lung Dis 2006;10(10): 1127–
1132.
86. Bieber J, Kavanaugh A. Consideration of the risk and treatment of tuberculosis in
patients who have rheumatoid arthritis and receive biologic treatments. Rheum Dis
Clin North Am 2004;30(2):257–270, v.
204
14
Adjunctive Management
Cleotilde H How, MD
Cristan Q Cabanilla, MD
Maria Nerissa A de Leon, MD
Anna Lisa T Ong-Lim, MD
OUTLINE
1 Overview
2 Corticosteroids
Anna Lisa T Ong-Lim, MD
3 Surgery
Cristan Q Cabanilla, MD
Maria Nerissa A de Leon, MD
4 Nutrition
Cleotilde H How, MD
205
OVERVIEW
There are several modalities employed in the management of tuberculosis, both phar-
macologic and non-pharmacologic: corticosteroids, surgery and nutritional support.
These have been considered to be of value in promoting healing and reducing the com-
plications and residual effects of the disease.
CORTICOSTEROIDS
The benefit of corticosteroids has been evaluated in the following forms of complicated
TB:
1. TB meningitis, where it acts by reducing vasculitis, inflammation, and intracranial
pressure, which may then lead to improved circulation of chemotherapeutic drugs
through the brain.1 Corticosteroids should be routinely used in HIV-negative pa-
tients with TB meningitis, to reduce death and residual neurologic deficit.2
2. TB pericarditis, where corticosteroids are recommended as adjunctive therapy dur-
ing the first 11 weeks of anti-tuberculosis therapy.3
3. TB pleural effusion, where a recent systematic review concludes that there is insuf-
ficient data to support routine use.4 Although steroids do not appear to reduce the
development of residual pleural thickening, some studies showed a significantly
more rapid resolution of symptoms.3
4. Endobronchial TB, where enlarged mediastinal lymph nodes cause respiratory diffi-
culty or a severe collapse-consolidation lesion in the middle or lower lobes, when
bronchiectasis is likely to be a troublesome sequela.1
In HIV-negative patients, corticosteroids may be a useful adjunct to the treatment of
tuberculosis, particularly when the host inflammatory response is contributing to tissue
damage or is impairing function.
The drug most frequently used is prednisone given 2 mg/kg daily, for 4 weeks. For the
most seriously ill children the dose may be increased to 4 mg/kg daily (maximum 60
mg/day). The dose of prednisone should be gradually tapered over 1-2 weeks before
finally being discontinued.5 In TB pericarditis, the adult dose for prednisone is 60 mg/
day given for 4 weeks, followed by 30 mg/day for 4 weeks, 15 mg/day for 2 weeks, and
5 mg/day for the last week (a total of 11 weeks). The dose for children is 1mg/kg body
weight, decreasing the dose over the treatment period as described for adults.3
206
REFERENCES
1. Cruz AT and Starke JR. 2014. Tuberculosis. In Cherry JD, Harrison GJ et al, eds.
Feigin and Cherry‘s Textbook of Pediatric Infectious Diseases 7th ed. Philadelph-
ia: Elsevier Saunders. p 1375.
2. Prasad K and MB Singh. Corticosteroids for managing tuberculous meningitis.
Cochrane Database of Systematic Reviews 2008, Issue 1. Art. No.: CD002244.
DOI: 10.1002/14651858.CD002244.pub3)
3. Centers for Disease Control and Prevention. Treatment of Tuberculosis. American
Thoracic Society, CDC, and Infectious Diseases Society of America. MMWR
2003;52(No. RR-11).
4. Engel ME et al. Corticosteroids for tuberculous pleurisy. Cochrane Database of
Systematic Reviews 2007, Issue 4. Art. No.: CD001876. DOI: 10.1002/14651858.
CD001876.pub2
5. World Health Organization. Treatment of Tuberculosis: Guidelines for National
Programmes. Third edition, 2003.
207
SURGERY
While the use of surgery is now rarely indicated because of the effectiveness of anti-
tuberculous drugs, there are certain conditions in which it may play a vital role in estab
-lishing the diagnosis and overall treatment of tuberculosis.
Diagnostic modality
Rapid and definitive diagnosis to facilitate early and appropriate treatment in TB has
been reported in the use of two surgical interventions, namely fine needle aspiration
biopsy and laparoscopy. Fine needle aspiration biopsy, with or without CT or ultra
-sound guidance, to obtain specimen for direct microscopy, nucleic acid amplification
test and culture has been recommended for cervical or axillary lymphadenopathy1,2,
soft tissue and bone lesion3,4and mediastinalmass5,6. Laparoscopy in suspected ab-
dominal TB has been reported in adults7-9and children10. Procedure-related complica-
tions were reported to be minimal1-6.
Treatment modality
Surgical intervention is absolutely necessary as a life-saving procedure (or as an
emergency approach, for without surgery death is imminent and unavoidable) in the
following conditions: massive hemoptysis; tension pneumothorax; pericarditis with res-
piratory or blood circulation insufficiency; intrinsic or extrinsic airway obstruction, sac-
cular bronchiectasis3,11 postmeningitic hydrocephalus12,13, tuberculoma in the lung pa-
renchyma resulting into compression3,11 or brain as a space-occupying lesion3, spinal
involvement with neurologic deficit,paravertebral abscess or instability due to kyphotic
deformity14,15,intestinal obstruction with or without perforation16,17 and hydronephro-
sis19=18. Masoud et al3 have included these other indications for surgery in TB:
Urgent approach for irreversible TB progression, despite adequate anti-TB chemo-
therapy and for recurrent haemoptysis that cannot be stopped by other treatment
methods.
Elective approach for (a) localised forms of cavitary TB with continuous Mycobacte-
rium tuberculosis excretion confirmed by bacteriological examination and DSST after 4
–6 months of supervised anti-TB chemotherapy; (b) MDR/XDR-TB characterised by
failure of anti-TB chemotherapy; (c) complications and sequelae of the TB process
(including MDR/XDR-TB) like spontaneous pneumothorax and pyopneu-mothorax;
pleural empyema with or without bronchopleural fistula; Aspergilloma; nodular–
bronchial fistula; broncholith; pachypleuritis or pericarditis with respiratory and blood
circulation insufficiency; post-TB stenosis of trachea and large bronchi; symptomatic
and chronic post-TB bronchiectasis.
For patients with drug-resistant TB, resection of the lung (as an adjuvant therapy) is
indicated for those with treatment failure.3
Marrone et al19 in a systematic review of 24 studies from 1975 to 2012 concluded that
treatment success rate was higher among patients undergoing surgery compared with
those treated with anti-TB drugs alone (OR 2.24, 95% CI1.68 – 2.97). All-cause mor-
tality is decreased by 24%. The positive effect of surgery on treatment success almost
208
doubled in the subgroup of XDR-TB patients (>400 cases, OR 4.55) compared with
MDR-TB patients (OR 2.27).
Surgery plays a limited role in the management of drug-sensitive tuberculosis and is
reserved for management of late complications, such as hydrocephalus, obstructive
uropathy, constrictive pericarditis and neurological involvement from Pott‘s disease 13.
Among patients with drug-resistant tuberculosis, surgery is most effective for patients
with localized pulmonary disease that can be completely resected. This includes pa-
tients whose sputum cultures remain positive for longer than three months despite ap-
propriate therapy, or those with isolates resistant to all of the first-line oral agents. Re-
section alone should not be considered curative; patients with multidrug-resistant TB
should continue anti-tuberculous therapy for 18 months following surgery20
209
REFERENCES
1. Coetzee L(1), Nicol MP, Jacobson R, Schubert PT, van Helden PD, Warren RM,
Wright CA.: Rapid diagnosis of pediatric mycobacterial lymphadenitis using fine
needle aspiration biopsy. Pediatr Infect Dis J. 2014;3):893-6.
2. Wright CA, Warren RM, Marais BJ. Fine needle aspiration biopsy: an undervalued
diagnostic modality in paediatric Mycobacterial disease. Int J Tuberc Lung Dis.
2009 13:1467-75
3. Masoud Dara, G Sotgiu, R Zaleskis and GB Migliori.Untreatable tuberculosis: is
surgery the answer? Eur Respir J 2015;45:577–582 DOI:
10.1183/09031936.00229514
4. Gupta S, Takhtani D, Gulati M, Khandelwal N, Gupta D, Rajwanshi A, Gupta S,Suri
S Sonographically guided fine-needle aspiration biopsy of lytic lesions of the
spine: technique and indications. J Clin Ultrasound. 1999;27:123-9.
5. Gulati M, Venkataramu NK, Gupta S, Sood BP, Sheena DM, Gupta SK, Suri S. Ul-
trasound guided fine needle aspiration biopsy in mediastinal tuberculosis. Int J Tu-
berc Lung Dis. 2000;4):1164-8.
6. Khan J), Akhtar M, von Sinner WN, Bouchama A, Bazarbashi M. CT-guided fine
needle aspiration biopsy in the diagnosis of mediastinal tuberculosis Chest.
1994;106):1329-32.
7. Guirat A), Affes N, RejabH,Trigui H, Ben Amar M, Mzali R:.[Role of laparoscopy in
the diagnosis of peritoneal tuberculosis in endemic areas Med Sante Trop. 2015
5:87-91
8. Krishnan P, Vayoth SO, Dhar P, Surendran S, Ponnambathayil S:.Laparoscopy in
suspected abdominal tuberculosis is useful as an early diagnostic method. ANZ J
Surg. 2008;78:987-9
9. Safarpor F, Aghajanzade M, Kohsari MR, Hoda S, Sarshad A, Safarpor D.: Role of
laparoscopy in the diagnosis of abdominal tuberculosis. Saudi J Gastroenterol
2007;13:133-5
10. Dinler G), Sensoy G, Helek D, Kalayci AG.: Tuberculous peritonitis in children: re-
port of nine patients and review of the literature. World J Gastroenterol.2008;14
(47):7235-9
11. Madansein R, Parida S, Padayatchi N, Singh N, Master I, Naidu K, Zumla A,
Maeurer M.: Surgical treatment of complicationsofpulmonary tuberculosis, including
drug resistant tuberculosis Int J Infect Dis. 2015 Mar;32:61-7.
12. Bajpai M, Nambhirajan L, Dave S, Gupta AK: Surgeryin tuberculosis Indian J Pedi-
atr 2000;67(2 Suppl):S53-7
210
13. Rajshekhar V: Surgery for brain tuberculosis: a review 2015 ActaNeurochir (Wien).
2015;14
14. Rasouli MR, Mirkoohi M, Vaccaro AR:Spinal Tuberculosis: Diagnosis and Manage-
mentAsian Spine J 2012;6:294~308
15. Moon MS, Kim SS, Moon YW, Moon H, Kim SS: Surgery-related complications and
sequelaein managementof tuberculosis of spine Asian Spine J. 2014;8:435-45.
16. Mukhopadhyay A, Dey R, Bhattacharya U. Abdominal tuberculosis with an acute
abdomen:our clinical experience. J ClinDiagnRes. 2014;8):NC07-9.
17. Awasthi S, Saxena M, Ahmad F, Kumar A, Dutta S.: Abdominal Tuberculosis: A Di-
agnostic Dilemma J ClinDiagn Res. 2015;9(5):EC01-3
18. Krishnamoorthy S, Gopalakrishnan G.: Surgical management of renal tuberculo-
sis.Indian J Urol. 2008;24:369-75
19. Marrone MT, V Venkataramanan, M Goodman, AC Hill, JA Jereb and SR Mase.
Surgical interventions for drug-resistant tuberculosis: a systematic review and meta
-analysis. Int J Tuberc Lung Dis 2013; 17(1):6-16
20. Schluger NW. Diagnosis, treatment, and prevention of drug-resistant tuberculosis.
In: UpToDate, C Fordham von Reyn (Ed), UpToDate, Waltham, MA. (Accessed on
27 Feb 2016).
211
NUTRITION
Overview
The interaction of nutrition and tuberculosis is important in the assessment and man-
agement of patients with tuberculosis.1,2 As a general measure in patient care, all as-
pects of therapy includes good nutritional support for patients with TB to improve out-
come of treatment and quality of life when the body is allowed to recover and normal
function restored. Children especially the young are more vulnerable than adults to TB
infection and nutritional neglect; extra care can make a difference in outcome.
Comorbidity
Undernutrition and TB are co-morbid conditions and this synergy is considered to mu-
tually impact each other adversely.1,2 Undernutrition increases the risk of TB and in turn
TB can lead to undernutrition with the underlying pathology affecting host defense. This
renders a child at risk for progression from TB infection to active disease, with in-
creased risk of poor treatment outcome, TB relapse and death.3
Nutrition
The WHO guideline1 aims to help improve health outcomes for people with TB, through
improved nutritional care and support. It provides guidance on nutritional assessment,
advice and treatment, and its integration into clinical care for people with TB. The guid-
ing principles and recommendations focus on nutrition assessment, counselling and
management to improve the clinical care of people with TB. Considered an essential
part of services for people with TB they can be adapted according to disease burden,
health-care infrastructure, including human resources.
Resource gap
Access of communities to adequate food supply and medical services and goods (e.g.
anti-TB drugs) has to be assured particularly in high- burden low-resource areas as
part of their regular TB care package. With the intensive global effort to improve TB di-
agnosis, treatment and control, TB prevalence and death rates have been reduced in
many countries where national TB programmes had been vigorous in their campaigns.
Still, in many other countries the incidence of TB is declining at a slow pace4 and TB
remains a major public health threat where resources are limited and the attendant
poverty and disease are aggravating factors.3
Barriers
There are barriers to the diagnosis of tuberculosis5 in common with undernutrition that
includes ignorance, fear, mistrust, shame. One of the six symptoms that should arouse
suspicion of TB in a young child with unexplained cough and fever is weight loss or fail-
ure to gain weight. It should trigger further clinical assessment of possible TB and se-
verity of nutritional deficiency for prompt intervention.6,7
Weight loss
Weight loss is a red flag that draws attention towards case-finding. Among children at
risk for TB faltering weight can be from poor intake of adequate amounts of food that
will provide the essential macronutrients and micronutrients. This can be due to loss of
appetite and metabolic changes attendant to the disease process. The resultant low
212
body mass index (BMI) (<18.5 kg/m2) and poor weight gain/weight loss is an important
parameter not only for diagnosis but for assessment of severity and for monitoring re-
sponse to treatment and for prognostication. A low value is associated with an in-
creased risk of death.8
Monitoring growth
The CDC recommends that the WHO growth charts be used to monitor the growth of
infants and children ages 0 to 2 years (in the US) and the CDC growth charts to monitor
growth for children age 2 years and older (in the US)9. The WHO BMI-for-age-and-sex
Z-score is used in children and adolescents aged 5–19 years. Weight-for-length or
weight-for-height Z-score is the recommended indicator for children who are less than 5
years of age, with midupper arm circumference being used to identify cases in need of
life-saving nutrition management.10 The PPS Preventive Pediatric Health Care Hand-
book (2016) has the BMI for age of boys and girls 2 to 5 years, and 5 to 19 years, to
provide a handy tool for monitoring growth. It also gives the weight for age, height for
age, and weight for length/height.11-14
Evidence for supplementation
The available literature cited in the WHO publication1 show that the evidence on the
clinical benefits for supplementation in patients with TB is insufficient. The standard of
care in the field – prescribing or providing nutritional support - is still the accepted norm.
The required dietary energy from macronutrients (e.g. protein, carbohydrate and fat), ,
is similar for people with active TB and for those without TB. It generally follows a rec-
ommended distribution that all people consume approximately 15–30% of energy as
protein,25–35% as fat and 45–65% as carbohydrate.15
Coping in disasters
In countries often visited by disasters, victims - particularly the children- are at risk for
catastrophic illnesses that come with food deprivation, interrupted health services
(including immunizations, nutrition programs, DOTS services) and the harsh physical
conditions with displacement from home and shelters, brought on by floods, typhoons
and earthquakes. In the section on TB during disasters the DOH Manual of Proce-
dures16 provides instructions following disaster situations ―to re-establish TB services to
reduce mortality and morbidity due to interrupted treatment arising from lack of follow-
up, drugs, facilities, and human resource‖. There are strategies to achieve this objective
and the algorithm to ensure that all existing TB patients can resume treatment after the
acute relief operations.
Integration
Patients with TB should be assessed regularly and receive advice and support ensuring
adequate nutrition particularly for children with a chronic debilitating illness like TB is
necessary for a holistic approach17,18. Children with TB and severe undernutrition will
be an extreme challenge requiring skills in a multidisciplinary and comprehensive ap-
proach, as provided in guidelines for child TB6,7. TB care can be integrated with other
services like maternal and child health services, nutrition and immunization programs.
There are still many aspects in the nutritional support of patients with TB that can im-
prove the outcome of treatment.19-22
213
REFERENCES
1. Guideline: Nutritional care and support for patients with tuberculosis. Geneva:
World Health Organization; 2013.
2. Scoping meeting for the development of guidelines on nutritional/food support to
prevent TB and improve health status among TB patients. Meeting report, Geneva,
2–4 November 2009. Geneva: World Health Organization; 2010 (http://
www.who.int/nutrition/publications/nutandtb_meeting_report/en/index.html, ac-
cessed 10 August 2016).
3. Hood MLH. A narrative review of recent progress in understanding the relationship
between tuberculosis and protein energy malnutrition. Eur J Clin Nutr.
2013;67:1122-1128; doi:10.1038/ejcn.2013.1432.
4. WHO Global tuberculosis report 2015 (WHO/HTM/TB/2015.22) (https://www.health
-e.org. accessed 10 August2016)
5. Chiang SS, Roche S, Contreras C, Alarcὀn V, del Castillo H, Becerra MC. Barriers
to the diagnosis of childhood tuberculosis: a qualitative study. Intl J TB Lung Dis.
2015;1144-52.
6. Guidance for national tuberculosis programmes on the management of tuberculosis
in children. Geneva, World Health Organization, 2014. (WHO/HTM/TB/2014.03).
7. Tuberculosis in Infancy and Childhood. Philippine Pediatric Society, 2016.
8. Zachariah R, Spielmann MP, Harries AD, Salaniponi FM. Moderate to severe mal-
nutrition in patients with tuberculosis is a risk factor associated with early death.
Trans. R. Soc. Trop. Med. Hyg. 2002;96:291–4. www.ncbi.nlm.gov/
pubmed/12174782.
9. CDC growth charts for the United States: methods and development.ISBN 0-8406-
0575-7 www.cdc.gov/growthcharts/cdc_charts.htm, accessed 10 August2016
10. World Health Organization. Growth reference data for 5–19 years (http://
www.who.int/growthref/en/, accessed 10 August2016).
11. PPS Preventive Pediatric Health Care Handbook. Philippine Pediatric Society,
2016.
12. World Health Organization, United Nations Children‘s Fund. WHO child growth
standards and the identification of severe acute malnutrition in infants and children.
A joint statement by the World Health Organization and the United Nations Chil-
dren‘s Fund. Geneva: World Health Organization and United Nations Children‘s
Fund; 2009 (http://www.who.int/nutrition/publications/
severemalnutrition/9789241598163_eng.pdf, accessed 10 August 2016).
13. World Health Organization. The WHO child growth standards (http://www.who.int/
214
childgrowth/en/, accessed 10 August 2016).
14. Physical status: the use and interpretation of anthropometry. Report of a WHO Ex-
pert Committee. Geneva: World Health Organization; 1995 (WHO Technical Report
Series 854) (http://www.who.int/childgrowth/publications/physical_status/en/, ac-
cessed 10 August 2013).
15. Institute of Medicine. Dietary reference intakes for energy, carbohydrate, fiber, fat,
fatty acids, cholesterol, protein, and amino acids. Washington, DC: National Acade-
mies Press; 2005. (In: Guideline: Nutritional care and support for patients with tu-
berculosis. Geneva: World Health Organization; 2013).
16. Manual of Procedures of the National Tuberculosis Program (5th edition). Depart-
ment of Health, 2014.
17. Guideline: Updates on the management of severe acute malnutrition in infants and
children. Geneva: World Health Organization; 2013.
18. Management of severe malnutrition: a manual for physicians and other senior
health workers. Geneva: World Health Organization; 1999 (http://www.who.int/
nutrition/publications/severemalnutrition/9241545119/en, accessed 10 August2016)
19. Papathakis P, Piwoz E. Nutrition and tuberculosis: a review of the literature and
considerations for TB control programs. USAID/Africa‘s Health for 2010. Washing-
ton DC: Agency for International Development; 2008 (http://pdf.usaid.gov/pdf_docs/
PNADL992.pdf.
20. Dodor E. Evaluation of nutritional status of new tuberculosis patients at the Effia-
Nkwanta regional hospital. Ghana Med. J. 2008;42(1): 22–8 (http://
www.ncbi.nlm.nih.gov/pmc/articles/PMC2423338..
21. Ramakrishnan CV, Rajendran K, Jacob PG, Fox W, Radhakrishna S. The role of
diet in the treatment of pulmonary tuberculosis. An evaluation in a controlled chem-
otherapy study in home and sanatorium patients in South India. Bull. World Health
Organ. 1961;25:39–59 (http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2555577/pdf/
bullwho00319-0062.pdf, 60.
22. Paton NI, Chua YK, Earnest A, Chee CB. Randomized controlled trial of nutritional
supplementation in patients with newly diagnosed tuberculosis and wasting. Am. J.
Clin. Nutr. 2004;80:460–5.
215
15
Prevention and Control
Josefina C Carlos, MD
Luis T Chan, Jr, MD
Ana Liza H Duran, MD
OUTLINE
1 Overview
2 Surveillance
3 Containment
216
OVERVIEW
Control of TB is based on the principles of surveillance and containment. Surveillance
is mainly concerned with case finding of TB and monitoring of cases, while contain-
ment involves treatment and prevention.1 TB control is a continuing process of collect-
ing, analyzing, interpreting and disseminating information about trends in disease and
infection with the ultimate goal of significantly reducing the burden of TB in the com-
munity.
The three basic strategies critical to the prevention and control of TB, in order of prior-
ity2:
1. Case finding with appropriate therapy
2. Contact tracing of exposed individuals for presence of infection or disease
3. Testing of groups with high risk for TB infection to identify those requiring treat-
ment for latent infection.
TB infection control program has become a crucial component of TB control due to
the increasing co-morbidity of TB and HIV cases and the emergence of multidru-
gresistant TB (MDR-TB) and extensively drug-resistant TB (XDR-TB). Effective TB
control policies require a sustained political, institutional and financial commitment
from both public and private sectors of the society.
SURVEILLANCE
Case Finding of TB Disease3
Case finding is the identification and diagnosis of TB cases among individuals with
presumptive signs and symptoms for tuberculosis. The three types of TB disease
case finding used by the National TB Program (NTP) are defined as follows:
Active case finding - involves the lead effort of a health worker to find TB cases in
the community or among those who do not consult with personnel in a DOTS fa-
cility
Intensified case finding – involves active case finding among individuals belonging
to special or defined populations (e.g., high-risk groups including those who con-
sult or find themselves at the facility for other purposes).
The NTP advocates detection of acid – fast TB bacilli using direct sputum smear mi-
croscopy (DSSM) for adults and children who can expectorate. DSSM is the easiest,
least expensive, readily accessible and the most rapid procedure to diagnose infec-
tious TB cases. It is also used to: a) monitor progress of patients with TB while they
are on anti–TB treatment; and b) confirm cure at the end of treatment.
217
TB bacilli isolation in young children is difficult to obtain due to their inability to expec-
torate sputum effectively. Consequently young children are also considered less infec-
tious than adults due to: 1) the inability of most young children to expectorate sputum
efficiently and 2) the few numbers of TB bacilli in their sputum i.e. paucibacillary.
Diagnosis of childhood TB is mainly achieved through a symptom based screening ap-
proach, history of contact to an infectious TB source and with tuberculin skin testing
(TST) and chest radiographs as diagnostic aids. The DOH/NTP recommends its two
entry points guideline for the recognition/case finding TB in children. See Chapter 11:
Diagnosis of TB in children.
The other tests endorsed by TB programs for diagnosing TB are TB culture and drug
susceptibility test, tuberculin skin test and rapid diagnostic test (e.g., Xpert MTB/RIF)
which shall be used for TB diagnosis among presumptive drug resistant TB, people liv-
ing with HIV (PLHIV) with signs and symptoms of TB, smear-negative adults with chest
x-ray findings suggestive of TB, smear-negative children and extra-pulmonary TB.
The screening and treatment of children with TB disease and TB infection forms an im-
portant component in the strategy and assessment of TB control programs since;
218
Figure 15.1 Concentric Circle Approach (Adapted from Iyanna Dary. TB Contact Investigation
Module 15. Health Resources and Services Administration [HRSA])
The Concentric Circle Approach is used to identify contacts who are high, medium, or
low risk.12 A close contact is a person who shared an enclosed space such as the
household, a social gathering place, workplace or facility, for extended periods within a
day with the index case during the three (3) months before commencement of the cur-
rent treatment episode11
A child with TB reflects ongoing transmission of TB cases and warrants prompt and
systematic contact investigation for an undiagnosed adult/adolescent source case. The
purpose of the investigation is to find the following: (1) contacts (and treat those) who
have TB disease in order to break the cycle of transmission; (2) those with latent TB
infection (LTBI); and (3) those at high risk of developing TB disease, who requires treat-
ment until LTBI can be excluded.6
Clinical evaluation of household and close contacts for active TB should be done on
the basis of their risk for having or developing active TB or for the potential conse-
quences of the disease if it develops. Priority should be given to contacts who are7:
219
Targeted Tuberculin Skin Testing (TST) for LTBI
Routine TST for TB screening is not recommended. Targeted Tuberculin skin testing
for LTBI is a strategic component of TB control that identifies persons who are at high
risk for developing TB disease and who would benefit from treatment of LTBI when de-
tected.8 The preferred method of screening for TB infection is the Mantoux TST. Tuber-
culin skin-testing programs should be conducted only among populations with high-risk
for progression of infection to disease and discouraged among persons with low risk.
In conducting tuberculin skin testing among targeted populations, cases warranting
treatment must be initiated and completed, with facilities for follow-up of cases diag-
nosed.
Interferon-gamma release assays (IGRAs) should not replace the tuberculin skin test
(TST) in low- and middle-income countries for the diagnosis of latent TB infection in
children or for the diagnostic work-up of children (irrespective of HIV status) suspected
of TB disease in these settings.8
World Health Organization (WHO) Recommendations for TST Screening3
Every household contacts identified to be a presumptive TB should be evaluated based
on the NTP case finding procedures.
All asymptomatic household contacts less than 5 years old of a bacteriologically con-
firmed index case shall undergo TST. If TST is negative, these contacts should be giv-
en IPT. If TST is positive, rule out TB disease with CXR before starting IPT. However, if
TST and CXR are not available, the child contact of a bacteriologically confirmed index
case can be given IPT based on the physician‘s clinical assessment.
All asymptomatic household contacts less than 5 years old of a clinically diagnosed in-
dex case shall undergo TST. If TST is negative, do not give IPT and advise to seek/
consult immediately if signs and symptoms of TB develop. If TST is positive, give IPT.
All asymptomatic household contacts 5 years old and above (with normal CXR findings,
if done) are advised to seek/consult immediately if signs and symptoms of TB develop.
Young children living in close contact with a source case of smear-positive pulmonary
TB are at particular risk of TB infection and disease. The risk of infection is greatest if
the contact is close and prolonged such as the contact of an infant or toddler with a
mother or other caregivers in the household. The risk of developing disease after infec-
tion is much greater for infants and young children under five years of age than it is for
children over five years old.
The best way to detect LTBI is through the TST. These tests should be used where
they are available to screen exposed contacts. If the TST and CXR are not readily
available, this should not preclude contact screening and management, as this
can be conducted on the basis of simple clinical assessment.
220
CONTAINMENT
Containment of TB concerns the stopping of spread of the infection and disease
through chemotherapy of patients with TB disease and treatment of LTBI. Preventive
therapy refers to the use of a simple regimen such as isoniazid to prevent the develop-
ment of active TB disease in persons known to have or likely to be infected with M. tu-
berculosis. However, it is not expected to result in true prevention (i.e., prevention of
infection in persons exposed to persons with infectious TB).8
Prevention
There are three essential measures embodied within the National TB Programs for
the prevention of TB in children. These preventive measures include:
1. Detection of sources of infection and the appropriate treatment of TB excretors
who are usually adolescents and adults;
2. BCG vaccination of newborns in countries where the transmission of TB is high;
and
3. Chemoprophylaxis for the prevention of infection among those at risk by expo-
sure to an infectious TB case or the prevention of overt disease among those al-
ready infected
(1997 National Consensus on Childhood TB)
221
facilities, poor access to DOTS facility due to geographical barriers, in debilitated or
bedridden patients and in indigenous peoples where cultural beliefs limit the choice of a
treatment partner. 3
Under the National TB Program, TB control services are provided mainly through public
primary health care facilities operated by local government units in a devolved set-up.
There are additional DOTS facilities within the NTP‘s network of service providers that
either refer diagnosed TB patients for treatment or directly provide TB treatment ser-
vices using DOTS strategy. These include private outpatient clinics; public and private
primary, secondary and tertiary care hospitals; workplaces; clinics under faith-based
organizations and community based nongovernmental organizations (NGOs); and pub-
lic institutions such as military facilities, jails and prisons.13 As of December 2015, there
are 1,739 Philhealth accredited DOTS facilities nationwide.14 The TB DOTS Referral
System is an effective two way referral system where a receiving facility provides feed-
back to a referring facility. Major reasons for referral are for TB diagnosis, registration
and initiation of treatment, continuation of treatment as when a patient initially diag-
nosed in one facility is referred to another facility near his/her residence, management
of serious side effects and complications, screening of MDR-TB and screeining of TB
among PLHIV.3
The TB registry contains a list of treatment for TB patients and their progress monitor-
ing towards cure. The TB recording and reporting system consists of a laboratory reg-
istry with a list of all patients with smear tests, treatment cards with details on intake of
medications, and follow-up of sputum examinations. These reporting forms are submit-
ted from the districts to the national level.
Since adopting the Directly Observed Treatment Short Course Strategy in all public fa-
cilities in 2003, the country has made significant progress in the battle against TB. 15 As
of 2014, national indicators for TB Case Detection Rate (CDR) and Treatment Success
Rate (TSR) are at 85% and 90%, respectively. These improved values show that the
country is on its way on achieving its targets.
Bacillus Calmette-Guerin (BCG) and Vaccine Development
Bacille Calmette Guerin (BCG) is a vaccine derived from a live attenuated strain of My-
cobacterium bovis. It is the only known vaccine in use against TB. BCG vaccination in-
duces artificial primary immunity to TB for prevention of subsequent illness caused by
virulent bacilli.16
At least 4 billion people were immunized with BCG since its discovery; making it the
most widely used vaccine worldwide.17 The use of BCG vaccine has shown varied dif-
ferences in protective efficacy results in different studies. The variation in protective ef-
fect from different trials ranged from 0% to 80%.This has been attributed to several fac-
tors: differences in BCG strains, differences in prevalence of infections with environ-
mental mycobacteria, genetic and physiological differences between the populations
and different immunological mechanisms against forms of disease with varying patho-
genesis.18
A meta-analysis on the efficacy of BCG vaccination in newborns and infants by Colditz,
222
et. al in 1995 concluded that BCG vaccination of newborns and infants reduces the risk
of TB by 50% on the average.19,20 Another meta-analysis of 10 randomized clinical tri-
als and eight case control studies analyzed the protective effect and confidence inter-
vals for pulmonary and meningeal/miliary disease. There was considerable variation in
the protective effect of BCG against pulmonary TB. The summary protective effect
against meningeal and miliary TB was 86% (95% CI:65,95) and in case control studies
75% (95%CI:64,84). According to the study, a more likely explanation is that immuno-
logical mechanisms operate against different forms of disease, with BCG being more
effective in preventing the hematogenous spread of tuberculous bacteria as in meningi-
tis and miliary forms.18 A recent finding shows that in a high burden country for TB,
BCG confers protection against incident TB disease among those with household expo-
sure in those younger than 10 years (RR, 0.35, 95% CI‘s, 0.19-0.66).22
The recommendation to give BCG at birth is supported by the theory that exposure to
certain varieties of mycobacteria can give rise to a cell-mediated response which op-
poses the protective effect of subsequent BCG vaccination. Therefore, the recommen-
dation is to give BCG at birth or soon thereafter before any exposure to environmental
mycobacteria.23
BCG is not recommended in countries where TB rates are low, since it would increase
tuberculin reactivity, thereby making the tuberculin test less accurate. Being a live at-
tenuated vaccine, BCG is not recommended to be given to immunocompromised indi-
viduals. Likewise the BCG accelerated test should not be used for TB screening.
According to WHO, those persons who have received BCG vaccination are not recom-
mended to be vaccinated again, since available evidence does not support such prac-
tice. Multiple vaccinations are not recommended for anyone.24
The best strategy to eliminate TB is through a new and more effective TB vaccine. 25
The ideal vaccine should be safe even when administered to immunocompromised per-
sons (e.g., persons with HIV infection) and not hypersensitive to PPD that will cross-
react with TST and thereby limiting its specificity. The vaccine should protect against
diseases resulting from subsequent infection (i.e. pre-exposure), as well as, endoge-
nous reactivation of an earlier infection (i.e. post-infection). Considering the limitations
of preventive therapy, an effective post-infection vaccine could be the most important
new tool to help eliminate TB.
In high-incidence countries, a post-infection vaccine could be administered even among
adults in high-risk groups (e.g. health-care workers), and its wide application could
have a major global impact. Until this ideal TB vaccine is available, the early detection
and completion of appropriate treatment (through DOTS) of LTBI and TB disease would
remain as the best strategy to effectively control TB.26
In recent years, there has been a dramatic increase in the number of candidate TB vac-
cines evaluated in research laboratories. Better understanding of the immunological
deficits of BCG and impressive progress in knowledge of mycobacterial genomics have
paved the way for promising new products. The main vaccine targets are prevention of
infection in naïve individuals, prevention of reactivation of latent infection, and thera-
223
peutic vaccines to prevent relapse in TB patients. Successful prevention-of-recurrence
trials may lead to the development of therapeutic vaccines that will either shorten the
duration of treatment or enhance response to therapy. Currently, the most favored re-
search strategies include recombinant modified BCG vaccines, attenuated strains of M.
tuberculosis, subunit vaccines, and DNA vaccines. 27, 44
Several hypotheses have been forwarded regarding the variable effectiveness of BCG
against PTB and these are, namely: differences between BCG daughter strains, inade-
quate dosage of BCG in some trials, interference by environmental mycobacteria, ge-
netic differences in human populations and geographic differences in clinical isolates of
Mycobacterium tuberculosis. The decrease in effectiveness of BCG against PTB has
been hypothesized to be a consequence of increased immune suppression by BCG
antioxidants. As BCG evolved through different passages, and genomic duplications/
mutations occurred in the process, BCG production of antioxidants is increased thereby
suppressing the host‘s oxidant dependent immune response.28 So a new model of BCG
evolution is hoped to be one that reduces BCG antioxidants, thereby enhancing its im-
munogenicity against PTB. Another finding that may explain the decreased effective-
ness of BCG against PTB is the loss of BCG strain T-cell epitopes as the BCG strain
underwent repeated human manipulation over the years. So the restoration of lost BCG
strain epitopes may be a useful future vaccine developmental strategy.29
Isoniazid Preventive Therapy (IPT)
The risk to develop active TB following infection is high, based on the age as well as
the immune status of the child. The younger the patient is, the higher is the chance to
develop active tuberculosis, 30-40% in <1, 10-20% in 1-2 years, and 5% in 2-5 years of
age. The progress to active TB usually occurs within 12 months of primary infection.30
WHO, recognizing the identification and treatment of LTBI as a tool of global TB con-
trol, recommends isoniazid preventive therapy for 6 months (10 mg/kg/day, range 7-15
mg/kg/day, maximum dose: 300 mg/day) in children <5 years of age, who are house-
hold or close contacts of people with TB and who, after an appropriate clinical evalua-
tion, are found not to have active TB (strong recommendation, high quality of evidence
based on WHO GRADE System).31
Likewise, IPT for 6 months is recommemded to children living with HIV who are >12
months of age and who are unlikely to have TB disease on symptom based screening
and have no contact with a TB case.31
The 2010 Cochrane Review on INH for preventing TB in non-HIV infected persons in-
volving 11 RCT‘s with N=73,373 patients, the authors found the difference in 6 months
IPT vs. 12 months IPT not statistically significant.32
For high income or upper middle income countries with low incidence TB (IR
<100/100,000), the 2014 WHO Guidelines on the Management of Latent Tuberculosis
Infection( a Cochrane Review involving 53 RCT‘s), reports 6H is preferred over 9H
based on resource requirements, feasibility and acceptability by patients. The same
Cochrane Review reported that 6 months of INH is equivalent to 9 months of INH, as
well as INH and rifapentine weekly for 3 months, or 3-4 months of INH and rifampicin,
224
or 3-4 months of rifampicin ( 6H~9H~INH + RPT weekly x 3 months~ 3-4 mos HR ~ 3-4
mos R) as other regimens for LTBI.33
Patients on IPT should be properly documented and registered, based on the NTP-
DOTS Program. Register the child using Form 4 (TB Treatment/IPT Register) and Form
9 (IPT register). Monitor and assess the child at least every 2 months (weigh monthly
and adjust dosage according to weight; clinical manifestations). If the child develops
any sign or symptom, evaluate accordingly, and if found to have TB disease, stop IPT
and start on treatment for TB disease and declare IPT outcome as failed. The out-
comes of IPT are, namely: (1) completed; (2) lost to follow-up (a child who interrupted
IPT for two months or more); (3) died (a child who dies for any reason during the
course of IPT); (4) failed ( a child who develops TB disease during the IPT), and (5) not
evaluated ( a child who has been transferred to another health facility with referral for
continuation of IPT and whose treatment outcome is not known).3
Structures for TB Control
An effective national TB program should have a high cure rate, a low level of acquired
drug resistance, with a high case detection rate.34 TB control programs should have
ready access in providing health care with the necessary personnel, infrastructure, fa-
cilities, services and goods. Every health promoter and caregiver has to understand the
problem and be familiar with its network to optimize use of all resources available ac-
cording to set targets.
World Health Organization (WHO) Global Tuberculosis Programme
The WHO Global TB Programme provides global leadership on matters critical to TB,
develops evidenced-based policies, strategies, and standards for TB prevention, care
and control, and monitors their implementation. Together with Member States, it pro-
vides technical support and builds sustainable capacity. It monitors the global TB situa-
tion and measures progress in TB care and control. It is also responsible for the gener-
ation, translation and dissemination of valuable knowledge on TB. It likewise facilitates
and engages in partnerships for TB action.35
The World Health Organization Global TB Programme and the European Respiratory
Society have jointly led consultation among broad cross section of stakeholders to
match TB needs with digital health solutions. In September 2015, WHO developed the
Digital Health for the End TB strategy: an agenda for action.The said agenda aims to
scale up TB response through information and communication technology and outlines
the strategic directions to promote the integration of digital health concepts into TB pre-
vention and care activities. 36
WHO‘s new End TB Strategy has three pillars namely: Pillar 1- Integrated patient cen-
tered TB care and prevention; Pillar 2- Bold policies and supportive systems and Pillar
3- Intensified research and innovation.
Among the initiatives are the use of video treatment support for TB patient via mobiles,
eHealth portal to improve TB care, use of electronic medication monitors for TB pa-
tients, digital dashboards for TB indicators, epidemiological trends and digital notifica-
tion of TB cases and web based training for health professionals.36
225
National Tuberculosis Control Program3
The National Tuberculosis Control Program (NTP) is one of the public health programs
being managed and coordinated by the Infectious Diseases for Prevention and Control
Division (IDPCD) of the Disease Prevention and Control Bureau (DPCB) of the DOH.
The NTP has the mandate to develop TB control policies, standards and guidelines,
formulate the national strategic plan, manage program logistics, provide leadership and
technical assistance to the lower health offices/ units, manage data and monitor and
evaluate the program. The program‘s TB diagnostic and treatment protocols and strate-
gies are in accordance with the global strategy of STOP TB Partnership and the poli-
cies of World Healh Organization (WHO) and the International Standards for TB Care
(ISTC)3.
The NTP works with various offices of the DOH and coordinates with the Philippine
Health Insurance Corporation( Philhealth) for TB –DOTS accreditation and utilization of
the TB- DOTS outpatient benefit package. 3
The various regional, provincial and city NTP teams of the DOH are responsible for the
management and control of tuberculosis in these areas. DOTS facilities which could
either be public health facilities or NTP engaged private facilities provide TB diagnostic
and treatment services that are in accordance with NTP protocol.
NTP closely works with the 17 government offices and five private organization in com-
pliance with the Comprehensive and Unified Policy(CUP) issued by the office of the
President in 2003.37 Under the framework of public-private collaboration in TB DOTS,
NTP collaborates with non-governmental organizations such as the Philippine Coalition
against TB(PhilCAT), a consortium of 60 groups , and the 100 year old Philippine TB
Society, Inc.(PTSI) and many other organizations. Various developmental partners and
their projects provide technical and financial support to NTP such as the WHO, United
States Agency for International Development (USAID) , Global Fund AIDS, TB and
Malaria (Global Fund) , Research Institute of TB/ Japan Anti TB Associations (RIT/
JATA), Korean Foundation for International Health (KOFIH) and Korean International
Cooperation Agency (KOICA).3
The NTP in cooperation with Knowledge Management and Information Technology
Service (KMITS) developed the Integrated Tuberculosis Information System (ITIS) in
September 2014. ITIS is the official electronic TB information system and is designed to
collect, consolidate and report TB data on time from all health facilities managing TB. 38
Local Government Units are also assisted by the NTP in organizing the TB Diagnostic
Committees (TBDC) to reduce over and underdiagnosis of TB and ensure that active
TB cases are placed on treatment. TBDC is composed of experts in the hospitals and
specialists with training in TB together with the PPMD unit head or City TB Coordinator
who meet once a week or twice a month to improve the quality of diagnosis of sputum
smear negative chest radiography suggestive of pulmonary TB (sn-PTB). Treatment
dilemma of certain TB cases are also referred and discussed during the meetings.
Case records are presented individually, the history is obtained from the TBDC referral
form and discussed by the TB Coordinator and consensus is reached by the TBDC
members on the appropriate management of the case. TBDC recommendations range
226
from treatment initiation to surveillance of a patient not categorized as an active case of
TB. These TBDC activities contribute to TB control in the country by ensuring the judi-
cious use of resources.39
1. Vision , goals , objectives and strategies of the NTP3
Based on the updated 2010 -2016 Philipine Plan of Action to Control TB (PhilPACT),the
NTP‘s vision, goals, targets and objectives are as follows:
VISION: TB free Philippines
Goal: By 2016, the following should have been achieved :
Incidence rate : 246/ 100,000
Prevalence rate: 414/100,000
Mortality rate: 23/100,000
Targets by 2016:
Case Detection Rate, all forms 90%
Treatment Success Rate, all forms 90%
Notification Rate of MDRTB 62%
0f estimated MDR among notified TB cases
Treatment Success rate of MDR-TB cases 75%
Table 15.1 Objectives and Strategies: 3
OBJECTIVE STRATEGY
1. Reduce local variation in 1. Localize implementation of TB control.
TB control performance 2. Monitor health system performance.
(Governance and health information).
2. Scale up and sustain coverage of 3. Engage both public and private health
DOTS implementation (Health service care providers.
delivery and human resource). 4. Promote and strengthen positive be-
haviour of communities.
5. Address MDR-TB, TB/HIV, and needs
of vulnerable population.
3. Ensure provision of quality TB ser- 6. Regulate and make available quality
vices (Regulation) TB diagnostic tests and drugs.
7. Certify and accredit TB care provid-
ers.
4. Reduce out-of-pocket expenses relat- 8. Secure adequate funding and improve
ed to TB care (Financing) allocation and efficiency of fund utiliza-
tion.
The Philippines has achieved its Millennium Development Goals in TB at the end of
2015.The Sustainable Development Goals (SDG) replaced the MDGs which expired at
the end of 2015. Under Goal 3 of SDGs, health is strongly reflected which aims to
―Ensure healthy lives and promote well-being for all at all ages.‖ The target on TB is
included under this which states that ―by 2030 end the epidemics of AIDS, tuberculosis,
227
malaria and neglected tropical diseases and combat hepatitis, water borne diseases,
and other communicable diseases.‖ The indicators for this SDG is TB incidence per
1000 person years which is the estimated number of new and relapse tuberculosis cas-
es arising in a given year, expressed as the rate per 100,000 population (WHO). All
forms of TB are included including cases in people living with HIV. Another indicator is
the number of TB deaths which is the number of deaths attributable to TB in a given
year excluding HIV positive TB deaths (WHO). Household surveys and health facility
data given during the annual reporting per country contribute much to the global data-
base.
228
a. Networking: The Role of Private Practitioners, NGOs and Volunteers
Notification of TB cases
Notifying the appropriate sectors of all TB cases seen and initiating treatment are key
factors for effective TB control. Reported TB cases to the national health authorities
each year account for less than half the estimated number of TB cases worldwide, and
private practitioners in countries with high burden of TB in general do not report TB cas-
es to their relevant national health authority. In the Philippines, public-private partner-
ships accounted for 9% of all notifications in 2007.3
Involvement of Private Practitioners in TB Control Programs and TB Education of
the Community
In the 2007 National TB Prevalence Survey, the following are the health seeking behav-
ior among TB symptomatic individuals; self medicated (43.4%); no action taken
(25.1%); and those who sought medical consult (34.6%). More than half (54.3%) who
sought consult went to public health care givers; with an almost equal number went to
the DOTS centers (26.7%) and public hospitals/clinics (26.4%).40
Although majority of TB symptomatic individuals now seek consult in government hos-
pitals and DOTS centers, the latest National TB Prevalence Survey showed private
practitioners still manage a substantial number of TB symptomatic cases, i.e. in private
clinics (21.7%) and private hospitals (16%).40 The active participation of private physi-
cians in advocating the current strategies for TB control of the National TB program and
the notification of TB cases seen should be encouraged and strictly implemented.
TB management practices of private practitioners may deviate greatly from the best
practice methods being advocated. Private practitioners rely mostly on chest x-ray in
diagnosing TB cases, with varied drug regimens used in treating TB infection and dis-
ease. Every physician, therefore, has to be updated on the prevailing TB status and its
implications, current trends in diagnosis and treatment, management of MDR-TB, HIV
and TB with the latest practices advocated for TB prevention, control and elimination.
As lead health care providers, physicians play a vital part in the social awareness of
TB, through adequate education of the public. The public should be informed on the
magnitude of the TB problem, its mode of transmission, clinical course, preventive
measures, availability of cure, importance of early recognition and detection of TB
through cost-effective tools with emphasis on the immediate institution and completion
of adequate anti-TB therapy.
Public-Private Mix41,42
The WHO initiated the Public-Private Mix (PPM) in 1995 as a strategy that would link all
health care entities within the private and public sectors including health providers in
other government ministries to national TB programs for expansion of DOTS activities.
The WHO advocates the NTPs to work with the private sector to initiate and sustain
productive collaboration. In 2008, case notifications increased by 18% in areas where
Public-Private Mix DOTS (PPMD) was implemented. Treatment success rates among
229
patients managed in PPMD units have been in the range 85–90%.
PPM initiatives have been implemented in 40% of the Philippines and account for 9% of
national notifications.3 PPM in the Philippines is known as PPMD (Public-Private Mix for
DOTS) and was adopted as a national strategy to increase case detection and improve
access to DOTS services in poor urban areas in 2003. Since then, a close collaboration
between the NTP and the Philippines Coalition Against Tuberculosis (PhilCAT) has pro-
duced impressive results. As Sub-Recipient (SR) of the Global Fund TB Project (July 1,
2012 to March 31, 2014), PhilCAT scales up Hospital DOTS, engaging 212 public and
174 private hospitals as partners for finding the ―missingTB cases‖, instilling a
―partnership in the referral system‖, contributing a total of 12,330 cases or about 5.6%
of the Philippines‘ total estimated cases (all forms).43
PPM helps increase TB case detection and reduces diagnostic delays by involving all
health care providers in timely referral and diagnosis, ensuring proper notification of all
diagnosed cases, and enrollment on appropriate treatment under program guidelines.
The engagement of private providers entails adequate training of these providers to
ensure that the quality of their services is aligned with the International Standards for
TB care. In addition, evidence shows that private providers are often the first point of
contact for health services. These providers (with adequate training) could play an im-
portant role in case detection and referral of patients to the public sector cutting down
significantly on diagnostic delays.
All health care providers have critical roles in TB control including: helping identify peo-
ple with TB; prescribing treatment; acting as treatment supervisors; tracing treatment
defaulters; providing information and spreading awareness, training and supervision of
health care staff; and management of drug supplies and equipment.
TB in the Community: Role of Private Practitioners in TB Control Program
The control of TB in the country can only be realized through collaboration. In the up-
dated 2010-2016 Philippine Plan of Action to Control Tuberculosis (2014 PhilPACT),
one of its strategies in scaling up and sustaining coverage of DOTS implementation is
to engage both public and private health care providers. The Philippine Ambulatory Pe-
diatric Association, Inc. (PAPA), whose majority membership involves pediatricians,
carries out its mission by providing holistic, innovative and socially responsible
healthcare for children in the home, school and community through one of its projects,
a Community Based Tuberculosis Control Project- ―Tb Rally‖ in collaboration with the
National Guidelines for TB in children. From 2001 to 2013, PAPA had been involved in
in 8 municipalities of the country. In doing these projects, PAPA had demonstrated that
control of childhood TB entails the collaboration and clear set-goals of all key players,
namely: the government through the local government unit (LGU) providing the logisti-
cal support, the municipal health officer and the RHU team, funding from the Non-
Government Organizations as well as the government (Office of Congress 2nd District of
Sorsogon) as well as the private pediatric practitioners from PAPA and the locality, the
latter assisting in providing the technical knowledge and expertise through training,
guidance, monitoring and community empowerment to follow-up and complete the pro-
gram. Other stakeholders like the Rotary, the church and other religious organizations
230
and the Department of Education assist in community mobilization thus localizing the
implementation of TB control.
In doing these projects, PAPA has demonstrated that the control of childhood tubercu-
losis entails more time, effort, funding, manpower and political will. A private-public
partnership and the collaboration of all the key players play a very significant role in
lessening the burden of TB, ensure success and future eradication of tuberculosis
among the Filipino children.
The TB infection control (TBIC) measures include three levels of hierarchy - ad-
ministrative controls, engineering controls and wearing of respiratory protective
equipment when necessary.
Administrative Controls
Administrative controls are the first line of defense; it impacts the largest number of
people and the most important level in the hierarchy of TBIC. This involves measures
that will reduce risk of TB transmission by preventing the generation of droplet
nuclei or reducing exposure to droplet nuclei; and has the greatest impact on pre-
venting the spread of TB. It is intended primarily to reduce the risk of uninfected per-
sons exposed to persons who have TB disease, with emphasis on prompt detection
and treatment of TB cases and observance of airborne precautions. Administrative con-
trol measures include:
231
Administrative measures involve creating guidelines in TB infection control suitable to
the needs of the population; training of staff in the prompt detection/recognition, preven-
tion and treatment of TB cases; education of patients and increasing community aware-
ness; and coordination and communication with the existing TB program.
Environmental Controls
The use of environmental control involves technologies that will significantly pre-
vent the spread by reducing the concentration or inactivation of infectious drop-
let nuclei in ambient air. It is considered as the second line of defense for preventing
the spread of TB and, in combination with appropriate administrative controls, will re-
duce the risk of infection.
Primary environmental controls manage the source of infection by using local exhaust
ventilation (hoods, tents, or booths) and dilute and remove contaminated air by using
general ventilation. Secondary environmental controls manage the airflow to prevent
contamination of air in areas adjacent to the source (airborne infection isolation [AII]
rooms formerly called negative pressure rooms) and clean the air by using high effi-
ciency particulate air (HEPA) filtration, or ultraviolet germicidal irradiation.
Cost-effective environmental control measures that could be used at the DOTS facilities
are natural ventilation and mixed mode mechanical ventilation (i.e., use of fans together
with natural ventilation).
For DOTS facilities, the following are practical and simple measures that could be
adopted:
Isolation rooms should have its own ventilation source and exhaust. Air that comes
from Isolation rooms should NOT recirculate into the general air circulation. Isolation
rooms meant to function as source isolation for infectious cases should be constructed
with the recommended ventilation of 12 air exchanges per hour.
232
make sure the mask would fit snuggly to the face and below the chin with the flexible
band of the mask fit to the nose bridge.
Particulate respirators are designed to protect health care workers and other individuals
from inhaling droplet nuclei and should not be worn by patients; while surgical masks
should not be worn by the health-care workers, these are designed to reduce the num-
ber of droplets being exhaled into the air by persons with infectious TB disease when
they talk, breath, cough or sneeze. Persons with suspected or confirmed infectious TB
disease should be given and encouraged to use a surgical mask to minimize the risk of
expelling droplet nuclei into the air.
The particulate respirators should meet or exceed the Centers for Disease Control and
Prevention/National Institute for Occupational Services and Health (CDC/NIOSH)-
certified N95 or CE-certified FFP2 standards (filter at least 95% of airborne particles ≥
0.3 μm in diameter).
233
REFERENCES:
234
15. Research monograph. PhilPACT Retrieved from http//www.doh.gov.ph/sites/default
16. Goh T, et al. A Comparative Study of Japanese and British BCG Vaccine Strains in
Newborn Infants. Asia Pacific Journal. 1989;3(1):32-40.
17. Asia-Pacific Vaccination Council. General Information on the Disease and the Vac-
cine. September 2002.
18. Rodrigues L, Diwan V. Protective Effect of BCG against TB Meningitis and Miliary
Tuberculosis: A Meta-analysis. International Journal Epidemiology. 1993;22
(6):1154-1158.
19. Colditz G, et al. The Efficacy of BCG Vaccination in Newborns and Infants in the
Prevention of Tuberculosis: Meta-Analysis of Published Literature. Pediatrics.
1995;96(1):29-35.
20. Brewer T. Preventing Tuberculosis with BCG Vaccine: A Meta-analysis of the Liter-
ature. Clinical Infectious Disease. 2000;31(Suppl)S:64-67.
21. Philippine Pediatric Society. Guide for Clinical Practice Guidelines: Childhood Tu-
berculosis. 2008:51.
22. Zelner JL, et al. Protection from BCG and IPT. Am J Respir Crit Care Med Vol 189
(7), Apr 1, 2014, pp853-859
23. Is BCG Vaccination Effective? Tubercle 62. 981;219-221.
24. World Health Organization. Weekly Epidemiological Record. Aug 1995;70(32):229-
231.
25. Advisory Council for the Elimination of Tuberculosis (ACET). June 1998; Develop-
ment of new Vaccines for Tuberculosis (Recommendations of the ACET). 21 Au-
gust 21 1998/47(RR13):1-6.
26. Weekly Epidemiological Record No. 4, 2004; 79,25-40.Available at http://
www.who.int/wer.
27. World Health Organization. Treatment of Tuberculosis: Guidelines for National
Programmes. 3rd ed. 2003
28. Kernodle DS, CID 2010: 51, (17 July), 177.
29. Wen Zhang, et al. Genomic Sequencing and Analysis of BCG Vaccine Strains.
PLOS One, Aug 2013.
30. Marais BJ, et al. The Natural History of Intra-thoracic TB. Int J Trans Lung Dis
2004; B(4):392-402.
31. WHO Guidance for NTP‘s on the Management of TB in Children, 2nd ed. 2014.
235
32. Smeija M, et al. Isoniazid for preventing TB in non-HIV infected persons. The
Cochrane Library, 2010, Issue 1.
33. WHO Guidance on the Management of Latent TB Infection (LTBI), 2015
34. World Health Organization. Treatment of Tuberculosis: Guidelines for National Pro-
grammes. 3rd edition. 2003
35. The Global TB Programme (2016, June) Retrieved from http://www.who.int/tb/
about/en/
36. Digital Health for the End TB Strategy (2015,Oct 7) Retrieved from http//
www.who.int/tb/areas-of-work/digital - health
37. Department of Health and Philippine Coalition Against Tuberculosis. Comprehen-
sive and Unified Policy for TB Control in the Philippines, Manila 2003
38. Utilization of the Integrated TB Information System (ITIS) in Quarterly National TB
Control Program (NTP) Reports. DOH Memorandum Order No. 2014-0284, Sep-
tember 2014
39. Queri et al. (2012) Tuberculosis Diagnosis Committees‘ Contribution to the National
TB Program in Manila and Quezon City. International Union Against Tuberculosis
and Lung Disease Journal Vol 2(3) pp.83-86
40. .Tropical Disease Foundation, Inc and Department of Health. Nationwide Tubercu-
losis Prevalence Survey 2007
41. World Health Organization website. Available from http:// www. Who. Int/ tb/
careproviders/ppm/en/index.html
42. Department of Health, Philippines, World Health Organization. Joint Tuberculosis
Programme Review. 2009
43. Mortera LL. Continuing the Legacy.KOALISYON 2013 pp.48 -49
44. 2016 TAG‘s Pipeline Report. Tuberculosis (TB) Edition. Progress in TB Vaccine De-
velopment, pp.19-23
236
APPENDIX 1A. DIAGNOSTIC ALGORITHM
237
APPENDIX 1B. SCREENING OF PEDIATRIC DRUG-
SUSCEPTIBLE HOUSEHOLD CONTACTS OF TB
238
APPENDIX 2. BCG IMMUNIZATION
CLEOTILDE H HOW, MD
BACKGROUND/RATIONALE
Mycobacterium bovis was first isolated by Nocard in 1902 but it was Calmette and
Guerin who developed the attenuated strain by allowing 231 passages in 13 years, be-
ginning in 1908. Although the first vaccine was given orally in 1912, the other forms of
the vaccine were later introduced intradermally (1927), by multiple puncture (1939) and
by scarification (1947).
The BCG vaccines at present that are used throughout the world in more than 100
countries may differ because of genetic changes that have occurred among the various
bacterial strains. This renders efficacy of different vaccination programs or trials highly
variable as well.
The immediate objective of BCG vaccination is to activate host cell-mediated immunity
through an innocuous primary infection, but not disease, with the live-attenuated bacil-
lus. Epidemiologic (e.g. case-control) studies and meta-analyses of published clinical
trials have shown the protective efficacy of BCG (up to 80%) against serious forms of
the disease such as meningitis and military tuberculosis. Evidence is not consistent to
support its efficacy against acquiring the infection among the exposed or progression to
primary disease once infected.
See Chapter 15 (Prevention and Control)
INDICATIONS
Use of the vaccine is part of the Expanded Programme on Immunization (EPI) of the
World Health Organization (WHO) for administration at birth or immediately thereafter.
It is recommended that infants be vaccinated soon after birth, just before discharge
from the nursery or lying-in clinic or at first consultation at the local health center or pri-
vate clinic. All previously unvaccinated children who consult at the health center are
given BCG as a preventive measure under the National TB Control Program.
BCG immunization of premature infants and those born with the condition, intrauterine
growth retardation (IUGR), are covered by the same guidelines of chronological age
and dosage requirement as their full-term counterparts with appropriate weight-for-age.
Prematurity or presence of IUGR should not be the reason for delay or withholding of
BCG.
Direct BCG inoculation without prior tuberculin testing has been shown to be safe
among these infants and children. In high-risk groups, however, where exposure and
disease are already suspected even in the very young, doing a tuberculin test is done
to prevent severe local reactions with BCG. This may uncover disease when correlated
with the other criteria for diagnosis of tuberculosis. BCG should also be considered in
older individuals such as health care workers who are still tuberculin negative and who
239
may be at great risk for acquiring infection and progression to disease once infected.
CONTRAINDICATIONS
The National Consensus on Childhood Tuberculosis (1997) and the earlier Consensus
in 1989 including the following conditions when BCG is not allowed: presence of im-
240
mune deficiency diseases or states where the immune system may be suppressed be-
cause of malignant disease or high-dose and/or prolonged therapy with corticosteroids
and other immunosuppressants; all HIV-infected children and adolescents, whether
asymptomatic or symptomatic. There is a new initiative to offer BCG even to HIV-
infected children who are asymptomatic, with normal CD4 and CD8 counts.
REFERENCE
1. Centers for Disease Control and Prevention. The role of BCG vaccine in the pre-
vention and control of tuberculosis in the United States: a joint statement by the Ad-
visory Committee for the Elimination of Tuberculosis and the Advisory Committee
on Immunization Practices. MMWR Morb Mortal Wkly Rep. 1996; 45 (RR-4): 1-18
241
APPENDIX 3. INTERNATIONAL CLASSIFICATION OF DISEASES
(ICD)
CODE DIAGNOSIS
A15 Respiratory tuberculosis, bacteriologically and histologically confirmed
A15.0 Tuberculosis of lung, confirmed by sputum microscopy, with or without
culture
A15.1 Tuberculosis of lung, confirmed by culture only
A15.2 Tuberculosis of lung, confirmed histologically
A15.3 Tuberculosis of lung, confirmed by unspecified means
A15.4 Tuberculosis of intrathoracic lymph nodes, confirmed bacteriologically
and histologically
A15.5 Tuberculosis of larynx, trachea, and bronchus, confirmed bacteriological-
ly and histologically
A15.6 Tuberculous pleurisy, confirmed bacteriologically and histologically
A15.7 Primary respiratory tuberculosis, confirmed bacteriologically and histolog-
ically
A15.8 Other respiratory tuberculosis, confirmed bacteriologically and histologi-
cally
A15.9 Respiratory tuberculosis unspecified, confirmed bacteriologically and his-
tologically
A16 Respiratory tuberculosis, not confirmed bacteriologically and histological-
ly
A16.0 Tuberculosis of lung, bacteriologically and histologically negative
A16.1 Tuberculosis of lung, bacteriological and histological examination not
done
A16.2 Tuberculosis of lung, without mention of bacteriological or histological
confirmation
A16.3 Tuberculosis of intrathoracic lymph nodes, without mention of bacterio-
logical or histological confirmation
A16.4 Tuberculosis of larynx, trachea and bronchus, without mention of bacteri-
ological or histological confirmation
A16.5 Tuberculous pleurisy, without mention of bacteriological or histological
confirmation
A16.7 Primary respiratory tuberculosis without mention of bacteriological or his-
tological confirmation
A16.8 Other respiratory tuberculosis, without mention of bacteriological or histo-
logical confirmation
A16.9 Respiratory tuberculosis unspecified, without mention of bacteriological
or histological confirmation
A17+ Tuberculosis of nervous system
A17.0 Tuberculous meningitis (G01*)
242
CODE DIAGNOSIS
A17.0 TB Meningitis
G01
A17.0+ Tuberculous meningitis
A17.1 Meningeal tuberculoma (G07*)
A17.1+ Meningeal tuberculoma (G07*)
A17.8 Other tuberculosis of nervous system
A17.8+ Other tuberculosis of nervous system
A17.9 Tuberculosis of nervous system, unspecified (G99.8*)
A17.9+ Tuberculosis of nervous system, unspecified (G99.8*)
A18 Tuberculosis of other organs
A18.0 Tuberculosis of bones and joints
A18.0+ Tuberculosis of bones and joints
A18.1 Tuberculosis of genitourinary system
A18.2 Tuberculous peripheral lymphadenopathy
A18.3 Tuberculosis of intestines, peritoneum and mesenteric glands
A18.4 Tuberculosis of skin and subcutaneous tissue
A18.5 Tuberculosis of eye
A18.6 Tuberculosis of ear
A18.7 Tuberculosis of adrenal glands (E35.1*)
A18.7+ Tuberculosis of adrenal glands (E35.1*)
A18.8 Tuberculosis of other specified organs
A19 Miliary tuberculosis
A19.0 Acute miliary tuberculosis of a single specified site
A19.1 Acute miliary tuberculosis of multiple sites
A19.2 Acute miliary tuberculosis, unspecified
A19.8 Other miliary tuberculosis
A19.9 Miliary tuberculosis unspecified
243
APPENDIX 4. REPUBLIC ACT NO. 10767
S. No. 2653
H. No. 5042
Republic of the Philippines
Congress of the Philippines
Metro Manila
Sixteenth Congress
Third Regular Session
Begun and held in Metro Manila, Monday, the twenty-seventh day of July, two thou-
sand fifteen.
244
pine Plan of Action to Eliminate Tuberculosis in consultation with appropriate public and
private entities. The Philippine Plan of Action shall consist of the following:
(a) The country‘s targets and strategies in addressing the disease;
(b) The prevention, diagnosis, treatment, care and support, and other components of
the country‘s response;
(c) The development and application of appropriate technologies to diagnose and treat
the disease;
(d) The strengthening of linkages with local and international organizations for possible
partnership in education, advocacy, research and funding assistance;
(e) The establishment of a review and monitoring system to gather data and monitor
the progress made in the elimination of tuberculosis; and
(f) The immediate mobilization of anti-TB services during and after natural and man-
made disasters through collaborative efforts of national and local governments and oth-
er entities.
SEC. 5. Strengthening of the National and Regional Coordinating Committees. – The
National Coordinating Committee (NCC) and the Regional Coordinating Committee
(RCC) of the DOH shall serve as the National TB Control Program‘s (NTP‘s) arm in
strengthening and supporting nationwide capacity for program operations and bridging
collaborative efforts between the public and private sector.
The Secretary of Health shall continue to improve the capability of the existing NCC
and RCC in ensuring efficiency in the implementation, monitoring and evaluation of the
Philippine Plan of Action and in the coordination of efforts of various sectors.
SEC. 6. Research, Demonstration Projects, Education and Training. – The Secretary of
Health shall, directly or through grants to public or nonprofit private entities, perform the
following activities:
(a) Conduct basic and clinical research based on the health agenda to be developed by
partners in the academe, health professional groups and other local health partners
with possible support from foreign organizations;
(b) Develop demonstration projects to generate evidence for responsive policies and to
develop regional capabilities for the prevention, detection, control, and elimination of
tuberculosis;
(c) Conduct nationwide public information campaign and education programs;
(d) Undertake education, training and clinical skills improvement activities for health
care providers;
(e) Provide support for model centers to sustain their initiatives under subparagraphs
245
(b), (c) and (d); and
(f) Collaborate with local and foreign organizations for partnership in various activities
and in providing technical and funding support.
SEC. 7. Strengthening of the Regional Centers for Health Development in the Provision
of Health Services to Eliminate TB. – The Secretary of Health shall strengthen the Re-
gional Centers for Health Development in the provision of health services to eliminate
TB by undertaking the following activities:
(a) Provide free laboratory services through the DOH retained hospitals;
(b) Provide reliable supply of drugs to patients for free by ensuring that local health cen-
ters, through coordination with local government units (LGUs) concerned, have suffi-
cient supply of medicines for the communities they serve;
(c) Undertake public information and education programs to train the public on basic
ways and means to prevent the spread of tuberculosis;
(d) Train and enhance the capability of health providers in both public and private hos-
pitals;
(e) Ensure the proper monitoring of tuberculosis cases in the country; and
(f) Ensure that monitoring services are extended as far as practicable, at the lowest lo-
cal level health unit.
SEC. 8. Education Programs. – The Secretary of Health, in coordination with the Com-
mission on Higher Education (CHED), shall encourage the faculty of schools of medi-
cine, nursing or medical technology and allied health institutions, to intensify infor-
mation and education programs, including the development of curricula, to significantly
increase the opportunities for students and for practicing providers to learn the princi-
ples and practices of preventing, detecting, managing, and controlling tuberculosis.
SEC. 9. Inclusion in Basic Education. – The Secretary of Health, in coordination with
the Secretary of the Department of Education (DepED), shall work for the inclusion of
modules on the principles and practices of preventing, detecting, managing and control-
ling tuberculosis in the health curriculum of every public and private elementary and
high school.
SEC. 10. Media Campaign. – The Secretary of Health, in coordination with the Philip-
pine Information Agency (PIA), shall encourage local media outlets to launch a media
campaign on tuberculosis control, treatment and management, using all forms of multi-
media and other electronic means of communication.
The media campaign shall include materials that would discourage the general public
from spitting in public places and exhibiting unhygienic behavior that tend to undermine
the overall effort of preventing the spread of the disease.
246
SEC. 11. Regulation on Sale and Use of TB Drugs. – The Food and Drug Administra-
tion (FDA) shall strengthen its implementation of the ―No prescription, No anti-TB
drugs‖ to regulate the sale and use of anti-TB drugs in the market. It shall also ensure
the quality of TB drugs distributed in the market.
SEC. 12. Notification on TB Cases. – All public and private health centers, hospitals
and facilities shall observe the national protocol on TB management arid shall notify the
DOH of all TB cases as prescribed under the Manual of Procedures of the National TB
Program and the Philippine Plan of Action on Tuberculosis Control.
SEC. 13. PhilHealth TB Package. – The Philippine Health Insurance Corporation, oth-
erwise known as the PhilHealth, shall, as far as practicable, expand its benefit package
for TB patients to include new, relapse and return-after-default cases, and extension of
treatment.
The PhilHealth shall enhance its present outpatient Directly Observed Treatment Short
Course (DOTS) package to make it more responsive to patients‘ needs. It shall likewise
increase the number of accredited DOTS facilities to widen target beneficiaries who
may avail of reimbursements.
SEC. 14. Report. – The Secretary of Health shall submit an annual report to the Com-
mittees on Health of the Senate and the House of Representatives on the activities car-
ried out to comply with the provisions of this Act.
SEC. 15. Appropriations. – The amount necessary to implement the provisions of this
Act shall be charged against the appropriations of the DOH, the DepED, the CHED and
the PIA under the General Appropriations Act.
SEC. 16. Implementing Rules and Regulations. – The DOH, in consultation with the
DepED, the CHED, the PIA, the LGUs, nongovernment organizations and other con-
cerned entities, shall issue the rules and regulations implementing the provisions of this
Act within ninety (90) days from its effectivity.
SEC. 17. Separability Clause. – If any provision or part hereof is held invalid or de-
clared unconstitutional, the other provisions which are not affected thereby shall contin-
ue to be in full force and effect.
SEC. 18. Repealing Clause. – Any law, presidential decree or issuance, executive or-
der, letter of instruction, administrative order, rule or regulation contrary to or incon-
sistent with the provisions of this Act is hereby repealed, modified or amended accord-
ingly.
SEC. 19. Effectivity. – This Act shall take effect fifteen (15) days after its publication in
the Official Gazette or in a newspaper of general circulation.
Approved,
247
(Sgd.) FELICIANO BELMONTE
(Sgd.) FRANKLIN M.
JR.
DRILON
Speaker of the House
President of the Senate
of Representatives
Senate Bill No. 2653 which was approved by the Senate on January 18, 2016 was
adopted by the House of Representatives as an amendment to House Bill No. 5042 on
January 19, 2016.
248