You are on page 1of 84

1 MllM?!~~I~~!

~/~[-
3 4067 02719 2565

1 C The
- Respiratory -
-
m .-

I
I
1 Structure of the respiratorysystem: lungs, airways and dead space

(a) Lung lobes (b) The airways

/ Nasal c a v ~ t y
/

,/'
Pharynx
Ep~glott~s
i i

Right lateral L e k lateral


aspect aspect
Sternal angle
(angle of Louis)
Trachea (generation 0 )

Bronchioles iqeneratlons 12-16)


Anterior aspect

Respiratory bronchioles
(generations 17-19)

(c) Bohr equation for measuring Anatomical dead space, End-tidal =


dead space Volume = VD alveolar gas
I In an exp~redbreath
none of t h e C0, exp~red
came from t h e dead space regon

M~xedexp~redgas Volume = V:, :.


Q u a n t ~ t yof CO,
M~xedexp~redC0, f r a c t ~ o n= FECo2 In m~xedexp~redalr = quantlty of CO,
from alveolar region

0 CO, - free gas CO, - c o n t a ~ n ~ ngas


g
End of expilation
End of inspiration

Lungs into three lobes: upper, middle and lower. The left lung has
The respiratory system consists of a pair of lungs within the an oblique fissure and two lobes (Fig. la). Vessels, nerves and
thoracic cage (Chapter 2). Its main function is gas exchange, but lymphatics enter the lungs on their medial surfaces at the lung root
other roles include speech, filtration of microthrombi arriving or hilum. Each lobe is divided into a number of wedge-shaped
from systemic veins and metabolic activities such as conversion of bronchopulmonary segments with their apices at the hilum
angiotensin I to angiotensin I1 and removal o r deactivation of sero- and bases at the lung surface. Each bronchopulmonary segment
tonin, bradykinin, norepinephrine, acetylcholine and drugs such as is supplied by its own segmental bronchus, artery and vein and can
propranolol and chlorpromazine. be removed surgically with little bleeding or air leakage from the
The right lung is divided by transverse and oblique fissures remaining lung.
10
The pulmonary nerve plexus lies behind each hilum, receiving muscle forming helical bands. Bronchioles start at about genera-
fibres from both vagi and the 2nd to 4th thoracic ganglia of the tion 12 and from this point on cartilage is absent. These airways
sympathetic trunk. Each vagus contains sensory afferents from are embedded in lung tissue, which holds them open like tent guy
lungs and airways and bronchoconstrictor and secretomotor effer- ropes. The terminal bronchioles (generation 16) lead to respira-
ents. Sympathetic fibres are bronchodilator. tory bronchioles, the first generation to have alveoli (Chapter 5) in
Each lung is lined by a thin membrane, the visceral pleura, which their walls. These lead to alveolar ducts and alveolar sacs (genera-
is continuous with the parietal pleura, lining the chest wall, di- tion 23), whose walls are entirely composed of alveoli.
aphragm, pericardium and mediastinum. The space between the The bronchi and airways down to the terminal bronchioles re-
parietal and visceral layers is tiny in health and lubricated with ceive nutrition from the bronchial arteries arising from the de-
pleural fluid. The right and left pleural cavities are separate and scending aorta. The respiratory bronchioles, alveolar ducts and sacs
each extends as the costodiaphragmatic recess below the lungs are supplied by the pulmonary circulation (Chapter 13). The res-
even during full inspiration. The parietal pleura is segmentally piratory epithelium is discussed in Chapter 16.
innervated by intercostal nerves and by the phrenic nerve, and
so pain from pleural inflammation (pleurisy) is often referred to Dead space
the chest wall or shoulder tip. The visceral pleura lacks sensory The upper respiratory tract and airways as far as the terminal bron-
innervation. chioles d o not take part in gas exchange. These conducting airways
Lymph channels are absent in alveolar walls, but accompany form the anatomical dead space (V,), whose volume is normally
small blood vessels conveying lymph towards the hilar bron- about 150mL. These airways have an air-conditioning function,
chopulmonary nodes and from there to tracheobronchial nodes warming, filtering and humidifying inspired air.
at the tracheal bifurcation. Some lymph from the lower lobe drains Alveoli that have lost their blood supply f o r example because of
to the posterior mediastinal nodes. a pulmonary embolus- no longer take part in gas exchange and
The upper respiratory tract consists of the nose, pharynx and form alveolar dead space. The sum of the anatomical and alveolar
larynx. The lower respiratory tract (Fig. 1 b) starts with the tra- dead space is known as the physiological dead space, ventilation
chea at the lower border of the cricoid cartilage, level with the of which is wasted in terms of gas exchange. In health, all alveoli
6th cervical vertebra (C6). It bifurcates into right a n d left main take part in gas exchange, so physiological dead space equals
bronchi at the level of the sternal angle and T415 (lower when up- anatomical dead space.
right and in inspiration). The right main bronchus is wider, shorter The volume of a breath or tidal volume (V,), is about 500 mL at
and more vertical than the left, so inhaled foreign bodies enter it rest. Resting respiratory frequency (f) is about 15 breathslmin, so
more easily. the volume entering the lungs each minute, the minute ventilation,
is about 7500mLlmin (=500x 15) at rest. Alveolar ventilation (V,)
Airways is the volume taking part in gas exchange each minute. At rest, dead
The airways divide repeatedly, with each successive generation ap- space volume = 150mL and alveolar ventilation is 5250mLlmin
proximately doubling in number. The trachea and main bronchi ( ~ ( 5 0 0 - 1 5 0 )15).
~
have U-shaped cartilage linked posteriorly by smooth muscle. The Bohr method for measuring anatomical dead space is based
Lobar bronchi supply the three right and two left lung lobes and on the principle that the degree to which dead space gas (0% C 0 2 )
divide to give segmental bronchi (generations 3 and 4). The dilutes alveolar gas (about 5% C 0 2 ) to give mixed expired gas
total cross-sectional area of each generation is minimum here, after (about 3.5%) depends on its volume (Fig. I c). Alveolar gas can be
which it rises rapidly, as increased numbers more than make u p for sampled at the end of the breath as end-tidal gas. The Bohr equa-
their reduced size. Generations 5-1 1 are small bronchi, the smallest tion can be modified to measure physiological dead space by using
being 1 mm in diameter. The lobar, segmental and small bronchi are arterial Pco2 to estimate the CO, in the gas-exchanging or ideal
supported by irregular plates of cartilage, with bronchial smooth alveoli.
2 The thoracic cage and respiratory muscles

(a) The sternum and ribs and their relationship t o the lungs and pleural cavities (b) Inferior aspect of a rib

the tubercle

Cardlac notch

( c ) An ntercostal space (d) Inferlor aspect of the diaphragm

Xiphisternum

9')
$31 Intercostal:

Innermost
~ntercostalmuscle
Internal
Intercostal muscle
External
~ntercostalmuscle

To avo~dthe reurovascular bundle, needles belng passed


through the Intercostal space (for example t o draln a
pleural infusion) should pass close t o the t o p of the rib
Thoracic cage psoas major and quadratus lumborum, respectively. The costal
The thoracic cage is composed of the sternum, ribs, intercostal part of the diaphragm is attached to the inner aspects of the 7th to
spaces and thoracic vertebral column, with the diaphragm di- 12th ribs and costal cartilages. The sternal part originates as two
viding the thorax from the abdomen. slips from the back of the xiphisternum. The phrenic nerves (C3,
The dagger-shaped sternum has three parts. The manubrium, 4, 5) supply motor fibres. Sensory innervation of the central di-
with which the first and upper parts of the second costal cartilage aphragm also runs in the phrenic, and pain from irritation of the di-
and the clavicle articulate (Fig. 2a), lies at the level of the third and aphragm is often referred to the corresponding dermatome for C4,
fourth thoracic vertebrae (Fig. 2b). The lower parts of the second the shoulder-tip. The lower intercostal nerves supply sensory fibres
and third to seventh ribs articulate with the body of the sternum to the peripheral diaphragm. The aorta, thoracic duct and azygos
(level with T5-T8). The angle between the manubrium and body at vein pass through the diaphragm at the aortic opening at the level of
the cartilaginous manubriosternal joint forms the sternal angle T12. The oesophagus, branches of the left gastric artery and vein
(angle of Louis) and this is a useful anatomical reference point. and both vagi pass through the oesophageal opening at the level of
The small xiphoid process (xiphisternum) usually remains carti- TI 0, and the inferior venacava and right phrenic nerve pass through
laginous well into adult life. an opening at the level of T8.
The first seven (true o r vertebrosternal) of the 12 pairs of ribs
are connected to the sternum by their costal cartilages. The hyaline Muscles of respiration
cartilages of the eighth, ninth and tenth (vertebrochondral) ribs Inspiratory muscles all act to increase thoracic volume, causing in-
articulate with the cartilage above, and the eleventh and twelfth are trapleural and alveolar pressure to fall to create an alveolar-to-
free (floating o r vertebral ribs). A typical rib (Fig. 2b) has a head mouth pressure gradient, drawing air into the lungs. The domes of
with two facets for articulation with the corresponding vertebra, the diaphragm, the main inspiratory muscle, move down when it
the intervertebral disc and the vertebra above. The rib also articu- contracts, by about 1.5 cm during quiet breathing and 6-7cm dur-
lates at the tubercle with the transverse process of the correspond- ing deep breathing. During quiet breathing, the first rib remains
ing vertebra. The two articular regions act like a hinge, forcing the fairly still and the external intercostals elevate and evert the suc-
rib to move through an axis passing through these areas. The flat- ceeding ribs, increasing both the anterior-posterior and transverse
tened shaft of the rib is weakest at the angle of the rib and this is diameters of the chest wall, the so-called 'bucket-handle' action.
where it tends to fracture in an adult. The upper two ribs, protected The expanded chest wall and lungs will recoil by themselves and
by the clavicle and the two floating ribs are least likely to fracture. quiet breathing uses no expiratory muscles.
There is a cervical rib attached to the transverse process of C7 in When ventilation or resistance to breathing is increased, accesso-
0.5% of people and the presence of this rib may cause paraesthesiae ry inspiratory muscles aid inspiration. These include the scalene
or vascular problems, due to pressure on the brachial plexus or sub- muscles, sternomastoids and serratus anterior. If the arms are
clavian artery. fixed by grasping the edge of a table, contraction of the pectoralis
Intercostal spaces contain external intercostal muscles whose major, which normally adducts the arm, helps expand the chest.
fibres pass downwards and forwards between the ribs. internal in- When ventilation exceeds about 40L/min, there is activation of
tercostal muscles whose fibres pass downwards and backwards expiratory muscles, especially abdominal muscles (rectus ab-
and an incomplete innermost intercostal layer (Fig. 2c). They are dominis, external a n d internal oblique), which speed up recoil of
innervated by intercostal nerves, which are the anterior primary the diaphragm by raising intra-abdominal pressure.
rami of thoracic nerves. Intercostal veins, arteries and nerves lie In quiet breathing ventilation is largely diaphragmatic, but when
in grooves on the undersurface of the corresponding rib, with the the phrenic nerves are damaged, normal ventilation can be main-
vein above, artery in the middle and nerve below. tained by the intercostal muscles. In a high cervical cord transection
The dome-shaped diaphragm (Fig. 2d) separates the thorax and all respiratory muscles are paralysed, but when the damage is below
abdomen and consists of a muscular peripheral part and a central the phrenic nerve roots (C3, 4, 5) breathing continues via the di-
tendon, which is partly fused with the pericardium. The muscular aphragm alone. In the newborn, ribs are horizontal, so rib move-
diaphragm takes its origin from the vertebrae and arcuate liga- ments cannot increase the volume of the chest and breathing is
ments, the rib cage and the sternum. The right crus arises from the entirely by the up-and-down action of the diaphragm or so-called
upper three lumbar vertebrae and the left crus from the upper two abdominal breathing. As the ribs become more oblique with in-
lumbar vertebrae. Their fibrous medial borders form the median creasing age, the movement of the ribs becomes more important to
arcuate ligament over the front of the aorta. The medial a n d lat- give thoracic breathing.
eral arcuate ligaments are thickenings of the fascia overlying the
3 Pressures and volumes during normal breathing

Funct~onalresidual (c)
Outward reco~l
o f chest wall \,
f----
--\

Pressure gradlent d ~ s t e n d ~ n g
I t h e lung (transmural =
\
/+' '\ I alveolap - ~ntrapleural)
'Negat~ve'
~ntrapleural
/ 4 " ) Pressure gradlent d r ~ v ~ nairg
along airways (mouth -alveolar

a lntrapleural

Alveolar pressure

,-'3

0.5
Volume
above FKC
(L)
0
lntrapleural -0.5
pressure
relative t o
atmospheric
(kPa) - 0.75

0.1
lntrapleural Alveolar
pressure, Oesophageal pressure 0
-0 5 kPa pressure, -0 (kPa) -0.1

Alveolar 0.5
pressure, 0 kPa
Airflow
(L/sec)
- 0.5
Inspiration Expiration Inspiration Expiration

- -
KV '
- - .-- ... .-- -
(VT) ( a t rest) 500mL Total lung capaclty (TLC)
/ V ~ t acl a p a c ~ t (VC)
y 5500mL Funct~onalres~dualcapaclty (FKC) 3500mL

/
l n s p ~ r a t o l yveserve volume (IKV) 3 3 0 0 m L
Exp~ratoryreserve volume (EKV) 1 7 0 0 m L
Residual volume (KV) IBOO~L /
-- -- _____.A
Functional residual capacity Pressures, flow and volume during a normal breathing cycle
The volume left in the lungs at the end of a normal breath is known During inspiration, the chest wall is expanded and intrapleural
as the functional residual capacity (FRC). At FRC, the respira- pressure falls. This increases the pressure gradient between the in-
tory muscles are relaxed and its volume is determined by the elastic trapleural space and alveoli (Fig. 3c), stretching the lungs. The alve-
properties of the lungs and chest wall. oli expand and alveolar pressure falls, creating a pressure gradient
The lungs are elastic bodies whose resting volume when removed between the mouth and alveoli, causing air to flow into the lungs.
from the body is very small. The natural resting position of the chest The airflow profile (Fig. 3d) closely follows that of alveolar pres-
wall, seen when the chest is opened surgically, is about 1 L larger sure. During expiration, both intrapleural pressure and alveolar
than at the end of a normal breath. pressure rise. In quiet breathing, intrapleural pressure remains neg-
In the living respiratory system, the lungs are sealed within the ative for the whole respiratory cycle, whereas alveolar pressure is
chest wall. Between these two elastic structures is the intrapleural negative during inspiration and positive during expiration. Alveolar
space, which contains only a few millilitres of fluid. When the res- pressure is always higher than intrapleural, because of the recoil of
piratory muscles are relaxed the lungs and chest wall recoil in oppo- the lung. It is zero at the end of both inspiration and expiration and
site directions, creating a subatmospheric ('negative') pressure in the airflow ceases momentarily. When ventilation is increased, the
space between them, and this tends to oppose the recoil of both the changes of intrapleural and alveolar pressure are greater and in ex-
lungs and chest wall. Functional residual capacity occurs when piration intrapleural pressure may rise above atmospheric. In forced
the outward recoil of the chest wall exactly balances theinward re- expiration, coughing or sneezing. intrapleural pressure may rise to
coil of the lungs (Fig. 3a). When the chest is opened, air enters the +8 kPa or more.
intrapleural space, the pressure becomes atmospheric and nothing
opposes the recoil of the lungs and chest wall. The lungs shrink to a Lung volumes
small volume and the chest wall springs out. If a subject breathes in and out of a simple water-filled spirome-
If the elastic recoil of either the lungs or chest wall is abnormally ter (Fig. 3e (i)), the drum falls and rises and the pen. attached by a
large or small. FRC will be abnormal. In lung fibrosis, the lungs are pulley system, produces a trace (Fig. 3e (ii)). The volume breathed
stiff and have increased elastic recoil, so the balance point, and in (or out) is known as the tidal volume and the trace shows sever-
hence FRC, occurs at a small lung volume. In emphysema, there is al resting tidal volumes, which are typically about 500mL. For the
lossof alveolar tissue and with it, loss of elastic recoil. When the res- fourth breath, the subject breathes in and out as fully as possible.
piratory muscles are relaxed, the reduced elastic recoil of the lungs This maximum tidal volume is the vital capacity. At the end of a
offers less opposition to the outward recoil of the chest wall and normal quiet inspiration, the subject could breathe in more and this
FRC is increased (barrel chest). Increased FRC can also occur be- is the inspiratory reserve volume. Similarly, the volume that he or
cause of 'air trapping' (see Chapter 7). she could exhale after a normal expiration is the expiratory reserve
volume. At the end of a maximal breath out, the volume remaining
lntrapleural pressure is the residual volume. Functional residual capacity and total
The space between the visceral pleura lining the lungs and the lung capacity are the volumes in the lungs at the end of a normal
parietaLpleura lining the chest wall is so small that measuring in- expiration and after a maximal breath in, respectively. Typical val-
trapleural pressure with a needle risks puncturing the lung. In- ues in an adult male are given in Table 1. Although a zero volume
trapleural pressure can be indirectly assessed from oesophageal line is shown (Fig. 3e (ii)), it is not possible to know where this actu-
pressure (Fig. 3b). The oesophagus is normally closed at the top ally is on a trace, because the subject cannot empty the lungs into the
and bottom except during swallowing and in the upright subject the drum. For this reason, although illustrated in Fig. 3e (ii), volumes
oesophageal pressure is the same as in the neighbouring intrapleur- on the right of Table 1 cannot be measured from a simple spirome-
al space. The subject swallows a balloon containing a little air and ter trace. They can be measured using helium dilution or body
its pressure is measured via a tube connected to a manometer. Grav- plethysmography (Chapter 18). The range of normal lung vol-
ity affects the fluid-lined intrapleural space and at FRC in an up- umes is large and an individual's volumes must be assessed with the
right subject, the intrapleural pressure at the apex of the lungs is aid of nomograms that give the predicted value of each volume for
about -0.5 kPa (-5cm H 2 0 )and about -0.2 kPa (-2cm H,O) at the the subject's age, sex and height.
bottom.
4 Gas laws and respiratory symbols

(a) Standard respiratory symbols

I ( Primary symbols I I
F = Fractional concentration of gas P = Pressure or partial pressure
C = Content of a gas in blood 5 = Saturation of haemoglobin with oxygen
V =Volume of a gas C2 =Volume of blood
A d o t over a letter means a time derivative, e.g. i/ =Ventilation (Llmin)
& = Blood flow (Llmin)
Secondary symbols

Gae: I = Inspired gas Blood: a = Arterial


E = Expired gas v = Venous
A = Alveolar gas c = Capillary
D = Dead space gas A dash means mixed or mean
T = Tidal e.g. J = Mixed venous
B = Barometric A ' after a symbol means end
ET = End-tidal e.g. c' = End-capillary

/ Tertiary symbolo Examples


I
1
O2 = Oxygen m2 = Oxygen consumption
C02 = Carbon dioxide P A C O=~Alveolar partial pressure
CO = Carbon monoxide of carbon dioxide

(b) Correction facto-s for gas volumes

-37
273 +
- P '
I
= volume
(
---------
Volume (BTps) r 4 7 f Pg and PHZO
are ln mmHg
+ t°C) PB - 6 3')

Volume (5TPD) =
i ,0: :72( (
volume (AT;rps)- -* 7 6 0 ~fPB and PHZOare n mmHg
PB~::P)

I
1 1 (c) Partla1 pressure of a gas n a lhquid
I
Liquid X containing dissolved gas, g, is exposed t o a gas phase
containing g a t three different partial pressures, PI, P2, P3. Only
when the Pg = P2 does the number of gas molecules leaving the
liquid per minute (t)equal the number entering the liquid (J-)
- i.e. t h e liquid and gas phases are in equilibrium.
:. Partial pressure of gas, 9, in liquid X (PXg) = P2

II Liquid Y also contains gas, g and is also in equilibrium with the


I I
4 gas phase when Pg = P2

p, :. Partlai pressure of gas, g, ~nI q u d Y (PYg) = P2

T However, the solublllty o f gas, g, ~nlhquld Y 1s less than n llquld X,


so a t the same partlal pressure, llquid Y contains a lower
concentration of g

Note In the bottom left flask, qas moves aqalnst ~ t s

II concentraton gradlent.
II
To understand the processes involved in respiration and how valid The effect of pressure and temperature on gasvolumes
nleasurenlents are made. it is important to understand the behav- The inverse relationship between the volume of a perfect gas and its
iour of gases in both gas mixtures and in liquids. pressure, described by Boyle's law (PC= 1IV) and the direct rela-
tionship between volume and absolute temperature (= 273+"C) de-
Fractional concentration and partial pressure 01 gases scribed by Charles' law (VC=T ) are important when measuring gas
in the gas mixture volumes. Gas collected in a bag or spirometer will shrink, both
Dalton's law states that when two or more gases, which do not react because of the direct effect of falling temperature (Charles'law) and
chemically. are present in the same container, the total pressure is because water vapour condenses as temperature falls. To enable
the sum of the partial pressures (the pressure that each gas would valid comparisons, volumes at ambient temperature and pressure
exert if isolated in the container). saturated with water (ATPS) are corrected to those they would
The total pressure exerted by the atmosphere was tradi- occupy under standard conditions. For measurements of lung
tionally measured by inverting a long mercury-filled glass tube volumes, this is to body temperature and pressure saturated with
over a mercury reservoir. At sea level, the height of the column water (BTPS). For O2consumption or CO, production. standard
supported is normally about 760mmHg (torr), which in SI units is temperature and pressure dry (STPD) (0°C 101.3kPa
about 101 kPa (1 kPa=7,50rnmHg). Dried air contains 2 1%oxygen. (760mmHg), PH~O=O) are usually used, so that each litre contains
The remaining gases are nitrogen, 78. I%, and inert gases such as the same number of molecules (1 mole =22.4L).
argon and helium, 0.9'%, although for convenience these physio- Boyle's law, Charles' law and the reduction of saturated vapour
logically inert gases are often pooled as 'nitrogen, 79%'. Air is con- pressure with temperature are combined in the equations for cor-
sidered to be C02-free,as the amount present (0.04%) is very small. recting volumes given in Fig. 4b.
Standardized symbols used in respiratory physiology are shown in
Fig. 4a. Gases dissolved in liquids
According to Dalton's law: If a gas is exposed to a liquid to which it does not react. gas particles
will move into the liquid. Henry's law states that the number of
Dry partial pressure oxygen in inspired air ( PIOJ molecules dissolving in the liquid is directly proportional to the
=oxygen fraction ( F o ~x) total barometric pressure (PB) partial pressu:e at the surface of the gas.
=0.21xl01(760)=21.2kPa (159mmHg) The constant of proportionality is the solubility of the gas in the
liquid and it is affected by the gas, the liquid and the temperature,
At altitude, the oxygen fraction of air is unaltered but barometric tending to fall as temperature rises.
pressure is reduced, being about 33.6 kPa (252 mmHg) on the top of
Everest. Content of dissolved gas X in a liquid Y
= solubility of X in Y x partial pressure of X at surface
Water vapour pressure
Air contains variable amounts of water vapour, depending on the The partial pressure of a gas in a liquid or gas tension, is a
water it has been exposed to and the temperature. The maximum or more difficult concept than that of partial pressure in a gas phase,
saturated water vapour pressure is higher in warm than cool air: where we can visualize the pressure of the molecules holding up a
at 20°C, it is 2.33 kPa (17.5mmHg). whereas at body temperature column of mercury. The molecules of the gas in the liquid phase will
(37"C), it is 6.3 kPa (47mmHg). The relative humidity (actual1 move about in the liquid and have a tendency to escape from the sur-
saturated water vapour pressurex 100%) of inspired air varies with face, which can be opposed by molecules of the same gas in a gas
climate; if it is 40'% at 20°C, water vapour pressure will be 0.9 kPa phase in contact with the liquid (Fig. 4c). If the partial pressure of
(7mmHg). The presence of water vapour means that ambient Fez the gas in the gas phase is altered until there is no net movement of
and FN* are usually a little lower than the dry fractions given above. gas between the gas phase and the liquid phase, the gas and liquid
Air passing down the airways quickly reaches body temperature are said to be in equilibrium. By definition, the partial pressure of a
(37°C) and 100'i/;1saturation. Total pressure remains close to baro- gas in a liquid is equal to the partial pressure of that gas in a gas
metric, so the added water vapour causes significant dilution of the phase with which it is in equilibrium. Partial pressure gradient (not
other gases. The available pressure for the other gases is therefore PB concentration gradient) always determines the direction of move-
-6.3kPa (P B -47mmHg). ment between phases such as a gas and liquid phase.
The partial pressure of moist inspired oxygen (PIo,)=0.21 x
(P B saturated vapour pressure at 37°C). At sea level, this is
- Note on time derivative symbols
19.9 kPa (=0.21 x(101-6.3)) or 149.7mmHg. At altitude. as PB Time derivatives are properly denoted by a dot over the symbol (e.g.
falls, the dilution of inspired gas with water vapour becomes rela- ' A, alveolar ventilation in Llmin, See Fig. 4a). However, for terms
?
tively more important. On the top of Everest, where P B is about such as the ventilation/perfusion ratio (VA/Q) the dots are often
33.6 kPa (252 mmHg), moist Pio, is 5.7 kPa (43 mmHg). omitted, and this convention is followed throughout this book.
5 Diffusion

1 (a) The alveolar-capillary membrane (b) Transfer of gases across alveolar-capillary membrane

Alveolar
N2O
I _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ _ - - -

(gap between

Alveolar
eoitheiium

Y
Endothelum
u co
Collagen and 1 I I I

elastin fibres 0 0.25 0.5 0.75


f Time along pulmonary capillary (5)
Mixed venous
Plasma
blood

(c) Diffusion through a sheet of tissue (e) The oxygen cascade: oxygen tension frum ambient air t o mitochondria

(d) The diffusion path t t ~ r o u g t h~ e alveolar-capillary membrane

Alveolar
epithelium

Interstitial
flu~d
Endothelium

$"'
i Red blood cell Capillary
+b5

Oxygen and carbon dioxide are transported in the body by The alveolar-capillary membrane (Fig 5a)
a mixture of bulk flow and diffusion. Bulk flow, generated by dif- There are about 300 million alveoli, approximately 0.2mm in diam-
ferences in total fluid pressure, is important in most of the airways eter, in an adult male. The wall between alveoli consists of two lay-
and in transporting blood containing these gahes betheen pul- crs of alveolar epithelium resting on two separate basement
monary and tissue capillaries. Diffusion, driven by partial pressure membranes enclosing the interstitial space, containing pulmonary
differences, is important in the last few millimetres of the airways. capillaries, elastin and collagen fibres. Together, the alveolar ep-
across the alveolar-capillary membrane and between tissue capil- ithelium and capillary endothelium form the alveolar-capillary
laries and mitochondria. membrane, through which gases diffusc. The alveolar-capillary
membrane is very thin (<0.4pm), except where collagen and elastin ance to diffusion, and so some prefer to call this constant the trans-
fibresareconcentrated, with a total surface area of about 85 m2. The fer factor (TLo~).
alveolar epithelium has two main types of cell. Type I cells line the Although measurement of DLo2is desirable, it is not possible be-
alveoli and are relatively devoid of organelles. Type I1 alveolar cells cause mean capillary Po, (P,o,) cannot be measured.
are rounded with large nuclei, microvilli and cytoplasm containing
striated osmiophilic lamellar bodies which store surfactant, an im- Carbon monoxide diffusing capacity, DLco
portant component of alveolar lining fluid (Chapter 6). C O uptake from the lungs (VCO)
-
P,co - Pzc0
Diffusion and perfusion limitation (Fig. 5b)
If gas containing the poorly soluble gas nitrous oxide (N20) is in- C O diffuses through the same pathway as 0, and its rate of dif-
haled, pulmonary capillary PN,Orises and quickly equilibrates with fusion is affected by the same factors that affect oxygen transfer.
alveolar PN20 With no alveolar-capillary partial pressure gradient However, unlike DLo2,DLcois measurable. Once C O arrives in the
remaining, diffusion ceases for the rest of the pulmonary capillary pulmonary capillary blood, it too combines with haemoglobin.
and uptake can only be increased by increasing pulmonary capillary Haemoglobin has approximately 240 times the affinity for CO than
blood flow. N,O uptake is said to be perfusion-limited. In contrast, it does for 0, and consequently as C O is transferred, almost all of it
when a mixture containing carbon monoxide (CO) is breathed, the enters chemical combination and the mean pulmonary capillary
CO combines so avidly with haemoglobin that pulmonary capillary P c o can be assumed to be zero.
Pco rises little. The pressure gradient driving diffusion is preserved This simplifies the equation to:
along the capillary. CO uptake would not be increased by increased
perfusion, but it would be if diffusion resistance was reduced by Carbon monoxide uptake from the lungs (VCO)
DLco=
reduced thickness or increased area of the alveolar-capillary mem- PAC0
brane. CO transfer is diffusion-limited. Oxygen transfer lies be-
tween these two extremes, but is normally perfusion-limited. Several methods are used for measuring DLco, but all involve
breathing a low level of C O (e.g. 0.3%). Helium is included as a
Factorsaffecting diffusion across a membrane (Fickand measure bf dilution by alveolar gas. By sampling exhaled gas, C O
uptake and mean alveolar Pco can be calculated. The normal value
Graham's laws)
For a sheet of tissue of area A and thickness T through which gas G depends on the method used, but is about 15-30mL/min/mmHg
(112-225 mLIminlkPa). DLco is sometimes divided by alveolar
is passing (Fig. 5c):
volume to give an index (Kco) that corrects for different lung
volumes.
A
Rate of transfer of gas G = - ( Q - 4)
T F a c t o r s affecting D L c o
DLco is reduced by a reduction in alveolar-capillary membrane
The constant of proportionality area in emphysema, pulmonary emboli or lung resection and by
- solubility of the gas in the membrane(s) increased thickness in pulmonary oedema. In pulmonary fibrosis,
4Molecular weight of the gas (mw) diffusion is impaired both by thickening of the alveolar-capillary
membrane and by the reduced area caused by reduced lung volume.
Although the molecular weight of CO, is about 1.4 times that of O,, Increased pulmonary blood volume, as occurs in exercise, increases
it is about 20 times more soluble, and so diffuses more easily. the effective area and DLco. DLcois increased with polycythaemia
For the alveolar-capillary membrane, the pressure gradient driv- and reduced in anaemia. DLco is therefore non-specific and not
ing diffusion is alveolar (PA)minus mean pulmonary capillary (P,). diagnostic of any particular condition. Its value lies in its sensitivity
The constants (s, mw, A and T) can be combined to give a single to abnormality, which it may reveal when other lung function tests
constant, the diffusing capacity (DLg) of the lungs for gas, g: are normal. In hypoventilation, reduced PAcocauses reduced CO
uptake and DLcois unaffected.
Rate of transfer of gas, g = DLg(PA- PC) The oxygen cascade (Fig. 5e) shows how Po, falls between air and
mitochondria. Mitochondria] oxidative phosphorylation will cease
Oxygen diffusing capacity, DLo2 when Po, falls below 1mmHg (0.13 kPa) and this ultimately limits
Oxygen uptake from the lungs ( ~ 0 ~ ) the capillary Po, that can be tolerated and therefore the amount of
-
PAo2- Pzo2 oxygen that can be removed as blood passes through the tissues.
Capillary Po, must remain high enough to drive diffusion to cells at
Oxygen uptake per unit partial pressure gradient (DLo2)is affected a rate sufficient to match oxygen consumption and maintain mito-
by the rate of combination with haemoglobin as well as by resist- chondrial Po, above this critical level.
6 Lung mechanics: elastic forces

(a) S t a t i c pressure-volume loop (b) Dynamic pressure-vo!ume loop

If intra-pleural pressure and volume are recorded continuously


(lower panel) a pressure-volume (upper panel) can be constructed
from pairs of simultaneous measurements of volume e.4. (b) with
pressure (b'). Alternatively t h e pressure and volume signals can be

cm H20
0 1 2 kPa

Transmural pressure
(= intra-pleural pressure since

-0.5 -0.6 0.7 -0.8 -0.9 -~l.O


Intra-pleural pressure relative t o atmospheric (kpa)

Volume above
(c) Surface tension FRC (litre)

Intra-pleural
pressure relative
t o atmospheric
T= Surface tension

I Pressure
Inspiration Expiration lnspiration Expiration

I above ambient = P
i
(d) Effect of surface area

:. When t a p is opened the small


bubble empties into the large
r Water molecule

R2 < R,b u t T2 < TI because surface concentration of surfactant


is higher when t h e alveolus is small
The fall in R is more than offset by the fall in T,
:. since P = L T , P does n o t rise, b u t falls a s the alveolus shrinks
R
To breathe in, the inspiratory muscles must contract to overcome The air-fluid interface lining the alveoli
the impedance offered by the lungs and chest wall. This is mainly in During inspiration, as well as stretching the collagen and elastin
the form of frictional airway resistance (Chapter 7) and elastic re- fibres, the surface tension forces at the air-alveolar lining fluid in-
sistance to stretching of the lung and chest wall tissues and the fluid terface must be overcome. At the surface of a bubble, the attraction
lining the alveoli. of the fluid molecules for each other creates a tension, which tends
to shrink the bubble (Fig. 6c). LaPlace discovered that a gas bubble
Assessing the stiffness of the lungs: lung compliance in a liquid would shrink until the pressure, P, within it reached a
The 'stretchiness' of the lung is usually assessed as lung compliance value of 2T/R, where T is a constant, the surface tension of the fluid
(CL)which is the change in lung volume per unit change in distend- and R the radius of the bubble. The law of Laplace (P=2TIR)
ing pressure (CL=AVIAP). The distending pressure, P, is the pres- predicts that if two bubbles are made of the same fluid, the smaller
sure difference across the lung, which equals alveolar - intrapleural bubble will have a higher pressure within i t s i n c e when the radius
pressure. of curvature is small, a greater proportion of the surface tension is
lntrapleural pressure can be assessed using an oesophageal bal- directed to the centre of the bubble (lower panel of Fig. 6 . 1 ~ )When
.
loon (Chapter 3). Alveolar pressure cannot easily be measured the two bubbles are connected, the small bubble empties into the
directly, but when no air is flowing alveolar pressure must equal large bubble as air flows down the pressure gradient.
mouth pressure (i.e. zero). The transmural pressure, P, is then equal The presence of an air-fluid interface creates several potential
to -intrapleural pressure. The subject breathes in steps and mea- problems:
surements are taken while the breath is held and plotted as a static 1 It reduces lung compliance and the higher the surface tension the
pressure-volume (P-V) curve (Fig. 6a). The curve flattens as the lower the compliance.
lung volume approaches total lung capacity. The inspiratory curve 2 The alveoli would be inherently unstable, with the smaller alveoli
is slightly different from the expiratory curve and this hysteresis is a tending to collapse completely.
common property of elastic bodies. Static lung compliance is the 3 As the fluid tends to shrink away from the alveolar cells, it would
slope of the steepest part of this static pressure-volume curve in the create a suction force tending to cause transudation of fluid from
region just above functional residual capacity (FRC). the nearby pulmonary capillaries.
Lung compliance is normally about 1.5 LIkPa, but as with lung The absence of these problems in normal adults is thought to be
volumes it is affected by the subject's size, age and sex. In restrictive partly due t o the presence in the alveolar lining fluid of surfactant.
disease, such as lung fibrosis, lung compliance is low. Like a stiff
spring, once stretched, fibrosed lungs have an increased tendency to Surfactant
shrink back to their resting position or increased elastic recoil. The Pulmonary surfactant is a mixture of phospholipids such as
loss of alveolar tissue in emphysema makes them easier to stretch sphingomyelin and lecithin produced by the type I1 alveolar cells
and lung compliance is increased. Although safe, swallowing an (Chapter 5). The presence of these substances in the alveolar lining
oesophageal balloon is not very pleasant or convenient. Fortu- fluid lowers the surface tension and increases compliance. The
nately, it is often possible to deduce that a patient has stiff lungs phospholipids have a hydrophilic end that lies in the alveolar fluid
from other measurements such as F R C (Chapter 3, 17 and 24), and a hydrophobic end that projects into the alveolar gas and as a
forced expiratory volume in 1 s (FEV,) and forced vital capacity result they float on the surface of the lining fluid. As an alveolus
(FVC) (Chapter 17). shrinks, its surface area diminishes and the surface concentration of
surfactant rises (Fig. 6d). As surface tension falls with increasing
Dynamic pressure-volume loops and dynamic compliance surface concentration of surfactant, the increased tendency for
A dynamic pressure-volume loop (upper panel of Fig. 6b) is ob- alveoli to collapse when they shrink is offset and stability improved.
tained from continuous measurements of intrapleural pressure and Alveolar stability is also aided by the connection and mutual
volume during a normal breathing cycle (lower panel of Fig. 6b). pull of neighbouring alveoli, a phenomenon known as alveolar
There are two points, at the ends of inspiration and expiration. interdependence.
where airflow and alveolar pressure are zero (a, a' and e, e') and the Surfactant production in the fetus gradually increases in the last
slope of the line joining these points is dynamic compliance. In third of pregnancy and may be inadequate in babies born prema-
health, its value is similar to the static compliance, but in some dis- turely, giving rise to the typical problems of neonatal respiratory
eases it may be lower, as stiff areas may fill preferentially during nor- distress syndrome (NRDS)-stiff lungs, areas of collapse and
mal breathing. Between the two zero flow points, the dynamic P-V transudation of fluid (Chapter 16).
loop appears fatter than the static P-V loop, as intrapleural pressure
must change more to drive airflow. In fact, the area of the
dynamic loop is a measure of the work done against airway
resistance (Chapter 7).
7 Lung mechanics: airway resistance

(a) Laminar and turbulent fiow (c) The effect of effort on inspiratory and expiratory aidlow
Effort dependent

Turbulent flow

(b) Main factors influencing bronchomotor tone

NANC (non-adrenergic C02 Sympathetic


non-cholinergic)
efferent (in vagus) I efferent
I

- - - - = Flow-volume curve for maximum effort from partly filled lungs


A = Peak expiratory flow rate with lungs filled t o t o t a l lung capacity
6 = Peak expiratory flow rate for partly fiiled lungs filled (KV + 3 litre)
TLC =Total lung capacity, KV = Residual volume

(e) Maximum flow-volume loops


Airway irritant Histamine,
receptors prostaglandins,
ieukotrines,
: serotonin

a
afferent
' 1 efferent

a
afferent
do
3 *t
A

M a s t cells,
eosinophils, etc

02 = 02 adrenergic receptor, VIP = vasoactive intestinal peptide


receptor, mus = muscuranic choinergic receptor

(d) Dynamic compression of airways

4 2 0
Beginning of Lung volume (L)
inspiration 600
- -,-Normal curve
-Obstructive airway disease o f smaller airways. Note:
concave appearance of forced expiratory curve
forced inspiratory flow affected less than forced expiratory flow
- - - Upper airway obstruction (e.g, tracheal stenosis). Note:
f l a t topped flowvolume curve
forced inspiratory fiow affected a s much a s expiratory flow
- Restrictive lung disease. Low peak flow rates are related t o low
volume. (Note: t h i s figure is drawn t o show t h e reiationship
between these traces by using absolute lung volume which cannot
Numbers are pressures in kPa (1 kPa = 7.5 mmHg) actually be obtained from a flowevolume loop alone).
rflow is driven by the mouth-to-alveolar pressure gradient gen- breathes between residual volume (RV) and total lung capacity
ted by the respiratory muscles (Chapters 2 and 3). (TLC). The inspiratory airflow at any volume increases progres-
sively with increasing effort (1 = minimum effort, 6 = maximum
AP (= mouth-alveolar pressure) effort). The flow-volume curves for progressively increasing expira-
RAW (=resistance of the airways) tory efforts (upper traces 1-6) are more complicated. In the early
part of expiration from TLC, flow is effort-dependent, but towards
r flow, gas particles move parallel to the walls, with cen- the end of the breath, as volume declines, the traces produced at dif-
moving faster than outer ones, creating a cone-shaped ferent effort levels come together. Expiratory airflow towards the
~ g 7a).
. The factors affecting laminar flow of a fluid of end of a breath is effort-independent and determined by lung vol-
osity, 11, in smooth straight tubes of length, I, and radius. r. are ume. Peak expiratory flow rate (PEFR) is seen to be reduced if the
ibed in Poiseuille's equation: lungs are only partially filled at the start of the forced expiration.
Effort-independent airflow is explained by dynamic compres-
sion of airways. Before the start of inspiration (Fig. 7d, upper
panel) pressure along the airways is zero, intrapleural pressure is
ng the radius of an airway increases its resistance 16-fold. negative (Chapter 3), and transmural pressure acts to hold airways
r, although the resistance of an individual bronchiole is open. Intrapleural pressure is negative during both quiet and forced
1. The total resistance of each inspiration and it remains negative in quiet expiration, so transmur-
ration of peripheral airways is normally low and the overall re- a1 pressure holds airways open. In a forced expiration, however, ex-
nce of lung airways is dominated by the larger airways. Outside piratory nluscle contraction raises intrapleural pressure well above
ung, the nose and pharynx contribute substantial resistance, atmospheric (e.g. 8 kPa, 60mmHg), increasing the pressure gradient
can be bypassed by mouth breathing in exercise. Peripheral from alveoli to mouth. This would be expected to increase airflow,
are often affected by disease, but since their resistance must but the increased intrapleural pressure also acts to compress air-
considerably to measurably affect airway resistance (RAW) ways. Airway pressure falls progressively along the airway and at
ey are known as the silent zone. some point-usually in the bronchi- the airway pressure will be
igher linear velocities, especially in wide airways and near sufficiently below intrapleural pressure for the airway to collapse,
points, flow may become turbulent. With turbulence, the despite its cartilaginous support. Pressure will then build up distally,
ont is square and flow = ~ A (notP AP), reflecting the dissipa- opening the airways again. The resulting fluttering walls can be seen
of energy in the formation of eddies. Normally. at rest, flow is on bronchoscopy and produce the brassy note audible on forced ex-
r throughout the airways, but in exercise it may become tur- piration in normal people.
, especially in the trachea, generating characteristic harsh
RAW in disease
Increased airway resistance is important in many diseases and can
ecting airway resistance be measured using a body plethysmograph. In health, RAW is about
s m o o t h muscle a n d epithelium 0.2 kPa/L/s (1.5 mmHg/L/s). More commonly, airway resistance is
nchial smooth muscle (Fig. 7b) receives a parasympathetic assessed indirectly from forced expiratory measurements, such as
oconstrictor nerve supply, which forms the efferent limb of forced expiratory volume in 1 s (FEV,). forced vital capacity
from airway irritant receptors. The airways contain p2- (FVC) and PEFR (Chapter 17). High airway resistance accentuates
nergic receptors, which cause relaxation when stimulated by the dynamic compression of airways by augmenting the pressure drop
rse sympathetic innervation or more importantly by circulating along airways. In addition, the airways may be less able to resist
ephrine (adrenaline) or drugs such as salbutamol. Parasympa- compression. in emphysema because of reduced radial traction and
c bronchoconstriction is inhibited by activation of airway in asthma because of bronchoconstriction. Collapse of small air-
tch receptors and C 0 2 has a direct bronchodilator effect. Pollu- ways may occur, leading to incomplete expiration, air trapping and
nts (e.g. sulphur dioxide, ozone) and substances released from increased functional residual capacity. Inability to produce high
ast cells and eosinophils can increase RAW via bronchoconstric- expiratory airflow impairs effective coughing, which can lead to a
on, mucosal oedema, mucus hypersecretion, mucus plugging and vicious cycle as secretions accumulate, further increasing RAW and
ithelial shedding. all of which are important in asthma (Chapter further reducing peak flow.
). Airway resistance can also be increased by chronic mucosal Expiratory wheezes (rhonchi), heard in asthma and other ob-
ertrophy in chronic obstructive pulmonary disease (COPD) structive diseases, are probably generated by oscillations in oppos-
hapter 22) and by material within the airways, such as inhaled ing airway walls near their point of closure. like sounds from the
reign bodies or tumours (Chapter 25). reeds of an oboe. A reasonable airflow is needed to generate such
sounds and when constriction becomes very severe, they disappear
ransrnural (airway -intrapleural) pressure gradient to give the ominous silent chest seen in life-threatening asthma.
scan have important effects on airway calibre, and this underlies Small airway collapse leads to characteristic shape of the flow-
effects of effort on airflow illustrated in Fig. 7c. Airflow is mea- volume curve in obstructive airway disease (Fig. 7e), which differs
red continuously and plotted against lung volume as the subject from that in upper airway obstruction and restrictive lung disease.
8 Carriage of oxygen

Haemoglobin structure

Haemoglobin is composed of four subunits, each containing o protein


chain (globin) and a haem group. Normal adult haemoglobin, HbA,
contains two identical a-chains composed of 141 amino acids and two
!.-chains composed of 146 amino acids. The haem group (0)
is
attached t c each chain a t a histidine residue and each has an iron
atom in the ferrous form, wk8ichbinds t o an oxygen molecule. The haem
groups lie in crevices in the crumpled ball o f globin chains. The exact 3 D
(or quaternary) structure of haemoglobin can change and alter the
accessibility of the oxygen binding site. Each molecule of haemoglobin
can bind up t o four molecules o f oxygen in a series of reactions which
can be summarized as:
Hb, + 4 0 2 @ H b,(02),

The oxygen - haemoglobin


20
dissociation curve, haemoglobin
on cent ration, 15g!dL

-
2

4
15
a = Normal a r t e r ~ ablood
'V"
l
,pnYe -- l Z" kPa (103 mmHg)

-c
2
E
O2contc:nt = 20 mL1dL
Oisaturat Ion = 97%
c 10
v = Rest~ngmixed verous
0
- Po2 = 5.3 kPa (4CmmHg)
L
m
0

2
O 5

0
0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 1 9 2 0 kPa
Po.,

Anaemia and carbon 20 Note: for simolicity the Bohr


monoxide pcisoning shift is ignored

-4
_i
15

-
i
E -
-
Hb=15g/dL
c Hb = 7.5gldL
2 10
0

c
u
m
2
0 5

0
0 2 4 6 8 10 12 14 16 kPa
Po2
1 I
0 70 40 60 80 100 120mmHg
At rest, an adult male consumes about 250mL oxygenlmin, which binding. The affinity of haemoglobin for oxygen and hence position
may rise to more than 4000mLlmin in exercise if he is very fit. Oxy- of the dissociation curve varies with local conditions. A reduced
gen diffuses from alveolus to blood until equilibrium is reached oxygen affinity, shown by a right shift in the curve, is caused by a fall
when pulmonary capillary Po, equals alveolar Po,. The solubility in pH. a rise in Pco, (the B o h r effect) or increased temperature
of oxygen in blood is low-0.000225mL oxygen per mL of blood (Fig. 8b). These changes occur in metabolically active tissues such
per kPa (0.00003 rnL/mL/mmHg)-so that at a normal arterial Po2 as exercising muscle and encourage oxygen release. In the lungs,
of 13.3kPa (100mmHg) there is only 0.3mL dissolved in each oxygen uptake is aided by the increasing affinity of haemoglobin for
lOOmL of blood. The main function of the red blood cell pig- oxygen, caused by falling Pco, and temperature and increased pH
ment, haemoglobin, whose structure is shown in Fig. 8a, is to and reflected by a left shift of the curve. The Po, at which the
carry the large quantities of oxygen needed by the tissues. haemoglobin is 50% saturated is known as the P,,. Under normal
Each gram of haemoglobin combines with up to 1.34mL oxygen, arterial conditions (pH =7.4, Pco2=5.3 kPa or 40 mmHg, temp =
so with a haemoglobin concentration, [Hb], of 15 gIdL, blood con- 37°C) PS0=3.5kPa(26.3mmHg); right shifts raise the P,, and left
tains a maximum of 20 mL1dL oxygen bound to haemoglobin. This shifts lower it. A rise in the concentration of 2,3-di(or bi)pho-
is known as the oxygen capacity, which varies with [Hb]. The sphogIycerate (2,3-DPG), which is a by-product of glycolysis in
actual amount of oxygen bound also depends on the Po,. The per- red cells, also causes a right shift. A rise in 2.3-DPG occurs in
centage of the available binding sites bound to oxygen is known as anaemia, causing a modest increase in PSo Blood bank storage
the oxygen saturation. causes progressive depletion of 2,3-DPG and an undesirable left
shift, but this can be minimized by storing the blood with citrate-
Oxygen saturation phosphate-dextrose.
Amount of oxvgen bound to In anaemia, at any given Po,, the oxygen content is reduced be-
cause of the reduced concentration of binding sites. Figure 8c shows
- haemoglobin (oxyg& content), mLldL x 1 OO%,
oxygen capacity, mLldL the dissociation curve for normal blood and for blood with [Hb]=
7.5gldL. Alveolar and arterial Po, is normal in anaemia and there-
The rate of rise of oxygen content with increasing partial pressure fore arterial 0, content is IOmLldL. At rest, the tissues need to re-
depends on the number of free haemoglobin binding sites remain- move abhut 5mLldL from the blood passing through them. To
ing and their affinity for oxygen. As each oxygen molecule binds in achieve this mixed venous content, Po, will need to fall to about
turn to the four haem groups, the quaternary structure alters and 5.3 kPa (40mmHg) (A in Fig. 8c) when [Hb]= 15gIdL and about
theaffinity of the remaining binding sites for oxygen increases. This 3.6kPa (mmHg) (B) when [Hb]=7.5gldL. The reduced venous and
cooperative binding increases the steepness of the oxygen- hence capillary Po, reduces the partial pressure gradient driving
haemoglobin dissociation curve in the middle (Fig. 8b), but diffusion of oxygen to the tissues. which may become inadequate in
the curve flattens again at partial pressures above about 8 kPa exercise when oxygen consumption increases.
(60mmHg) because there are few remaining sites. Figure 8c also shows the dissociation curve for blood that has
In arterial blood, Po, is normally about 13 kPa (l00mmHg) and 50% of oxygen-binding sites occupied by carbon monoxide (CO,
oxygen saturation about 97'%, and with a normal [Hb], an oxygen dashed line). Arterial oxygen content is IOmLIdL. but there is also
content about 20mLldL. Rises or modest falls in Po, from 13 kPa an altered shape and leftward shift of the dissociation curve, be-
(lOOmmHg), for example during hyperventilation or mild hypoven- cause C O binding increases the affinity of the remaining (CO-free)
tilation, cause little change in the arterial oxygen content, as the d ~ s - sites for oxygen. This impairs oxygen release in the tissues. Mixed
sociation curve is flat in this region. More severe reductions in PO?. venous Po, will now have to fall to 2 kPa (15 mmHg) (point C) to re-
to levels in the steep region, are associated with significant reduc- lease the 5 mL1dL required and this will greatly reduce the pressure
tions in oxygen saturation and content. Consequently, breathing gradient for diffusion. At about 50-60% carboxyhaemoglobin,
oxygen-enriched air may significantly raise arterial oxygen content symptoms of impaired cerebral oxygenation (headache, convul-
and hence exercise capacity at altitude and in patients with chronic sions, coma and death) develop, whereas anaemic patients with the
hypoxic respiratory disease, but has little effect on a normal person same arterial oxygen content are typically asymptomatic at rest.
at sea level. Haemoglobin has a high affinity for C O (about 240 times that for
Low Po2 in tissue capillaries causes oxygen release from haemo- oxygen), so breathing even low concentrations causes a progressive
globin, whereas the high Po, in pulmonary capillaries causes oxygen increase in the cherry-red carboxyhaemoglobin.
Carriage of carbon dioxide

-
.

( a ) CO, dlssoc~attoncurve

Red blood cell


I
(b) C02 uptake and 0, dellvery ~nthe tissues

Chlcr~des h ~ f t

TA $
CO; -+CO, + HO
, +H2C03 +H+ + C I f-- CI-

20 30 40 50 60 vmHg
I I I I I I
3 4 5 6 7 8 kPa
Ro, CA = carbon c anhydrase
I1 A = Ncrmal a r t e r ~ apolnt
l
H b = haem aroup
Hb4(02)+ = oxygenated haemoglob~n

I V = Ncrmal mixed venous potnt


I

Carbon dioxide (CO,) is produced by tissues and transported in the the tissues are producing. Conversely, oxygenation of haemoglobin
blood to the lungs, where it is expired. The amount of CO, that can in the lung assists the unloading of CO, from the blood. This is
be carried in the blood is much greater than that of O,, as seen in the illastrated by Fig. 9a and equation 2.
COz dissociation curve (Fig. 9a). The CO, dissociation curve is
also more linear and does not reach a plateau. CO, is transported in
I the blood as bicarbonate ions, as carbamino compounds combined Note that as a consequence of all the above, deoxygenated red
with proteins o r simply dissolved in the plasma. cells have a higher intracellular osmolality and water enters, causing
" I
Bicarbonate: About 601%of CO, is transported in the form of then1 to swell slightly. In the lung, CO, is given off, osn~olalityfalls
bicarbonate. CO, and water combine to form carbonic acid and the red cells shrink again.
(H,CO,) and thence bicarbonate (HCO,): Carbamino compounds: CO, combines rapidly with terminal
amino groups on proteins to form carbamino compounds:

CO, + Protein,NH, w Protein.NH.COOH (3)


The left-hand side of the equation proceeds slowly in plasma, but
is accelerated dramatically by the enzyme carbonic anhydrase In blood. the most prevalent protein is haemoglobin, which con]-
(CA). which is present in red blood cells. Ionization of carbonic acid bines with CO, t o form carbaminohaemoglobin. Reduced haemo-
to bicarbonate and H+is rapid in the absence of any enzyme. Bicar- globin forms carbamino con~poundsmore readily than oxygenated
bonate is therefore formed preferentially in the red cells, from which haemoglobin and this also contributes to the Haldane effect (Fig.
it easily diffuses out into the plasma. The red cell membrane is, how- Yb). Around 30%)of the CO, expircd by the lungs is carried as car-
ever, impermeable to H+ ions and they remain within the cell. In banlino compounds.
order to maintain electrical neutrality, CI- ions diffuse into the cell, CO, in solution: CO, is -20 times more soluble in water than 0 2 .
an effect known as the chloride shift (Fig. 9b). A build-up of H+ in A significant proportion (-10'!41) of the C 0 2exhaled is therefore car-
the red blood cell would impair further movement of equation 1 to ried to the lung dissolved in the plasma.
the right, thus limiting formation of bicarbonate. However, H+
binds avidly to reduced (deoxygenated) haemoglobin, i.e. haemo-
globin acts as a buffer, so the rise in H+concentration is limited and Hypoventilation and hyperventilation
more bicarbonate can be formed. Oxygenated haemoglobin does Ventilation is normally closely matched to the metabolic require-
not bind H+ so well, as it is more acid. This contributes to the ments of thc body and this can be estimated from the rate of CO,
Haldane effect, which states that, for any givcn Pco2, the CO, con- production (Chapter 12). The partial pressure of CO, in the alveoli
tent of deoxygenated blood is greater than that of oxygenated (PAco2)is proportional to the amount of CO, exhaled per minute
blood. As a result, when blood gives up oxygen to respiring tissues, (VCO?)as a fraction of total alveolar ventilation (V,), i.e. PAco, =
i.e. becomes deoxygenated, it is able t o take up more of the C 0 2that Vco2/[/A. The gas in the alveoli is in equilibrium with arterial blood,
so P,co, estimates the partial pressure in the blood (P,co,). At any ter 21) unless the attack is severe or prolonged enough to lead to ex-
given metabolic rate, doubling the alveolar ventilation halves alveo- hausion. Hypoventilation is difficult to achieve voluntarily, as the
lar and arterial Pco?, and halving alveolar ventilation doubles respiratory centres create an overwhelming desire to breathe.
P,co, and Pi,co,. Changes in alveolar ventilation also affect alveo- Hyperventilation can be induced voluntarily and in states of high
lar Po,, but the relationship is not as simple because 0, is present in anxiety or pain.
both inspired and expired gas. Thus doubling alveolar ventilation Hypoventilation leads to hypercapnia (high P,co2) and hypoxia
will halve the difference between the inspired and alveolar O2 frac- (low P,o,). Increasing severity of hypercapnia causes peripheral va-
tion. Hypoventilation (under-ventilation) and hyperventilation sodilatation, muscle twitching and hand flap, confusion, drowsiness
(over-ventilation) are therefore defined in terms of P,coS, so that a and eventually coma; there is a concomitant respiratory acidosis
patient is h1~puventilutingwhen P,coS>45mmHg (5.9 kPa) and hy- (Chapter 10). The effects of hypoxia are dealt with elsewhere
pervrnriluting when the PAco2<40mmHg(5.3kPa). Note that the (Chapter 8). A low P,co, as a result of hyperventilation causes light-
C 0 2content of the blood will be affected more slowly by hypo- or headedness, visual disturbances due to cerebral vasoconstriction.
hyperventilation than the O2content, as the C 0 2stores in the body paraesthesia ('pins and needles') and muscle cramps, especially car-
(e.g. as HCOi) are -75 times greater than those for 0, (e.g. haemo- popedal spasm.
globin, myoglobin). Also, although hyperventilation increases arte-
rial Po,, in a healthy patient it has little effect on O2content as the Respiratory gas exchange ratio
haemoglobin is normally close to saturation (Chapter 8). Respiratory gas exchange ratio (R) is the ratio of CO, production to
Hypoventilation may occur when the respiratory drive is im- 0, consumption as measured at the mouth. In the steady state CO,
paired by head injury. or drugs such as morphine or barbiturates production and O2consumption reflect tissue metabolism. Metabo-
which suppress the respiratory centres. It may also be caused by res- lizing carbohydrates produces a volume of CO, equal to the volume
piratory muscle weakness or severe chest trauma. Hypoventilation of 0, consumed, whereas metabolizing fats and proteins produces a
is sometimes a feature of severe chronic obstructive airways disease smaller volume of CO, than O2 consumed. For an average mixed
(COPD; Chapter 23), but is not usually a feature of asthma (Chap- diet R = 0.8.
L
10 Control of acid-base balance

Carbonls
snr~ydrase
(a) CO, + H,O t, HC
, O, s HC3,- + H+

[HCO,-] x [H'! H
[ CO]; x [H+]
K= ---------- +K,= +log KA= log[Yt] + log +-log[H'] = -109 K A + log -
[H~CO,I [c3,1

I From ldw of Ma84 actton


6.I d ~ s m i i a i i om
A t equtlibrurn, kdO,] = [ H , C O j
. ~n ~ t a n i Kl icorrected Cii.*iiit\on conbtant
Take 1095
and rearrrang?
Henderson-Hassclbslch
equatlon

Pco, Pco,
6OmmHg 40mmHg
(7.8 kPa) '53 iPa)

Pco,
20mmlig
(2.6 kPaj

(d) Causes of acid-base disorders ( e ) Flenley acid-base nomogram

[ H ' l (nM) pH

II
I
Respiratoryacidosis Reepiratary alkaloele

P,~rwayobstruct~on Hlgh levels of anx~ety - Metabolic


Ketipiratoy muscle d~seaae Pa~n acidosis
Head trauma Alt~~ude Acute
Excesswe mechan~cal respiratory
ventla~~on

Metabolic acidolsie Mstabolic alkaloeie L Normal A

Renal failure o r t u b ~ l adamage


r
Lactic acidosia (hypoxia, sepsi5)
Ketoacidosis (diabetes, mineraloccrticoids
starvation)
1 alkaloeis

I Paio, (mmHg:~
The pH of arterial blood is normally -7.4 ( [ H + ] = 4 0 n ~Regulation
). hypoventilation (e.g. acute respiratory failure) will decrease the
of acid-base status so that blood pH remains between 7.35 and [HCO;] : Pco, ratio and consequently pH (see above). This respira-
7.45 (45-35 n ~is )vital for the correct functioning of the body. Car- tory acidosis is represented by a move from A to B (Fig. 10c);A to
riage of CO, in blood and its removal in the lungs (Chapter 9) has C represents a respiratory alkalosis (e.g. hyperventilation). A sus-
an important influence on acid-base status, as around 100 times tained respiratory acidosis caused by chronic respiratory failure
more acid equivalents are expired per day in the form of CO,/car- (Chapter 20) can be partially compensated by excretion of H+ (as
bonic acid than are excreted as fixed acids by the kidneys. phosphate and ammonium) and reabsorption of HCO; in the kid-
Buffers bind or release H+ according to the pH; this limits the neys. The [HCO;] : Pco, ratio is thus largely restored and pH returns
change in pH that occurs when acid is added. The relationship be- towards normal. This renal compensation is described by the
tween the amount of acid equivalent added to a solution containing arrow between B and D (Fig. 10c). Conversely, a respiratory
a buffer and the resultant change in pH is known as the buffer alkalosis may be compensated by increased renal excretion of
curve. Buffers are most effective when pH is close to their pK, (log HCO; (C to E).
of dissociation constant, KA; see Fig. IOa). The most important The term metabolic acidosis (or alkalosis) is used when acid-
buffers in blood are haemoglobin and bicarbonate (HCO;). CO, base status is disturbed by changes in HCO; rather than C 0 , a s a
combines with water to form carbonic acid (H,CO,), which dissoci- result, for example, of renal disease or increased H+ production
ates to HCO; and H+ (Chapter 9). The relationship between pH, (Fig. IOd). A metabolic acidosis (G) may be partially compensated
Pco, and [HCO;] is described by the Henderson-Hasselbalch by increased ventilation and a reduction in Pcoz ( G to E), initiated
equation (Fig. 10a): by detection of acid pH by the chemoreceptors (Chapter 12). There
can be little respiratory compensation for metabolic alkalosis
(F), as this may require unsustainable falls in ventilation.

Base excess
pK, is 6.1 and [CO,] can be calculated as Pco,xCO, solubility, Measurement of pH alone gives little indication of acid-base status
which is 0.03 mmol.L.mmHg-' (0.23 mmol.L.kPa-I). In normal (Fig. IOd); although pH may be normal, Pco2and [HCO;] may not
blood, [HCO;] is 2 4 m and ~ Pco, 40mmHg (5.3 kPa) and pH cal- be (D, ~ ) . ' ~ e a s u r e m e n tofs blood pH, Pco2 and Po, are always
culates as 7.4. Whatever their actual values, if the ratio [HCO;]: taken clinically. Base excess or base deficit (negative base excess) is
[CO,] remains constant at 20, then pH will remain at 7.4. Although the millimole per litre of acid or alkali needed to titrate the blood
the pK, of the bicarbonate system (6.1) is further away from blood back to a pH of 7.4. It is normally 0 +2mmol/L. In a pure metabol-
pH (7.4) than would seem ideal for a buffer, the fact that Pco, and ic acidosis, it is greater than the difference between the actual and
HCO; can be independently controlled by ventilation (Chapter 9) normal HCO;, as haemoglobin and buffers must also be titrated.
and the kidneys, respectively, means that in practice it makes an Base excess is normally calculated from the pH and Pco, automati-
effective buffer system. cally by clinical blood gas analysers, corrected for haemoglobin.
Haemoglobin is an important buffer, particularly when deoxy- The example in Fig. 10c is for a fully compensated respiratory aci-
genated (Chapter 9) and significantly improves the buffering capac- dosis (D) and gives an indication of the degree of renal compensa-
ity of whole blood compared with plasma (Fig. lob; the steeper the tion (i.e. after titration back to pH7.4). Base excess may be useful
line, the better the buffering). All other blood proteins combined for diagnosis, but should be used with caution as a basis for treat-
have <201%, of the buffering capacity of haemoglobin. ment, as the whole-body buffer line may differ significantly from
that of blood ill vitro. due to contributions from interstitial fluids
Acidosis, alkalosis and compensation (Fig. lob).
The relationship between pH, HCO; and Pco, can be portrayed Metabolic and respiratory acid-base disorders may often be com-
using a Davenport diagram (Fig. lob). HCO; is plotted against bined, making diagnosis ditlicult. A common example is respiratory
pH for given values of Pco?. The line marked BAC is the buffer line failure (Chapter 20), where concomitant hypoxia can cause meta-
for whole blood; in the absence of other changes (e.g. anaemia, bolic acidosis in addition to the primary respiratory acidosis. A use-
polycythaemia), changes in Pco2 alter HCO; and pH along this ful diagnostic aid is the Flenley nomogram (Fig. IOe). Only one
line. Point A represents normal conditions (pH7.4, HCO; 2 4 m ~ , type of disturbance is likely if the patient's arterial pH and Pco, fall
Pco, 40mmHgl5.3 kPa). An acute rise in Pco, (hypercapnia) due to within a band (95% confidence limits).
11 Control of breathing I: neural mechanisms

fo~low~ng
hed l~nes

Chemoreceptors
Lung receptors I
(via vaausl

Control of breathing involves a central controller in the brairistem thought the pre-Botzinger complex, in the rostra1 part of the vcn-
that sets the basic rhythm and pattern of ventilation and controls tral respiratory group, forms the kernel of the rhythm generator
the effectors (respiratory muscles). It is modulated by higher cen- for normal breathing, as neurones within it still exhibit cyclical
tres and feedback from sensors, including the chemoreceptors, firing when the complex is isolated from the rcst of the brainstem.
which match it to metabolism (Chapter 12) and mechanoreceptors The medullary respiratory centre is modulated by the pneumo-
in the lung (lung receptors). The neural networks involved are taxic centre, located in the nucleus parabrachialis of the pons;
complex and not fully understood, reflecting the need to coordinate sectioning between the pons and medulla alters the rhythm and pat-
ventilation with functions such as coughing and vocalization. tern of breathing, but does not abolish it. The pneumotaxic centre
receives both ascendinginput from the medullary respiratory centre
Brainstem and desccnding input from the hypothalamus and higher centres.
The pons and medulla contain several diffuse groups of neurones Factors such as emotion and temperature affect breathing via this
that are associated with generation uf the pattern and rhythm of route, but the basic pattern of normal breathing is maintained fol-
breathing. Sectioning between the medulla and spinal cord abolish- lowing sectioning above the pons, although voluntary control is
es breathing (Fig. I 1). The medullary respiratory centre is located lost. Voluntary control of breathing is mediated by motor neurones
in the reticular formation of the ventrolateral medulla, beneath from the cortex contained in the pyramidal tract, which bypasses
the Aoor of the 4th ventricle. This consists of a dorsal respiratory both the pneumotaxic and medullary respiratory centres (Fig. 11).
group in the nucleus tractus solitarius, containing inspiratory Certain rare lesions in the brainstem can therefore lcave the volun-
motor neurones leading to the respiratory muscles and a ventral tary pathways intact whilst impairing brainstem mechanisms, such
respiratory group in and around the nucleus ambiguus, contain- that ventilation may cease when the patient falls asleep ('the curse of
ing mainly expiratory neurones. There is reciprocal innervation Ondine'). Some books refer to an apneustic centre in the lower
between inspiratory and expiratory neurones, such that activity in pons, as sectioning here causes a characteristic gasping response
one inhibits activity in the other. In eupnoea (normal breathing) ex- There is, ho*ever, little evidence that this is of physiologica
piration is passive and cxpiratory neurones show little activity. It is relevance.
The medullary respiratory centre receives ascending input receptors is depression of somatic and visceral activity, which may
through the tractus solitarius from the central and peripheral be appropriate for serious lung damage.
chemoreceptors, the latter via the glossopharyngeal nerve Irritant receptors: located throughout airways between epithe-
(Chapter 12) and from the lung receptors mainly via the vagus. lial cells, with rapidly adapting afferent myelinated fibres in the
vagus. Receptors in the trachea lead to cough; those in the lower
Lung receptors and reflexes airways lead to hyperpnoea. They also cause reflex bronchial and
Stretch receptors: located in the smooth muscle of the bronchial laryngeal constrictions. Irritant receptors are stimulated by irritant
walls. These are mostly slowly adapting (continue to fire with sus- gases, smoke and dust (Chapters 17 and 31), but also by rapid large
tained stimulation), but there is a small rapidly adapting (transient) inflations and deflations, deformation of the airways, pulmonary
component. Their afferent nerves ascend via the vagus. Stimulation congestion, inflammation and disease. Irritant receptors are re-
of stretch receptors causes breathing to be shorter and shallower sponsible for the deep augmented breaths seen every 5-20min at
and delays the next inspiratory cycle. These receptors are largely rest. which reverse the slow collapse of the lungs that occurs in quiet
responsible for the Hering-Breuer inspiratory reflex, where lung breathing. These receptors may be involved with first deep gasps of
inflation inhibits inspiratory muscle activity. Conversely, the defla- the newborn ('first breath') and are probably important for the
tion reflex augments inspiratory muscle activity on lung deflation. Hering-Breuer deflationary reflex.
These reflexes are weak and unimportant during normal breathing Proprioceptors (positionllength sensors): located in the Golgi
in man. but are of more relevance when tidal volume is large (>I L, tendon organs, muscle spindles and joints of the respiratory mus-
e.g. in exercise) and in the neonate. cles. Afferents lead to the spinal cord via the dorsal roots. Stimu-
Juxtapulmonary o r 'J' receptors: so called because they are lated by shortening and load in respiratory muscles, though not
located on the alveolar and bronchial walls, close to the capillaries. diaphragm. They are important for coping with increased load and
Their afferents are small unmyelinated (C-fibre) or myelinated achieving optimal tidal volume and frequency. Note that input from
nerves in the vagus. Activation causes apnoea (cessation of breath- non-respiratory muscles and joints can also stimulate breathing.
ing) or rapid shallow breathing, falls in heart rate and blood pres- Other receptors that may modulate respiration:
sure, laryngeal constriction and relaxation of skeletal muscles via Pain receptors: stimulation often causes brief apnoea followed
spinal neurones. J receptors are stimulated by increased alveolar by increa;ed breathing.
wall fluid, oedema, pulmonary congestion, microembolisms and Receptors in the trigeminul regiorz and larylzx: stimulation may
inflammatory mediators such as histamine. All these stimuli are give rise to apnoea o r laryngeal spasm.
associated with many types of lung disease. The general action of J Arterial baroreceptors: stimulation depresses breathing.
12 Control of breathing II: chemical mechanisms

E f f e c t o f C02, pH a n d O2 o n ventilation:
Normal Po,
Normal pH -13 k f a

Alveolar PCO, (kPa) Alveolar Pco2 (kPa) Po, (kPa)


li

(d) Central chemoreceptors


Central

Yon5 1 chemoreceptor

Blood-brain barrier

Medulla -
oblongata
iarbon~c
anhydrase

[Ht] a t chemoreceptor = aPco, 1 [HCO,-]


Pco,from blood, and [HCO, ] from CSF
a = constant

(f) Peripheral chemoreceptors Groups o f cells surrounded by fenestrated


sinuaoidal c a p ~ l l a r ~ e s
Aortic Carotld
Glomus
nerve bodies slnus
nerve
Sheath
(type 11)
cells
, ,
i <. Carotld
I \ Common - body
\\<
Heart
', carotid (not p a r t
-... artery o f sinus) . nerve fibres
-..-.._--/'l
Chemical control of ventilation is mediated via chemorecep- The peripheral chemoreceptors are the carotid and
tors, which detect arterial Pco?, Po? and pH and modulate ventila- aortic bodies. The carotid body is a small (-2 mg) structure located
tion via the respiratory centre in the medulla (Chapter 11). PCO,is at the bifurcation of the common carotid artery, just above the
the most important factor. Ventilation is thus closely matched to carotid sinus. It is innervated by the carotid sinus nerve, leading to
metabolism-large changes in metabolic rate result in very small the glossopharyngeal (Fig. 12f). The aortic bodies are found on the
changes in arterial P r o z and Poz aortic arch and are innervated by the vagus. In humans, they are less
Normal alveolar P r o z (PAcoz)is -5.3 kPa (40 mmHg). lncreasing important than carotid bodies. The carotid body contains glomus
PAco2causes minute ventilation (litres ventilated per minute) to rise (type I) cells and sheath (type 11) cells (Fig. 12g). Glomus cells are
in an almost linear fashion (Fig. 12a), by -15-25 Llmin for each kPa responsible for chemoreception; they have dense granules contain-
rise in PAco2(-2.7 L/min/mmHg). There is considerable variation ing dopamine and contact axons of the carotid sinus nerve. The
between individuals. and athletes and patients with chronic respira- function of sheath cells is unclear.
tory disease often have a reduced response to PAco2(Chapters 24 Carotid bodies respond to increased PCO,or [H+]and decreased
and 40). If PAco2increases above 10 kPa, ventilation decreases due Po, (not blood 0, content) by increasing firing rate in the carotid
to direct suppression of the respiratory centre. A metabolic acido- sinus nerve, and thus ventilation. They have a high blood flow and
sis (an increase in [H+]caused by reduced [HCO,-1; see Chapter 10) consequently a small arteriovenous difference for Pco, and PoZ
shifts the COz-ventilation response curve to the left, whereas a They respond rapidly (seconds) and are sufficiently fast to detect
metabolic alkalosis shifts it to the right (Fig. 12a). Note that a rise small oscillations in blood gases associated with breathing. The
in [H+]caused by increased Pco2 is called a respiratory acidosis. mechanisms by which changes in Pco,, p H and Po, are detected are
Increasing PAo2from the normal value of -13kPa (-100mmHg) unclear, but for Po, they are believed to involve inhibition of K+
has little effect on the C0,-ventilation response curve, but if the channels in the glomus cell, with consequent depolarization and
PAo, is reduced, the slope of the relationship becomes steeper and Ca2+ entry.
ventilation increases more for any given rise in PAco2(Fig. 12b).
When the effect of PAo2is investigated independently (at constant Adaptation: chronic respiratory disease and altitude
PAco2),there is little increase in ventilation until the PAo2falls When hypercapnia (raised arterial Pco,) is prolonged, for example
below -8 kPa (-60 mmHg) (Fig. 12c). The effect of reducing PAozis. in chronic respiratory disease, CSF p H gradually returns to normal
however, potentiated if the PAco2is raised-i.e. there is a synergis- due to an adaptive increase in HCO:, transport across the
tic (more than additive) relationship between the effects of PAoz blood-brain barrier. The drive to breathe from the central chemore-
and PAco2. ceptor is consequently reduced, even though Pco, is still high. As-
sociated with this, there is occasionally a loss of sensitivity to
The chemoreceptors further increases in P,co,, and the patient's ventilation is then
The central chemoreceptor consists of a diffuse collection of neu- primarily controlled by the level of Po, (hypoxic drive). Care
rones located near the ventrolateral surface of the medulla, close to must be taken with such patients, as giving high concentrations
the exit of the 9th and 10th cranial nerves (Fig. 12d). It is sensitive of Oz in order to increase blood 0, saturation may raise the Po,
to the pH of the surrounding cerebrospinal fluid (CSF) and does sufficiently to depress the hypoxic drive and hence ventilation.
not respond to Po,. CSF is separated from blood by the Normally, -2428% 0, is given to such patients. This leads to
blood-brain barrier, a tight endothelial layer lining the blood ves- a sufficiently small rise in Paco2as to have little effect on the hypo-
sels of the brain. This barrier is impermeable to polar molecules xic drive, but because of the steep slope of the 0, dissociation
such as H+ and HCO,, but CO, can diffuse across it easily. The pH curve (Chapter 8) it can result in a significant improvement in 0,
of CSF is therefore determined by the arterial Pco2 and the CSF content.
[HCO;] (Chapter 10) and is not directly affected by changes in At high altitudes, ventilation is stimulated by the low atmos-
blood pH (Fig. 12e). CSF contains little protein, so its buffering ca- pheric Po,. This leads to hypocapnia and alkalosis (as CO, is
pacity is low; therefore a small change in Pco2 will cause a large blown off), which depress ventilation. Over some days, the p H of
change in pH. Stimulation of the central chemoreceptor by a fall in CSF returns to normal due to HCO, transport out of the CSF, even
CSF pH (rise in blood Pco,) causes an increase in ventilation. The though the Pco, remains low and ventilation increases again. Over
central chemoreceptor is responsible for -80% of the response to a longer period blood p H returns to normal due to renal compensa-
COz in humans. It has a relatively slow response time (-20 s), as CO, tion (Chapter 10). These processes form part of the acclimatization
has to diffuse across the blood-brain barrier. to altitude.
13 Pulmonary circulation and anatomical right-to-left shunts

(a) Pulmonary and systemic circulation and normal (b) The initial effects o f a 2 0 % right-to-left shunt I
anatomical right-to-left shunts 0, and C02 contents and partial pressure

Pulmonary capillary pressure:

Pulmonary a r t e r y '0;: content = 15 mLldL


*C02 content = 5 2 mLldL

I '
80 20
Arterial Oilcontent = - x 2 0 + - x 15
100 100
80 20
Arterial C 0 2 c o n t e n t = - x 48 + - x 52
100 100

"Note: t h e mixed venous contents used are normal \

abnormal arterial contents would lead t o abnormal


contents so t h i s simple analysis underestimates t t
arterial contents.
: Aortic
5 y s t e m i c capillary pressure: ' pressure:
Venous end Arterial end i
I
120170,
mean
The Po2and Pco2 t h a t result from these 0, and CO:
found from t h e O2 and CO, dissociation curves:
: 90mmHg

........ 70
LO2 dissociation cur

VCM = venae cordis minimae (Thebesian veins)


- 60
--I

I -5 50
E
2 40

pressures and contents

20% mixed venous blood


with 8 0 % blood undergoing

-1 1 3 5 7 9
Po21Pco2(kPa)
Pulmonary vascular resistance is more evenly distributed in the the pulmonary vein. This part results in deoxygenated blood from
microcirculation and pulmonary capillary flow remains pulsatile. the airways contaminating blood returning from alveoli (Fig. 13a).
Local systemic resistance and blood flow are controlled by In addition, a small amount of the coronary venous blood drains di-
sympathetic nerves, metabolites and other substances acting on ar- rectly into the left ventricular cavity via the venae cordis minimae
terioles. Both sympathetic and parasympathetic nerves innervate (Thebesian veins). These additions of deoxygenated (right-sided)
pulmonary vessels, but their influence is weak in most circum- blood to oxygenated (left-sided) blood are known as anatomical
stances. Systemic arterioles dilate in response to hypoxia, increasing right-to-left shunts. In normal people, they are equivalent to 2% or
flow and hence oxygen delivery. In contrast, hypoxic vasoconstric- less of the cardiac output, but they explain why arterial Po, is less
tion occurs in the pulmonary circulation. This response, which is than alveolar Po, even though pulmonary capillary blood equili-
accentuated by high Pco?, improves gas exchange by diverting brates with alveolar gas.
blood from underventilated to well-ventilated regions (Chapter 14). In disease, right-to-left shunting of blood may be much larger.
The response is unhelpful in the presence of global lung hypoxia, at Atelectasis (lung collapse) or consolidation in pneumonia will re-
altitude or in respiratory failure, where it may contribute to the de- sult in pulmonary arterial blood supplying the affected region fail-
velopment of pulmonary hypertension and right heart failure. ing to undergo gas exchange. Right-to-left shunts are also the cause
As cardiac output increases in exercise, pulmonary vascular resis- of reduced arterial oxygenation in cyanotic congenital heart dis-
tance falls, as vessels are recruited and distended and the rise in pul- ease such as tetralogy of Fallot. Atrial or ventricular septa1 de-
monary arterial pressure is small. The pulmonary circulation acts as fects d o not usually cause impaired gas exchange and cyanosis, as
a blood reservoir and the volume it contains varies, being about the higher left-sided pressures give rise to left-to-right shunts in
450mL when upright and 800mL lying down. Inspiration also in- which some oxygenated blood is pumped again through the lungs.
creases pulmonary vascular volume.
Fluid balance across capillaries is determined by hydrostatic and The effect of right-to-left shunts on arterial blood gases
oncotic pressures (the Starling forces; see The Cardiovascular Sys- In the right-to-left shunt shown schematically in Fig. 13b, 20% of
tem at a Glance, Chapter 20) across capillary walls. Capillary on- blood fails to pass through functioning alveoli and its 0, and CO,
cotic pressure opposes filtration and is about 27mmHg in both contents remain at mixed venous levels of 15 and 52 mLldL, respec-
circulations. Although hydrostatic pressure is low in the pulmonary tively. Eighty per cent of the blood undergoes normal gas exchange,
capillaries (about IOmmHg), net filtration of fluid occurs in pul- emerging with normal 0, and CO, contents of 20 and 48 mLldL, re-
monary capillaries as it does in systemic capillaries. Other factors spectively. The initial effect on arterial gas contents is calculated
favouring filtration are interstitial oncotic pressure, which from a weighted average of the contents in these two blood streams.
is relatively high in the lungs (about 18mmHg) and interstitial This gives an arterial 0, content 1 mLIdL below normal and CO,
hydrostatic pressure, which is negative (about -4 mmHg). Pulmo- content 0.8mLldL above normal. From the flat part of the oxygen
nary oedema occurs when these forces are altered to increase net dissociation curve, it can be seen that the resulting arterial Po,
filtration above the rate that can be cleared by the pulmonary lym- is about 9kPa (68mmHg) compared with the normal 13kPa
phatics. For example, it may occur when pulmonary capillary pres- (97mmHg). The much steeper CO, dissociation curve means the
sure is increased in mitral stenosis and left ventricular failure. rise in PCO,is small, from the normal value of 5.3 kPa (40mmHg)
Inspiratory crepitations (crackles) on auscultation in these condi- to about 5.5 kPa (41 mmHg).
tions are probably caused by popping open of airways in lungs stiff- If the respiratory system is otherwise normal, the reduced P,o,
ened by congestion with blood. They are most obvious at the bases, and increased Pace, simulate ventilation via the chemoreceptors
where hydrostatic pressure is highest. Pulmonary congestion and and C 0 2 washed out of the functioning areas restores arterial CO,
oedema are worsened by the increase in pulmonary blood volume content and Paco2to normal. In contrast, increased ventilation has
lying down. little effect on arterial oxygen content and PO,, as blood draining
the ventilated areas of the lung was already saturated. If hypoxia is
Anatomical or true right-to-left shunts severe, the stimulation in ventilation is often great enough to reduce
Ideally, all venous blood emerging from tissues would return to the PAco2below normal. Typically, in a right-to-left shunt there is a low
right side of the heart to be pumped through gas-exchanging lung. PAo, with a normal or low PAco2.
In fact, part of the blood draining the bronchial circulation joins
14 Ventilation-perfusion mismatching

(a) (b) Var~atronof v e n t ~ l a t ~ oVA,


n , perfus~on,
Different types of VAlQ regions Q and vent~lat~on-perfusonratlo,
VA/Q w ~ t hv e r t ~ c ahl e ~ g h In
t the
Normal Dead space Dead space effect Shunt effect Truelanatomical shunt
S? u p r ~ g hlung
t
VA = Normal VA = Normal VA = Normal VA = Low VA = 0
Q = Normal Q= 0 Q = Low Q = Normal Q = Normal
VA/Q = Normal VAIQ = m VA/Q = H~gh VA/Q = Low V,/Q =0
(close t o 1)

Apex
Pog and 02contents of blood from these regions breathing air and oxygen
, .
0, content, mLldL O2 content, mL1dL O2 content, mLldL O2 content, mL1dL

2:E
20E
00 2:E 10 20 0 10 20 0 10 20
i

Normal No blood Normal • Low • Low Alveoli a t s t a r t Blood vessels


0 Unchanged draining t h i s 0 Unchanged 0 Increased 0 Unchanged and end of a t different
region breath heights

(c) The effect of a mixture of high and low P A / dregions on arterial blood gases (d) Alveolar air equation

Small flow with: This predicts t h e PO, in t h e functioning or 'ideal'alveol~

u Pao, Z n o 2 -&o,

R = The respiratory gas exchange ratio = C02 production


0, consumption

(R is usually about 0.8)

n o 2 = Inspired 0, partial pressure


Paco, = Arterial C02 partial pressure (= alveolar)

At rest, alveolar ventilation and pulmonary blood flow are similar, close to 1. Ventilation-perfusion mismatching or inequality is said
each being around 5 Llmin. Ventilation (VA)and perfusion (Q) may to occur when regional VAIQ ratios vary, with many being much
vary in different lung regions, but for optimal gas exchange they greater or less than 1 (Fig. 14a). A right-to-left shunt from complete
must be matched. Areas with high perfusion need high ventilation collapse or consolidation of a region (Chapter 13) has VA/Q= 0, and
and ideally, local ventilation-perfusion ratios (VA/Q) should be can be viewed as an extreme example of ventilation-perfusion mis.
matching. At the other extreme, alveolar dead space from a pul- airways are partly blocked by bronchoconstriction, inflammation
monary embolus is a ventilated region without perfusion and VA/Q or secretions and high VA/Q in emphysematous areas where capil-
= M. Regions where VA/Q is much greater than 1 have excessive ven- laries are lost. Hypoxic vasoconstriction (Chapter 13) helps re-
tilation or dead space effect and blood from them has a high Po, duce the severity of ventilation-perfusion mismatching.
and a low Pco,. Regions with VA/Q much less than I behave quali-
tatively like shunts and are sources of shunt effect or venous ad- Effect of ventilation-perfusion mismatching on arterial
mixture. Blood draining them has undergone some gas exchange, blood gases
but Po, is lower and Pco, higher than normal. The effect on Po, and Blood emerging from areas with high VA/Q might be expected to
0, content draining different VA/Q regions both during air breath- compensate for blood from areas with low VA/Q. This is not the
ing and during oxygen breathing is shown in Fig. 14a (lower panel). case, for two reasons (Fig. 14c). Firstly, although Po, will be in-
creased in high VA/Q regions, oxygen content is raised little, as
blood is normally nearly saturated. Blood draining regions with low
Effect of the upright posture on perfusion, ventilation and VA/Q and low Po, (especially if <8 kPa, 60 mmHg) will have signifi-
V,/Q (Fig. 14b) cantly reduced oxygen content. In addition, these areas contribute
Hydrostatic pressure in all vessels varies with vertical height above more blood than areas with high VA/Q, which are typically caused
or below the heart, because of the weight of blood. On standing, the by reduced perfusion. The net effect of mixing blood from areas
increased pressure at the lung bases distends vessels, increasing flow. with a wide range of ventilation-perfusion ratios is a low arterial 0,
Pressures generated by the right heart are low and higher up the content and P,02. CO, content is less severely affected because
lung vascular pressures in diastole may fall below alveolar pressure the overventilated areas d o lose extra CO, and partly compensate
at the venous end of the pulmonary capillary. In such regions, flow for low VA/Q regions. Moreover, any abnormalities of Pao2 and
is reduced and determined by the difference between arterial and P,co, will lead to a reflex increase in ventilation, which usually cor-
alveolar pressure. There may be regions at the apices--especially in rects or overcorrects the raised Paco2 whilst being less effective at
haemorrhage or positive-pressure breathing-where alveolar pres- raising Pao2.The final arterial blood gas picture, a low P,o, and a
sure also exceeds pressure at the arterial end of the pulmonary normal or low Pace,, is similar to that resulting from anatomical
capillaries. The vessels collapse completely for part of each cardiac right-to-left shunts (Chapter 13).
cycle, giving low, intermittent flow. The net result is a blood flow One difference is that arterial hypoxia caused by ventilation-per-
per unit volume of lung tissue that falls progressively from base to fusion mismatching improves much more with oxygen therapy than
apex. that caused by a shunt. In shunts, the oxygen-enriched air fails to
Gravity also affects intrapleural pressure, which is less negative at reach the shunted blood. In VA/Q mismatching, increased oxygen
the base than the apex. As a result, at functional residual capacity fraction can increase local Po, in areas of low VA/Q(Fig. 14a), giv-
apical alveoli are more expanded-with less capacity for further ex- ing rise to significant improvement in arterial oxygen content and
pansion during inspiration-than at the bases. Consequently, venti- pressure.
lation is also higher at the base than the apex. The effect of gravity
on ventilation is less marked than on perfusion and so VA/Q is Assessment of ventilation-perfusion mismatching
higher at the apex than the base. In young people, this modest de- Regional ventilation and perfusion can be visualized by inhalation
gree of mismatching has little effect on blood gases. The scatter of and infusion of appropriate radioisotopes (Chapter 19). A simple
ventilation-perfusion ratios increases with age and contributes to but useful index of the degree of mismatching is the difference be-
the reduction in P,o, seen in the elderly. tween Po, in gas-exchanging or 'ideal' alveoli and in arterial blood.
Ideal alveolar Po2can be calculated from the alveolar air equation
Ventilation-perfusion matching in disease (Fig. 14d). An increased A-a Po, gradient (A = alveolar Po,, a =
Increased ventilation-perfusion mismatching is an important cause arterial Po,) is usually caused by ventilation-perfusion mismatch-
of gas exchange problems in many respiratory diseases, including ing or anatomical right-to-left shunts. In normal young people,
asthma, chronic obstructive pulmonary disease (COPD), pneumo- there is a small A-a gradient (<ZkPa) arising from the normal
nia and pulmonary oedema. Regions of low VA/Q may arise when anatomical right-to-left shunts discussed in Chapter 13.
15 Development of the respiratory system and birth

(a) Branching morphogenesis PI Week 4 --+Week 5 - Week 6 Week 8

Trachea

Factors released by mesoderm cells cause the


epithelium t o grow inwards towards them as a bud; Bronch~al Embyon~c Secondary
inhibitory factors prevent budding either side buds oesophaqus bronch~
/ '~n,dod~rml
Mesoderm ep~thel~um

5eg;ental
bronchi
I
The embryological origins of the lung are primitive endoderm of 17th week, most of the major structures of the lung have formed
the foregut. which eventually forms the epithelium and glands of and are lined with glycogen-rich columnar epithelial cells. The gas
the larynx, trachea and lungs and splanchnic mesoderm, which exchange surfaces have not yet developed and fetuses delivered dur-
forms cartilage, smooth muscle, lung parenchyma and connective ing this period are therefore not viable.
tissue. In common with many glandular organs, the lung develops Canalicular period (16th-25th weeks): bronchial cartilage,
by branching morphogenesis (Fig. 15a), with budding and smooth muscle, abundant pulmonary capillaries and connective
branching of the endodermlepithelium into mesoderm. The process tissue develop from the mesoderm. There is progressive differentia-
requires reciprocal signalling between epithelium and mesoderm, tion and thinning of epithelial cells. The bronchi will have sub-
with the mesoderm being primarily responsible for programming divided -17 times after 24 weeks, finally forming the respiratory
development of adjacent epithelium into the relevant structures. bronchioles which themselves divide into three to six alveolar ducts
Many signalling molecules are vital for the orchestration of branch- and some thin-walled terminal sacs. These are lined by very thin
ing morphogenesis during lung development, including fibroblast type I alveolar pneumocytes (squamous epithelium), which to-
growth factors (FGF), epidermal growth factor (EGF), bone mor- gether with endothelial cells from capillaries form the future alve-
phogenetic proteins and sonic hedgehog. Development of the respi- olacapillary membrane (gas exchange surface). There are a few
ratory system is generally divided into five stages or periods. type I1 alveolar pneumocytes, secretory epithelial cells that pro-
Embryonic period: the tracheobronchial tree originates from duce surfactant. This reduces surface tension and allows expansion
the laryngotracheal tube, below the 4th pharyngeal pouch at the of the terminal sacslalveoli (Chapter 6). but although it is present in
caudal (tail) end of the primordial pharynx. The laryngotracheal small amounts from about the 20th week, there is insufficient to sup-
tube starts to appear just prior to the 4th week of development, port unaided breathing until after 26 weeks (see neonatal respira-
after the heart begins to beat. By the end of the 4th week, its end has tory distress syndrome, Chapter 16). Some gas exchange can
bifurcated into two bronchial buds, progenitors of the two main occur at the end of this period, as there are both thin-walled termi-
bronchi and bronchial tree (Fig. 15b). nal sacs and good vascularization, but the general level of immatu-
Pseudoglandular period (5th-17th weeks): the bronchial buds rity means that fetuses born before the end of the 24th week
have now developed into the primordial left and (slightly larger) normally die despite intensive care.
right primary bronchi, which subsequently divide by branching Saccular (terminal sac) period (24th week-parturition): associ-
morphogenesis into five secondary bronchi (three right, two left). At ated with rapid development in the number of terminal sacs and the
the 7th week, these have started to branch progressively into 10 pulmonary and lymphatic capillary networks. Budding from the
(right) or eight to nine (left) segmental (tertiary) bronchi, each of terminal sacs and walls of terminal bronchioles and thinning of
which eventually forms a bronchopulmonary segment. By the type 1 pneumocytes leads to formation of immature alveoli from
around week 32. Sufficient surfactant and vascularization are nor- Birth
mally present between the 24th and 26th week to allow survival of The lungs are initially 50% full of fluid at birth, which is replaced by
some premature fetuses, though this is very variable (see Chapter air. During and immediately following birth. fluid is removed via the
16). Surfactant increases significantly in the two weeks before birth. pulmonary and lymphatic circulations and through the mouth as a
Alveolar period (late fetal to childhood): clusters of immature result of squeezing during delivery. Expansion and filling of the
alveoli form during the early part of this period; mature-type alve- alveoli with air is critically dependent on the presence of surfactant
oli do not appear until after birth. Fetal breathing movements are to lower surface tension. Before birth, the vascular resistance of the
present before birth, with aspiration of amn~oticfluid, and these pulmonary circulation is higher than that of the systemic and blood
stimulate lung growth and respiratory muscle conditioning. Lung is consequently shunted via the ductus arteriosus and foramen ovale
development is impaired in the absence of fetal breathing. inade- (see Tlze Cardiovusculur System ut u Glunce Chapter 25). At birth.
quate amniotic fluid (oligohydramnios) or space for lung growth the increased arterial Po, rapidly reduces the pulmonary vascular
(see Chapter 16). The increase In lung size over the first 3 years is due resistance below that of the systemic and constricts the ductus arte-
primarily to an increase In number of alveoli and respiratory bron- riosus. Blood therefore starts to flow through the lungs, the pressure
chioles; thereafter, both the number and slze of alveoli increase. gradient across the foramen ovale reverses, causing closure, and
More than 90% of alveoli are formed after b ~ r t h reaching
, a maxi- blood flow takes its adult course.
mum after 7-8 years. At the end of lung development, there are ap-
proximately 23 generations of airways, w ~ t -17
h million branches.
16 Complications of development and congenital disease

( (a) Relationship between prematurity and development of NRDS (d) 5ome genetic diseases in which t h e lung is a primaty site of injury 1 1
Diewase inheritance Pathogenesis Lung pathology
I I
Alphal-anttypsin AD Protease-antiprotease Emphysema
deficiency imbalance
Ciliary dyskinesia AR lrnpdlrcd mucoc~liary A~rway~nfect~on,
clearance bronchiectasis
Cystic Abros~s AR Abnormal chlor~de A~rwayinfection,
transport bronchiectasis
Familial idiopathic AR Unknown Diffuse f ~ b r o s ~ s
T-L .--
Lipoid proteinosis Hyalinized or
(Urbach-Wiethe deposition in upper granular deposte
eyndrome: respiratory t r a c t in t h e tracheo-
Gestat~onalage: <30 / wee~s

I
3u-34 3-3
n , , . m L nr n
A
causing mucosal bronch~al
< I L ~ <I500 <I7004
1 th~cken~ngand alrway submucosa
I
1 II
obstruction

'
Other factors s ~ c a
h s socioeconomic status, maternal health, Tracheobroncho- Saccular bulges Recurrent airway
race and sex also affect incidence of NRDS meqaly (Moun~er- beLween cart~laqe
. . mfect~ons
Kuhn's syndromej
I I rings resulting from
atrophy of elastic
and smooth muscle
(b) Congenital diaphragmatic hernia tissue and causing
~ m p a ~ r emucocil~ary
d
clearance
Congenital cartilage 7 Deficiency of Kecurrent airway 1
deficiency subsegmental infections,
Hypoplaatic

Heart
&p compressed
I I _ ^
(
(Williams-Campbell
syndrome) 1 I
bronchial cartilage
with airway collapse (
bronchiectaeis

Diaphragm
17- J3

Oesophageal atresia and


tracheosophaqeal flstu a (85%)
Tracheosophageal fistula (5%)
1
Problems associated with premature birth compl~anceis low. NRDS is most commonly caused by lack of suf-
Neonatal respiratory distress syndrome (NRDS), otherwise ficient quantities of surfactant and consequent high surface tension
known as hyaline membrane disease, occurs in -2% of all births and in the alveoli and small airways. Incidence therefore increases
is characterized by rapid, laboured breathing and often sternal re- sharply with degree of prematurity (Fig. 16a), although other fac-
traction due to partial collapse of the lungs after each breath. Lung tors may also reduce production of surfactant. When a premature
birth is anticipated, the expectant mother can be treated with corti- fuse with the membranes on the thoracic and peritoneal wall leads
costeroids (betamethasone) to speed fetal lung development and to a posterolateral defect, most commonly occurring on the left side
surfactant production. Treatment with exogenous surfactant in (-85%), through which the abdominal viscera pass (herniate) into
the first 30min after birth, either of natural origin or artificial, has the thorax (Fig. 16b).This often includes the stomach, spleen and
also proved to be beneficial. Survival of neonates with NRDS often much of the intestines. The presence of the resultant mass severely
requires high positive pressure mechanical ventilation and high restricts lung development and later inflation, leading to a signifi-
levels of oxygen. cantly reduced lung volume and life-threatening breathing diffi-
The large majority of NRDS cases are related to prematurity, culties. The latter are the prime cause of death in congenital
with some due to other causes including damage to type I1 pneu- diaphragmatic hernia and most infants will die because the lungs
mocytes. A very few cases are due to a congenital absence of are insufficiently developed to support life outside the uterus. Al-
pulmonary surfactant protein B. These patients d o not respond though surgical correction of the defect is possible both before and
to any form of therapy and tend to die in the first few months of after birth, the mortality rate is very high. A related but very much
life. less common condition is eventration of the diaphragm, where
Bronchopulmonary dysplasia (chronic lung disease of the new- half the diaphragm lacks adequate muscle and bulges (eventrates)
born) is a long-term consequence of NRDS, primarily as a result of into the thoracic cavity. The viscera are forced into the pocket so
treatment with high positive pressure ventilation combined with formed, again restricting lung development.
high levels of oxygen (hyperoxia). The condition is characterized by Tracheoesophageal fistula (an opening between oesophagus
alterations in the structure and function of airways and pulmonary and trachea) is the most common abnormality of the lower respira-
blood vessels, including increases in airway and vascular smooth tory tract itself, with an incidence of about one in 4000 births. Its
muscle and obliteration of some microstructures. This leads to origins are located in the 4th week of development, when the
poorly reversible airway obstruction and sometimes pulmonary embryonic respiratory tract starts to develop and divide from the
hypertension (high pulmonary blood pressure). Survivors may re- embryonic oesophagus (see Chapter 15). Eighty-five per cent of
tain symptoms for many years, if not for life. There are several simi- cases are associated with the descending part of the oesophagus
larities to chronic obstructive pulmonary disease (COPD, Chapter having a blind ending (oesophageal atresia) (Fig. 16c); the lower
23) and chronic severe asthma in adults. part of the oesophagus'joins instead to the base of the trachea. As a
Several techniques have recently been designed to minimize the result, normal feeding is impossible and the gut becomes distended
incidence of bronchopulmonary dysplasia in infants with NRDS. with air. There are also consequences in utero, as normally amniot-
These include extracorporeal membrane oxygenation (ECMO), ic fluid is ingested by the fetus. Thus oesophageal atresia is com-
where blood is circulated via external apparatus for gas exchange; monly associated with excess amniotic fluid (polyhydramnios),
mechanical ventilation and hyperoxia are therefore not required which can lead to severe defects in the central nervous system. Some
and some success has been reported. Conversely, ECMO has not 5%)of cases of tracheoesophageal fistula show no atresia but only a
been found useful in adults with acute respiratory distress syndrome fistula, and the remainder less common variations. Rare defects in-
(ARDS, Chapter 35). Partial fluid ventilation, where the lungs are volving blockage or narrowing of the trachea itself (tracheal atre-
ventilated with fluids containing oxygen-carrying perfluorocar- sialstenosis) are nearly always accompanied by various types of
bons, has also been reported to be beneficial. Fluid ventilation tracheoesophageal fistula.
circumvents problems associated with high surface tension by Congenital influences o n respiratory disease: several impor-
removing the air-liquid interface and allows small airways to open tant respiratory diseases that are discussed in detail in other chap-
and contribute to gas exchange. ters have definite or implied genetic components. including asthma
(Chapter 21), chronic obstructive pulmonary disease (Chapter 23).
Congenital diseases emphysema (Chapter 23), cystic fibrosis (Chapter 30) and primary
Congenital diaphragmatic hernia is the most common cause of pulmonary hypertension (Chapter 24). Other genetically linked
lung hypoplasia (inadequate development of the lung), with an in- diseases that cause pathological problems primarily in the lung are
cidence of about one in 2000 births. Failure of the diaphragm to listed in Fig. 16d.
17 Lung defence mechanisms and immunology

Secretory
I4A Gel phase
! Lumen

I
Mucus
501 phase
Epithelium
Secretory Basement
component membrane
Goblet-
cell &
' 5ubmuGo5al gland
'c_u_y

Phagocyte

-Lamina propria

Inhalation of air also allows ingress of dust. irritant particles and and trachea, whether inhaled or transported from distal regions by
pathogens. The huge surface area of the lungs provides multiple op- mucociliary transport, stimulate irritant receptors (Chapter 1 l),
portunities for damage, and the warm, humid environment provides provoking sneezing and coughing that eject foreign matter.
ideal conditions for bacterial and other infestations. The respira-
tory tract. however, has a range of powerful defence mechanisms. Mucus and airway secretions
Dysfunction of these mechanisms underlies many respiratory dis- The respiratory epithelium is covered with a 5-10pm layer of gelati-
eases, for example asthma (Chapter 21) and fibrosis (Chapters 27 nous mucus (gel phase) floating on a slightly thinner fluid layer (sol
and 31). phase) (Fig. 17). The cilia on epithelial cells beat synchronously,
and as they do so their tips catch in the gel phase and cause it to
Physical and physiological defences move towards the mouth, transporting particles and cellular debris
The nostrils and nasopharynx provide a physical barrier to particles with it (mucociliary transport or clearance). It takes -40min for
>10pm. in the form of hairs and mucus to which particles adhere. mucus from large bronchi to reach the pharynx and from respira-
Mucociliary transport (see below) subsequently transfers these to tory bronchioles several days. Many factors can disrupt this mecha-
the pharynx. where they are ingested. Only particles less than 5pm nism, including an increase in mucus viscosity or thickness, making
generally get further than the trachea. The nasopharynx also pro- it harder to move (e.g. inflammation, asthma), changes in the sol
vides important humidifying and warming functions for inhaled phase that inhibit cilia movement or prevent attachment to the gel
air, preventing drying of epithelium. Irritant particles in the nose phase and defects in cilia activity (cilia dyskinaesia). Mucociliary
transport is reduced by smoking, pollutants, anaesthetics and infec- tion of potent antimicrobial agents including reactive oxygen
tion, and in cystic fibrosis (Chapter 30) and the rare congenital im- species. Phagocytosed organic material is usually digested, whereas
motile cilia syndrome. Reduced mucociliary transport causes inorganic material is sequestered inside the cell. As alveolar epithe-
recurrent respiratory infections that progressively damage the lium does not have cilia, alveolar macrophages are key to remov-
lungs--for example, bronchiectasis, where the bronchial walls are ing material and are the major cell present in the alveoli. Other
thickened, permanently dilated and inflamed (see Chapters 30 and functions include clearance of surfactant proteins and suppres-
40). sion of unnecessary immune responses by production of anti-
Mucus is produced by goblet cells in the epithelium and submu- inflammatory cytokines such as interleukin-10 (IL-10) and
cosal glands. The major constituents are carbohydrate-rich glyco- transforming growth factor (TGFP). However, in more severe
proteins called mucins that give mucus its gel-like nature. The infections, they can initiate inflammatory responses and by release
fluidity and ionic composition of the sol phase is controlled by epi- of chemoattractants such as leukotriene B, promote neutrophil
thelial cells. Mucus contains several factors produced by epithelial infiltration from the plasma. They can also act as antigen-
and other cells or derived from plasma: anti-proteases such as a,- presenting cells (see below).
antitrypsin inhibit the action of proteases released from bacteria
and neutrophils which degrade proteins, and a,-antitrypsin defi- Development of immunity
ciency predisposes to disruption of elastin and development of T-lymphocytes and B-lymphocytes migrate to lymph nodes,
emphysema (see Chapters 17 and 23). Surfactant protein A, apart tonsils and adenoids and diffuse patches of bronchus-associated
from its actions on surface tension, enhances phagocytosis by coat- lymphoid tissue (BALT) within the lamina propria. Here they
ing or opsonizing (literally 'making ready to eat') bacteria and interact and are programmed. Antigen is presented to CD4+
other particles. Lysozyme is secreted in large quantities in the air- T-lymphocytes (T-helper or T Hcells) by antigen-presenting cells.
ways and has antifungal and bactericidal properties; together with The most important are dendritic cells, highly specialized mono-
the antimicrobial proteins lactoferrin, peroxidases, and neutrophil- nuclear phagocytes (see Fig. 17). Macrophages, B-lymphocytes
derived defensins, it provides non-specific immunity to the respira- and some epithelial cells can also act as antigen-presenting cells.
tory tract. Secretory immunoglobulin A (IgA) is the principal On presentation of antigen, T H cells release cytokines such as
immunoglobulin in airway secretions and with IgM and IgG agglu- IL-2, IL-4, IL-13 and interferon-y (IFN-y). IL-2 activates CD8+
tinates and opsonizes antigenic particles: it also restricts adherence T-lymphocytes (cytotoxic or Tc cells), which kill infected cells.
of microbes to the mucosa. Secretory IgA consists of a dimer of two IL-4, IL-13 and IFN-yactivate B-lymphocytes in the presence of
IgA molecules produced by plasma ceIls (activated B-lymphocytes. antigen binding to surface immunoglobulins (IgM) (see Fig. 17).
see below) and a glycoprotein secretory component. The latter is Activated B-lymphocytes proliferate and differentiate into plasma
produced on the basolateral surface of epithelial cells, where it cells that re-enter the bloodstream. These secrete large amounts of
binds the IgA dimer (see Fig. 17). The secretory IgA complex is then antigen-specific antibody (immunoglobins). Binding of antibody to
transferred to the luminal surface of the epithelial cell and released antigen may neutralize some toxic molecules, but more commonly
into the bronchial fluid (see Fig. 17). I t can account for 10% of the activates secondary mechanisms. either directly by opsonization,
total protein in bronchioalveolar lavage fluid. allowing recognition and phago-cytosis by macrophages and neu-
trophils, or by activation of complement. When activated, com-
Lung macrophages plement can: kill pathogens by lysis (bursting the cell membrane);
Macrophages are mobile mononuclear phagocytes that are found opsonize the antibody-antigen complex; and recruit inflammatory
throughout the respiratory tract. They act as sentinels in the air- cells. For more detailed information on immunity, see I/l~n~itnology
ways, providing innate protection against inhaled microorganisms at a Glunce.
and other particles by phagocytosis (ingesting them) and produc-
18 Pulmonary function tests

I (a) Volume-time spirograms during forced expiration from t o t a l lung capacity


I

Time (5) Time (5) (c) The body plethysmograph


FEV, = Forced exp~ratolyvolume in I s A = Normal respiratory system for measurlng lung volumes
FVC = Forced v ~ t acapacity
l 6 = Obstructive alrway disease
FEF25-S
i = Mean forced expiratoly flow from C = Restrictive lung disease
25-75%of FVC

(b) Helium diiution for measurlng Function Res~dualCapac~ty*

Sp~rometerf~lledwlth volume, Vl Volume = V,+ FRC


and hel~umconcentration, [He], C, [He] = C2

Volume = FRC -

Starting a t t h e end of a normal expiration, (lung volume = FRC), t h e subject The subject inhales a g a ~ n sat closed shutter
bveathes In and o u t from t h e splrometer u n t ~equilibr~um
i is reached S ~ n c ehel~um
is poorly soluble In blood. Lung volume expands from V1t o Vl t AV
AV can be deduced from t h e rise in box pressure, ,P
,,
Vl x C1 = (V,t FKC) x C, (cal~bratedwlth known volumes)
Mouth (= alveolar) pressure fall from PI t o P,
From Boyle's law. Vl x P, = P2(V1+ AV)
"Note. To measure TLC or RV t h e subject 1s asked t o breathe In fully or breathe
Hence t h e oriq~nalvolume in t h e lungs, Vl can be found
o u t fully before breath~ngt h e hellurn gas mixture.

Accurate assessment of defects in airflow, lung volume or gas height and sex by standard nomograms derived from large cross-
exchange is essential to the diagnosis and management of many sectional studies.
respiratory disorders. It is important to note that these tests charac- Airway resistance can be measured using a body plethysrno-
terize 'defects'; the clinician has to diagnose 'diseases'. The normal graph (Chapter 7 and Fig. 18c) to measure alveolar pressure. Lung
range of many lung function tests is very wide and it is essential to compliance can be measured using an oesophageal balloon to
compare measured values with those predicted for the subject's age, measure intrapleural pressure (for details, see Chapter 6). More
commonly, abnormalities of airway resistance (in obstructive on effort and not very reproducible, but it may correlate well with
airway disease) are assessed indirectly from forced expiratory subjective dyspnoea.
manoeuvres and abnormalities of compliance (in restrictive lung
disease) are assessed indirectly from lung volume measurements. Lung volumes
Restrictive ventilatory defects (RVDs) are characterized by a re-
Forced expiratory tests duction in TLC. Lung volumes such as TLC, RV and FRC can be
Peak expiratory flow rate (PEFR) is frequently measured, despite measured by helium dilution (Fig. 18b) or by body plethysmo-
its inability to distinguish between different types of ventilatory de- graphy (Fig. 18c). The gas dilution method is simpler for patients,
fect and its dependence on patient effort (Fig. 7c). It is reduced in but it is sensitive to gas leaks and will underestimate TLC in the
obstructive disease, respiratory muscle weakness and often in re- presence of extensive bullous o r cystic lung disease. RVDs may be
strictive lung disease (secondary to reduced volume). Its main value caused by parenchymal lung disease (pulmonary fibrosis, sclero-
lies in monitoring diseases, especially asthma, once the diagnosis derma, pulmonary oedema), chest wall disease (kyphoscoliosis.
has been made. massive obesity) or weak respiratory muscles (myasthenia gravis,
In contrast. plots of volume against time (spirogram) or air- muscular dystrophy). RV and FRC can help distinguish between
flow against volume during a forced expiration can help to distin- these conditions, as FRC and RV are usually reduced in lung dis-
guish between different types of defects. The patient is asked to ease; whereas FRC is usually normal and RV elevated in muscle
inhale to total lung capacity (TLC) and breathe out as hard and fast weakness. FVC and TLC usually decline in parallel, therefore once
as possible to residual volume (RV). A plot of volume against time an RVD has been established by measurement of TLC, the progress
(Fig. 18a) can be produced by continuously measuring volume, of the disease may be followed with FVC from spirometry.
either with a spirometer o r by integrating a flowmeter output. If a Measurement of lung compliance (Chapter 6) and trans-
flowmeter is used, it is also possible to compute a flow vs. volume diaphragmatic pressure (Pd,) may distinguish further between
plot from the same forced expiration (Fig. 7c). Flow-volume RVD due to parenchymal lung disease or muscle weakness. By using
plots show characteristic shapes with different defects (see Fig. 7e). two small balloon-tipped catheters, one measuring oesophageal
such as the 'scooped out' appearance seen in obstructive airway (PP,,,,,,) pressure and the other gastric (Pabd) pressure. Pd, (=
disease. Pabd-Ppleural) can be measured during a maximal inspiration or sniff
Forced vital capacity (FVC) and forced expiratory volume in from FRC. Typically, in parenchymal lung disease lung compliance
't' seconds (FEV,) can be read off the volume vs. time plot (Fig. is low, elastic recoil pressure high and Pdinormal; whereas in
18a).FEV, is extremely reproducible and correlates well with func- respiratory muscle weakness lung compliance is relatively normal,
tion and prognosis. It is normal for FVC and FEV, to peak in adults elastic recoil pressure low, and Pdilow.
in the third decade and then decline by approximately 30mLlyear Diffusing capacity is a measure of the ability of gas to diffuse
(Fig. 23b). FEV,IFVC is normally 0.75-0.90, but higher values may from the alveolus into pulmonary capillary blood. As discussed in
occur in normal children. FEV,/FVC helps distinguish between ob- Chapter 5, D,co is used as a surrogate for DLo2,since it is simple to
structive and restrictive ventilatory defects. Typically, in obstructive measure and carbon monoxide diffuses across the lung in a fashion
lung diseases (e.g. COPD, acute asthma) the FEV,/FVC is less than similar to oxygen. It often helps interpretation to normalize D,co to
0.70. If the airway obstruction is due to asthma, FEV,, FVC and the alveolar volume (VA) by calculating the coefficient, K c o =
FEV,/FVC may all increase after the inhalation of bronchodilators. DLcolVA.D,co is reduced by reduced alveolar surface area, thick-
In restrictive lung disease (e.g. lung fibrosis), absolute values of ened alveolarxapillary membrane, reduced capillary blood volume
FEV, and FVC are reduced, but FEV,/FVC is normal or high. or anaemia. Reductions in the DLcocan be caused by a variety of
Forced mid-expiratory flow (FEF,,-,,) is the average forced parenchymal diseases (idiopathic pulmonary fibrosis, emphysema,
expiratory flow rate over the middle 50% of the FVC. It may be pneumonia) or vascular diseases (pulmonary hypertension, pul-
especially affected by small airway disease, but the normal range is monary oedema), such that the test is sensitive but not specific.
wide. Reductions in the D,co below 50% predicted for age, sex and height
Maximal voluntary ventilation (MVV) is measured by asking are often associated with oxygen desaturation during exercise.
the subject to breathe as hard and fast as possible into a spirometer Severe reductions in DLco( 4 0 % predicted) may result in resting
for 15 s, with theventilation expressed in Llmin. It is very dependent hypoxaemia.
19 Chest imaging and bronchoscopy

II Evaluation d t h e CXR includes all the followlng: Chest radiograph intsrpretation Normal chest x-ray
I
I

Chest mdiograph Interpretation Normal l ~ b r ax-ray


l

liudioy-aphy of [lie chrbt was one of the first medical utilizations tion-perft~sionscans). Specilic raclingraphic :\bnnrni:ilities arc dis-
of X-ray imaginp and it is still itscd daily to detect. diagnose o r cussed in later chapter.5,
follou, rnorpliologic abnormalities in the clicst. Standard two- Posteroanterior (PA) and lateral chest radiograplrs (CXRs)
dimensional chest X-rays are still Llle mainstay of chest radiogra- allow two-drmcnsionitl visu:ilization of thc lungs, great vc~scls.
phic proccdurcs. Rerent inriovations h a w included digital imaeing. heart. diaphrapni and mcdi;trlinurn. F'A lilms should he pcrforliied
three-dimensional ilnaginp (computed tomography scans) and seen in ( 'SRand basic C'XR
upright at total lung capacity. Fc;lt~~ms
physiologic itnages (positron emission loniography and vcntilit- interpretation are show11in tlic Fig. 19. I n paticnts undcr 40 years
of age with no suspected lung disease. lateral films need not be equivocal VIQ scans, pulmonary hypertension and pulmonary vas-
performed for screening purposes. Portable films shot anterior- cular disease. including vasculitis and arteriovenous malformations.
posterior (AP) in patients unable to stand magnify the heart and These studies are often preceded by echocardiography to visualize
mediastinum and d o not allow detailed visualization of lung right ventricular function and estimate pulmonary artery pressure
parenchyma. using Doppler imaging.
A standard PA and lateral CXR should allow visualization of Positron emission tomography (PET) utilizing a fluorinated
both lungs, including the diaphragmatic position, as well as the analogue of glucose (FDG) gives images of the lung that highlight
normal trachea. main carina, mainstem bronchi, major and minor areas of increased glucose metabolism. Malignant cells have in-
fissures, aorta, main pulmonary arteries and heart. Understanding creased glucose uptake and appear as increased densities on PET
of the normal anatomy of a CXR is essential to allow recognition images. Recent studies have demonstrated that F D G PET is useful
of abnormal lung parenchymal infiltrates, enlarged lymph nodes in distinguishing between benign and malignant solitary pu monary
adjacent to the trachea or in the hila, enlarged pulmonary arteries, nodules and in detecting small nodal metastases that may not be
volume loss of a lobe or segment or cardiac enlargement. In the apparent on C T scanning. For these indications, PET has a sensitiv-
case of a suspected pleural effusion, lateral decubitus films allow ity and specificity from 80 to 97% with false positive scans seen in
visualization of as little as 50mL of free-flowing fluid. Digital cases of infection or granulomatous inflammation. Whole body
CXRs are being developed that allow more detailed views of the F D G PET was recently used to detect clinically inapparent distant
denser portions of the thorax and show finer detail of the lung metastases.
parenchyma. Bronchoscopy enables direct visualization of the endobronchial
Computed tomography (CT): a limitation of standard CXR tree. Chest physicians perform most bronchoscopies, as day cases
imaging is that the two-dimensional image obscures details and under local anaesthetic in the sedated but awake patient, using a
averages densities in the third dimension (anterior-posterior on the flexible fibre-optic instrument. It has the advantages of visualiza-
PA film). C T allows thin slice axial images and fine-detailed exami- tion of the upper lobes and is a safe technique with a low complica-
nation of intrathoracic structures. Administration of intravenous tion rate. Saturation and heart rhythm should be monitored and
contrast allows imaging of the pulmonary blood vessels (such as in supplemental oxygen administered during the procedure. Facilities
suspected pulmonary embolus) and demonstration of abnormal for resuscitation should always be immediately available. Thoracic
lymph nodes (such as in evaluation of malignancy or infection). C T surgeons may use a rigid bronchoscope in the fully anaesthetized
also permits resolution of interstitial lung infiltrates as well as pre- patient. This instrument allows larger biopsies and better suction-
cise localization of infiltrates, masses, cavities, bulla, fluid collec- ing, and is the method of choice when removing inhaled foreign
tions and airway abnormalities. Examples of C T scans are shown in bodies. Bronchoscopy is most frequently performed to investigate if
several chapters. Newer technology allows complete axial scanning a shadow on a chest radiograph is due to a lung cancer (Chapter 36).
of the thorax with a single breath-hold. If an endobronchial tumour is seen, biopsies for histological analy-
Ventilation-perfusion (ViQ) scans are mostly performed in the sis and washings and brush samples for cytological analysis can be
evaluation of pulmonary embolism (PE) (Fig. 25). Gamma cameras taken. In addition, information regarding the operability of the tu-
can visualize radiopharmaceuticals either injected into the venous mour can be obtained. Bronchoscopy can also be used to diagnose
blood (perfusion) or inhaled (ventilation). Thromboembolism clas- parenchymal lung disease using the technique of transbronchial
sically causes a VIQ mismatch, with absence of perfusion in the biopsy, which obtains parenchymal and bronchial tissue for
presence of ventilation. Unfortunately, the value of VIQ scans is histological examination. Collection of bronchoalveolar fluid
limited by the observation that many PEs result in indeterminate (BAL, bronchoalveolar lavage) is useful in diagnosing alveolitis
VIQ scans that show small mismatches or matched VIQ deficits. In (raised lymphocyte count in sarcoidosis), infection in the immuno-
these cases, other studies must be utilized to demonstrate throm- compromised patient (e.g. Pneumocj2stis carinii pneumonia) and
boemboli. Contrast C T scans are increasingly used as screening tuberculosis. Bronchoscopy also aids investigation of collapsed seg-
tools for PE and are being investigated as possible replacements for ments or lobes. Therapeutically, bronchoscopy is used to remove in-
VIQ scanning. Quantitative VIQ scans may be used in preparation haled foreign bodies. to aspirate sputum plugs and secretions, to
for lung resection surgery. to assess regional lung function and esti- relieve stenosis by placement of stents and during treatment
mate the amount of residual lung function. of endobronchial tumours with laser or endobronchial radio-
Pulmonary angiography, where the vasculature is visualized therapy. Haemorrhage, pneumothorax and cardiac arrhythmia,
following injection of contrast medium (see Chapter 25), may be although uncommon, are the main complications of fibre-optic
required in some patients with suspected pulmonary emboli but bronchoscopy.
20 Respiratory failure

I (a) Causes of respiratory failure (b) Mechanisms of arterial hypoxia (low Pao2)
Central drive Normal
Cns depressant drugs Normal alveolar-
(e.4. barbiturates) c a p ~ i l a ymembrane
Yead injuy Normal alveolar vent~iatlon
Cerebrovascular accident >98%cardlac
Primary alveolar hypoventilat:on output pa5s1ng
througti gas-
Airway obstuction exchang~ngalveol~
Foreigq body or tumour
Matching of ventilation and petfusion
Asthma
throughout the lung5
Chest wall Spinal cord
Crush ~n,ur?,
-flail chest (apnoea lf above 23: 1. Low inspired Pop - e.g. altitude (low Pg) or low
inspired O2concentration + lo^ alveolar Po2

2. Hypoventilation - Inadequate alveolar


ventilation +low alveolar Por
Pne~rnonla

Pneumothorax 3 Diffusion impairment - Pulmonay cap~liary


Atelectasls Respiratory muscles blood falls t o reach equli~br~um wlth alveolar gas
NRD514RD5 Muscular dystrophies Gulllaln-Bard +low puimonary e n d - c a p ~ l l aP~
o2

Neuromuscularjunction
Myasthenla gravls
Other Muscle relaxants
Cyanot~ccongen~talheart d~sease 4 Ventilation-perfusion mismatching - Blood
Oulmonargl embo,~ f r o v areas w ~ t hh~ghk41d mlxes with biood from
. .
Pulmonary oedema low V A G dreas +low pulmonary venous Po2

5. Right-to-left shunt - Shunted blood fa115


t o undergo gas exchanges, mlxes wlth
pulmonary cap~llaryblood+low pulmonary

1 (c) Effects of hypox:a and hypercapnia


velouslleft /entr~cularPo2

opoietin from hypoxic kidney +polycythaemia + f oxygen


carriage despite low Pao, but i f excessive (haematocrit >55%)the
Central cyanosis (not very sensitive; may be fviscosity impairs tissue blood flow
absent in anaemia) Polycythaemia+florid complexion; increased cyanosis
Cardiac arrhythmias "Pulmonary hypertension-, right ventricular hypertrophy
Hypoxic vasoconstriction' of pulmonary vessels Fluid retentionlright heart failure (cor pulmonaleg)+ peripheral

impaired CNSImuscle function: irritability, confusion,

Cardiac arrythmlas vessels and therefore contributes t o the development of cor pulmonale
Respiratory failure is usually said to exist when arterial Po, falls 85% if haemoglobin concentration is normal (15gIdL) and the
below 8 kPa (60mmHg) when breathing air at sea level. In type 1 resulting central cyanosis is visible in the tongue and mucus mem-
respiratory failure, the arterial hypoxia is accompanied by a nor- branes of the mouth. It appears at higher oxygen saturations in
mal or low arterial Pco,, whereas in type 2 o r ventilatory failure, polycythaemic patients, whereas in severe anaemia central cyanosis
arterial Pco, is increased above 6.7kPa (5OmmHg). Respiratory may be impossible, as it would require an O2 saturation incom-
failure may be acute or chronic. In chronic respiratory failure, there patible with life.
are permanent abnormalities in blood gases, which typically worsen
periodically (acute o n chronic). This strict definition excludes Respiratory failure in asthma
some patients whose respiratory systems might otherwise be con- Hypoxia in a severe asthma attack is primarily due to VA/Q mis-
sidered failing. Some patients have disabling dyspnoea (breathless- matching. Paco2 usually falls as the attack worsens, because periph-
ness) of respiratory origin but maintain Po,>8 kPa. eral chemoreceptor and pulmonary receptor stimulation produce a
Some of the many causes of respiratory failure are listed in Fig. reflex increase in ventilation despite the increased work of breath-
20a. Symptoms and signs clearly depend on the underlying cause. ing. A raised or even apparently normal Paco2 (e.g. 5.3kPa.
Dyspnoea and tachypnoea (increased respiratory rate) will be 40mmHg) in a severe hypoxic asthma attack is a cause for concern,
prominent in severe asthma but absent in conditions with reduced as it may indicate the onset of exhaustion and potentially life-
central drive. threatening asthma.

Respiratory failure in chronic obstructive


Mechanisms leading to hypoxia and hypercapnia pulmonary disease
Of the five causes of hypoxaemia (Fig. 20b), only hypoventilation
The clinical picture of severe chronic obstructive pulmonary disease
inevitably causes increased P,co,.
(COPD) is variable, but two extreme patterns- the pink puffer and
the blue bloater- are recognized and described in Chapter 23. The
Vco2
P,cq C=Z -(Chapter 9) blue bloater is associated with type 2 respiratory failure. He has a
VA chronically low Pao, and high Paco2and these worsen with acute
infections, which precipitate acute on chronic respiratory failure.
If hypoxia is out of proportion to the hypercapnia and the A-a
Patients with chronic hypercapnia typically have a near-normal
Po2 gradient (see Chapter 14) is increased, one of the other
arterial p H owing to an efficient compensatory metabolic alkalosis
mechanisms (see 3-5 in Fig. 20b) must also be present. The primary
via renal generation and retention of bicarbonate. During an acute
effect of right-to-left shunts and ventilation-perfusion mis-
exacerbation, Paco2may increase further and pH then falls signifi-
matching is to raise arterial CO, content, but this is usually cor-
cantly. as renal adjustments are slow. Arterial pH can therefore
rected or over-corrected by a reflex increase in ventilation (Chapters
indicate the proportions of acute and chronic hypercapnia. Patients
13 and 14).
with chronic hypercapnia are at risk of respiratory depression
Thickening of the alveolar~apillarymembrane in lung fibrosis
and a further, potentially fatal, increase in Paco2if given high in-
may give rise to diffusion impairment, preventing equilibration of
spired oxygen (Chapter 39). This may be due to loss of hypoxic drive
pulmonary capillary blood with alveolar gas, especially in exercise,
in the presence of reduced CO, sensitivity, but other mechanisms
when time in the capillary is reduced. However, in many conditions
may contribute, including increased VA/Q mismatching by the
thought to cause diffusion impairment, there is also substantial
removal of hypoxic vasoconstriction. As these patients are on the
VA/Q mismatching, and this is probably the main cause of the
steep part of the oxyhaemoglobin dissociation curve, significant
hypoxia.
improvements in arterial oxygen content can usually be achieved by
small increases in FIo2(to 24 or 28%). The resulting small improve-
Effects of hypoxia and hypercapnia ment in P,02 does not cause respiratory depression (see Chapter
The direct effects of hypoxia and hypercapnia, together with the 12).
compensations and complications that occur in chronic respiratory
failure, are shown in Fig. 20c. Management
Although hypoxia usually offers the greatest threat to vital or- All patients suspected of having respiratory failure will need ar-
gans, hypercapnia and especially acidosis are also important and terial blood gas measurement, as the severity is difficult to assess
they often accentuate the adverse effects of each other. Hypoxia and clinically. A chest X-ray helps detect possible causes and aggravat-
hypercapnia are better tolerated when they develop slowly in ing factors such as pneumonia or pneumothorax. Other investiga-
chronic respiratory failure because of adaptations such as poly- tions, including lung function tests, will depend on the clinical
cythaemia and compensatory metabolic alkalosis. situation and likely underlying disease. Management will include
Cyanosis is a greyish-blue tinge seen when a tissue's microcircu- airway maintenance, clearance of secretions, oxygen therapy
lation contains a high concentration of deoxygenated haemoglobin. (Chapter 39) and in some cases mechanical ventilation (Chapter
It may occur with impaired blood flow, for example in the hands and 38). Specific therapies, such as bronchodilators and antibiotics, are
feet in circulatory shock, when it is known as peripheral cyanosis. directed at the underlying cause or aggravating factors. Abnor-
When the arterial blood contains more than about 1.5-2g/dL of de- malities in haemoglobin concentration, fluid balance and cardiac
oxygenated haemoglobin, the concentration in the microcirculation output should be treated to improve tissue oxygen delivery and in-
reaches the critical level for cyanosis to be observable even in well- crease mixed venous oxygen content, which in turn will also reduce
perfused tissues. This occurs with an arterial saturation of about the effects of venous admixture on arterial oxygenation.
21 Asthma: pathophysiology

(a) Causes and precipitating factors of asthma (c)

Globlet cells, Epithelial cell


allergens mucus
I

El-
Drugs
(NSAIDs,
Exercise
P-blockers)

(b) Typical responses t o inhaled allergens

" response response response


1-

IL-3, 4, 5, 13 = interieukins, LTD4/C4 = cysteinyl leukotrienes,


PgD, = prostaglandin D,, MBP = major basic protein, ECP = eosinophil
cationic protein, GM-CSF =granulocyte macrophage colony stimulating
factor, PAF = platelet activating factor
1 Hours Days Chemoattractants include PAF, LTB4, and RANTES I

Asthma is an inflammatory disease of the airways. Patients suffer flow. Damage to the epithelium (epithelial shedding) is reflected by
from episodes of cough, wheezing, chest tightness andlor dyspnoea whorls of epithelial cells (Curschmann's spirals) in the mucus, which
(breathlessness), which are often worse at night or early in the also contains eosinophil cell membranes (Charcot-Leyden
morning. There is considerable variation in the severity and crystals). In chronic severe asthma, remodelling of the airways
frequency of attacks. Asthma can be usefully defined as 'increased occurs, including increased bronchial smooth muscle content. This
responsiveness of the bronchi to various stimuli, manifested by causes irreversible narrowing of the airways and limits the
widespread narrowing of the airways that changes in severity either effectiveness of bronchodilators.
spontaneously or as a result of treatment'.
The major characteristics of asthma are: Prevalence
1 Narrowing of the airways and impeded air How, commonly Asthma is increasing in prevalence, particularly in the Western
reversible spontaneously or following treatment. world, where >5% of the population may be symptomatic and
2 Increased sensitivity to bronchoconstricting stimuli receiving treatment. There has been a concomitant increase in
(hyperresponsiveness). mortality, despite improved treatment. In the UK, one in seven of
3 Increased numbers of inflammatory cells (eosinophils, mast the population has allergic disease and over 9million people will
cells, neutrophils, T lymphocytes) in the bronchi. have wheezed in the last year. The number of teenagers with asthma
There is also hypersecretion of mucus, blockage of airways with has nearly doubled over the last 12 years. Prevalence of asthma
mucus plugs and swelling of mucosa due to inflammation- varies greatly geographically, being least common in the Far East
associated vascular leak and oedema, all of which further limit air and most common in the UK. Australia and New Zealand. There is
50
some correlation with Westernized life-styles, including living Occupational a s t h m a
conditions that favour house-dust mites and atmospheric pollution. Many materials may give rise to occupational asthma. Some are
allergens (e.g. flour, grain, animals, certain commercial enzymes),
Classification whereas others are not, including isocyanates (present in industrial
Asthma can be classified as extrinsic, having a definite external and polyurethane coatings) and fumes from welding or soldering.
cause and intrinsic, where no external cause can be identified. Twenty per cent of the working population may be susceptible to
Extrinsic asthma most commonly occurs as a result of an allergic occupational asthma.
response, with development of I g E antibodies to specific antigens
(allergic or atopic asthma) and tends to start in childhood with Drug- associated a s t h m a
symptoms becoming less severe with age. Intrinsic asthma generally Aspirin and other non-steroidal anti-inflammatory drugs
appears in adults and does not improve. (NSAIDs) promote asthmatic attacks in 5% of asthmatics. They
inhibit the cyclooxygenase (COX) pathway that synthesizes
Atopic a s t h m a prostaglandins and shift arachidonic acid metabolism from COX
Individuals who readily produce IgE to common antigens are prone towards the lipoxygenase pathway and production of LTC, and
to allergic asthma. Major antigens include fecal pellets from house- LTD,. Aspirin-induced asthma is reversed by antileukotriene
dust mite--the most common cause of asthma worldwide, grass therapy (Chapter 22).
pollen and dander from domestic pets. Genetic factors, The bronchi are innervated by parasympathetic nerves that
atmospheric pollution and maternal smoking in pregnancy all release acetylcholine, which causes bronchoconstriction and
predispose to raised IgE levels and later development of asthma and stimulates mucus production and non-adrenergic, non-cholinergic
airway hyperresponsiveness (Fig. 2 1 a). (NANC) nerves. Parasympathetic activity may increase in asthma
Inhalation of allergens by atopic individuals initiates an due to axon reflexes from irritant receptors in the bronchi (Chapter
immediate response (bronchoconstriction) that usually subsides 11). There is little sympathetic innervation, although circulating
within 2 h (Fig. 21 b); this is reversible with bronchodilators such as epinephrine (adrenaline) acting via P,-adrenoceptors on smooth
the P2-adrenoceptor antagonist salbutamol (Chapter 22). This is muscle causes bronchodilation. Consequently P-adrenoceptor
often followed 3-12h later by a late-phase response, including antagonists such as propranolol can cause bronchoconstriction in
bronchoconstriction and development of airway inflammation and asthmatics. This may even occur with nominally PI-selective drugs
hyperresponsiveness, which is less susceptible to bronchodilators. and treatment of cardiovascular disease with such agents should be
Some materials such as isocyanates cause only an isolated late avoided in asthmatics.
phase. The increase in airway hyperresponsiveness associated with
the late phase may promote recurrent asthma attacks over several O t h e r factors
days. Asthmatics have hyperresponsive airways and irritant gases and
The immediate response is caused by antigen1IgE-induced mast dusts that d o not affect healthy individuals can precipitate
cell degranulation and release of histamine, prostaglandin D, asthmatic attacks or worsen symptoms. Such factors include
(PgD?) and leukotriene C , and D, (LTC,, LTD,); these cause tobacco smoke, exhaust fumes and pollutants such as nitrogen
bronchoconstriction, increased mucus production and vascular dioxide, sulphur dioxide and ozone. Exercise and inhalation of
leak (Fig. 21c). In the late phase, mediators from mast cells and cold air often precipitate wheezing in asthmatics, probably via
activated T lymphocytes cause infiltration of neutrophils and drying and cooling of the bronchial epithelium. This is common in
eosinophils. Eosinophils are present in large numbers in asthmatic children. Emotional stress can also induce an asthmatic attack.
bronchi and release leukotrienes, platelet-activating factor (PAF), Certain viral infections (rhinovirus, parainfluenza, respiratory
major basic protein (MBP) and eosinophil cationic protein syncytial virus) are also associated with asthma attacks. The
(ECP). MBP and ECP contribute to epithelial cell damage, with pathogenesis of asthma is complex; Fig. 21c shows only a
consequent increased permeability to allergens, release of simplified scenario.
eosinophil chemoattractants (e.g. RANTES) and cytokines such as
granulocyte macrophage colony-stimulating factor (GM-CSF) and
exposure of C-fibre afferent nerve endings. The latter release
proinflammatory tachykinins.
22 Asthma: treatment

(a) Step-wise approach t o asthma therapy I

Short-acting broncho-
dilator a5 required
(inhaled P2 agonist)

Used more t h a n once daiiy

need for additional long-

(b) Drugs used in asthma therapy 1


Pp-adrenoreceptor I Muecarinic receptor Xanth,nee
Type
agoniste I
antagonists
Cortlco~osterolds Cromones I
I
Anti-leukotrienes
I ---- - - - - - .

Inhaled. oral and lV Inhaled Oral and lV Inhaled Inhaled Oral


S h o r t actlng lpratrop~umbrom~de Theophyll~ne Beclomethasone propr~onate, Sodum Receptor antagonlets
Salbutamol (aibuteroi) Ox~trop~umbrom~de Amnophyl~ne Fiut~casonepropr~onate, cromoglycdte Montelukast,
Terbutal~ne,Rtmeterol Enprofilline Budeson~de (cromoiyn) Prarlukast,
Fenoterol, P~rbuterol
I 5iow release Oral
Nedocroml Zaf~rlukast
oxygenaee inhibitors:
I

ne aigni icant described


r, though zafrlukaat
enn aaacciated with

duced steroid dose


her than the drug dose
narrow therapeutic

Pressurized metered
Remove t h e cap and shake t h e inhaler
Tilt the head back slightly and exhale

- Position t h e inhaler in t h e mouth ( or preferablyjust in front of t h e open mouth)


During a slow inspiration, press down t h e inhaler t o release the medication
Continue inhalation t o full inspiration
Hold breath for 10 seconds
Management of asthma should encompass: assessment of ticosteroids are the mainstay of long-term asthma therapy. They
severity and efficacy of therapy; identification and removal of pre- can, however, have significant side effects, including oral candidiasis
cipitating factors; therapy to reverse bronchoconstriction and in- (5%) and hoarseness. Growth may be retarded in children receiving
flammation; patient and family participation and education. high-dose inhaled corticosteroids. O r a l corticosteroids such as
prednisolone may be required in patients whose asthma cannot be
Assessment controlled by inhaled steroids. The danger of adverse effects is much
L u n g function: Asthma is diagnosed when inhaled bronchodila- greater and excess corticosteroids may permanently suppress the
tors cause >15'/;1 improvement in forced expiratory volume in 1 s hypothalamic-pituitary axis. Combination therapies containing
(FEV,) or peak expiratory flow rate (PEFR) (Chapter 18). The ab- both steroid plus long-acting P2-agonists are useful for moderate1
sence of improvement does not rule out asthma- the disease could severe asthmatics.
be in remission and chronic severe asthma is poorly reversible. Air- Cromoglycate and nedocromil inhibit release of inflammatory
way resistance has a circadian rhythm. being least at midday and mediators and prevent activation of mast cells and eosinophils.
greatest at 3 4 a.m. Serial measurements of PEFR in the morning, They may also suppress sensory nerve activity and release of neu-
midday and on retiring are useful for identifying the enhanced vari- ropeptides (Chapter 21). Prophylactic use reduces both immediate
ation in airflow limitation characteristic of asthma and for assessing and late phases of the asthmatic response and hyperresponsiveness.
response to therapy over time. Poorly controlled asthma shows a They are less powerful than steroids and only effective in mild and
characteristic morning fall in PEFR ('morning dipping'). Occupa- exercise-induced asthma. However, they have few side effects and
tional asthma is suggested when PEFR improves after a break from are often the drug of choice for children. Use has declined since the
work. Lung function tests are often coupled with exercise tests in introduction of safer, low-dose steroids, which are cheaper, more
children. who often exhibit exercise-induced asthma. effective and do not need to be taken so often.
Bronchial provocation tests are used to determine hyperre- Xanthines such as theophylline have bronchodilatory and some
sponsiveness when asthma is suspected but PEFR measurements anti-inflammatory actions and are taken orally. They inhibit phos-
are not diagnostic. Patients inhale increasing doses of histamine or phodiesterases that break down CAMP.Limitations include numer-
methacholine (acetylcholine analogue) until FEV, declines by 20%. ous side effects and a narrow therapeutic range; these are partially
The dose at which this occurs (PD2,,FEV,) is greatly reduced in asth- overcome by slow-release preparations. Xanthines are used as
matics, who are always hyperresponsive. second-line drugs in asthma, particularly where P2-agonists are in-
Skin prick tests are essential for identifying extrinsic factors. effective at controlling symptoms and in steroid-resistant asthma.
Development of a wheal around the prick site indicates allergen Antileukotriene therapy has not been fully characterized and
sensitivity. Exposure to identified allergens should be immediately comes in two forms: cysLT receptor (LTC,lD4) antagonists such as
minimized (e.g. replacement of furnishings to reduce house-dust montelukast, and 5-lipoxygenase inhibitors such as zileuton. Both
mite; removal of pets), as once extrinsic asthma is established it may have equal efficacy for bronchoconstriction caused by allergens, ex-
not be reversible. Only 50% of patients with occupational asthma ercise and cold air, with -50% reversal. They are also effective in
are cured by avoidance of the precipitating factor. aspirin-sensitive asthma, indicating the key role leukotrienes have in
this condition (Chapter 21). Antileukotrienes improve lung func-
Therapy tion in mild and moderate asthmatics, but the greatest benefit may
The goal is long-term control and all patients except those with be for very severe asthmatics taking steroids. Both drug types are
the mildest symptoms should receive anti-inflammatory drugs as taken orally and are relatively long-lasting, with few adverse effects.
well as bronchodilators. International guidelines favour step-wise Histamine antagonists have not proved useful in asthma, al-
treatment regimens (Fig. 22a). Asthma therapy is centred on in- though newer non-sedating antihistamines such as terfenadine may
haled compounds (Fig. 22b). Inhalation maximizes bronchial de- alleviate mild allergic asthma.
livery whilst minimizing systemic side effects. Metered dose inhalers
are the most commonly used delivery systems, although only -15% Problems with treatment
of the dose may reach the lungs (Fig. 22c). Failure to control asthma is often related to poor compliance with
P,-Adrenoceptor agonists such as salbutamol are rapid and treatment regimens-for example, due to peer pressure in children.
powerful bronchodilators and are of first choice for alleviating Compliance may also be poor when asthma is apparently con-
acute symptoms. They activate adenylate cyclase to increase cyclic trolled. so patients stop therapy (e.g. steroids, cromoglycate) be-
adenosine monophosphate (CAMP)and may also reduce mediator cause they are 'cured'. Poor inhaler techniques are common. Patient
release from inflammatory cells and airway nerves. Long-acting P2- education and training is key to asthma therapy.
agonists such as salmeterol allow twice-daily dosage regimens.
Long-term use of P,-agonists is associated with reduced effective- Severe uncontrolled asthma -'status asthmaticus'
ness (tolerance). This requires immediate treatment and hospitalization. Indica-
Muscarinic receptor antagonists such as ipratropium prevent tions: inability to complete sentences ('telegraph speaking'), high
acetylcholine released by parasympathetic nerves from causing respiratory rate, tachycardia, PEFR <50% predicted.
bronchoconstriction and hypersecretion of mucus. They are less Dangerously life-threatening when bradycardialhypotension,
effective than P2-agonists but longer-lasting and more effective cyanosis or comalexhaustion are present andlor PEFR <30%
against irritants than allergens. May be additive to P2-agonists. predicted. Treatment: Immediate nebulized P2-agonists delivered
Corticosteroids such as beclomethasone are the most important in oxygen and intravenous steroids, with subsequent oral pred-
anti-inflammatory drugs. They reduce eosinophil numbers and acti- nisolone. In unresolving cases, intravenous P2-agonists or xanthines
vation and activity of macrophages and lymphocytes. Inhaled cor- and ventilation may be required.
23 Chronic obstructive pulmonary disease

(a) Typical signs and syvptoms of COPD

Chronic bronchitis

Chronic cough, producing sputum Chron~cbreathlessness (dyspnea)


Hypoventilation, little respiratory effort Cyanoss unusual; normoxlc a t rest, hypoxlc on exerclse
Cyanosis, hypoxaemia with seconda ry polycythaemia Barrel cLlest (hyperlnflat~on).~ n d e r w e ~ g h t
C02 retentionlchronic hypercapnia
- leading t o peripheral vasodilation and bounding pulse Rarely exhibit oedema or cor pulmonale
Oedema Increased TLC, KK lung compliance
Cor pulmonale Reduced DLCO
Normal lung volumes, DLCO,lung compliance

N o t e Many p a t ~ e n t smay present w ~ t hboth chron~cbronch~tlsand emphysema

(b) Influence of smokng on a~rflowlhmltat~on (c) S p ~ r o m e t y

Never smoked or
Normal

? - ..- - -..---
COPD
- Smoked regularly
-
9 ..---...-.---

D~sablllty

1 i
25 50 75 1 5
Age (years) Tme (5)

Chronic obstructive pulmonary disease (COPD) is a group of noea (breathlessness) at rest or on exertion. Many asymptomatic
chronic diseases characterized by reduced expiratory airflow and smokers have lung function abnormalities that predate symptoms,
increased work of breathing. Other terms are COLD and COAD which may be prevented by early smoking cessation. Screening
(chronic obstructive lunglairway disease). The reduced airflow can healthy smokers to identify subclinical airway obstruction is, how-
be due to decreased lung elastic recoil, increased airway resistance ever, controversial.
or both in combination. 'Typical patients show a progressive decline Chronic bronchitis is a clinical diagnosis requiring symptoms of
in lung function, interposed with intermittent acute exacerba- chronic mucus hypersecretion. These symptoms include cough and
tions, eventually leading to progressive respiralory symptoms, excessive mucus production (associated with hypertrophy of mucus
disability and respiratory failure (Chapter 20). COPD encom- glands) for most days out of 3 months for 22 successive years, in the
passes chronic bronchitis and emphysema, which often present absence of airway tumour, acutelchronic infection or uncontrolled
together (Fig. 23a). Asthma is generally not classified as COPD. cardiac disease. Excessive airway mucus leads to increased airway
Chronic hypoxaemia in COPD can lead to pulmonary hyperten- resistance and obstruction. Most patients have normal total lung
sion (Chapter 24). capacity (TLC), functional residual capacity (FRC), residual vol-
Pathogenesis: cigarette smoking is the single most important ume (RV), D,co (diffusing capacity) and static lung compliance
risk factor for development of COPD. Expiratory airflow normally (Chapter 18). Patients with advanced chronic bronchitis often fit
decreases with age; cigarette smoking accelerates this decline (Fig. into the 'blue bloater' (type B non-fighter) morphology (see table),
23b). Other risk factors include increasing age, male gender, child- with marked hypoxaemia, polycythaemia, CO, retention
hood respiratory infections, airway hyperreactivity, low socio- and cor pulmonale (fluid retentionlheart failure secondary to lung
economic status and a,-antitrypsin deficiency (see Chapter 17). disease). Hypoxaemia is mostly due to VA/Qmismatch (Chapter 14)
COPD is diagnosed by airflow obstruction indicated by a and responds well to supplemental 02. There are no radiographic
reduced FEV,IFVC ratio of <0.70 or FEV, two standard devia- signs diagnostic of chronic bronchitis.
tions below predicted provided restrictive disease is excluded (Fig. Emphysema is caused by progressive destruction of alveolar
23b; Chapter 18). Patients with COPD have symptoms of dysp- septa and capillaries, leading to development of enlarged airways
a n d airspaces (bullae), decreased lung elastic recoil and the benefits on lung function and survival are controversial. 0,
increased airway collapsibility. The pathophysiology of emphy- therapy prolongs life in patients with resting daytime hypoxaemia
sema may involve an imbalance between inflammatory cell by slowing progression of the disease. O 2should be utilized as much
proteases and antiprotease defences (Chapter 17). Centrilobular as possible, as there is a dose relation with increasing time. Patients
emphysema is associated with cigarette smoking and predomi- with nocturnal or exercise desaturation benefit from supplemental
nantly involves the upper lung zones. Panacinar emphysema is 0, at night or during exercise. O r a l corticosteroids (to reduce
associated with a,-antitrypsin deficiency (Chapter 17) and pre- inflammation) improve function in <25'% of COPD patients and
dominantly involves the lower lung zones. Patients with emphysema due to the risk of side effects should not be used routinely without
typically have airflow obstruction with elevated TLC, FRC and RV, objective demonstration of benefit. Inhaled corticos~eroidsd o not
reduced D,co and increased static lung compliance. Clinically, such convincingly improve lung function.
patients tend towards the 'pink puffer' (type A fighter) morpholo- Symptomatic therapy for COPD includes inhaled bronchodila-
gy, with tachypnoea and dyspnoea at rest, signs of hyperinflation tors, theophylline, mucolytics and pulmonary rehabilitation. Beta-
and malnutrition including thin body and barrel chest, purse-lipped agonists and anticholinergics improve symptoms and lung
breathing using accessory muscles and distant breath sounds with a function, possibly having additive effects when combined. Theo-
prolonged expiratory phase. Blood gases are normal at rest, with phylline has negligible effects on spirometry, yet may improve
marked O2desaturation during exertion. Radiographically, emphy- exercise performance and blood gases. Patients producing large
sema may appear as hyperinflated lungs with a large retrosternal air- amounts of sputum may benefit from mucolytics. Pulmonary
space and flat diaphragms. When the condition is advanced, there rehabilitation improves quality of life, exercise tolerance and hospi-
may be areas with a lack of vascularity or visualization of bullae. talizations without an effect on lung function.
High-resolution computed tomography (CT) is useful to demon- Prevention of acute C O P D exacerbations include pneumo-
strate enlarged airspaces and air trapping. coccal and influenza vaccination. Patients with any combination of
Therapy: there is no specific therapy to reverse COPD other than increased dyspnoea, increased sputum o r purulent sputum benefit
to prevent disease progression, minimize chronic symptoms and from antibiotics targeted against common respiratory pathogens
prevent acute exacerbations. Patients with advanced COPD may (Haemophilus influenzae. Moraxellu cutrrrrhulis, Streptococcus
be candidates for lung transplantation. Lung volume reduction pneunzoniae). Short courses of oral corticosteroids improve lung
surgery has been reported to improve lung function in emphysema, function and hasten recovery in patients with acute exacerbations.
but long-term efficacy and criteria for patient selection are under Overall prognosis for COPD patients is dependent on the sever-
investigation. ity of airflow obstruction. Patients with a FEV, <0.8 L have a
Smoking cessation slows disease progression (Fig. 23b). Unfor- yearly mortality of -25%). Patients with cor pulmonale. hypercap-
tunately, prolonged quit rates are usually ~ 3 3 %The
. most effective nia. ongoing cigarette smoking and weight loss have a worse prog-
program combines physician advice, nicotine replacement and anti- nosis. Death usually occurs from infection, acute respiratory failure,
depressant treatment. In a,-antitrypsin deficiency, replacement pulmonary embolus or cardiac arrhythmia.
therapy can increase plasma and lung antiprotease levels; however.
24 Pulmonary hypertension

(a) Causes of pulmonary hypertension (b) Evaluation of suspected pulmonary hypertension

I Embolic oblteratlve dlsease I


I Chronlc thromboembol~cdlsease I Dyspnoea,
Loud P2

Congenital heart dsease


I Primary lung disease: I Other studles

Pulmonary arterial
I COPD (Chapter 22) 1 Autoant~bod~es Valvular dlsease
HIV
Llver funct -
stud;"" 1 1 Chest X-ray/
cT kparenchymal lung dlsease
I

Collagen vascular

"
Flbroslng m e d a s t l n ~ t l s
Left atrlal myxoma R ~ g hheart
t
Veno-occlus~vedlseases : catheterlzatlon

Pulmonary hypertension is defined as a mean pulmonary arterial (Chapter 27) such as pulmonary fibrosis, scleroderma or sarcoidosis
(PA) pressure greater than 25mmHg at rest or during exercise. It is cause capillary distortion, as lung parenchyma is affected. Destruc-
usually slow to develop and presents with non-specific symptoms, tion of capillaries occurs in emphysema (Chapter 23) or pneu-
including dyspnoea on exertion, shortness of breath, palpitations, monectomy. Arterial resistance may increase due to vasospasm,
chest pain, light-headedness and syncope. Signs are difficult to remodeling or mechanical obstruction. Alveolar hypoxia is
elicit early and may only include an increased pulmonic component a potent stimulus for arterial vasoconstriction and may cause
of the second heart sound. With more severe hypertension, right pulmonary hypertension in COPD or other chronic lung diseases
ventricular dysfunction will be apparent, including jugular venous (Chapter 23), or at high altitude. Remodeling of pulmonary arte-
distension, right ventricular heave, pedal oedema and hepatic en- rioles is characteristic of idiopathic pulmonary arterial vasculopa-
largement. Patients with pulmonary hypertension usually die from thy (PAV, formerly termed primary pulmonary hypertension),
progressive right heart failure (see the section on heart failure in but may also be seen in chronic hypoxia and chronic left-to-
The Cardiovascular System at a Glance, Chapter 43). Detection of right shunting due to congenital heart disease. PAV may be
pulmonary hypertension requires a high index of suspicion, since due to chronic arterial vasospasm, as some patients demonstrate
signs and symptoms are non-specific and the diagnosis requires fur- a reduction in PA pressure after administration of vasodilators
ther testing. (e.g. diltiazem or prostacyclin). Acute and chronic venous
Since PA pressure is a function of pulmonary vascular resist- thromboembolism causes pulmonary hypertension by mechanical
ance, cardiac output and back-pressure (left atrial pressure), many obstruction of the pulmonary arterial bed. In acute thromboem-
kinds of abnormality can result in pulmonary hypertension (Fig. bolism, a component of vasospasm is also present, as the platelet-
24a). Increased left atrial (LA) pressure, most commonly due to left rich thromboembolus releases vasoactive mediators such as
ventricular dysfunction as in congestive heart failure, leads to thromboxane, serotonin or platelet activating factor. Abnormali-
elevation of PA pressure by increasing back-pressure through the ties of pulmonary veins are an uncommon cause of pulmonary
lungs. Mitral insufficiency or stenosis may also increase PA pres- hypertension, but may occur in pulmonary veno-occlusive disease
sure enough to cause hypertension. In these cases, patients will often or in fibrosing mediastinitis secondary to chronic histoplasmosis or
have signs of pulmonary capillary hypertension such as crackles. tuberculosis (Chapter 33).
Echocardiography should demonstrate LA enlargement. Increases in cardiac output alone seldom cause pulmonary
Increases in pulmonary vascular resistance may occur in the hypertension, as the lungs have a large capacity to accommodate
veins, capillaries or arteries. Increases in capillary resistance are increases in flow through recruitment and distension of the pul-
common and may occur in any lung disease that causes capillary monary vasculature. Up to two-thirds of normal lung can be re-
distortion or reduction in surface area. Interstitial lung diseases moved (increasing flow through the remaining lung by three times)
56
before pulmonary hypertension will develop. However, chronic Treatment
elevations of cardiac output (e.g. congenital cardiac defects, hyper- Patients with pulmonary hypertension die of right heart failure;
thyroidism) may cause pulmonary hypertension, as the chronic high severe right heart failure with right atrial pressure >20mmHg has
flow causes arterial remodelling and increased resistance. a 1-year mortality greater than 75%. Therefore therapy for pul-
monary hypertension in most patients is directed at the underlying
abnormality. to relieve right ventricular strain. There is generally
Evaluation of patients with suspected pulmonary hypertension no specific therapy for pulmonary hypertension in cases of left
(Fig. 24b) begins with echocardiography, allowing calculation of ventricular dysfunction, pulmonary venous disease or pulmonary
right ventricular systolic pressure and visualization of left atrium, parenchymal diseases. Hypoxaemic patients with parenchymal lung
mitral valve, right ventricle and congenital abnormalities. If pul- disease benefit from 0, therapy to diminish hypoxic vasoconstric-
monary hypertension is found in conjunction with an enlarged LA, tion. Patients with thromboembolic disease (Chapter 25) should
it is most likely due to either left ventricular or mitral disease. Chest receive anticoagulation and evaluation for surgical thromboem-
radiology, pulmonary function testing and measurement of arterial bolectomy. Patients with PAV should also receive anticoagulation
oxygen allow detection of parenchymal disease or hypoxia. In the to prevent microthrombi or the devastating effect of an acute
absence of LA enlargement or pulmonary parenchymal disease, thromboembolus (Chapter 25). PAV is the only disease where
further evaluation of pulmonary arteries is necessary. Ventila- therapy directed at the pulmonary arterial abnormality has been
tionlperfusion scanning is most useful to demonstrate chronic shown to benefit. Most patients with PAV require chronic infusions
thromboemboli. Right heart catheterization is the definitive test or frequent nebulization of prostacyclin to improve survival.
for the assessment of pulmonary hypertension, as PA pressure can Prostacyclin has acute vasodilator properties, but an effect on pul-
be measured directly and LA pressure estimated from the pul- monary vascular remodelling or endothelial function more likely
monary capillary wedge pressure (see The Cardiovascular System at explains its positive effect on patient long-term function and sur-
a Glance, Chapter 32). vival. New therapies including endothelin antagonists or nitric
Patients with pulmonary hypertension without an elevated LA oxide are under investigation. Lung transplantation may be consid-
pressure and no apparent pulmonary venous, lung parenchymal, ered for patients with pulmonary hypertension due to parenchymal
chronic thromboemboli or congenital heart disease are assumed to lung disease or PAV.
have PAV. PAV is most often idiopathic, but may be associated with
autoimmune diseases such as scleroderma. There is a familial form
of PAV with autosomal dominant inheritance and associations
between PAV and use of anorexigenic drugs.
25 \lenous thrornhoernholisrn and pulmonary embolism

Fig.25 I'ulmclnary;tnqi~)pr;lms(;I.rl):~nal:t~llr~'irlr V/Q sc:\nu(h. c -vtt>til;tlioli qciln: C. I' per1'11sio11


sciln) iri il no1'1n31patielit :tnd a lxtticnt witti ;I
masrivi. pulnir)n;rr>c.niboli\m -i-l~r an~iogram(dl rl~r~\v~cc-rmple~c blrlck ul' [he riplit pulrnonal.! artery (conlpitrc \vrtli a ) .1 his i.; r~:!lcc~tcrlhy 11iu
Ios,oi riglil I r ~ n gpct'lasrr~n(1.1. hut not vcnttl;trron (e).

Virchtrw'x triad vcnr\llc.;t:rqi.;,Iiyp!rco:~~uillii~.


~rntlv i ~ \ c ~ ~ iti1ul.y
l : t r are prcdictnrs uf incr1::lcccl riqk for 1)V'I'IPE
-- - - .- -- - --
. - -- --. - - .-.- -
General Trauma c . ~ . Low flm.states i?.g. Inherited tieficiencies in:
Smc-]king Surgcn~fi'.g. Iiiplkr~t.~.
rep1:pl;lcclncnt) C't~ri>nit:
heitrt i1icc.t.;~ Prolein S, Protein < '
l'rczn,~n cy Spln;~lcilril injury lnimoI~rli;.;\rion 'I~ltithrornhi~ll l l . l'laarnirlc~ptlri
( I r a I cont r:tccplivcs AnlipIiv>pl~olipid;trlt~lx>tlic.:
:\gc Disease c.g. Damare to endnthrlium e . ~ . Other inherited:
Obeity il.lnlicnancy !'rit)r tllron~llr,si.i T?esi,t:mc.cto pr.r)~tili C' (ItctorV ILcidcn
Ncl~ItroLpl;lc
hyndro~nc Int~t\~:tsci~lur catheters P~-o\I~rorlih~ti
v:~ri:ttit20; 10
Athcn>cclcrosi.; 1 Ivac.rliornoc\~tei~r;tertiia

Venous thmmboernlrolism and i1.i ntorr sipriificant cr~mp1ic;ttilm. Irrlnw tlrc krlcc ~\pprc)rirntttcly15 ??kt will prtrpag:ltc prouirnslly
pulmonary embolism (PE), are comlnon clinicul disorder5 th~il 11 50% 1 - 1 4 ul'cmholi~inp
to tlic Scmnritl aricl ilr;tc ;trrericc : ~ n d1iav~~
Iiatvc ;I substantial impact 1211 patient ~ni)rhidit!inntl rnort:~lily:Table to thc lun!~.Thrombi niay develop in the uxill;trv and quhcl;~vi;un
I .;how r ~ s kI';tctor.c, I'E il.i mi)sl ahcn a 2nmplic:~~icrn01' deep vcins, usu;tlly rlrtc to uurgcry or Intravenous c:~rliclcr~ but tliesc are
venous thrombosis ( D V T ) . Hr~ltidisorders usually przsent with usuallv smallcr. with lcsa risk of c:~tactrophrccrjnscqucnces 11' thcy
non-specific sign5 or symptoms. and are comrncinly undcrdiiic- emboli~c.Soor1 aflcr thrnrnl~i~s I'ormittlo~l.IIIC intrimic tibrinoly~ic
noscd: fl-rcy theri.forcrcrluirc all itppmpri;iteclinic:tI cuspicic~nand a cascade begins t o orp;1tii7e 11icIllmmhus. T l i c rihk o f it thrombus
systcin:~licdiagnostic apprc~ach. tmboli~ingis gro;ltcst eilrly (luring o n ~ o i ~ rprolili.ratinn
g nnrl dc-
cwasr..: once i t I \ nr,v;tni7ed.
Deep venous thrombosis
Ne:lrly all c l ~ n ~ c a l\~gnilic:tnt
lr c;isc\ of PF arke from DVT in the Pulmonary embolism
1owt.r c\trurnities xnd p e l \ i ~+17it~cthrnnih~typically or~pinntc \Vlicn ;t tlironibus cmho1irt.s lo the lung. rcqpircitnry or circulatnry
Table 2 Strategies for prophylaxis against DVT. standard, although VIQ scanning is usually the initial test for as-
sessment of PE, as it is non-invasive, safe and clinically useful (Fig.
Risk of 25: see Chapter 19). A negative perfusion scan effectively rules out
DVT Patient Regimen
PE and a 'high probability' scan (multiple segmental perfusion de-
Low <40years Early ambulation fects with normal ventilation) has a >85% probability of PE (Fig.
(<Ii%)) Minor surgery (<I h) Compression stockings 25). With a high clinical suspicion, a high-probability V/Q scan has a
Minimal immobility positive predictive value >95%. Unfortunately, most V/Q scans are
Moderate >40 years Low-dose: non-diagnostic or indeterminate, with a 15-50% likelihood of PE,
(5-1 0'%1) General surgery (> 1 h) Unfractionated heparin or necessitating further diagnostic testing. Non-invasive imaging of
Cardiac, medical condition Low molecular weight heparin the lower extremity deep veins with Doppler imaging or impedance
Cerebrovascularaccident
'! Inherited hypercoagulability plethysmography is useful, since the presence of thrombosis requires
treatment similar to PE. In patients with underlying cardiac or pul-
High Extended surgery Full-dose:
monary disease, pulmonary angiography is indicated if the above
(>I 5'%) Kneelhip joint Low molecular weight heparin
Hip fracture Warfarin (Coumadin) tests are not diagnostic. In patients with no underlying cardiac or
Trauma pulmonary disease, serial lower extremity imaging may be per-
formed and treatment withheld in the absence of a positive result.
A negative d-dimer assay may be useful to rule out DVTIPE.
abnormalities occur due to sudden occlusion of a pulmonary artery Spirallhelicalcomputed tomography (CT) has been found to have
or arteriole. Occlusion of regional perfusion causes an increase in a sensitivity for PE of 70-95% (higher for more proximal emboli)
dead space, necessitating an increase in minute ventilation to and a specificity >90% when interpreted by experienced readers. C T
maintain normal P,co,. Surfactant production in the lung region scanning also allows visualization of parenchymal abnormalities
distal to the embolus may be reduced after 24 h. resulting in atelec- and may be particularly useful in patients with chronic obstructive
tasis. Most patients with PE have hypoxaemia or a wide A-a gra- pulmonary disease (COPD) or extensive chest X-ray abnormalities,
dient, mostly due to V,IQ mismatching (Chapter 13). Pulmonary where VIQ scanning is often indeterminate. Echocardiography
infarction occurs in less than 25%)of cases of PE. Circulatory com- may reveal RV dysfunction in PE and rule out pericardial tampon-
plications arise from obliteration of the pulmonary vascular bed ade or severe left ventricular (LV) dysfunction. Transoesophageal
and a reduction of cardiac output. The severity of complications is echocardiography may visualize thromboemboli in the main pul-
related to the amount of lung embolized and the pre-existing state monary arteries. but not in the lobar or segmental arteries.
of the pulmonary vasculature and right ventricle (RV); a single large
embolus can be catastrophic, whereas multiple small emboli can Prevention in patientsat risk
cause 'pruning' of the smaller arteries. Compensatory mecha- Depending on the level of risk (low. medium or high) prophylaxis
nisms to maintain cardiac output include pulmonary vascular with pneumatic compression devices, low-dose aspirin or heparin,
distension and recruitment, increased heart rate, catecholamine- adjusted-dose heparin, low molecular weight heparin or warfarin
induced increased venous return and RV contractility. With >50% are effective (Table 2).
obstruction circulatory collapse may occur. Less severe emboli may
be fatal to patients with pre-existing lung or heart disease. Treatment
The cornerstone of therapy for DVTIPE is anticoagulation. which
Signs and symptoms stops propagation of existing thrombus and allows organization.
The signs and symptoms of DVTIPE are non-specific. Lower Therapy should be instituted immediately to patients with a high
extremity pain, swelling, erythema and Homan's sign (pain in the suspicion of disease, because further embolization may be life-
calf on dorsifexion of the foot) occur in a minority of patients with threatening. Unfractionated heparin ( U F H ) or low molecular
DVT. Most patients with PE have dyspnoea, pleuritic chest pain, weight heparin ( L M W H ) for 5-7 days, followed by warfarin for
apprehension and tachypnoea. Tachycardia, cough, crackles, 6 months, is standard therapy. U F H and warfarin must be moni-
haemoptysis, diaphoresis, syncope and chest pain are less common. tored, as subtherapeutic effects increase the risk of recurrent throm-
With severe PE, signs related to RV failure (e.g. hypotension, jugu- boembolism. LMWH is more bioavailable and does not require
lar venous distension) may occur. Most patients with PE have non- monitoring. Patients with inherited or acquired hypercoagulability
specific abnormalities on chest X-ray, including atelectasis or small may require lifelong therapy. Patients with contraindications to
effusions. Non-specific electrocardiographic (ECG) abnormalities anticoagulation (recent surgery, haemorrhagic stroke. central ner-
are common; with RV strain, the ECG may show an SIQ3T3pattern, vous system metastases, active bleeding) or recurrent PE while on
right axis deviation (RAD) or right bundle-branch block (RBBB). therapeutic anticoagulation should receive an inferior vena cava
Arterial blood gas abnormalities are very common, usually con- (IVC) filter to prevent fatal PE.
sisting of widened A-a gradient, hypoxaemia and hypocapnia Although activation of fibrinolysis with thrombolytics hastens
(despite increased dead space). None of these tests will diagnose PE; resolution of perfusion defects and RV dysfunction, there is lack of
however, their results may be useful in assessing clinical likelihood. convincing evidence for benefit. Moreover, thrombolytics cause
increased bleeding complications, including a 0.3-1.5% risk of in-
Diagnosis tracerebral haemorrhage. Therefore thrombolytics are only recom-
Deep venography or pulmonary angiography are the diagnostic mended for life-threatening PE with compromised haemodynamics.
26 Pulmonary involvement in vasculitis

Fig. 26.1 CT scan of patient with Wegener's gr;inulomatosis,showing Fig. 26.2 Histological sect ion showing necrobiotic regions with multi-
large cavitating masses. nucleate giant cells (arrows).

Since the entire cardiac output traverses the lung through the pul- Granulomatoses
monary circulation, it is not surprising that a variety of disorders Wegener's granulomatosis (WG) is a systemic vasculitis that pre-
characterized by vascular inflammation involve the lung. al- dominantly involves the upper and lower respiratory systems and
though such pulmonary vasculitis is relatively uncommon. These the renal glomeruli. The vascular inflammation may involve arte-
disorders may be secondary to systemic collagen vascular disease-- rioles, capillaries and venules. Patients are generally aged 40-60 and
such as rheumatoid arthritis, scleroderma or systemic lupus present with upper respiratory symptoms usually involving the
erythematosus (SLE)-or primary vasculitides that involve pul- sinuses (sinusitis) or nasopharynx (ulcers, septa1perforation, saddle
monary blood vessels (Wegener's granulomatosis, microscopic nose deformity). Radiographic abnormalities in the chest are com-
polyangiitis, lymphomatoid granulomatosis, allergic granulo- mon, even in the absence of cough or haemoptysis. Most com-
matosis and angiitis). Antiglomerular basement membrane monly, they appear as nodules or masses, often with cavitation (Fig.
disease (Goodpasture's syndrome), while not a vasculitis, is 26. l), but they may appear as parenchymal infiltrates. Renal disease
included, since its clinical presentation may be similar to that of is usual and consists of glomerulonephritis with haematuria, pro-
pulmonary vasculitis. Most of these disorders cause systemic symp- teinuria and red blood cell (RBC) casts. The c-ANCA (antipro-
toms and depending on the blood vessel involved may cause pul- teinase 3; antineutrophil cytoplasmic antibody) has a 60-90%
monary infiltrates, masses or alveolar haemorrhage. sensitivity and >90% specificity for WG. Transbronchial lung
biopsies are seldom sufficient to diagnose WG. Larger amounts
Collagen vascular diseases of parenchyma from open or thoracoscopic biopsy are necessary
Rheumatoid arthritis may cause vasculitis and pulmonary to demonstrate granulomatous inflammation in arterial walls or
hypertension (Chapter 24); however, this complication is far less perivascular spaces. Biopsies of the paranasal sinuses or kidneys
frequent than pleural disease (Chapter 28) or diffuse parenchymal may also be diagnostic in the proper clinical context. WG may also
disease (Chapter 27). Patients may develop Caplan's syndrome as involve the ears (otitis media), eyes (conjunctivitis, uveitis), heart
a result of dust inhalation (e.g. coal dust) (Chapter 31). Limited (coronary arteries), peripheral nervous system, skin or joints.
cutaneous scleroderma often spares lung parenchyma and causes Microscopic polyangiitis (MPA) is also a vasculitis of small
pulmonary hypertension by direct involvement of the pulmonary vessels, which has microscopic similarities to WG and polyarteritis
arterioles. While not common, pulmonary capillaritis causing nodosa. In contrast to WG, MPA does not involve the nasopharynx
alveolar haemorrhage due to SLE is a devastating complication of and sinuses and is usually associated with p-ANCA (antimyeloper-
the disease with a high mortality rate. Patients generally have a pre- oxidase) rather than c-ANCA. It is often seen in patients with
existing diagnosis of SLE. usually with renal involvement. Rarely, hepatitis B or C infection. Treatment with corticosteroids and
SLE may cause pulmonary hypertension by direct involvement of cyclophosphamide substantially reduces mortality.
the pulmonary vasculature. Clinically, this is indistinguishable from Lymphomatoid granulomatosis (LG) is a systemic vasculitis
pulmonary arterial hypertension (Chapter 24). of the lungs, kidneys, central nervous system (CNS) and skin. LG
Table 1 Pulmonary vasculitides.

Disease Blood vessel Comment

Collagen vascular diseases


Rheumatoid arthritis Arterieslarterioles Uncommon
Scleroderma Fibrosis in arterioles CREST syndrome
SLE Capillaritis Pulmonary hemorrhage
Vasculitides
Wegener's granulomatosis Granulomatous inflammation Pulmonary hemorrhage common
Arteriolarlvenular vasculitis c-ANCA (90%))
Fibrinoid necrosis
Capillaritis (113)
Microscopic polyangiitis Arteriolelvenule vasculitis Related to Wegener's and PAN
Capillaritis, fibrinoid necrosis p-ANCA, hepatitis B, C
Lymphomatoid granulomatosis Angiocentriclangiodestructive lymphocytes, Epstein-Barr Virus
plasma cells, atypical lymphocytes Lymphoproliferative
Allergic granulomatosis and angiitis Necrotizing vasculitis in small and Asthma, eosinophilia
medium-sized arteries, arterioles, venules 66'% p-ANCA or c-ANCA
Granulomas, eosinophils
Fibrinoid necrosis
Anti-GBM disease Intra-alveolar haemorrhage Smoking, recent infection
Linear IgG in basement membrane
Minimal inflammation
ANCA, antineutrophil cytoplasmic antibody; CREST, calcinosis, Raynaud's phenomenon, esophageal involvement, sclerodactyly and
telangiectasia; GBM, glomerular basement membrane; PAN, polyarteritis nodosa; SLE, systemic lupus erythematosus.

is strongly associated with, and may be a late complication of, eosinophilia, migratory pulmonary infiltrates and neuropathy.
Epstein-Barr virus (EBV) infection. It behaves like an indolent Most patients respond to corticosteroids. Cyclophosphamide or
lymphoproliferative disease and may transform into a B-cell lym- azathioprine may be added in resistant cases. Patients who respond
phoma. Patients typically have fever, malaise, cough, dyspnoea and to therapy seldom relapse. Patients with an onset of asthma imme-
apapular rash. Radiographic abnormalities usually consist of mul- diately before or concurrent with vasculitis have a poorer prognosis.
tiple lower lobe nodular densities. Lung biopsy shows angio- Overall, the survival is >70%, with mortality usually due to cardiac,
centriclangiodestructive mixed cell infiltration with lymphocytes, CNS, renal or gastrointestinal involvement.
plasma cells and atypical lymphocytes. Vascular occlusion and Anti-glomerular basement membrane (GBM) disease is
necrosis are common. L G is considered to be a lymphoproliferative caused by antibodies directed against the glomerular membranes
disorder and is treated with chemotherapy and corticosteroids. causing glomerulonephritis. In Goodpasture's syndrome these
Without treatment, the disease progresses and is usually fatal. antibodies sometimes cross-react with alveolar basement mem-
Allergic granulomatosis a n d angiitis (Churg-Strauss syn- brane, causing alveolar haemorrhage. Alveolar haemorrhage seems
drome) is a mediumlsmall vessel granulomatous vasculitis of the to occur predominantly in patients who smoke cigarettes, or who
lung, skin, heart, nervous system and kidney. It is probably the sec- have a recent respiratory infection which may alter alveolar perme-
ond most common pulmonary vasculitis after WG. Most patients ability. Patients present with rapidly progressive glomerulonephri-
have a history of allergic rhinitis andlor asthma and peripheral tis, haemoptysis, anaemia and diffuse alveolar infiltrates on
eosinophilia that may predate the vasculitis by up to a decade. Pa- radiographs. In contrast to the primary vasculitides, prolonged sys-
tients will present with worsening asthma, fever, malaise, subcuta- temic symptoms such as fever, malaise or rash are uncommon. Pul-
neous tender nodules, mononeuritis multiplex and radiographic monary function testing demonstrates elevated DLco from
infiltrates. There may also be pericarditis, abdominal pain and extravasated haemoglobin in the lung. Serial measurement of DLco
glomerulonephritis. Radiographic abnormalities are most often may show a decline as extravascular haemoglobin saturates with
patchy, fleeting infiltrates, but may include cavitating nodules or CO. Diagnosis requires demonstration of anti-GBM antibodies in
masses, interstitial infiltrates or pleural effusions. Chest computed serum or linear IgG in glomerular or alveolar basement mem-
tomography (CT) may show ground glass opacities or peribronchial branes. c-ANCA or p-ANCA may be positive. Patients with Good-
thickening. p-ANCA or c-ANCA may be positive. Lung biopsy pasture's syndrome should be treated with plasmapheresis,
will show perivascular granulomatous inflammation, small artery cyclophosphamide and corticosteroids. Therapy may control alveo-
and vein vasculitis, prominent eosinophils and necrosis. The lar haemorrhage; however, renal disease is usually irreversible if the
diagnosis may be made without biopsy in the presence of asthma, presenting creatinine is >5 mgldL.
27 Interstitial lung disease

Acute eosinophilic pneumonla


Acute n t e r s t ~ t i apneumonia
l Dermatomyosit~slpolymyosit~s
1 (Hamman-Rich syndrome) Rheumatold a r t h r ~ t i s / Cyclophosphamide Coal worker's pneumocon~osis I

I
Cryptogenic organizing pneumon~a
Eosinophil~cgranuloma
ldiopath c pulmonary fibros~a
Scleroderma
Systemic lupus erythematosus
Sjogren's syndrome
1
I
/
Methotrexate
Nitrofuranto~n
/
I
/
Farmer'5 lung
Hard metal d~sease
Extr~nsicallergic alveolltis I
I / (hypersensitivity pneumonit~s)
Lymphang~olyomyomato~i~
Lymphangitic carclnomatoaia /
Oxygen
Penicillamine 1 P~geonfancier's lung
Lymphocytic inter5titial pneumonia / Radiation / 5111cos15

(a) Cellular basis of fibrosis (not t o scale)

fibres, fibronectin), Fibroblasts and Borne inflammatory cells

\ infiltration of
Growth factor% infiammatorv cells
TGF-P, FGF,
PDGF Eo5inophil5
4 Neutrophils
Recruitment, growth and J'
activation of fibroblasts Elastase,
proteolytic enzymes
J J
Production of Degradation of
reticulin, collagens elastic components
Note: Fibrosis is complex, and t h i s diagram is highly simplified.
TGF-P = transforming growth factor, FGF = fibroblast growth factor,
PDGF = platelet derived growth factor. TGF-P is a key mediator of fibrosis
Fibmi. A
(c) CXR of patient with sarcoidosis
(see text)

1 (b) CT scan showing typical honeycomb


lung appearance and interstitial
infiltrates with some dorsal
consolidation (d) CT scan of sarcoidosis
-
Interstitial lung disease (ILD) refers to an extensive variety of histological examination of the lungs in patients with clinical
acute and chronic clinical disorders, characterized by inflammation IPFICFA.
or fibrosis of alveolar-capillary units and distal airways (Fig. 27a). Sarcoidosis is a multiorgan granulomatous disease of unknown
Thesediseases are not limited to the interstitium, but may involve all aetiology. but with characteristic clinical and pathologic features.
of the matrix components of the lung. such that 'diffuse parenchy- Activated CD4 lymphocytes appear to participate in the patho-
mal lung diseases' is a more accurate description of their morphol- physiology. Sarcoidosis is usually asymptomatic, presenting with
ogy. The symptoms of I L D are most often dyspnoea and cough, unsuspected bilateral hilar adenopathy on X-ray in young ( 2 0 4 0
usually indolent over months. Signs include digital clubbing and years) people. Symptomatic sarcoidosis most often involves the
diffuse inspiratory crackles. With advanced disease. there may be lung with ILD, but may also be present in the eyes, skin, joints, cen-
hypoxaemia and right ventricular failure. Pulmonary function tral nervous system (CNS), heart or liver. Radiographically, lung
tests (Chapter 18) reveal reduced total lung capacity (TLC), func- disease manifests as any combination of hilarlparatracheal
tional residual capacity (FRC) and residual volume (RV) due to de- adenopathy, reticulonodular infiltrates or advanced fibrosis with
creased compliance and increased elastic recoil of the lungs. cysts (Fig. 27c). Pleural disease is uncommon. Patients with signifi-
D,co is reduced because of a reduction in surface area for gas cant radiographic parenchymal abnormalities may have a normal
exchange secondary to reduced lung volume. With exercise. there chest physical examination. The classic histological feature of sar-
is rapid shallow breathing and O2 desaturation. Most (>90%) coidosis is non-caseating granulomas, which are commonly
of patients with ILD will have an a b n o r m a l chest X-ray, with found on transbronchial biopsy even in the absence of radio-
any combination or alveolar, interstitial or mixed infiltrates, graphic abnormalities. Angiotensin-converting enzyme is com-
typically predominating in the lower lobes. Small (0.5-2cm), monly elevated in sarcoidosis. but this test is neither sensitive nor
thick-walled cysts denote advanced fibrosis and give rise to a typical specific for diagnostic purposes. Sarcoidosis is usually a self-limited
radiological appearance known as 'honeycomb lung' (Fig. 27b). disease without therapy. U p to 50% of cases resolve without treat-
High-resolution computed tomography (HRCT) allows a far more ment within 3 years; however, some patients will develop progres-
accurate distinction of parenchymal involvement and may have sive ILD involvement. Patients should be followed with periodic
diagnostic or therapeutic implications. Bronchoalveolar lavage radiographs and pulmonary function tests. Therapy is indicated for
(BAL) may be useful to diagnose malignancy, eosinophilic pneu- significant organ involvement or significant impairment on pul-
monia, alveolar proteinosis or alveolar haemorrhage. BAL fluid monary function testing. Oral corticosteroids are standard therapy
containing increased inflammatory cells (Chapter 17) reflects and most patients achieve remission within months. The optimal
alveolitis and correlates with ground glass infiltrates on HRCT; duration of therapy is uncertain and is individualized for each pa-
this may identify patients with rapidly progressive o r potentially tient. Immunosuppressive or immuriomodulatory medications have
reversible disease. O p e n or thoracoscopic lung biopsy is usually been reported to benefit selected patients, but no controlled trial has
necessary to determine a diagnosis and therapeutic plan. demonstrated superiority over corticosteroids. Lung transplanta-
ILD may be classified clinically or histologically. Clinically, ILD tion is an option for patients with advanced fibrosis.
may be caused by occupational or environmental exposures (Chap- Interstitial lung disease is a common complication of autoim-
ter 31) drugsltherapies, autoimmune diseases and primary pul- m u n e o r collagen vascular diseases such as scleroderma, rheuma-
monary conditions or may be idiopathic (see Table). Histological toid arthritis, systemic lupus erythematosus (SLE), polymyositisl
patterns include usual interstitial pneumonia (UIP), desquamative dermatomyositis (PMIDM) and Sjiigren'ssvndrome. It is important
interstitial pneumonia (DIP), non-specific interstitial pneumonia to distinguish this manifestation of the underlying disease from a
(NSIP), acute interstitial pneumonitis (AIP, Hamman-Rich syn- complication of drug therapy or opportunistic infection. These pa-
drome), lymphocytic interstitial pneumonia (LIP), organizing tients have signs, symptoms, radiographs and pulmonary function
pneumonia. eosinophilic pneumonia, granulomatous disease or tests similar to patients with IPFICFA or AIP. Some patients with
honeycomb lung. The histological patterns are usually not disease- symptomatic disease will respond to aggressive therapy with
specific. Knowledge of clinical and histological associations is nec- chemotherapeutic agents plus corticosteroids. Up to 70% of pa-
essary to diagnose and treat these disorders. tients with scleroderma may have interstitial fibrosis clinically or
Idiopathic pulmonary fibrosis (1PF)lcryptogenic fibrosing radiographically. Patients with a ground-glass pattern on HRCT or
alveolitis (CFA) typically occurs in men in their 60s, with sub- alveolitis on BAL have more rapid deterioration of lung function.
pleural fibrosis in the lower lobes. U I P is the most common his- The median survival of 8years is better than that in patients with
tological pattern of IPFICFA, although it may represent a point IPFICFA. Interstitial lung disease is also common in patients with
in the progression of D I P or NSIP to end-stage honeycomb lung. rheumatoid arthritis, with males, smokers and patients with severe
U I P is a patchy disease with areas of normal lung, interstitial in- joint disease at higher risk. Acute and chronic interstitial pneu-
flammation and fibrosis with fibroblast proliferation (Fig. 27a). The monitis occurs in 4 0 % of patients with SLE. While PMIDM is an
median survival in patients with U I P is less than 5 years and corti- uncommon disease, up to 50% of patients will develop interstitial
costeroids have minimal effect. DIP and NSIP are diagnosed at lung disease and it is a frequent cause of mortality. Interstitial
younger ages, have more diffuse, more cellular, less fibrotic involve- disease is often associated with the presence of antisynthetase
ment and respond well to corticosteroids. The mean survival in pa- autoantibodies (e.g. anti-Jo-1). LIP is the most common inter-
tients with D I P and NSIP is >10 years and complete recovery is stitial Iung disease in Sjogrm's syndrornp and may evolve to pul-
attainable with therapy. These differences highlight the utility of monary lymphoma.
28 Pleural diseases

/ Causes of pleural effusions

r p eural LDti >0.66of t o p norrna 5 e r ~ mv a l ~ e )


- . . . . . - -.- -. .....-. -- - . .. - . . . - .... . .- .-
- - - - -- - - -- . . . . .. . . -.

Infectious Autoimmunelcollagen vascular Miscellaneous


-- .. .. . - --
Para-wneumonlc 1 System~clupus erythematosus Pulmonary embol~sm

Asbestos exposure
Empyema Haemothorax
Tuberculosis
Parastic
amoeba
ech~nococcus
paragon~mus
VIra l

Clrrhoslslhepatlc hydrothorax Nephrotlc dlsease


Congestive heart fallure Pertoneal dlalysls
Myxoedema

I CXR of effusion in left lung


The potential space between the parietal and visceral pleurae For example, extremely low glucose is typical for empyema, malig-
serves as a coupling system between the lung and the chest wall, and nancy. tuberculosis (Chapter 33), rheumatoid arthritis, systemic
normally contains a small amount of fluid. A negative pleural pres- lupus erythematosus (SLE) or oesophageal perforation. If clini-
sure is maintained by the dynamic tension between the chest wall cally indicated, a specific diagnosis may be obtained from micro-
and the lung (Chapter 3). Both pleurae have a systemic blood sup- biologic stains and culture, cytopathology, amylase, triglycerides
ply and lymphatics, although lymphatic drainage of the pleural and measurement of antinuclear antibody (ANA) titre. Although
space is predominantly via the parietal pleura. Fluid flux through all patients with SLE have a positive ANA titre in the pleural fluid,
the pleural space is determined by Starling's relationship between it is also present in a significant proportion (-1 5%) of other effu-
microvascular pressures, oncotic pressures, permeability and sur- sions; these may be related to malignancy.
face area. Normally, there is net filtration of transudative (protein- Pneumonia (Chapter 32) commonly causes parapneumonic
poor) fluid into the pleural space that is balanced by resorption via pleural effusions. These effusions are usually sterile exudates with a
the parietal lymphatics. Pneumothorax is an important condition neutrophilic leukocytosis and require only treatment of the pneu-
that occurs when air enters the pleural space and pleural pressure monia to resolve. However, if bacteria invade the pleural space, a
rises to atmospheric pressure; it is discussed in detail in Chapter 29. complicated parapneumonic effusion or empyema will develop.
Chylothorax is due to accumulation of triglyceride-rich lymph in These effusions are characterized by a low pH and extensive fibrin
the pleural space, generally as the result of damage to the thoracic deposition causing fluid loculation and require adequate open or
duct causing leakage into the pleural space, for example due to closed drainage for healing. Streptococcus pneurnonicle, Stuphylo-
trauma or carcinoma. Empyema is accumulation of pus. Pleurisy coccus aureu.r, Gram-negative bacteria and anaerobes commonly
is a term commonly used to describe the sharp localized pain arising cause complicated effusions.
from any disease of the pleura. It is made worse by deep inspiration Tuberculosis pleurisy occurs when a subpleural focus of pri-
and coughing. mary infection ruptures into the pleural space, causing a delayed
Most diseases of the pleura present with pleural effusion. Effu- hypersensitivity response. Subsequently, an exudative effusion
sions are due to excessive fluid formation or inadequate fluid clear- with a lymphocytic leukocytosis. a paucity of macrophages and an
ance. Symptoms develop if the fluid is inflammatory or if elevated adenosine deaminase will develop. Patients develop fever,
pulmonary mechanics are compromised. Thus, the most common dyspnoea, pleuritic pain and a positive tuberculin response (see
symptoms of a pleural effusion are pleuritic chest pain, dull Chapter 33). Granulomatous inflammatiop is seen on pleural biop-
aching pain, fullness o f the chest or dyspnoea. Physical exami- sy and culture of pleural tissue has the highest diagnostic yield.
nation reveals decreased breath sounds, dullness to percussion, de- Primary lung malignancies or metastases to the lung may
creased tactile or vocal fremitus. If there is inflammation, there may cause pleural effusions by direct invasion or by obstruction of
be a friction rub. Compressive atelectasis may cause bronchial parietal lymphatic drainage. Malignant effusions are mostly exuda-
breath sounds. tive (90%), often with a very high LDH, low pH and low glucose.
Pathophysiology: it is useful to categorize pleural effusions as Cytology of the pleural fluid has a high diagnostic yield. Sympto-
transudative or exudative (Fig. 28a). Transudative effusions matic pleural effusions may respond to therapy for the underlying
are usually due to an imbalance in Starling's forces across normal malignancy, although palliative obliteration of the pleural space
pleural membranes, have protein-poor fluid, are often bilateral and (pleurodesis) is often necessary to relieve dyspnoea or chest pain.
are not associated with fever, pleuritic pain or tenderness to palpa- Mesothelioma is an uncommon malignancy that originates in
tion. The most common cause of a transudative effusion is conges- the pleura and/or peritoneum. Over 75%)of cases develop 20-30
tive heart failure. Other causes include cirrhosis with ascites, years after occupational asbestos exposure. Asbestos may also
nephrotic syndrome. pericardial disease or peritoneal dialysis. cause benign pleural effusions or calcified plaques on the parietal
Exudative effusions imply disease of the pleura or the adjacent pleura in the lower lungs or along the diaphragmatic surface.
lung and are characterized by an increased protein, lactate dehydro- Mesothelioma typically develops in men aged 50-70, presenting
genase (LDH), cholesterol or white blood cell count (WBC) (Fig. with insidious dyspnoea and aching chest pain. Chest X-rays
28a). The differential diagnosis of exudative effusions is broad, usually show unilateral pleural effusion (Fig. 28b), and computed
including: infection. malignancy, autoimmune disease, oesophageal tomography (CT) shows fibrotic encasement of the pleural space.
perforation and pancreatitis. Pleural fluid cytology is not usually diagnostic. Thoracoscopic
The diagnostic evaluation of a pleural effusion should include biopsies have the highest yield. Treatment is generally palliative, in-
measurement of cell count with differential, pH, protein, LDH, cluding pleurodesis. The prognosis is poor, with a median survival
cholesterol and glucose. These studies will usually distinguish exu- of approximately 1 year.
dates from transudates and will often suggest a specific diagnosis.
29 Pneumothorax

1 (a) Pneumothorax Complete pneumothorax I

Moderate pneumothorax Small pneumothorax 1

1 Degree of collapse (d) Chest draln ~nsertlon Edge of 61 , 1


Management
Complete Moderate / Small

Pr~mary Asp~ratel Asprate Observe


chest draln I

I Secondary / Chest dram Chestdraln Chest draln I


Traumatcl Chest draln Chest drain Observe1
latrogenlc chest dram

(c) Aspiration
Local anaesthetic

Plastic cannul

Aspirate using 3-way t a


Definition Clinical evaluation
Pneumothorax is a collection of air between the visceral and pari- A pneumothorax should be considered in any patient who
etal pleura, causing a real rather than potential pleural space (Fig. suddenly becomes breathless for no obvious reason, particularly if
29a). there is underlying lung disease. Sudden onset of sharp pleuritic
pain and breathlessness are the commonest presenting symptoms.
Causes, epidemiology and prognosis Most primary spontaneous pneumothoraces are small, defined as
Pneumothorax may be categorized into the following types. occupying less than 30% of the diameter of the hemithorax (Fig.
Primary spontaneous pneumothorax (PSP): characteristi- 29b), and cause few symptoms other than pain. Clinical signs can be
cally affects tall. narrow-chested, young (2040-year-old), male surprisingly difficult to detect, but in larger pneumothoraxes re-
(M: F 5 : 1) adults. It is the commonest type of pneumothorax duced air entry and hyperresonant percussion over one hemithorax
(prevalence 81100 0001year. rising to 2001100 0001year in subjects are characteristic and may be associated with tachypnoea and
>1.9m in height). There is usually no antecedent history of chest cyanosis. Occasionally, air may track into the mediastinum (pneu-
disease, and underlying lung disease is unlikely. It occurs following momediastinum), into the subcutaneous tissue at the root of the
rupture of small apical subpleural air cysts ('blebs'). Often the neck or along the tract of a chest drain, causing subcutaneous em-
pneumothorax is small and in some cases it can be managed without physema. Subcutaneous emphysema can cause extensive facial and
drainage or with simple aspiration (see below) on an outpatient body swelling and has a characteristic cracking sensation on palpa-
basis. Ipsilateral recurrence is common: after a second and third tion. It usually subsides without complications following adequate
pneumothorax, recurrence rates are 60% and SO%, respectively. drainage of the pneumothorax.
Pleurodesis (fusion of the visceral and parietal pleural) by medical
(pleural insertion of bleomycin or talc) or surgical (abrasion of the Investigation
pleural lining) means is recommended after the second pneumotho- Routine monitoring procedures may detect tachycardia, hypoten-
rax (or first, if the risk of a second pneumothorax would be high as sion and desaturation at the onset of a pneumothorax. Routine
in a mountain climber). It is usually curative. blood tests are normal. although blood gases demonstrate hypox-
Secondary pneumothorax is associated with respiratory dis- aemia and hypocapnia. A chest radiograph in inspiration will con-
ease, most commonly obstructive (COPD, asthma). fibrotic firm the diagnosis. Localized pneumothoraxes following trauma or
(cryptogenic fibrosing alveolitis, CFA) or infective (tuberculosis. mechanical ventilation may only be visible on a chest computed
pneumonia) and occasionally rare or inherited disorders tomography (CT) scan. Other investigations are only of value in
(lymphangioleiomyomatosis,Marfan's, cystic fibrosis) that dis- determining the nature of underlying lung disease.
rupt lung architecture. Secondary pneumothorax is more serious
than PSP, because the patient has less respiratory reserve to deal Management
with the lung collapse due to underlying lung disease. These patients Immediate supportive therapy includes supplemental oxygen by
require admission to hospital and even a small pneumothorax will face-mask and analgesia. Treatment of a pneumothorax is depen-
need intercostal tube drainage (Fig. 29b). dent on the cause, size and symptoms and is summarized in Fig.
Traumaticliatrogenic pneumothorax: often follows blunt 29b. A tension pneumothorax must be drained immediately (see
(road traffic accidents) or penetrating (fractured ribs, stab wounds) above). A small PSP (<30%) may be managed without drainage on
chest trauma. Intensive-care unit patients with lung disease are at an outpatient basis. Follow-up radiography will confirm gradual re-
particular risk of pneumothorax due to the high pressures (baro- absorption of pleural air. Moderate (>30%) and even large PSP,
trauma) and alveolar overdistension ('volutrauma') associated with significant symptoms, may be successfully aspirated. Follow-
with mechanical ventilation. 'Protective' ventilation strategies using ing infiltration of local anaesthetic, a 16-G cannula is inserted into
low-pressure, low-volume ventilation reduce this risk. Therapeutic the second intercostal space in the midclavicular line. The needle is
procedures (line insertion, chest wall surgery, pleural biopsy) are the withdrawn and air is aspirated through the cannula using a 50-mL
commonest iatrogenic causes of pneumothorax. syringe and a three-way tap connected to an underwater seal (Fig.
A tension pneumothorax may complicate primary, secondary 29c). Aspiration is discontinued when resistance is felt, if excessive
or traumatic pneumothoraces, but is most common during me- coughing occurs or if more than 2.5 L have been aspirated. Success-
chanical ventilation and following trauma. It occurs when air ac- ful aspiration is confirmed on a repeat chest radiograph. Following
cumulates in the pleural cavity faster than it can be removed. The aspiration, all patients should be admitted overnight. Intercostal
increase in intrathoracic pressure causes mediastinal shift, compres- chest drainage (Fig. 29d) may be required for moderate or large PSP
sion of functioning lung, inhibition of venous return and shock due with hypoxaemic respiratory failure or if aspiration is unsuccess-
to reduced cardiac output. It is a medical emergency and fatal if ful. In general, all secondary and traumatic pneumothoraces will
not rapidly relieved by drainage. Detection is a clinical diagnosis; require admission to hospital and a chest drain. Suction
awaiting chest X-ray (CXR) confirmation may be life-threatening. (5-50cmH,O) should be applied using a high-flow system (i.e. 'wall'
Immediate drainage with a 14-G needle in the second intercostal suction) if a persistent leak develops. The correct pressure is that
space in the midclavicular is essential. A characteristic 'hiss' of which opposes the visceral and parietal pleura, allowing pleurode-
escaping gas confirms the diagnosis. A chest drain is then inserted sis. Early thoracic surgical advice should be obtained if the pleural
(Fig. 29b). surfaces cannot be opposed, as this suggestsdevelopment of a bron-
chopleural fistula, which may require surgical correction.
30 Cystic fibrosis and bronchiectasis

- ~ ~~-~

(a) Mean survival of CF patients (G) Other conditions associated with CF

(
-'
1
1
--
Condition

Delayed development, puberty


% of CF patent6

100% I
- . --

Male infertility (absentlobstructed


vas deferens and epididymis)
-
1 98%

Female ~nfertility

Nasal polyps
m o s t in 2nd decade

Symptomatic sinusitis

0 25 50
Age (years) Rectal prolapse
Rare in adults
(b) Development o f respiratory problems in CF
Bone demineralization
(vitamin D deficiency)
--.--
~ ~ h ~ o s t e ~ a r t h r o ~ a, t h y 15%adults
- . .-
.
--I
Dysfunctional gallbladder
or gallstones
.+

B ~ l ~ acirrhosis
ry 5% adults

(d) Some conditions associated with bronchiectasis

Allergic bronchopulmonary aspergillosis (Chapter 31)


Alpha,-antitryps~ndef~cency(Chapters 17, 23)
Bronchial obstruction (fore~gnbod~es,mucus, tumour)

3damage,

1-1 lnflammat~onand bronchiectasis Stero~ds 1


Congen~talcartilage deficiency (Williams-Campbell syndrome)
Cystic fibrosis
Fibrot~cdisease and alveol~tis(Chapters 27, 31)
HIV and ~mmunodefic~ency (Chapter 3 4 )
Infection (e g measles, pertuss~s),pneumona (Chapter 32)
Lung transplant
Primary c l ~ a r ydyskines~a(Kartagener's syndrome, Chapter 17)
Rheumatoid arthritis

Impaired lung funct~on


Progresa~verespiratory failure
I-
-
_
;-
#
~rinLhidi
*r-
Tuberculos~s(Chapter 33)
Tracheobronchomegaly (Mounier-Kuhn's syndrome)

Cystic fibrosis (CF) is the primary cause of severe chronic lung dis- There is usually an increased salt content of sweat. Figure 30c
ease in children, although 90% of children now survive into their shows some associated disorders.
second decade (Fig. 30a). C F is characterized by chronic bron- C F is an autosomal recessive trait that is the most common
chopulmonary infection and airway obstruction (Fig. 30b) and by genetic cause of morbidity and mortality in the white population,
exocrine pancreatic insufficiency with consequent effects on gut with a prevalence of around one in 2000 live births; nearly 5% of
function, nutrition and development. The key feature of C F is in- whites of European descent are heterozygous carriers. Prevalence is
creased viscosity and subsequent stasis of epithelial mucus. far less in others. being -1 in 17000 for those of African descent. C F
is due to mutations in a gene on chromosome 7 encoding for the antibiotic is determined following identification of infecting organ-
cystic fibrosis transmembrane conductance regulator (CFTR), isms. The dose should be higher in C F patients and the course
a cyclic adenosine monophosphate (CAMP)-regulated epithelial longer. Development of resistance is a key problem and is transfer-
chloride channel that can also alter activity of other ionic trans- able; segregation of patients is thus advisable. Adequate immuniza-
porters. Dysfunction of CFTR impairs epithelial chloride, sodium tion for measles, pertussis and influenza is important, as these
and water transfer and thus causes reduced mucus hydration and organisms are particularly dangerous in CF.
increased viscosity (Chapter 17). Over 800 mutations in the CFTR Clearance: training by physiotherapists in postural drainage
gene have been described, but the most common, found in -65% of (tipping the body so that the infected lobe is uppermost) is vital,
patients with CF, is deletion of the phenylalanine codon at position coupled with chest percussion to mobilize secretions to the upper
508, the hF508 mutation. airways where they can be coughed up. Such treatment is prescribed
one to four times a day. Recently introduced therapies include
Clinical features inhalation of DNase, an enzyme that breaks down DNA from
The lungs of neonates with C F are often normal, but rapid devel- dead cells which contributes to mucus viscosity. Inhalation of
opment of respiratory symptoms, including refractory cough and aerosolized saline may improve mucus hydration, as may blockade
infections, is usual. C F patients nearly always have an increased of sodium reabsorption with amiloride or stimulation of chloride
lung volume and finger clubbing (increased curvature of the nail secretion with nucleotide triphosphates. Cough should never be
and loss of normal angle between nail and nail bed). Recurrent suppressed, as it is an important method of clearance.
bronchopulmonary infections, primarily as a result of defective Other therapies: bronchodilators (P-agonists) may improve lung
mucus clearance, are rarely cleared once established and eventually function and corticosteroids may assist inflammation in some
result in bronchiectasis (see below), extensive lung damage and patients. A potential therapy under intense investigation is gene
dysfunction. Spontaneous pneumothorax (Chapter 29) and transfer of the normal CFTR gene. In end-stage respiratory disease
haemoptysis (spitting blood; see Chapter 40) are not uncommon. a lung transplant should be considered.
About 10% of neonates present with meconium ileus (failure to pass Nutrition: most patients with C F require pancreatic enzymes
meconium), which can cause death in the first days of life; 20% of with meals, supplemented with vitamins. High-calorific foods
older patients exhibit a similar ileal obstruction (meconium ileus should be advised.
equivalent, MIE). Eighty-five per cent of patients have steator-
rhoea (high fat stools) as a result of pancreatic insufficiency. Some Bronchiectasis
patients have only mild respiratory symptoms for many years, but Bronchiectasis is an abnormal and permanent dilation of proximal
this is inevitably followed by a characteristic increase in the fre- (>2mm) bronchi due to inflammation and subsequent destruction
quency and severity of periods of exacerbation of symptoms of the elastic and muscular components of their walls (see also
(cough, dyspnoea, loss of appetite). Eventually, severe restrictions Chapter 40). It is normally associated with defects in mucociliary
in activity herald the end-stage disease, followed by respiratory fail- clearance (Chapter 17) and persistent respiratory infections.
ure, hypoxaemia, pulmonary hypertension and death. Onset is often in childhood, following pulmonary infections com-
plicating measles or pertussis. Since the introduction of antibiotics,
Diagnosis the most common cause of bronchiectasis is now C F (Fig. 30d),
Several factors need to be taken into account, including a family except in poorly resourced countries. Symptoms depend on the
history of the disease and the presence of typical respiratory and severity and location of diseased bronchi, but commonly include
gastrointestinal disorders (Fig. 30c). A sweat chloride o r sodium persistent productive cough, with large quantities of foul-smelling
concentration above 60mmollL is diagnostic when coupled with purulent sputum as the disease worsens. Severity has been corre-
such disorders, although -1% of C F patients may have normal lated with the volume of sputum produced, but not with dyspnoea.
sweat electrolytes. DNA analysis can detect known mutations (e.g. Haemoptysis and recurrent pneumonia or abscesses are common;
AF508), but is limited by the high number of unknown mutations. haemoptysis is normally mild, but can become life-threatening,
In later disease chest X-rays can detect bronchiectasis (see below). particularly in C F patients. Fever, anaemia and weight loss may
Neonates can be screened for C F by blood immunoreactive trypsin, accompany the disease. Patients often develop finger clubbing,
which may detect many, but not all cases. metastatic abscesses, respiratory failure and amyloidosis. Chest X-
rays and high-resolution computed tomography (HRCT) can often
Management detect the dilated and thickened bronchi (Chapter 40). Manage-
The primary objectives of treatment are to control infection, pro- ment is similar to that for CF, though without the nutritional
mote mucus clearance and improve nutrition. Early antibiotic requirements.
therapy is crucial to inhibit progression of the disease. Choice of
31 Occupational and environmental-related lung disease

1 (a) Common examples of irritant gases and other agents causing lung-specific resporses 1
Agent 5oune Reeponee
-- -
-
1 Ammonia Industrial refrigeration leaks, fertilizers
.... . ..
.. - . -..... -
Low exposure
Exacerbations of asthma and COPD
Chiorine gas Industrial leakage, water purification including swimming pools, Enhanced response t o allergen
household bleach (liquidlpowder) interactions Moderate exposure
Mild mucosai irritation
Airway inflammation and bronchiolitis
N~trogend ~ o x ~ d e Vehicle exhausts, welding, power stations, oil refineries, gas and oil Severe exposure
N~trogenoxdes burning equipment, organic decomposition, structural or polymer fibres Epithelial damage leading t o diffuse alveolar damage
-- Pulmonary oedema and ARDS
Ozone Vehicle exhausts, welding, copiers, ozone generators, bleaching, water In some cases - late response (2-8 weeks)
treatment, plasma welding Bronchiolitis obliterans a f t e r initial recovery
~. . .- ........................................

Sulphur dioxide Combustion of fossil fuels, power stations, oil refineries, smelters, oil
burning heaters, mining, ore refining, cement manufacturing, Also: direct bronchoconstrlction, especially in
refrigeration plants asthmatics
. .-
. .. . . -- ............. --. .....................................

Acrolein, aidehydes Structural or wildland fires, other combustion Also: strongly pro-inflammatory (esp. acroiein)

I Diesel particulates
(<lO~ml
Diesel engines Airwaylalveolar inflammation
Increased deaths in elderly

A
. - - ........... .-.......-...- .....................

zing, metal cutting, metal reclamation Acute pneumonitis 12-24 hours after exposure

-- .-

Paraquat ! Ingestion of herbicides /Accelerated, chemically induced pulmonary


! f~bros~s

(b) Typical causes of allergic alveoiitis

Dieeaeeloccupatlon Material Caueative agent


--- --
Thermophllic actinomycetes bacteria
(5accharopolyspora rectivirgula,
Bagassosis Thermoactinomyces species)
1 Mushroom workers / Compost

.-A/ Contam~natedwater
+
- Also K/ebsieIla oxytoca, amoebae

1
.--- . . .

Pgeon fanc~er's(breeder's) lung


-- -
Avtan protelns -
- -- I
L
!
Farmers, sawm~ll,tobacco, esparto Fungal contamlnatlon o f mater~als j Primarily Aspergiilus species
grass and brewery workers
- --.
- .

Household ngal infestations of damp walls and woodwork Multiple fungal species
-.

- o t h e r bacterial causes ntamination of water, wood shavings etc. Bacillus subtilis, Klebsieiia, Epicoccum
! nigrum, non-tubercular mycobacteria
The most common form of occupational and environmental lung Mesothelioma (Chapter 28) can develop up to 40 years after
disease is asthma (Chapters 21 and 22). See Chapter 36 for lung light exposure. and is invariably fatal. Milder forms of asbestos-
cancer. induced pleural disease produce dyspnoea and restrictive defects
coupled with pleural thickening, pleural plaques (distinctive par-
Response to acute lung irritants tially calcified lesions generally on parietal pleura) or effusions, with
Inhaled irritants (Fig. 31a) cause exacerbation of asthma and scattered fibrotic foci. N o treatment is effective for asbestos-related
chronic obstructive pulmonary disease (COPD), coughing and dys- disease, as the stimulus remains in the lungs. Asbestos-related
pnoea through activation of irritant receptors (Chapter 1 I), and ir- lung cancer is discussed in Chapter 36.
ritation of mucus membranes. Highly soluble agents (e.g. ammonia, Silicosis is a fibrotic disease caused by inhalation of silica. Occu-
sulphur dioxide) cause immediate irritation in the upper airways, pations at risk include mining, quarrying, stoneworking, manufac-
whereas less soluble agents (e.g. chlorine, ozone) favour deeper pen- ture of abrasives, foundry work and glassworking. Its prevalence in
etration to alveolar epithelial cells, which are particularly suscepti- developed nations is low. Silica is very toxic to macrophages and
ble to injury. High concentrations lead to extensive lung injury, thus highly fibrogenic. Chronic silicosis (over decades) is character-
primarily by damage to epithelium, consequent inflammation and ized by silicotic nodules of collagen around a cell-free core, first
pulmonary oedema. Development of acute respiratory distress developing in hilar lymph nodes. In acute silicosis due to heavy ex-
syndrome (ARDS) is common, and treatment is similar (Chapter posure, severe dyspnoea may develop over months. The clinical fea-
35). Some patients who initially recover from moderate or severe tures of silicosis are similar to PMF.
exposure may subsequently develop bronchiolitis obliterans Berylliosis is caused by systemic poisoning by inhaled beryllium
(obliteration of bronchioles by fibrous growth) after 2-8 weeks. (electronics, high tensile alloys), giving rise to symptoms similar to
Although steroids may slow progression, prognosis is often poor. sarcoidosis (Chapter 28). Industrial regulations have made this a
rarity.
Inhalation of mineral dusts (pneumoconiosis)
C o a l workers' pneumoconiosis (CWP) is caused by inhalation of Inhalation of organic material
coal or carbon dust. In simple CWP, the upper lobes of the lung Extrinsic allergic alveolitis (or hypersensitivity pneumonitis) is
contain small (<4mm), round opacities (coal macules) consisting of a diffuse inflammatory disease of small airways and alveoli caused
dust, dust-laden macrophages and fibroblasts. These may enlarge to by allergens, primarily microbial spores, that are small enough to
coal nodules, which are fibrosed. Weakening of bronchiolar walls reach the alveoli (Fig. 31b). The most common example is farmer's
leads to focal emphysema, which together with macules is charac- lung, caused by dust from mouldy hay or plants contaminated with
teristic of CWP. Simple CWP is often described as symptom- thermophilic actinomycetes bacteria, which thrive in warm moist
less, with no discernible change in lung function. It can, however, conditions. Typically, symptoms occur several hours after exposure,
develop into progressive massive fibrosis (PMF), with black fibrotic and include fever, dyspnoea and cough. Although early removal
masses from 1 cm to several centimeters in diameter that may have of antigen exposure results in rapid recovery, continuous exposure
necrotic cavities. Obliteration and disruption of airways results in leads to progressive interstitial fibrosis (Chapter 27), with infiltra-
emphysema. Patients show irreversible airflow limitation, loss of tion of inflammatory cells and formation of granulomas (chroni-
lung volume and elastic recoil, and reduced D,co, with significant cally inflamed tissue masses characterized by multinucleate giant
breathlessness on exertion. Treatment is limited, and similar to cells). Patients present with dyspnoea, restrictive defects and de-
other progressive fibrotic diseases (Chapter 27). Caplan's syn- creased D,co. Fluffy nodular shadowing or ground glass opacity
d r o m e is a nodular form of CWP associated with the defective im- may be shown in X-ray, with honeycomb lung (Chapter 27) in severe
munology of rheumatoid disease; it may also occur with asbestosis cases. Detailed histories are required to establish antigens, with con-
or silicosis. firmation by precipitating antibodies in serum. Bronchiolar lavage
Asbestos is a fibrous mixture of silicates that is highly resistant to or even lung biopsy may be required where diagnosis is not clear.
degradation. The fibres are 1-2pm wide, but up to 50pm (blue as- Management centres on abolishing antigen exposure. High-dose
bestos; crocidolite) or 2cm (white asbestos; chrysotile) long. They oral corticosteroids can regress early disease, but established disease
are thus easily trapped in the lung. Blue asbestos is far more dan- with fibrosis is irreversible and can progress to respiratory failure.
gerous. Regulations have reduced exposure since the 1980s, but the Differential diagnosis includes asthma (Chapter 21), sarcoidosis
presence of asbestos in buildings and the long interval between ex- (Chapter 27). viral and mycoplasma pneumonias (Chapter 32) and
posure and disease development mean that asbestos-related disease mycobacterial infections.
will be encountered for some time. Asbestos bodies (protein- Byssinosis occurs in workers handling raw cotton, flax and
covered fibres) in the lungs are indicative of exposure, but not dis- hemp. It is characterized by chest tightness, cough and/or shortness
ease. The type and extent of disease largely depend on exposure. of breath on the first day back at work, with recovery as the week
Asbestosis is a fibrous lung disease developing up to 10 years after progresses. It is primarily due to acute bronchoconstriction, pos-
heavy exposure. Patients present with progressive dyspnoea, basal sibly related to contaminating bacterial endotoxins; byssinosis is
crackles on inspiration, and sometimes finger clubbing. There is not induced by processed cotton or linen (flax). Long-term expo-
a restrictive lung function defect and reduced D,co, with diffuse sure causes a disease similar to chronic bronchitis (Chapter 23),
streaky shadows on X-ray and thickening of visceral pleura; honey- with chronic productive cough, progressive decline in lung function
comb lung is often prominent in the lower lobes. Prognosis is poor. and disability.
32 Pneumonia

(a) Pathogenesis of hospital-acquired pneumonia

Swaliowing difficulty, vomiting

(b) Seasonal peak of respiratory pathogens

Respiratory syncytial viru

(c) Radiographic features of pneumonia

Chronic heart, lung and renal 1 support


, Postoperative patients:
Immunosuppression: treatment, Obesity
disease, HIV infection Male sex
Aspriation: alcohol dependency, Prolonged ventilation
epilepsy and dysphagia Cigarette smoking
Recent influenza or viral illness Upper abdominal surgery
Environmental factors: Thoracic surgery
Bird (psittacosis) and farm Prolonged surgery
animal (Q fever) exposure
Travel abroad (tularaemia,
paragonomiasis etc)

t o air conditioning
Pneumonia is defined as an acute lower respiratory tract illness, Clinical: age >60 years, respiratory rate >30/min. diastolic
usually due to infection, associated with fever, focal chest symptoms blood pressure <60mmHg, new atrial fibrillation. confusion.
(signs) and recently developed shadowing on CXR. It is caused by a Laboratory: raised serum urea >7mmol/L, serum albumin
wide variety of microorganisms and pathological insults, some of <35 g/L, hypoxaemia (Po2<8 kPa), leucopenia (white cell count
which are shown in Table I. <4x 109/L), leucocytosis (>20x 1OY/L),multilobar involvement
and bacteraemia.
Classification Complications of pneumonia include pleural effusion/
Classification of pneumonia by radiographic appearance (lobar empyema (Chapter 28), lung abscess, respiratory failure (Chapter
pneumonia or bronchopneumonia) gives little practical informa- 20), bacteraemia and meningitis, and rarely pericarditis. myocardi-
tion about cause or appropriate treatment. Classification by micro- tis or cholestatic jaundice.
biological pathogen (streptococcal, viral, fungal) most accurately Investigations: early identification of likely pathogens aids man-
defines the course, clinical picture and outcome, but is not practical agement, although no microorganism is isolated in 33-50% of pa-
in the clinical situation where microbiological diagnosis is often tients, due to previous antibiotic therapy or inadequate specimens.
delayed. The following classification scheme is widely accepted: Chest X-rays (Fig. 32c) and CT scans aid diagnosis and detect com-
1 Community-acquired pneumonia is common, and one in plications. Routine blood tests: white cell count (>I5 x 109/L=
1000 of the population require admission for pneumonia each year. pneumonia, <3x 109/L = poor prognosis), haemolysis and cold
The most likely pathogens in these cases are Streptococcus agglutinins may indicate mycoplasma infection. Liver function
pneumoniae (60-75%), Mycoplasma pneumoniae (5-1 8%), injluenza tests may indicate Legionella or Mycoplasma infection. Blood
A (7-8%), Haemophilus influenza (4-5%) and Legionella (2-5%). gases identify respiratory failure. Blood cultures, sputum for
Mortality is -5-1 8% of admissions. acid-fast bacillus (AFB), Gram stain, culture and sensitivity
2 Hospital-acquired (nosocomial) pneumonia is any lower res- identify the pathogen. Serology detects Legionella and Myco-
piratory tract infection developing two or more days after hospital plasnza pneumonia. Bronchoscopy provides specimens for micro-
admission and not apparent at admission. Pneumonia is the biology. Pneumococcal antigen detection (serum, urine) and
3rd commonest nosocomial infection after wound and urinary urine enzyme-linked immunoassay (ELISA) for Legionella
tract infections, and occurs in -4 per 1000 hospital patients; its infection are occasionally useful.
pathogenesis is outlined in Fig. 32a. Likely pathogens are Gram-
negative bacilli (65-70%) including Klebsiella. Psc~udomonas, Management
Escherichia coli, Proteus and Staphylococcus (10-1 5%). Mortality Supportive measures include O2by face mask to maintain Pao2> 8
is -20-30% (50% with bacteraemia). kPa or O2 saturation > 90%, and intravenous fluids and inotropic
3 Other pneumonias include aspiration and anaerobic pneumo- drugs may be required for haemodynamic stability. Ventilatory
nia, pneumonia in the immunocompromised patient (e.g. Pneu- support: continuous positive airway pressure (CPAP). non-invasive
mocystis carinii in human immunodeficiency virus (HIV) patients, positive pressure ventilation (NIPPV), or mechanical ventilation
Chapter 34) and recurrent pneumonia due to cystic fibrosis or may be required with respiratory failure (Chapters 20 and 38).
bronchiectasis (Chapter 30). Antibiotic therapy: as microbiological results may not be avail-
able for 24h, antibiotic therapy is initially given 'blind'; appropriate
Risks therapy depends on how the pneumonia presents, and must be
Common risk factors are shown in Table 2, and are affected by amended as soon as the microbiological results and antibiotic sensi-
season and locality. Pneumonia is more common in winter due to tivities become available.
increased incidence of viral infections (influenza, respiratory syncy- Community-acquired pneumonia: In the hospitalized patient,
tial virus): seasonal peaks of common respiratory pathogens are antibiotic therapy must cover streptococcal pneumonia and atypi-
shown in Fig. 32b. Variations may occur due to 3-yearly epidemics cal pneumonias (Mycoplasrna, Legionella). Erythromycin with or
of Mj~coplasrnapneumoniae and irregular epidemics of influenza without cefuroxime is recommended. Staphylococcal infection is
Epidemics of Legionella pneumophila (Legionnaires disease) and common after influenza, and Haernophilus influenzae is associated
Coxiella burnetii (Q fever) are usually localized. with chronic lung disease.
Hospital-acquired pneumonia: antibiotic therapy for
Gram-negative bacilli and staphylococci is required. Broad-
Diagnosis spectrum antibiotics, including combinations of cephalosporins.
General features include malaise, fever, rigors, myalgia, cyanosis, quinolones, gentamicin and antistaphylococcal drugs, are
tachycardia and a respiratory rate >20/min. Specific symptoms in- recommended.
clude dyspnoea, pleuritic chest pain, cough, wheeze and sputum Aspiration pneumonia: anaerobes must be covered in addition
production, with focal signs of dullness, crepitations, bronchial to other microbes (add metronidazole).
breathing and pleuritic rub. In the very young or old and those with Pneumonia in the immunocompromised: broad-spectrum an-
atypical pneumonia (Mj1coplasma, Legionella, psittacosis), non- tibiotics are required after chemotherapy. HIV-infected patients
respiratory features may predominate (headache, confusion, diar- may require specific therapy for viral (cytomegalovirus) and proto-
rhoea, rash, cyanosis). zoal infections; Pneumocystis carinii pneumonia is treated with
Severity should be assessed. In community-acquired pneumo- steroids and high-dose co-trimoxazole (Septrin).
nia, the following features are associated with increased mortality,
and indicate the need for transfer to a critical care unit:
33 Pulmonary tuberculosis

Abscess T B with cavitation I


G ~ a ncells
t
(mult~nucleate)
Central
caseat~on
('cheesy pus')

Lymphocytes

Acd fast
,&-I bacilli

'Primary complex'

Tuberculosis

Neurological TB

Fever and we~ghtloss Cerebral abscess


1 (c) P u l m o n a ~complications
Nlght sweats Nerve lesions

Mycetoma Apical cavities


I I ,Pneumothorax

Miliary T B
Malaise
Weight loss
Low-grade fever

Epidemiology and aetiology lus, Mycobacterium tuberculosis. The inhaled bacillus infects well-
Worldwide, tuberculosis (TB) affects 10 million people and causes 3 ventilated, poorly perfused upper lung lobes subpleurally. A granu-
million deaths each year. In developed countries, it is an uncommon loma forms (Fig. 33a) known as the G h o n focus, and with the
disease affecting approximately one per 10000 population. Pul- enlarged hilar lymph nodes draining the affected lung is known as
monary tuberculosis is most common in Asian, Chinese and West the 'primary complex' (Fig. 33a). This process occurs over 3-8
Indian people. Airborne transmission and close contact with an weeks, and is accompanied by the development of an inflammatory
infected person spread the disease. Those who are elderly, maln- reaction to the injection of tubercular protein (tuberculin) into the
ourished or immunosuppressed (HIV infection, diabetes mellitus, skin, which can be used as a diagnostic test (Heaf or Mantoux
corticosteroid therapy, alcoholism, intercurrent lymphoma) are test). Complete healing usually follows. with fibrosis and calcifica-
more susceptible. Improved housing and nutrition reduce the inci- tion of the Ghon focus and immunity to further infection.
dence of tuberculosis. Post-primary pulmonary tuberculosis occurs if the Ghon
focus fails to heal due to poor host defences, or following reac-
Pathogenesis tivation at a later date. It is potentially fatal. Local dissemination
Primary pulmonary tuberculosis is caused by the acid-fast bacil- causes tuberculous pneumonia and pleural effusions. Blood-
borne spread may affect the meninges or individual organs. In a few Prevention
cases, widespread infection involves many tissues and is known as Vaccination of non-immune subjects, assessed by Heaf testing at
miliary tuberculosis. age 12-13, with B C G (bacille Calmette-Guerin), a non-virulent
strain of bovine TB, produces immunity and reduces the risk of pul-
Clinical features monary tuberculosis by 70%.
Primary pulmonary tuberculosis usually occurs at an early age.
Although often asymptomatic with no clinical signs, it may cause Drug therapy
a mild febrile illness, erythema nodosum (painful, indurated shin The prognosis in tuberculosis is good if the patient is not immuno-
lesions) and small pleural effusions. Bronchial compression by compromised. Good nutrition, reduced alcohol consumption and
lymphadenopathy may cause wheeze and occasionally lobar compliance with drug therapy are important factors in successful
collapse followed by late bronchiectasis (Chapter 30). treatment. Uncomplicated pulmonary tuberculosis is treated for 6
Post-primary tuberculosis develops over months, with malaise, months. Initially. at least three drugs are used, to prevent the devel-
anorexia, weight loss, night sweats and a productive cough. Breath- opment of resistant strains. The recommended regime is rifampicin.
lessness, chest pain, haemoptysis and cervical lymphadenopathy pyrazinamide and isoniazid for 2 months, followed by rifampicin
may occur. Clinical signs of pneumonia and pleural effusion are and isoniazid for 4 months. Additional pyridoxine prevents
common, whereas lupus vulgaris (an indolent skin infection) is less isoniazid-induced peripheral neuropathy. Liver function tests
frequent. Miliary tuberculosis presents with a non-specific pyre- should be monitored, as rifampicin and pyrazinamide can cause
xial illness, malaise and weight loss. Sparse clinical signs include liver dysfunction. If drug resistance is suspected (TB recurrence in a
hepatomegaly and choroidal tubercles in the retina. non-compliant patient) then a four-drug regimen (adding ethambu-
tol) may be initiated. When culture results are available, alternative
Investigation drugs replace those to which the mycobacterium is not sensitive.
Blood tests may detect anaemia, decreased sodium and increased Ethambutol (monitor colour vision for optic neuritis), strepto-
calcium. mycin (monitor plasma levels to avoid hearing impairment) or
Mantoux test: strongly positive in post-primary pulmonary ciprofloxacin may be used.
tuberculosis (>5 mm skin induration with 10 units of intradermal In some organs (e.g. bone), tuberculosis is treated for longer,
tuberculin injection). The test is often negative in miliary TB (re- often with additional drugs. In meningeal or cerebral tuberculosis, a
duced host response) and HIV-infected patients (reduced cellular four-drug regime for 12 months with additional steroids is recom-
immunity). mended. to ensure adequate brain penetration and to prevent
Heaf test (screening test): A ring of six pinpricks is made cranial nerve compression by meningeal scarring.
through a tuberculin solution placed on the forearm. No response
at 4-7 days (grade 0) demonstrates lack of immunity; four to six Complications
discrete nodules (grade I) or a ring formed by coalition of the Reactivation of old tuberculous scars may occur when a patient is
six pinpricks (grade 2) indicate immunity. A single nodule formed immunocompromised (Fig. 33c). Chemoprophylaxis with isoniazid
by infilling of the ring (grade 3) represents recent contact or early is often given before immunosuppressive treatment (chemotherapy,
tuberculous infection, and a nodule >5-7mm with surface vesicles organ transplantation). Bronchiectasis and lung cavities with sec-
or ulceration (grade 4) suggests infection. ondary fungal infections (mycetoma), cranial nerve lesions and
Microbiology: the acid-fast bacilli may be detected in sputum or renal tract obstructions may develop due to scarring associated with
lung washings using the Ziehl-Neelsen stain. However, bacilli are healing after tuberculosis. Surgery may be required to correct these
slow-growing, and culture and drug sensitivities take 4-6 weeks. defects. Non-compliance or inadequate treatment results in multire-
Bone marrow or cerebrospinal fluid (CSF) culture may confirm the sistant strains of mycobacteria that may be very difficult to eradi-
diagnosis of miliary tuberculosis. cate. Compulsory supervision and isolation of these patients may be
Histopathology: pleural aspiration with biopsy confirms tuber- required.
culosis in -90%) of patients with pleural effusions. Liver biopsy will
isolate miliary tuberculosis in -60% of cases. Contact tracing
Chest radiography (Fig. 33b): upper lobe shadowing is sugges- Community health services must be notified when a patient is
tive of tuberculosis. Apical cavities, pleural effusions and pneu- diagnosed with TB, to trace contacts and prevent spread of the
mothoraces may occur. In miliary tuberculosis, widespread small disease. Contacts are screened with a Heaf test. If this suggests a
nodules (2-3mm diameter) are diffusely spread throughout the risk of infection, then chest radiography and appropriate follow-up
lungs (miliary shadowing), and are easily missed. is arranged.
34 Respiratory infections in HIV infection

(a) Common respiratory infections in HIV-infected individuals (b) CXR of pneumocystis carinii pneumonia, showing diffuse
alveolar infilfcrate
Microbe Typical CD4+ cell count (per mm3)
-
I

I Bacteria 1 I
I 5. pneumonlae / Ary I
Haemophius species / Any
I? aeruginosa ~ 2 0 0
L. pneumophila

Mycobacteria
!
Ii
Any

M. tuberculos~s Any (extrapulmonay o r atyp~cal


, presentat~onwhen 1 2 0 0 )
Dlssemlnated MAC / ~ 5 0
--

TZo0
- --
Fungi
Pneumocystls carin11
Cyptococcus neoformanl 1200

Respiratory compl~catlons oi- human ~mmunodefic~encyvirus Bacterial pneumonias (Chapter 32) are extremely common In
(HIV) infection are related to the degree of immunosuppression patients with HIV infection just as in immunocompetent patients.
and the development of acquired immune deficiency syndrome Streptococcus pneumoniae and Haeinophilus infiuenzae are most
(AIDS). Highly active antiretroviral therapy (HAART) allows common, but in many communities, Staphylococcus aureus and
expansion of the immune system and has improved the survival of Pseudoinonas aeruginosa are also common. Symptoms, signs and
patients with AIDS, partly by decreasing the rate of respiratory in- radiographs are generally similar to those in immunocompetent
fection. Nevertheless, HIV-infected individuals are at increased risk patients (Chapter 32). The presence of high fever, purulent sputum.
of infection with mycobacteria, Pneurnocystis carinii, or other rapid onset of symptoms or pleuritic chest pain help distinguish
fungi (Fig. 34a). bacterial pneumonia from PCP. Legionella infections are more
Pneurnocystis carinii pneumonia (PCP) remains a common common in patients with HIV infection.
cause of respiratory illness in patients with HIV, despite the wide- Mycobacterial infections from Mycobacterium tuberculosis
spread use of prophylaxis. PCP rarely occurs in Africa, however: it (MTB) or M. avium complex (MAC) are significant concerns in pa-
seems to be a disease of developed countries. PCP may be transmit- tients with HIV. Approximately one-third of HIV-infected patients
ted via inhalation rather than by reactivation of dormant infection. exposed to MTB will develop primary disease, and patients with
Patients with PCP have progressive dyspnoea on exertion, malaise, prior exposure have a 1 O%/year chance of developing reactivation
fever, non-productive cough and hypoxia. In contrast to patients disease. Tuberculin skin tests should be performed on all patients
with other causes of immunosuppression. PCP in the HIV-infected with HIV infection. Induration of >5 mm is consistent with prior
is indolent over weeks. Radiographs usually show diffuse interstitial MTB infection, and requires prophylactic treatment in the absence
infiltrates early and alveolar infiltrates late (Fig. 34b). Spontaneous of active disease. False-negative tuberculin reactions are usual once
pneumothorax (Chapter 29) is common, particularly in patients CD4+ counts are <100/mm3. The clinical manifestations of MTB
receiving aerosolized prophylaxis. PCP rarely occurs in patients infection depend on the degree of immunosuppression. When
with CD4+ cell (Chapter 17) counts >300/mm3, and most patients CD4+ counts are >300/mm3, the clinical presentation is similar
have counts <100/mm3. Diagnosis requires demonstration of cysts to that in patients without HIV infection, including fever. non-
or trophozoites in induced sputum or bronchoalveolar lavage. productive cough and upper lobe cavitary disease. As immunosup-
Trimethoprim-sulfamethoxazole is the most effective therapy. Cor- pression advances, mediastinal adenopathy, diffuse o r miliary
ticosteroids are co-administered to patients with hypoxaemia to pulmonary infiltrates, and extrapulmonary (nodal, bone mar-
avoid respiratory deterioration often observed in the first days of row, genitourinary, central nervous system) involvement become
therapy. more typical. Patients with MTB treated with HAART may devel-
o p worsening systemic symptoms, fever and adenopathy due to culture of the fungus. Blood cultures are usually positive in cases of
immune reconstitution. HIV-infected patients with MTB should re- histoplasmosis.
ceive the standard drug therapy for their community for 29 months. Cytomegalovirus (CMV) is a major cause of pneumonia in pa-
The high prevalence of MTB disease in patients with HIV infection tients immunosuppressed after organ transplantation, but it is not
has contributed to the spread of multidrug resistance in many a major respiratory pathogen in HIV-infected individuals. CMV is
regions. often recovered in respiratory samples by culture or antigen detec-
M A C infection presents as a disseminated systemic wasting tion, but it does not cause primary pneumonia until CD4+ cell
disease in patients with CD4+ cell counts <50/mm3. Patients have counts are very low. More commonly, CMV is found in conjunction
malaise, fever, night sweats, abdominal pain, diarrhea, weight loss with another pathogen. Diagnosis of CMV pneumonitis in an HIV-
or non-productive cough. Anaemia and elevated alkaline phos- infected individual requires evidence of parenchymal invasion or
phatase are the most common laboratory abnormalities. Radi- cytopathological effect in a symptomatic patient without evidence
ographs are often normal, or have nodular infiltrates. The use of of other infection.
prophylactic antibiotics in patients with low CD4+ cell counts is Malignancies involving the respiratory system may be confused
effective in reducing the risk of disseminated MAC infection. with infections in HIV-infected individuals. Kaposi's sarcoma
Other fungal infections: the combined humoral and cellular (KS) and non-Hodgkin's lymphoma (NHL) are the most com-
immunodeficiency due to HIV infection also leads to an increased mon malignancies. KS is a tumour of vascular origin related to in-
risk of infection due to fungi, many which are ubiquitous in the en- fection with h u m a n herpesvirus 8. Patients with lung involvement
vironment. Most fungal infections cause disease predominantly usually have concurrent skin or upper respiratory tract involve-
outside the respiratory tract but respiratory symptoms are not un- ment. Its clinical manifestations range from an asymptomatic
usual. Cryptococcus often has concurrent respiratory manifesta- incidental finding on radiograph to fulminant disease causing respi-
tions with meningitis. Respiratory symptoms are non-specific, and ratory failure. N H L in the lung may present radiographically as
radiographs usually show interstitial or nodular infiltrates. Cultur- nodules, mass(es), or effusions. Symptoms are typical of a systemic
ing fungus or demonstration of cryptococcal antigen in blood and or respiratory illness. NHL typically occurs in patients with ad-
CSF are diagnostic. In endemic areas, histoplasmosis and coc- vanced immunosuppression and CD4+ cell counts <100/mm3.
cidioidomycosis may cause respiratory disease in HIV-infected NHLs are typically aggressive B-cell or Burkitt's lymphoma, sug-
individuals. Both cause a systemic illness with fever, weight loss, gesting a relationship with pre-existing herpesvirus infections. HIV-
dyspnoea, and non-productive cough. Coccidioidomycosis is usu- infected individuals may be at greater risk of developing advanced
ally seen in patients with CD4+ cell counts <300/mm3. Radiograph- lung cancer at young age, with a poor prognosis for survival. Many
ic abnormalities range from diffuse alveolar/interstitiaI infiltrates to of these patients have early HIV infection without a history of
localized nodules/masses or adenopathy. Diagnosis requires stain or opportunistic infection.
35 Acute respiratory distress syndrome (ARDS)

(a) Acute respiratory distress syndrome CXR of trauma-induced ARDS

Pathophysiology Radiology

Loss of
surfactant
and Increased
response
permeabll~ty
sepslsltrauma)

damage
alveolar collapse
(asplrat~onl
and dependent

surfactant Endothelial
with alveolar and alveolar
CT scan of ARDS
collapse cell damage

(b) Ventilator-induced damage

Normal
Surfactant alveol~

Increased damage Damaged


wlth repeated alveol~dlfflcult
alveolar coliapse t o Inflate

Increased mean
alrway pressure (d) VIQ matching

PEEP

Nltrlc oxlde (NO)

Alveolar Pneumothorax Vent~iationaim


recruitment
and preventon Low PIP

Reducesshunt
(c) Common causes of ARDS

D'rect p~lmonar) 1na;rect


- - -- -. -
. -.- . - - - - - - - - - -- - - -- -- - - - - - - ---- - - . . - . - - . . . . -
infective (pneumonla, tuberculosis) i Sepsls Consolidated
Pulmonary trauma / Non-thoraclc trauma lung
Near-drownlng / Burns
TOXIC gas lnhalatlon Haemorrhage, mult~pletransfusion
Smoke Post arrest
NO2, NH, C12 Bowel lnfarctlon
Normal lung
Phosgene Analphyiactlc
Front
Oxygen toxlclty (F,02> 0 8) / Pancreatltls Prone position
lnhalatlon of gastlc contents (pH c 2) / Uraemla, toxlns, eclampsia improves VIQ match More blood supply
8 Drugs (sal~cylates,barbiturates) and mechanics of ventilation a t base
- reduces shunt = Improved VIQ match
A R D S is most simply defined as 'leaky lung syndrome' or 'low sured to assess fluid balance and ensure adequate tissue oxygen
pressure (i.e. non-cardiogenic) pulmonary oedema'. It describes an delivery. Serial blood gas measurements are used to monitor gas
acute, diffuse inflammatory lung injury, often in previously healthy exchange. Early detection of secondary pulmonary infection re-
lungs (Fig. 35a) in response to a variety of direct (i.e. inhaled) or in- quires microbiological examination of sputum or bronchoalveolar
direct (i.e. blood borne) insults. washings. Radiological: serial CXRs identify progression of dif-
The internationally agreed criteria for diagnosis of ARDS are: fuse bilateral pulmonary infiltrates. Similarly, early computed
1 Severe hypoxaemia, Pao21FIo2< 200, (+/-PEEP) e.g. tomography (CT) scanning can identify diffuse patchy infiltrates
P,o, (55 mmHg)/FIo2(80% inspired 0,)= 55/0.8=(75). with dependent consolidation; later scans reveal pneumotho-
2 Bilateral diffuse pulmonary infiltrates o n chest X-ray. races, pneumatoceles and fibrosis.
3 Normal o r only slightly elevated left atrial pressure (pul-
monary artery occlusion pressure <I 8 mmHg). Management
Acute lung injury (ALI) is the precursor to ARDS. Apart from The key to successful management of ARDS is to establish and
a lesser degree of hypoxaemia (Pao21F,02<300), the criteria for treat the underlying cause. In the early stages, oxygen therapy
diagnosis are the same. and physiotherapy may suffice. With progressive respiratory failure,
non-invasive ventilation -with continuous positive airway pressure
Epidemiology and prognosis (CPAP) or non-invasive positive pressure ventilation (N1PPV)-or
The incidence of ARDS is -2-8 cases per 100000 population per full mechanical ventilation and high-inspired oxygen concentra-
year, but its precursor ALI is much commoner. ARDS mortality is tions may be required to maintain adequate ventilation and oxy-
generally high (>SO%) but is determined by the precipitating condi- genation. The high airways pressures needed to achieve normal
tion (-35% for trauma, -60% for sepsis, and -80% for aspiration tidal volumes during mechanical ventilation often result in lung
pneumonia). Age (>60 years) and sepsis are also associated with damage (barotrauma), including pneurnothorax and lung cysts.
increased mortality. Early diagnosis and treatment may improve This ventilator-induced lung injury and oxygen toxicity (F,02>0.8)
outcome. The cause of death is multiorgan failure (MOF), usually must be prevented, as these contribute to mortality and MOF.
due to a combination of tissue hypoxia and overwhelming sec- The basic principles of mechanical ventilation are to limit
ondary infection. Less than 20% of patients die from hypoxaemia pressure-induced damage, optimize oxygenation and avoid
alone. circulatory compromise (reduced cardiac output and blood pres-
sure due to high intrathoracic pressures; see also Chapter 38). A
Pathogenesis (Fig. 35a) and causes (Fig. 3 5 ~ ) 'protective lung ventilation strategy' of low tidal volumes (6 mL/kg)
During the acute inflammatory phase of ARDS, cytokine- and low peak inspiratory pressures (<30cmH20) reduce lung dam-
activated neutrophils and monocytes adhere to pulmonary age, complications and mortality. Alveolar recruitment (of col-
endothelium or alveolar epithelium, releasing inflammatory media- lapsed alveoli) is achieved with high positive end-expiratory
tors and proteolytic enzymes (Chapter 17). These damage the in- pressures (PEEP >IOcm H20) or long inspiratory-to-expiratory
tegrity of the alveolar-capillary membrane, increase permeability times. The CO, retention ('permissive hypercapnia') resulting from
and cause alveolar oedema. Reduced surfactant production causes this strategy of low tidal volume ventilation can be tolerated for
alveolar collapse and hyaline membrane formation. The loss of long periods.
functioning alveoli and ventilation/perfusion mismatch leads to Excessive fluid loading must be avoided, as this increases
progressive hypoxaemia and respiratory failure. The subsequent the alveolar flooding characteristic of ARDS. The aim must be to
late healing fibroproliferative phase results in progressive pul- maintain adequate perfusion of other organs whilst using the
monary fibrosis and reduced compliance (stiff lungs). Associated lowest possible left atrial pressures. In the acute situation, diuretics
pulmonary hypertension is partially due to activation of the coagu- may be essential to correct hypoxaemia by reducing extravascular
lation cascade, with pulmonary capillary microthrombosis and lung water. Thereafter, combinations of pulmonary and systemic
regional hypoxic vasoconstriction. vasodilators (before and after load reduction of the left heart),
inotropes and vasoconstrictor agents may be used to achieve ade-
Clinical features quate cardiac output and perfusion pressures at low left atrial filling
The acute inflammatory phase lasts 3-10 days and results in pressures.
hypoxaemia and MOF. It presents with progressive breathlessness, Essential general measures include good nursing care, physio-
tachypnoea, central cyanosis, hypoxic confusion and lung crepita- therapy, nutrition and infection control. Reducing fever (shivering)
tions. These symptoms and signs are in no way diagnostic and are and controlling anxiety with sedation decreases metabolic demand.
shared with many other pulmonary conditions. During the later N o drug therapy has been consistently beneficial in early ARDS,
healing, fibroproliferative phase, pulmonary fibrosis (lung scar- including steroids, anti-inflammatory agents, anticytokines or sur-
ing) and pneumothoraces (Chapter 29) are common. Secondary factant therapy. However, 7-10 days after onset, steroid therapy
chest and systemic infections complicate both phases. may prevent the development of subsequent pulmonary fibrosis.
Inhaled nitric oxide and nursing the patient in the prone position
Investigation improves gas exchange by increasing perfusion to ventilated areas
Monitoring: routine measurements include temperature, respira- of lung, but no survival benefit has been demonstrated (Fig. 35d).
tory rate, O2 saturation and urine output. In addition, the arterial Extracorporeal membrane oxygenation (ECMO) techniques to
and central venous pressures, the cardiac output and occasionally oxygenate blood or remove CO, are effective in children, but the
the left atrial pressure (using a pulmonary artery catheter) are mea- benefit in adults has not been established.
36 Lung cancer

(a) Mass on CT: A>3cm spiculated mass is seen in (d) Staging system for non-small cell lung cancers
upper lobe of t h e right lung

IA
I0
IIA
z
(tumour) (node) (metastasis)

f:5 3 c m without division


T2: >3cm, or invasion of main bronchus
>2cm from main carina, o r invades
110 visceral pleura, or bronchus causing
obstruction
T3: Invades chest wali or pleura, or main
bronchus <2cm from main carina
T4: Invades adjacent structure,
malignant effusion, satellite nodules

(b) Fibreoptic bronchoscopy showing tumour IIIA NO: No lymph node metastasis
invading bronchus N1: lps~lateralhilar lymph nodes
1110 N2: I p s ~ a t e r am
i e d ~ a s t n aor
l subcar~nal
lymph nodes
N3: Contralateral, scalene or supra-
clav~cularlymph nodes
--
MO: No d ~ s t a n m
t etastas~s
MI: Any d ~ s t a n metastasis
t

(e) Survival for non-small cell cancer

(c) CXR showing squamous cell tumour in hilar region

Post-resection
p a t h o l o g i c a l diagnosis

IIB
lllA

I I I A Inoperable
y F } c l i n i c a i diagnosis
"
0 Years
More people die in the US and Europe from lung cancer than from antigens. SC carcinoma is associated with most paraneoplastic
breast, prostate and colon cancer combined. Furthermore, the syndromes including Cushing's syndrome. syndrome of inapprop-
number of cases is likely to increase in the next 25 years due to con- riate secretion of anti-diuretic hormone (SIADH). Lambert-Eaton
tinued use of cigarettes. particularly in women. Lung cancer has syndrome, cerebellar ataxia, o r idiopathic orthostatic hypotension.
a worse prognosis than other common cancers, with an overall Squamous cell cancer may cause hypercalcaemia from release of
5-year survival of 13%. parathyroid hormone-related peptide.
Physical findings in the lung are related to disease extent. Small
Risks parenchymal nodules are undetectable by physical examination.
Cigarette smoking accounts for the vast majority of lung cancer Focal findings may be due to atelectasis, airway invasion, pleural
cases. Risk is directly related to the duration and number of ciga- effusion (Chapter 28) or supraclavicular adenopathy. lnvasion
rettes smoked, age of initiation, depth of inhalation and levels of tar of adjacent structures may cause superior vena cava syndrome
and nicotine. In heavy smokers (>20 pack-years) the lifetime risk (obstruction), Horner's syndrome (autonomic overactivity), or
of lung cancer is 10"/0, 10-30 times greater than for lifelong non- brachial plexopathy. Digital clubbing or hypertrophic pulmonary
smokers (<0.3'%). After quitting cigarettes, risk gradually declines osteoarthropathy may be present.
over 15 years, but remains 2-5 times greater than in non-smokers.
Passive smoking in non-smokers may increase the risk by -1.5%. Evaluation
Asbestos exposure is the most common occupational risk for Evaluation of patients with suspected lung cancer should in-
lung cancer (Chapter 31). Tobacco smoke is synergistic with as- clude demonstration of malignancy, staging and suitability for
bestosis, increasing the relative risk to 6-60 times that of a non- therapy. Radiographs provide information regarding the size and
smoker. R a d o n gas, found naturally in rocks, soil and ground location of the tumour. benign calcification, involvement of adja-
water, may also increase risk. cent structures, atelectasis, pleural effusion, and adenopathy (Fig.
36c). Computed tomography (CT) scans are superior to plain
Classification X-rays. If a focal lesion does not change in two years, it is unlikely to
Lung cancers are divided pathologically into small ceIl (SC. be malignant. Positron emission tomography (PET) scanning
20-30% of total) and non-small cell (NSC. 70--80% of total) types. has a high sensitivity for distinguishing benign from malignant nod-
N S C types are grouped due to their similar biology, treatment and ules and for detecting nodal or distant metastases.
prognosis, and include squamous cell (30%). large cell (l5%), and Staging is assessment of the extent of the tumour, and largely de-
adenocarcinoma (33%), which is increasing in prevalence, espe- termines treatment options and prognosis. Separate staging systems
cially in women. Adenocarcinomas typically present as a peripher- are used for SC and NSC cancers. S C cancer is staged as either
al nodule (<3cm) or mass (>3 cm); they are the most common type limited or extensive disease. Limited disease describes tumour
in non-smokers, and mainly arise in areas of pulmonary scarring. confined to one hemithorax, including malignant pleural effusion
Bronchoalveolar cell carcinoma is an adenocarcinoma variant with and supraclavicular lymph node metastasis. Extensive disease
low metastatic potential. Squamous cell carcinomas arise from describes metastatic spread beyond the hemithorax. SC cancer is
the bronchial epithelium, and generally present as a central mass generally an incurable disease. Standard therapy for limited disease
with tumour visible in the airway (Fig. 36a,b), often with symptoms (33%) is combination chemotherapy and radiotherapy, with re-
due to local tumour invasion (cough, haemoptysis, chest pain and sponse rates approaching 90%; median survival with therapy is -1 8
hoarseness). Large cell carcinoma is undifferentiated, and lacks months. Standard therapy for extensive disease (66%) is chemother-
the histological features of adenocarcinoma or squamous cell carci- apy. The response rate is -70%, treatment prolonging median sur-
noma; it generally presents as a large peripheral mass, often with vival from -3 months to -1 year.
metastases. S C carcinomas arise from neuroendocrine cells in the N S C cancer staging is based on the tumour (T). node (N), and
bronchial submucosa, and typically present as a central mass with metastasis (M) classification system (Fig. 36d). T 3 tumours invade
lymph-node enlargement. These are aggressive tumours that invade thoracic structures that are potentially resectable, and T4 tumours
lymphatics and blood vessels. Nearly all have metastasized at include malignant effusions or tumours invading non-resectable
diagnosis. structures. Summation of T N M categories determines the stage
of disease and treatment, and predicts survival (Fig. 36e). In func-
Presentation tional patients with stage I or I1 disease and adequate pulmonary
Less than lo'%of lung cancers are discovered incidentally in asymp- reserve (postoperative FEV, >800mL), surgical resection is
tomatic patients. Most patients are 50-70 years of age, with non- optimal. Some patients with stage IIIA disease are surgical candi-
specific symptoms including new unresolving cough, haemofit'ysis, , dates. Patients with stage IIIB or IV disease are not candidates for
chest pain, hoarseness, dyspnoea on exertion, malaise and weight curative resection. Unresectable disease is generally treated with
loss. Symptoms due to haematogenous extrathoracic metastasis chemotherapy and radiation therapy, or radiation alone. Stage
to bone, liver, bone marrow, adrenals and brain are present in IV disease is incurable (median survival 6-12 months). Treatment
around one-third of patients at diagnosis. options are palliative. Painful bone metastases, brain metastasis, or
Paraneoplastic syndromes- signs or symptoms associated airway obstruction may improve with directed therapy. The benefit
with lung cancers that are not related directly to metastatic of aggressive chemotherapy for patients with advanced disease
tumour -may precede radiographic demonstration. They may be is modest. Platinum and taxol-based chemotherapy regimens are
due to secretion of hormones or hormone-like substances from currently most common for NSC cancer.
tumours, or serum autoantibodies (e.g. anti-Hu) related to tumour
37 Sleep-disordered breathing

-- -
Polysomnogram
(a) Normal (b) Obstructive sleep apnoea (c) Central sleep apnoea
l r l ' l r ' ' ' r l l ' l ' ' " ' r ~ r ' ' ' l ~l l~" '" L
" "l l " l " ' l l ' l r ' r l " l l ' l " " r c " l l l l r r r " ' l r l l l l ' r l l l ' ~ l ~ l T ~ r r ~ ' ~ l ' ~ ~ ~

EOG (L) 0
EOG (R)

C3-A2

C3-01

ECG

EMG (L)
Desaturation
5a0,
Airflow
(cannula)

No airflow
Aidlow
(thermistor) --s,w=*-
Thoracic

Abdomen --J\iVu--"V!J4- 1
EOG = electrooculogram from both eyes (eye movement), C3-A2, C3-01 = electroencephalogram from standard locations,
ECG = electrocardiogram, E M G = electromyogram of leg (movement), SaO, = blood 0, saturation,
Airflow = measured by cannula and thermister placed in airflow, ThoraciclAbdomen = breathing movements

Sleep-disordered breathing is a common problem, with a vast po- Obstructive sleep apnoea (OSA) is characterized by the ab-
tential for improvement in the patient's quality of life. Normal sleep sence of airflow with continued respiratory effort (Fig. 37b). Up to
(Fig. 37) consists of rapid eye movement (REM) and non-rapid eye 4% of the general population and approximately 90% of patients
movement (NREM) sleep. REM sleep normally comprises about with sleep apnoea have OSA. OSA is far more common in
25-30% of total sleep, and is characterized by an awake- males than females, and is associated with alcohol consumption, in-
pattern electroencephalography (EEG), voluntary muscle atonia creasing age, obesity. increased neck circumference, hypertension
and dreaming. NREM sleep is divided into light (Stages 1,2) and and hypothyroidism. Obstruction typically occurs in the upper air-
deep (Stages 3,4) sleep. Ventilatory drive is normally diminished in way and pharynx, and is related to the normal decreases in upper
REM compared to NREM sleep, causing a slight fall in P,02 and a airway muscle tone and increased airway resistance that occur in
rise in P,co2, the magnitude of changes depending on starting con- deep or REM sleep. It may also occur in children with inflamed or
ditions. Normal duration and architecture of sleep results in the ab- enlarged tonsils or adenoids. These factors, in conjunction with
sence of daytime hypersomnolence, which can be quantified by individual anatomy, extraluminal tissue and posture. result in air-
sleep latency or wakefulness testing. Daytime hypersomnolence due way collapse/closure during inspiration, when airway pressure is
to sleep deprivation and sleep fragmentation may lead to an overall reduced. Apnoea resolves with arousal and restoration of muscle
decrease in quality of life, including increased risk of motor vehicle tone. Although many hundreds or thousands of episodes of apnoea
accidents and cognitive dysfunction. It may be due to a variety of and arousal may occur each night, patients are often unaware of
causes. including obstructive sleep apnoea and more rarely cen- them; their sleep partners commonly report loud snoring, snort-
tral sleep apnoea. ing or apnoea. Patients develop daytime hypersomnolence, loud
Sleep-disordered breathing is diagnosed using polysomnogra- snoring and weight gain. Their physical examination may demon-
phy (Fig. 37), which records the EEG for sleep patterns, movements strate pedal oedema, nasal congestion, enlarged tongue, shallow
of abdomen and thorax to assess breathing, oronasal flow and palate, enlarged uvula or retrognathia. Most patients have normal
oximetry for O2saturation. arterial blood gases and haemoglobin. In chronic obstructive pul-
monary disease (COPD) patients, nocturnal hypoxia can be severe cations. O2 alone may decrease o r eliminate hypoxia, but will not
even with mild OSA, and gas exchange is already compromised and treat obstruction o r eliminate arousals.
they may be already hypoxaemic with a low O2 saturation. Central sleep apnoea (CSA) is characterized by cessation of air-
In OSA, polysomnography reveals repeated episodes of OSA or flow during sleep without evidence of respiratory effort, and is due
hypopnoea (reduced flow with oxygen desaturation or arousal), to a loss o r inhibition of central respiratory drive (Fig. 37c).
which terminate with arousal (Fig. 37b). These episodes are quanti- Patients with CSA may be subdivided into those with daytime
fied by the apnoea plus hypopnoea index (AHI, episodeslh). hypercapnia or normocapnia. CSA with daytime hypercapnia is
Normal sleep may have an AH1 < 10. Severe OSA usually has usually due to central alveolar hypoventilation, neuromuscular
an AHI>40. A m~norityof patients with very severe OSA may disease or chest wall disease (e.g. kyphoscoliosis). Central alveolar
de-velop obesity hypoventilation syndrome (Pickwick syn- hypoventilation may be primary (Ondine's curse) or secondary to
drome). Obstructive episodes can cause pulmonary hypertension brainstem disease (see Chapter 11). Neuromuscular causes include
(Chapter 24) from hypoxic pulmonary vasoconstriction. Systemic muscular dystrophy, phrenic nerve dysfunction, myositis (muscle
blood pressure increases during apnoeas, possibly due to sympa- inflammation) and myasthenia gravis. Patients with a CNS cause
thetic stimulation, and LV afterload increases during obstructive for hypoventilation ('won't breathe') may benefit from a respiratory
apnoeas due to the marked fall in pleural pressure during episodes. stimulant. Patients with weakness or chest wall disease ('can't
There is a strong association between OSA and daytime systemic breathe') benefit from assisted mechanical ventilation. Positive
hypertension, although causality has not been established. pressure ventilation with a nasal or face mask is most commonly
Therapy for OSA requires relief of obstruction. Moderate prescribed (Chapter 38). Tracheostomy with assisted positive pres-
weight loss (>LO'%) will often result in substantial improvement in sure ventilation is also effective.
patients with mild OSA. Nasal continuous positive airway pres- Cheyne-Stokes breathing (CSB) is characterized by oscillatory
sure (CPAP, Chapter 38) is the most commonly prescribed therapy, ventilation due to instability in ventilatory control. During sleep,
and is effective in compliant patients. Unfortunately, about 50% of patients will alternate periods of hyperpnoea with hypopnoealap-
patients will not comply with nasal CPAP long-term. Some patients noea. CSB is most commonly due to congestive heart failure or neu-
will respond to oral appliances or uvulopalatopharyngoplasty rological disease. CSB may be treated with O,, respiratory
(surgery). Tracheostomy bypasses the obstruction, and will relieve stimulants (theophylline) or assisted ventilation. Congestive heart
OSA; however, it is associated with psychological and local compli- failure related CSB improves with diuretics and afterload reduction.
38 Mechanical ventilation

1 (a) indicatjons for mechanical ventilation or support in aduks


1
1 Surgery / Cervical cord damage above C4 I
I General anaesthesia with neuromuscular blockade / Neck fractures I
Post-operative management following major surgery
Neuromuscular disorders - when VC < 2 0 - 3 0 mL1kg
Respiratory centre depression
Guillain-Barrd
Usually when Paco2 >7-8kPa (50-60mmHg)
Myasthenia gravis
Head injury
Poliomyelitis
Drug overdose: e.g. opiates, barbiturates
Polyneuritis
Kaised intracranial pressure: cerebral haemorrhagel
tumourslmeningit~slencephalitis
Chest wail d~sorders
5 t a t u s epilepticus
Kyphoscol~os~s
Lung disease / Trauma espec~allyf l a ~segment
l (mult~pler b fractures
Pneumonia / +section of chest wall unattached)

I Acdte respiratory distress syndrome (ARDS)


1 Severe asthma attack I Other I
Acute exacerbation of chronic obstructive pulmonay Cardlac arrest
disease (COPD), cystic fibrosis / Severe c~rculatoryshock
I Trauma-lung contusion / Resistant hypoxia in Type 1 respiratory failure (reduces I
Pulmonary oedema oxygen consumpt~on)

1 (b) Nasal mask and NIPPV (c) Airway pressure profiles in different types of ventilation

5V IPPV IPPV + PEEP 5IMV


INW CPAP

20

0,
I
5
0

5V = spontaneous vent~lat~on,INPV = ~ n t e r m t t e n negat~ve t pressure vent~lat~on, CPAP = cont~nuous


p o s ~ t ~ alrway
ve pressure, BiPAP = b ~ p h a s ~continuous
c p o s ~ t ~ valrway
e pressure, IPW = ~ n t e r m ~ t t e n t
posltlve pressure v e n t ~ l a t ~ o(=n CMV), CMV = controlled mechan~calvent~lat~on, PEEP = posltve end-
expiratory pressure, 51MV = synchron~zedl n t e r m t t e n t mandatory ventllaton If a spontaneous breath
occurs In t h e t ~ m ~ nwindow
g ~t t r ~ g g e r sa synchron~zedventlator breath and if n o t a mandatory breath
1s glven soon a f t e r the t ~ m ~ nw~ndow
g

1 (d) Complications of mechanical ventilation


I
I Risks during endotracheal intubation o r tracheostomy
I
/ Risks associated with sedation and paralysis I
I Myocard~aldepress~onfrom anaesthetic / Cardlac depress~on I
I A s p ~ r a t ~ oofn g a s t r ~ ccontents : Depress~onof resp~ratorydr~ve(delays wean~ng) I
I Fall in Pao2 during apnoea / Increases danger of disconnectionlventilator failure
I
--

Risks associated with mechanical ventilation


intubation of the oesophagus
High airway pressure- barotrauma
intubation of a bronchus / Alveolar overdistension- volutrauma:
Blockagelaccidental extubation 1 Pneumothorax, pneumomediastinum
Laryngealltracheal damage or stenosis
Infection
/ Subcutaneous emphysema (= air in skin)
----
Risks associated with high inspired oxygen (see Chapter 39) i Structural damage t o lung, airways and capillaries
Bronchopulmonary dysplasia ( a e Chapter 16)
Mechanical ventilation is usually used to prevent or treat type 2 turn, and occasionally the fall in cardiac output can reduce tissue
respiratory (ventilatory) failure. The main indications in adults are oxygen delivery despite the increased P,02. The increased mean air-
listed in Fig. 38a. way pressure caused by PEEP also increases the risk of barotrauma.
A good compromise is to use the minimum PEEP required to keep
Types of mechanical ventilation (Fig. 38c) Po2 at an acceptable level (>8 kPa, 60mmHg) when breathing
Inspiratory muscle paralysis by poliomyelitis was a common reason 50-60% oxygen.
for mechanical ventilation in the first half of the 20th century. It was
usually performed by intermittent negative pressure ventilation Non-invasive respiratory support
(INPV), which is still occasionally used today. Patients are placed Non-invasive ventilation avoids the use of tracheal intubation or
inside a tank ventilator sealed at the neck, and tank pressure is in- tracheostomy. An example is INPV (above). but this is no longer
termittently lowered, expanding the chest and lowering intrapleural widely used. In contrast, non-invasive positive pressure techniques
pressure as in spontaneous breathing. Disadvantages of this 'iron using either a nasal mask (Fig. 38b) or sometimes a full face mask
lung' include claustrophobia, discomfort, difficult nursing care and are increasingly being used.
the bulk and expense of the equipment. Jacket a n d cuirass venti- In continuous positive airway pressure (CPAP), a standing
lators produce a negative pressure just around the chest, but diffi- pressure of 5-1 OcmH,O is applied to a nasal or face mask in a spon-
culty in achieving a satisfactory seal limits their use to patients only taneously breathing patient (Fig. 3%). This has several potential
needing ventilatory augmentation. beneficial effects. First, it helps prevent upper airway collapse in ob-
From the 1950s, intermittent positive pressure ventilation structive sleep apnoea. In interstitial diseases such as A R D S , it
(IPPV; controlled mechanical ventilation, C M V ) quickly re- recruits alveoli, reducing VA/Q mismatching. FRC is increased, and
placed INPV for most purposes. Air is driven into the lungs by rais- this may increase lung compliance by moving the patient onto the
ing airway pressure. usually via an endotracheal or tracheostomy steep part of the pressure-volume curve (Chapter 6). CO, retention
tube. Expiration is achieved by allowing pressure to fall to zero. This may be a problem during CPAP, and this may be improved by alter-
simple form of IPPV is used during routine surgery. Typical initial nating the pressure level between high and low values (biphasic
adult settings for IPPV are: positive pressure ventilation, BiPAP) to periodically increase the
emptying of the lungs.
Tidal volume, VT = 8-12mLkg CPAP may improve oxygenation and may aid the patient's own
respiratory efforts, but it cannot produce ventilation by itself. In
Respiratory frequency, f = 8-14 breathslmin contrast. non-invasive intermittent positive pressure ventilation
(NIPPV) is IPPV delivered by face or, more usually, nasal mask.
Minute ventilation, V(= VT x f ) = 6000 mL1min For it to be used successfully, the patient must be cooperative and in-
troduced to the technique gradually, to allow synchronization of his
Inspiratory time:expiratory time = 1:2-1:3 or her breathing with the ventilator. Its use includes nocturnal ven-
tilation of patients with chronic respiratory failure due to neuro-
Minute ventilation is adjusted to maintain P,co2 at about 5 kPa muscular disease or thoracic deformity. It is being used increasingly
(37mmHg). A slightly lower P,co, may be used initially in the to treat acute exacerbations of chronic obstructive pulmonary
presence of raised intracranial pressure. Accepting a higher P,co, disease (COPD), avoiding the need for intubation and improving
(permissive hypercapnia) may prevent the need for excessively survival.
high airway pressures. P,o, is maintained above 10 kPa (75mmHg) In summary, the main beneficial effect of CPAP is recruitment of
by adjusting inspired Fo,. The lowest concentration needed is used, alveoli. The reduction in collapse sometimes also gives rise to a
usually in the range 30-60%. It may be preferable to accept a slight reduction in the work of breathing. In contrast, N I P P V is used to
lower Po, than to use ~ 6 0 %for long periods. reduce the work of breathing rather than to recruit alveoli. This
Microprocessor control of ventilators has permitted develop- may be of benefit in the tired (e.g. COPD) patient.
ment of numerous variations of IPPV. For example, in non- Weaning the patient off the ventilator following surgery is usual-
paralysed patients, the positive pressure may be synchronized with ly achieved by reversing neuromuscular blockade and lightening the
spontaneous breaths and a mandatory breath given if no sponta- anaesthetic level. In intensive-care patients, weaning may be more
neous breaths occur in a preset time (synchronized intermittent difficult. Several techniques are used, including removing mecha-
mandatory ventilation, SIMV). In another form, the ventilator nical ventilation for progressively longer periods, or by using a
operates only where spontaneous ventilation falls below a preset spontaneously breathing mode (e.g. SIMV), and pressure support
minimum (mandatory minute ventilation, M M V ) . in which support is progressively reduced. CPAP applied via the
If, instead of allowing airway pressure to fall to zero, a small endotracheal tube may also help the weaning process.
positive pressure is maintained throughout expiration (positive Problems are hard to predict accurately, but are most likely fol-
end-expiratory pressure, PEEP), there is a reduction in VA/Qmis- lowing prolonged ventilation, in debilitated patients, or in those
matching and an improvement in P,02 in some conditions, such as with neuromuscular o r chronic respiratory disease. A pattern of
acute respiratory distress syndrome (ARDS). This occurs because rapid shallow breathing 5min after disconnection from the ventila-
PEEP increases functional residual capacity (FRC) and reduces the tor is one of the more useful predictors of failure.
closure of airways and alveoli towards the end of expiration. Un- Complications o f mechanical ventilation are numerous, and
fortunately, intrathoracic pressure is raised, impairing venous re- are listed in Fig. 38d.
39 Acute oxygen therapy

Oxygen delivery systems High-flow (venturi) face mask


0
LS L/
8568
High-flow (venturi) face mask Total
The O2 flow through the j e t mixing venturi valves draws the correct gas flow from
3 0 Llm~n
proportion of oxygen t o produce the required O2 concentration 3 0L l m I i n, ~ into mask
Delivers more mixed gas t o the mask than the patient needs (e.g. 30Llmin)
Inspired 02concentration unaffected by patient's breathing pattern
Safest in COPD patients in respiratoryfailure Jet of
oxygen

14LIm1n
air I I air oxygen
Entrained
air

Low-flow masks Low-flow face mask


Oxygen flows a t a s e t rate into the mask and is supplemented by air
drawn into the mask n
F102 achieved depends on ventilation:
Ventilation = 5LImin 3 Llmin air
drawn into
Oxygen fiow = 2 ~ 1 m i nAir (21% 02) = 3Llmin
F102 = (2 + 0.21 x 3 ) 15 x 100 = 5 3 %
2LImn
Ventilation = 25LImin -5Llmin
oxygen
Oxygen fiow = 2 Llmin Air (21% 02) = 2 3 Llmin into mask inspired
Fro2 = (2 + 0.21 x 23) 125 x 100 = 27%
Not recommended where accurate control of F102 desirable e.g. patients oxyaen
with COPD and chronic hypercapnia

Nasal prongs Nasal prongs


Delivers a constant oxygen flow therefore Pro2 varies with ventilation
More comfortable than mask
No need t o remove t o expectorate or eat
02delivered by nose is probably inhaled even in mouth breathing

Rebreathing and anaesthetic mask


High concentration oxygen mask (F102 > 60%)
Incorporate a degree of rebreathing .'. risk of C02 retention

With non-invasive ventilation: may help where oxygen supplementation


alone is n o t enough (see Chapter 3 8 )

Correctly administered oxygen therapy can be life-saving. The failure, emboli); low haemoglobin concentration (anaemia); ab-
wrong concentration and inadequate monitoring can have severe normal oxygen dissociation curve (haemoglobinopathies, CO
consequences. poisoning); and poisoning o f intracellular oxygen usage (e.g.
cyanide, sepsis). Tissue hypoxia occurs within 4 m i n of failure of
Pathophysiology any of these systems, because tissue and lung oxygen reserves are
Tissues require oxygen for survival. Oxygen utilization is dependent small. Successful treatment requires early recognition. but the clini-
on delivery by the circulation of blood containing adequate quanti- cal features are often non-specific, including altered mental state,
ties of oxygen in a readily releasable form to tissues capable of using dyspnoea, hyperventilation, arrhythmias and hypotension (for de-
the delivered oxygen. Tissue hypoxia can be caused by: low arter- tails, see Chapter 20). The effects of anaemia and abnormal dissoci-
ial Po, (hypoxaemia); inadequate tissue blood flow (cardiac ation curves are discussed in Chapter 8.
Measuring tissue hypoxia (Pao2>6.65kPa) without decreasing arterial pH below 7.26. Non-
Arterial oxygen saturation (Sao2)is measured with a pulse oxime- invasive positive pressure ventilation and respiratory stimulants
ter, and partial pressure of oxygen (Pao2) by blood gas analysis. may improve ventilation and prevent CO, retention. Pulmonary
These are the principal clinical measurements used for initiating, oxygen toxicity: Fro2>60%may damage the alveolar membrane if
monitoring and adjusting oxygen therapy. However, these measures inhaled for > 2 4 4 8 h. Fire: facial burns and deaths occur when pa-
can be normal when tissue hypoxia is caused by low cardiac output tients smoke whilst using oxygen. Absorption collapse: if an air-
states, anaemia and failure of tissue oxygen use. In these circum- way becomes blocked, oxygen in the trapped alveolar gas is rapidly
stances, mixed venous oxygen partial pressure (Pqo2),which is absorbed, but N 2 absorption is slow, as alveolar PN,is in equilibri-
measured in blood taken from a pulmonary artery catheter, approx- um with mixed venous PNZ If N2 is replaced by 0, during oxygen
imates to mean tissue Po,. Severe hypoxia in a single organ (e.g. due therapy, temporary blockage of airways by secretions is more likely
to an arterial embolus) may be associated with a normal Pdo2,S,O, to lead to collapsed alveoli, which may be difficult to reopen.
and Pvo2.Measurement of individual organ hypoxia is difficult, and
requires specialized techniques (e.g. tonometry). Efficacy of acute oxygen therapy
Arterial hypoxaemia
Right-to-left shunting (e.g. pneumonic consolidation): when
Acute oxygen therapy
shunt is >20°!. hypoxia persists despite high F,oZ
The main indications for instituting oxygen therapy are:
Alveolar hypoventilation (e.g. drug overdose, neuromuscular
H y p o x a e m i a (P,,02<7.8kPa, S,02<90%).
disorders): oxygen rapidly corrects hypoxaemia, but the high P,co,
H y p o t e n s i o n (systolic blood pressure <100 mmHg). remains. The primary aim is to improve ventilation.
L o w cardiac o u t p u t a n d metabolic acidosis (bicarbonate VIQ mismatch (in COPD or asthma, Chapter 14): improved
<18 mmollL). oxygenation, but the response will vary between patients.
Respiratory distress (respiratory rate >24/min).
The airway should be checked before starting oxygen therapy. and Tissue hypoxia without arterial hypoxaemia
arterial blood gases analysed as soon as possible to assess Pao2, Low-output cardiac states (congestive cardiac failure, CCF; my-
P,co2, pH and bicarbonate. ocardial infarction, MI): oxygen solubility is low, and even at F,o,
In the acute situation, the dose o f oxygen may be critical. In- loo%, only 30% of total oxygen requirement is carried dissolved in
adequate oxygen accounts for more death and disability than can be blood. In the absence of hypoxaemia, high oxygen can only margin-
justified by the small risks associated with high-dose oxygen. Short ally improve tissue oxygenation by a small increase in dissolved oxy-
periods of high-dose oxygen may preserve life until more specific gen. This may preserve some cells with critical oxygen supply, but
therapy (e.g. antibiotics, thrombolytics) can be instituted. When it must not delay correction of the primary clinical problem (e.g.
blood gas results are available, the dose of oxygen can be adjusted to restoring tissue blood flow).
maintain the P,02 between 8 and 10 kPa. The recommended initial Chronic lung disease: relief of breathlessness is reported in
oxygen concentrations as a fraction of oxygen in inspired air (FroZin some patients without hypoxaemia, and a trial of oxygen may be
percentage) are: warranted.
C a r d i a c or respiratory arrest, 100'%1. C a r b o n monoxide poisoning: a high F,02is essential despite a
H y p o x a e m i a w i t h Paco,<5.3 kPa, 40-60%. normal P,o,, because oxygen competes for haemoglobin binding
H y p o x a e m i a w i t h P,co,>5.3 kPa, initially 24%. sites and reduces the half-life of carboxyhaemoglobin from about
The important features. advantages and disadvantages of different 320 to 80min.
oxygen delivery systems are shown in Fig. 39.
Monitoring oxygen therapy
Dangers of oxygen therapy Careful monitoring of oxygen therapy by oximetry (Sao2)and arte-
C a r b o n dioxide retention: high-dose oxygen given to the 10-1 5x1 rial blood gas measurement is essential. Oximetry has the advantage
of chronic obstructive pulmonary disease (COPD) patients with of continuous oxygen measurement, whereas blood gases are re-
type 2 respiratory failure (Chapter 20) will reduce hypoxic drive to quired to monitor P,co2 and pH.
breath. increase V/Q mismatching, and may cause C 0 2 retention
and respiratory acidosis that may be lethal. Fortunately, as these pa- Stopping oxygen therapy
tients are on the steep part of the oxygen dissociation curve, small Oxygen therapy should be stopped when the arterial oxygenation is
increases in P,o, cause worthwhile improvements in arterial oxygen adequate with the patient breathing room air (P,oz>8kPa,
content. The initial oxygen concentration should be low (24-28%) S,02>900/u). In patients without arterial hypoxaemia, oxygen
and progressively increased if repeat blood gas analysis shows should be stopped when the acid-base state and clinical assessment
little or no increase in PacoZ The aim is to correct hypoxaemia are consistent with resolution of tissue hypoxia.
40 Case studies

Case 1: Asthma h o w do you think they would they compare with the normal for a
Tom, who is 15 years old, attends his GP's asthma clinic. He has had boy of his age and size?
asthma since early childhood. On two occasions, when he was 8 and 5 If you had had your stethoscope with you, what would you have
10 years old, he had attacks severe enough to require hospital ad- heard on examining his chest?
mission. At present, his asthma is well controlled on regular inhaled
beclomethasone dipropionate 200 yg twice daily and inhaled salme- At 18 years of age, Tom goes to college in London. He stops taking
terol50 yg twice daily. Today, his peak flow is 510 Llnlin (predicted regular medication, as he feels he has 'grown out' of his asthma. He
value for his age and height 530Llmin). On auscultation, there is keeps a salbutamol inhaler in his room 'just in case'. During the first
vesicular breathing and no other sounds. term he is well, apart from a couple of wheezy episodes while play-
ing football. In the second term, he develops a heavy cold, and over
Questions
24h he becomes progressively more breathless despite frequent
1 Is this peak flow normal? Apart from the nomogram values, can
puffs of salbutamol. His friends call out his GP, who finds the fol-
you think of any other peak flow reading with which it would be
lowing: Tom is fully alert, but talking in broken sentences because
useful to compare today's clinic reading?
he is very breathless. He is not cyanosed. He is using his accessory
2 If you measured the following:
muscles of respiration. On auscultation, there are wide-spread expi-
FEV,lFVC
ratory rhonchi (wheezes). BP is 115180, heart rate I10 beatslmin,
Airway resistance respiratory rate 30breathslmin, peak flow 200 Llmin.
Functional residual capacity 6 Which observations suggest that this is a fairly severe attack?
Lung compliance 7 If the G P had measured airway resistance. FRC, lung compli-
Arterial Po, a n d arterial Pco, ance, arterial Po? and Pco,, how would they compare to the pre-
h o w would they compare with the normal for a boy of his age and dicted values?
size?
On a school trip to a countryside park, Tom becomes a breathless His G P decides that this attack warrants hospital admission. and he
running across a field. His teacher is alarmed by his noisy breathing calls an ambulance. Unfortunately, owing to heavy traffic it is 40min
and asks you, a passing medical student, to assess whether they need before he arrives at the local Accident and Emergency Department.
to get him to hospital. By this time, Tom is confused, too breathless to talk and unable to
3 What simple observations can you make that will help you decide produce a peak flow reading. The casualty officer notices he is now
how severe this attack is? In his backpack he has a salbutamol cyanosed, although the widespread rhonchi noted in the GP's letter
inhaler, a saln~eterolinhaler, a sodium cromoglycate inhaler and a have now disappeared. Arterial blood gases show arterial Po2=7
beclomethasone inhaler. Which should he use? kPa and arterial Pco2=5.5 kPa while breathing 60'%/;,oxygen.
4 If you had been able to measure the following during his episode 8 Discuss the features which suggest this asthma attack is life-
of breathlessness: threatening. D o the reduced rhonchi on auscultation contradict the
FEV,lFVC other findings?
Peak flow rate 9 What is the cause of the low arterial Po2? Was the inhaled oxygen
Airway resistance helpful, and if so was the correct concentration used? Is this Pco,
normal, and how does it affect your assessment of the severity of
Functional residual capacity
this attack?
Lung compliance
Arterial P o 2 and arterial PCO?

Case 2: Severe breathlessness (point of maximal impulse [PMI], scaphoid abdomen. no he-
A 38-year-old man is seen for evaluation of severe exertional dys- patomegaly. but with a trace of pedal oedema. His haematocrit was
pnoea. Two years ago, he had been able to play squash regularly. but 45% and other laboratory tests were normal.
he stopped 6 months ago because of dyspnoea and fatigue during Chest radiograph shows flat diaphragm with increased radiolu-
exercise. He now reports dyspnoea after climbing one flight of stairs. cency at the lung bases.
He has no cough, sputum or wheeze. He smoked one pack of ciga-
rettes a day for 10 years and quit 7 years ago. He has no allergies or Lung function tests Measured '%I Predicted
pets, and has not travelled outside of Europe or the USA. One of his
six siblings died at age 25 with an unknown progressive lung ailment. FEV, (L) 0.80 20
FVC (L) 3.0 60
Physical examination was notable for a thin male, BP= 110175, FEV,lFVC 0.27 33
H R = 104, respiratory rate = 22, oxygen saturation = 96% at rest; TLC (L) 8.1 120
chest with diminished breath sounds and a prolonged expiratory D,co (mL/min/mmHg) 14 45
phase, slightly elevated jugular venous pressure, midline apex beat
Questions 5 What will happen to the patient's oxygenation with exercise?
1 What would you expect the patient's FRC and RV to be? Why?
2 Why is the cardiac PMI shifted to the midline? 6 Why is the patient's jugular venous pressure elevated?
3 Why is the FVC low? 7 What are the most likely diagnosis and pathophysiology of his
4 Why is the DL(.()low? disease?

Case 3: Restrictive ventilatory defect Patient A Patient B


Two 60-year-old patients are being evaluated for dyspnoea. On ex- Lung function
amination, both patients have an oxygen saturation of 88%, small tests Measured %I Predicted Measured ' X Predicted
lung volumes to percussion and normal cardiac examinations. Pa-
tient A has diffuse bilateral inspiratory crackles and digital club- FEV, (L) 1.1 26 1.1 26
bing. Patient B has clear lungs and difficulty in rising from his chair FVC (L) 1.3 26 1.3 26
and raising his hands over his head. FEV,/FVC 0.80 100 0.80 100
TLC (L) 3.0 43 3.8 54
Questions FRC (L) 2.0 54 3.1 87
1 What patterns of abnormalities d o these patients exhibit?
R v (L) 1.7 65 2.5 120
2 Based on the lung function results, what is the most likely patho-
physiology explaining each patient's symptoms? DLco 18 50 36 100
(mL/min/mmHg)
3 What is the likely explanation for the differences in FRC and RV
between the two patients?
4 What is the differential diagnosis for Patient A?
5 What is the differential diagnosis for Patient B?
6 Both patients have hypoxemia. Which patient is more likely to
have hypercapnia?

Case 4: Haemoptysis secretions. She had been investigated by several doctors, but chest
A 31-year-old married Vietnamese woman presented to the emer- radiographs were normal, and she had been reassured that the
gency room following an episode of haemoptysis in which she had bleeding was from the upper respiratory tract. For the 6 months be-
expectorated 250mL of fresh red blood. Nasopharyngeal examina- fore her presentation to casualty, she had coughed up small quanti-
tion by the ENT surgeons was normal, and a chest radiograph (Fig. ties of blood every 2-3 weeks (<50mL), but there was no associated
40) in casualty was unhelpful, although bronchial wall thickening fever, wheeze or breathlessness on these occasions. Two weeks be-
was noted behind the heart (arrow). There was no further bleeding, fore presentation to the emergency room, she developed a cough
and she was discharged with an outpatient appointment. Over the productive of purulent sputum and night-time sweating. As a child,
next few days, she continued to expectorate small clots of blood she had suffered with whooping cough, but there was no other past
mixed with discoloured phlegm. medical history of serious chest illness or tuberculosis. She was a
At her outpatient appointment, she reported a 10-year history of non-smoker.
recurrent, intermittent haemoptysis in which she had expectorated Examination was normal. She did not have finger clubbing.
small quantities of fresh red blood, sometimes mixed with bronchial anaemia, cyanosis or lymphadenopathy. Chest examination was un-
remarkable. The breath sounds were vesicular, and there were no
crackles or wheezes. Routine blood tests, including an ESR and C-
reactive protein and sputum microbiology including examination
for tuberculosis, were normal. The grade 1 Heaf test was consistent
with immunity to tuberculosis.

Questions
1 What are the commonest causes of haemoptysis, and from which
circulation does bleeding occur?
2 Is haemoptysis life-threatening, and how is the severity of bleed-
ing classified?
3 What are the clinical features that may help establish the diagno-
sis? What is the most likely cause in this case?
4 What investigations would you perform to establish the diagnosis
in this case?
5 What is bronchiectasis, and what causes it?
Fig. 40 Chest radiograph. 6 How should a large haemoptysis be managed?
41 Case studies: answers

Case 1: Asthma
1 For males. peak flow should be no more than 100 Llmin below the breath has been fully exhaled, or air may be trapped behind col-
predicted; Tom's peak flow is therefore within the normal range for lapsed airways. This would lead to a raised FRC. The volume1
his age and size. The most useful value to compare it with would be pressure curve flattens as TLC is approached. i.e. compliance is
his own best peak flow rate. reduced. Consequently, with this severity of attack. the work of
2 All of these should be normal. Asthma, especially in the young, is breathing is increased not only because of increased work against
reversible, and usually between attacks patients have normal airway airway resistance, but because of increased elastic resistance. In ad-
resistance. compliance, lung volumes and blood gases. Peak flow dition, with increased FRC, the inspiratory muscles may not be at
and auscultation (but see note in answer 9) suggest little evidence of their optimum working length and hence efficiency is impaired.
airway obstruction today. Some ventilation-perfusion mismatching would be expected, as
3 Simple observations that can be made in these circumstances: bronchoconstriction and inflammation will result in underventila-
How breathless is he? Inability to talk in complete sentences is an tion of some regions, and with the resulting shunt effect there is
indication of a severe attack. likely to be some degree of arterial hypoxia. Increased total ventila-
Respiratory rate (>25 breaths per minute suggests a severe attack). tion will usually lower the Pco,, resulting in a final blood gas picture
Cyanosis (very severe attack). of low Po, and low Pco?
Pulse rate (>I I0 beats per minute suggests a severe attack). 8 The cyanosis indicates severe hypoxia, and is probably responsi-
If he has his peak flow meter with him a value 4 0 % of his best or ble for his confused mental state. The inability to talk and produce a
predicted suggests severe attack. peak flow reading are also signs of life-threatening asthma. The dis-
Even in the absence of the above signs of a severe attack, it is im- appearance of rhonchi is consistent with very poor air movement.
portant to monitor the response to treatment to ensure that im- Rhonchi are a characteristic feature of airway obstruction, but they
provement rather than deterioration is occurring. He should use a are not a reliable indicator of severity. In life-threatening asthma,
short-acting P,-adrenoreceptor agonist (salbutamol), which relaxes the normal vesicular breath sounds are also absent. A silent chest in
bronchial smooth muscle (see Chapter 22). an asthma attack is an ominous sign.
4 He now has definite bronchoconstriction; we would expect both 9 The low Pao2is caused by ventilation-perfusion mismatching.
peak flow and FEV,lFVC to be reduced. In this mild attack, he Hypoxia is what kills in severe asthma, so it is appropriate to give
would probably be able to exhale completely. so FRC is likely to be high inspired oxygen, which should significantly raise alveolar oxy-
normal. There is at present no reason why lung compliance should gen tension in poorly ventilated regions of his lung, and so improve
be altered. Although he will be working harder than normal, he arterial oxygenation. In this patient, there is no need to worry about
should be achieving a normal alveolar ventilation and his blood ventilatory drive, so as high as possible is the correct emergency
gases should be normal. A reduced arterial Pco, may be caused by treatment. With a face mask, the maximum achievable is -60%.
anxiety and consequent hyperventilation. Although a Pco, of 5.5 kPa would usually be considered 'normal',
5 Expiratory rhonchi (musical sounds caused by vibration of the in the presence of severe hypoxia it should be regarded as worrying.
sides of collapsing airways). With this degree of hypoxia, the drive to breathing should be in-
6 The broken sentences, use of accessory muscles, high heart and creased, with increased ventilation and low Pco,. Here the failure to
respiratory rate are all important. The peak flow is only about 40% raise ventilation appropriately is likely to indicate exhaustion. The
of his best value. patient may deteriorate rapidly-a further fall in ventilation will
7 This is clearly a severe asthma attack, and airway resistance worsen hypoxia, and this may be fatal. In the presence of significant
would be greatly increased. It is likely that air trapping would occur, hypoxia, a 'normal' or high arterial Pco2 should be regarded as a
as initially expiration is affected more than inspiration. As expira- serious finding.
tion is slowed, the subject may be forced to breathe in before the last

Case 2: Severe breathlessness


1 The obstructive ventilatory defect (low ratio of FEV,/FVC) cou- chest wall, with an axis close to 90'. For a similar reason, the apex
pled with a reduced diffusing capacity for carbon monoxide would beat may be quiet.
suggest emphysema. Emphysema is characterized by a reduction in 3 The FVC is low because the RV is high. Airways close prema-
lung elastic recoil, increased lung compliance and floppy airways. turely in emphysema, which increases RV. Furthermore, because of
Therefore, FRC and RV are both likely to be elevated. the marked decrease in maximal expiratory flow rate due to the
2 The hyperinflation of the lung and the increase in FRC pulls the decreased lung elastic recoil, patients' spirometry traces may not
apex of the heart caudally and to the middle. This can be seen radi- plateau. indicating that the lung was still emptying at very low flow
ographically as a small midline heart. The ECG will show low volt- rates when the FVC manoeuvre was terminated.
age due to the increased amount of air between the heart and the 4 Diffusing capacity is influenced by the alveolar-capillary surface
area for gas exchange. Emphysema is characterized by a loss of the Any degree of hypoxaemia during exercise will exacerbate the
alveolar-capillary units that are utilized for diffusion. There need pulmonary hypertension by superimposing hypoxic pulmonary
not be a defect in transfer of gas from the alveolus to the capillary to vasoconstriction on the already increased resistance.
reduce the DLro; a reduction in surface area is adequate to cause 7 The presence of early-onset emphysema, the family history
abnormality. and the history of cigarette smoking make the diagnosis of a,-
5 With exercise, the patient's oxygen saturation will fall because of antitrypsin (AAT) deficiency most likely. He probably has the
the diffusion defect. With this magnitude of diffusion defect, the red homozygous ZZ genotype that causes a marked reduction of AAT
cells have adequate time to equilibrate with alveolar oxygen as they levels to <15% normal. The radiograph is consistent with panacinar
traverse the alveolar-capillary membrane. However, during exer- emphysema and alveolar destruction predominantly at the bases.
cise, when cardiac output rises, red cells traverse the alveolarxapil- The lung destruction is due to release of proteolytic enzymes from
lary membrane at rest more quickly and do not equilibrate with neutrophils and other inflammatory cells in response to environ-
alveolar oxygen tension at the end-capillary segment. This results in mental stimuli. These enzymes are normally neutralized by the
deoxygenated blood entering the systemic circulation when cardiac antiproteases in the lung to prevent lung destruction in the presence
output is increased. This exercise-induced desaturation will be ac- of mild inflammatory stimuli. In AAT deficiency, the proteases are
centuated when alveolar oxygen is reduced, such as at high altitude. not neutralized, and induce panacinar emphysema under 'normal'
The threshold for significant oxygen desaturation with exercise is circumstances. Cigarette smoking induces a neutrophilic response
approximately D,co< 50%)predicted. in the lung that accelerates the decline of lung function in AAT de-
6 The patient probably has a component of pulmonary hyper- ficiency. Intravenous replacement of AAT in patients with reduced
tension due to the emphysema. The loss of capillary units raises lung function may slow the decline in FEV,.
pulmonary vascular resistance and increases right heart work.

Case 3: Restrictive ventilatory defect


1 Both patients have restrictive ventilatory defects based on the low lung volumes. However, this presupposes adequate expiratory
reduced TLC. FEV, and FVC are reduced proportionally, so the muscle strength to actively lower lung volume below FRC (which
FEV,/FVC is normal; therefore there is no obstructive ventilatory can be reached from TLC passively). Patient A has normal muscle
defect. Patient A has reduced DLco,signifying a gas transfer defect. strength and airways that resist collapse due to the parenchymal
2 Restrictive ventilatory defects may be due to stiff lungs, stiff chest lung disease, resulting in a reduced RV. Patient B has weak expira-
wall, or weak respiratory muscles. Diseases causing stiff lungs will tory muscles, resulting in an elevated RV.
reduce all lung volumes/capacities simultaneously, including TLC, 4 The differential diagnosis is long, and includes the disorders dis-
FRC, and RV. Most parenchymal lung diseases will also cause a cussed in Chapter 27. Briefly, these would include occupational/
reduced DLco, whereas chest wall disease and respiratory muscle environmental disorders, connective tissue/autoimmune diseases,
disease will not. An increased RV is also not compatible with stiff drugltreatment-induced diseases, primary lung disorders or idio-
lungs. Thus, Patient A seems to have a problem with stiff lungs. The pathic disorders. Idiopathic pulmonary fibrosis or cryptogenic
relatively normal FRC and DLcoin Patient B suggest that the lungs fibrosing alveolitis is likely in a 60-year-old with lung crackles, club-
and chest wall are normal. Either a stiff chest wall or weak muscles bing, no significant past history, no signs or symptoms of extrapul-
may cause an increased RV. Patient B's lung function and difficulty monary disease, and the lung function shown for Patient A.
in rising out of a chair and raising his arms suggest a muscle disease. 5 Respiratory muscle weakness may be due to a variety of neuro-
Diseases causing weak respiratory muscles will reduce TLC, be- muscular diseases that can involve the spinal cord, motor nerves,
cause the patient cannot inspire deeply. neuromuscular junction or skeletal muscles:
3 The FRC is determined by the balance between the inward pull of Spinal cord: tumour, syringomyelia, polio, amyotrophic lateral
the lung elastic recoil pressure and the outward pull of the chest sclerosis, tetanus.
wall. Therefore, FRC will be reduced if either the net lung recoil Motor nerves: brachiallphrenic nerve neuritis, trauma.
pressure increases (due to stiff, low-compliance lungs), or if the net Neuromuscular junction: myasthenia gravis, botulism, organ-
outward pull of the chest wall decreases (for example when scarring ophosphate poisoning.
of the chest wall produces an added inward recoiling force). Since Skeletal muscle: muscular dystrophy, myositis, mitochondria1
FRC is determined by the balance between two opposing static disease, myopathy (nutritional, drug, metabolic, inherited).
forces, respiratory muscle weakness should not influence FRC. 6 Hypoxaemia in interstitial lung diseases is usually due to ventila-
However, in clinical practice, patients with respiratory muscle weak- tion-perfusion mismatching. Patient A likely has hypocapnia, be-
ness often have a slightly reduced FRC. The mechanism for this cause patients with interstitial lung disease tend to hyperventilate in
finding is probably related to the lack of deep breaths or sighs caus- response to stiff lungs and hypoxia. This will lower arterial carbon
ing microatelectasis that will increase lung recoil and decrease dioxide tension. In contrast, Patient B likely has hypoxaemia due to
compliance. alveolar hypoventilation, and is therefore hypercapnic. Patient B is
RV is the amount of gas remaining in the lung at the end of max- also more likely to develop acute respiratory failure with the limited
imal expiration. In adults, RV is determined by airway collapse at ventilatory reserve due to the muscle weakness.
Case 4: Haernoptysis possibility of an alveolar haemorrhage syndrome. In this case, the
1 The table below illustrates that most cases of haemoptysis are due age of the patient, the long history of minor haernoptysis and the
to infection (-80°K) i n c l u d i n g tuberculosis, pneumonia. lung ab- symptoms of purulent sputum and night-time fever suggest a diag-
scess and bronchiectasis. Only a minority are due to malignancy nosis of bronchiectasis, although other potential causes include
(-20%)).Pulmonary embolism and trauma are other potentially im- recurrent pulmonary emboli, vasculitis and a benign adenoma.
portant causes. The bronchial (rather than the pulmonary) circula- 4 Routine blood tests (white cell count raised in infection), includ-
tion is the usual source of bleeding. ing ESR (raised in vasculitis) and C-reactive protein (raised in in-
2 Approximately 3 5 4 0 % of cases of haemoptysis are classified as fection). Specialist blood tests (D-dirners for pulmonary emboli,
trivial (flecks of blood in sputum). 45-50% as moderate (<500mL Aspergillus precipitans, vasculitis screen) may be required. Sputum
or 0.5-2 cups daily) and only 10-20% as massive (>500mL or more microbiology may isolate infective organisms (pneumonia, abscess,
than 2cups of blood daily). Mortality is directly related to the rate Aspergillus) or acid-fast bacilli (tuberculosis). A screening Heaf test
and volume of blood loss and the underlying pathology. In patients may detect tuberculosis. Chest radiography should be obtained
expectorating >500mL of blood within a 4-h period. the mortality in all patients. It may provide important diagnostic information
is -70%, compared to 5% in patients expectorating the same quan- including evidence of a mass, cavity, or abscess. C T scans with
tity over 1 6 4 8 h. Death results from asphyxia, caused by flooding contrast may detect the site of bleeding, tumours, vascular malfor-
of the alveoli and only rarely from circulatory collapse. mations and other structural abnormalities. In this case, the CT
3 A good history is essential. and may indicate the cause of scan demonstrated a grossly dilated bronchus (> IOmm) consistent
haemoptysis. The characteristic clinical picture of diseases like tu- with bronchiectasis in the anteromedial segment of the left lower
berculosis, bronchiectasis and bronchogenic carcinoma may direct lobe (Fig. 41a). Bronchoscopy is often required to detect endo-
subsequent investigation and management. Chest examination may bronchial lesions and inhaled objects (e.g. tooth). Combinations of.
reveal localized crepitations or consolidation but widespread soil- bronchoscopy and C T scanning have the highest diagnostic yield.
ing of the tracheobronchial tree with blood (due to coughing), often Bronchial arteriography may be required to detect the site of
results in diffuse clinical signs. Examination of expectorated blood bleeding (Fig. 4 1 b).
may provide clues. Food particles suggest the possibility of hae- 5 Bronchiectasis is described in Chapter 30.
matemesis, but blood in the nasogastric aspirate does not differen- 6 The key aspects of management of massive haemoptysis are to
tiate between haematemesis and haemoptysis, as coughed-up blood maintain a patent airway and oxygenation (oxygen therapy). As-
is often swallowed. Purulent material in the sputum may indicate phyxia (not bleeding) is the greatest immediate risk to the patient.
bronchiectasis or a lung abscess. Associated haematuria raises the Promote drainage of blood and prevent alveolar 'soiling' by posi-
tioning the patient slightly head down in the lateral decubitus posi-
tion, with the 'presumed' bleeding side down. Determine the cause,
site and severity of the bleeding (as above): haematemesis and upper
Infective (-80'%,) Malignant (-20%) Other airways bleeding (e.g. nose) may be confused with haemoptysis.
Treatment of the underlying cause is essential if the haemoptysis
Tuberculosis Lung cancer Pulmonary infarction
is to be controlled (antibiotics for pneumonia or a lung abscess).
Pneumonia Metastatic cancer Adenoma
Lung abscess Lymphoma Traumatic Avoid excessive chest manipulation, including physiotherapy, as
Bronchiectasis Alveolar haemorrhage this may increase or restart bleeding. Cough suppression with
A.~pergillu.r Vasculitis codeine 30-60mg every 6 h may be helpful. Institute appropriate
antibiotics and bronchodilators.
Immediate control of haemoptysis is achieved at bronchoscopy
by directing boluses of iced saline with epinephrine (IOmL; 1:
10000 dilution) at the bleeding site.
Bronchial angiography a n d embolization are the established
therapeutic techniques for the initial control of haemoptysis. This
procedure is initially successful in 70-100% of cases. The best re-
sults are described in patients with dilated bronchial arteries (e.g.
bronchiectasis). Rebleeding often occurs (-40%1), and infarction of
the anterior spinal artery with paraplegia is reported (-5'%,). Most
studies agree that surgical therapy is associated with the best long-
term outcomes for isolated lesions. Primary medical management
may be mandatory because bleeding cannot be localized (wide-
spread Aspergillus infection) o r is not amenable to surgical resec-
tion of a pulmonary segment. In other patients, surgery will be
contraindicated because of end-stage lung disease (FEV, <40')/a
predicted), poor cardiac reserve, unresectable cancer, or severe
bleeding diathesis.
(b) Final diagnosis: bronchiectasis of the anteromedial segment of
Fig. 41 (a) CT scan, (b) bronchial arteriography. the left lower lobe.

You might also like