35
INTRODUCTION Cystic fibrosis (CF) is a multisystem disorder affecting children and, increasingly, adults. 1 CF is characterized chiefly by chronic airways obstruction and infection and by exocrine pancreatic insufficiency with its effects on gas- trointestinal function, nutrition, growth, and maturation. This condition is the most common life-threatening genetic trait in the white population. 2 Numerous mutations of a single gene are responsible for the CF syndrome and for variations in its severity. The gene encodes a membrane protein called the cystic fibrosis transmembrane regulator (CFTR). CFTR functions in many tissues as a kinase- regulated Cl - channel. In some tissues, CFTR also regulates the activity of other ion channels. Typically, mutations in CFTR affect both of these functions. Cystic fibrosis is an important medical problem for a number of reasons. It is the major source of severe chronic lung disease in children and has become an important cause of morbidity and mortality from chronic lung disease in young adults. CF is responsible for most cases of exocrine pancreatic insufficiency in childhood and early adulthood and for many cases of nasal polyposis, pansinusitis, rectal prolapse, nonketotic insulin-dependent hyperglycemia, and biliary cirrhosis in these age groups. Therefore, CF enters into the differential diagnosis of many pediatric and young adult patients. Finally, research advances have introduced the challenge of designing pharmacologic and gene trans- fer therapies to combat the broad range of manifestations and complications. Central to CF diagnosis and care is a carefully integrated and closely monitored network of approximately 115 refer- ral centers in the United States sponsored by the Cystic Fibrosis Foundation. The Foundation also supports a smaller number of multidisciplinary research centers aimed at elucidating the molecular pathophysiology and improv- ing the quality of life for patients with CF. Similar care and research centers are found in Canada and in many European countries. HISTORICAL PERSPECTIVES Cystic fibrosis was first described as a distinct clinical entity in the late 1930s. However, numerous references to infants and children with meconium ileus and characteristic pan- creatic and lung diseases are sprinkled throughout the lit- erature from as early as 1650. Of interest are references in European folklore to the association of salty skin and early demise. 3 Dorothy Andersen, a pathologist at Babies Hospi- tal in New York City, is usually credited with the first com- prehensive description of CF in 1938. 4 She coined the term cystic fibrosis of the pancreas. In 1945, Farber suggested that CF is a disease of exocrine glands, characterized largely by failure to clear their mucous secretory product. 5 He intro- duced the term mucoviscidosis, which was used for a number of years. Chronic infection of the lungs was recognized early as a major contributing factor, and antibiotics were first used for the treatment of CF in the 1940s. At the same time, an autosomal-recessive inheritance pattern for CF was 38 Introduction Historical Perspectives Epidemiology Genetic Basis Pathology Lung Pathology Other Respiratory Tract Pathology Nonrespiratory Pathologic Features Pathophysiology Cystic Fibrosis Transmembrane Regulator Protein: Structure, Metabolism, and Function Abnormal Airway Mucosal Salt and Water Transport Active Ion Transport Properties of Airway Epithelia Mucin Macromolecule Secretion in the Cystic Fibrosis Airway Pathophysiology of Infection Clinical Manifestations Lower Respiratory Tract Disease Upper Respiratory Tract Disease Complications of Respiratory Tract Disease Gastrointestinal Manifestations Pancreatic Disease Hepatobiliary Disease Genitourinary Tract Abnormalities Sweat Gland Dysfunction Diagnosis Treatment Ambulatory Care Hospital Therapy Course of the Disease and Prognosis Summary 1217 Cystic Fibrosis Richard C. Boucher, M.D., Michael R. Knowles, M.D., James R. Yankaskas, M.D.

Cystic Fibrosis

Embed Size (px)

Citation preview

Page 1: Cystic Fibrosis

INTRODUCTION

Cystic fibrosis (CF) is a multisystem disorder affecting children and, increasingly, adults.1 CF is characterizedchiefly by chronic airways obstruction and infection and by exocrine pancreatic insufficiency with its effects on gas-trointestinal function, nutrition, growth, and maturation.This condition is the most common life-threatening genetictrait in the white population.2 Numerous mutations of asingle gene are responsible for the CF syndrome and forvariations in its severity. The gene encodes a membraneprotein called the cystic fibrosis transmembrane regulator(CFTR). CFTR functions in many tissues as a kinase-regulated Cl- channel. In some tissues, CFTR also regulatesthe activity of other ion channels. Typically, mutations inCFTR affect both of these functions.

Cystic fibrosis is an important medical problem for anumber of reasons. It is the major source of severe chroniclung disease in children and has become an important causeof morbidity and mortality from chronic lung disease inyoung adults. CF is responsible for most cases of exocrinepancreatic insufficiency in childhood and early adulthoodand for many cases of nasal polyposis, pansinusitis, rectalprolapse, nonketotic insulin-dependent hyperglycemia, andbiliary cirrhosis in these age groups. Therefore, CF entersinto the differential diagnosis of many pediatric and youngadult patients. Finally, research advances have introducedthe challenge of designing pharmacologic and gene trans-fer therapies to combat the broad range of manifestationsand complications.

Central to CF diagnosis and care is a carefully integratedand closely monitored network of approximately 115 refer-ral centers in the United States sponsored by the CysticFibrosis Foundation. The Foundation also supports asmaller number of multidisciplinary research centers aimedat elucidating the molecular pathophysiology and improv-ing the quality of life for patients with CF. Similar care and research centers are found in Canada and in many European countries.

HISTORICAL PERSPECTIVES

Cystic fibrosis was first described as a distinct clinical entityin the late 1930s. However, numerous references to infantsand children with meconium ileus and characteristic pan-creatic and lung diseases are sprinkled throughout the lit-erature from as early as 1650. Of interest are references inEuropean folklore to the association of salty skin and earlydemise.3 Dorothy Andersen, a pathologist at Babies Hospi-tal in New York City, is usually credited with the first com-prehensive description of CF in 1938.4 She coined the termcystic fibrosis of the pancreas. In 1945, Farber suggested thatCF is a disease of exocrine glands, characterized largely byfailure to clear their mucous secretory product.5 He intro-duced the term mucoviscidosis, which was used for a numberof years. Chronic infection of the lungs was recognized earlyas a major contributing factor, and antibiotics were first usedfor the treatment of CF in the 1940s. At the same time, an autosomal-recessive inheritance pattern for CF was

38

IntroductionHistorical PerspectivesEpidemiologyGenetic BasisPathology

Lung PathologyOther Respiratory Tract PathologyNonrespiratory Pathologic Features

PathophysiologyCystic Fibrosis Transmembrane Regulator Protein:

Structure, Metabolism, and FunctionAbnormal Airway Mucosal Salt and Water TransportActive Ion Transport Properties of Airway EpitheliaMucin Macromolecule Secretion in the Cystic Fibrosis

AirwayPathophysiology of Infection

Clinical ManifestationsLower Respiratory Tract DiseaseUpper Respiratory Tract DiseaseComplications of Respiratory Tract DiseaseGastrointestinal ManifestationsPancreatic DiseaseHepatobiliary DiseaseGenitourinary Tract AbnormalitiesSweat Gland Dysfunction

DiagnosisTreatment

Ambulatory CareHospital Therapy

Course of the Disease and PrognosisSummary

1217

Cystic FibrosisRichard C. Boucher, M.D., Michael R. Knowles, M.D.,

James R. Yankaskas, M.D.

38.qxd 1/25/05 2:47 PM Page 1217

Page 2: Cystic Fibrosis

1218 Section J • OBSTRUCTIVE DISEASES

suggested by Andersen and Hodges.6 In 1953, di Sant’Ag-nese and colleagues investigated salt depletion in childrenwith CF during a summertime heat wave and concludedthat excessive loss of salt occurred via sweat.7 Subsequently,they documented that sodium and chloride levels in sweatare elevated in virtually all (>98%) persons with CF. Thisobservation led to a description by Gibson and Cooke ofthe pilocarpine iontophoresis method for sweat testing,8 amethod that remains the diagnostic standard to this day. Bythe late 1950s, CF was reported occasionally in older chil-dren and young adults. Soon thereafter, comprehensive andaggressive approaches to the care of patients were institutedin many treatment centers, and these approaches have beencredited with the survival into adulthood of a steadilyincreasing number of patients with CF. In the past 40 years,a markedly refined description of the CF syndrome and themany related complications has emerged.

Several recent observations have resulted in partial under-standing of CF pathogenesis at a molecular level. In theearly 1980s, epithelial physiologists described abnormalitiesof both sodium and chloride transport by CF respiratoryepithelia9 and the chloride impermeability of sweat glandducts in patients with CF.10 These observations focusedattention on a pathogenetic role for abnormal electrolyteand water movement across CF epithelia. From 1985 to1987, geneticists, using restriction fragment length poly-morphism analysis, located the CFTR gene on the long armof chromosome 7.11–14 Shortly thereafter, the CFTR genewas isolated, cloned, and sequenced,15 and the major muta-tion of this gene was characterized.16 Transfer of a wild-type(normal) gene into CF cells corrected the chloride trans-port defect.17,18 The product of the CF gene, the CFTR,was studied and found to be both a Cl- channel19–21 and aregulator of other channels.22,23 Studies of the metabolismof CFTR suggested that mutations could lead to abnormalfolding and mislocation of the protein.24–26 Knockout of theCFTR gene in transgenic mice has provided an animalmodel that possesses several physiologic and clinical simi-larities to human CF.27–30 These observations provided adetailed understanding of CFTR structure and function andhave laid the groundwork for development of more specifictherapeutic interventions, including gene therapy.

EPIDEMIOLOGY

Cystic fibrosis is recognized in approximately 1:250031 and1:17,00032 live births in white and black populations,respectively, in the United States. The range of reportedincidence figures worldwide varies from 1:56933 in a confined Ohio Amish population to 1:90,000 in an Asianpopulation of Hawaii.34

Generally, mutations of the CF gene are most prevalentin northern and central Europeans and in persons whoderive from these areas. An intermediate incidence is likelyalthough less well documented in non-European whites. CFis considered rare in American Indians, Asian populations,and black natives of Africa. It has been suggested that therelatively low frequency in populations living in tropical andsemitropical geographic locations is related to adverse con-sequences in the past from excessive salt loss in heterozy-gotes as well as homozygotes for the CF gene. In white

populations, 2% to 5% are carriers of a CF gene mutation.These people have no clinical stigmas of CF. Although anumber of chemical or physiologic alterations have beendescribed in heterozygotes, these alterations can be identi-fied only on a statistical basis.

GENETIC BASIS

Cystic fibrosis is an autosomal-recessive trait resulting frommutations at a single gene locus on the long arm of chro-mosome 7.2,31 This locus spans approximately 250 kB ofDNA, contains at least 27 exons, and codes for a largeprotein that has several transmembrane domains, two cyto-plasmic nucleotide (ATP) binding folds, and numerousphosphorylation sites containing a cytoplasmic regulatory(R) domain (Fig. 38.1). The primary and secondary struc-ture of the protein product of the CF gene resembles othermembrane proteins that act as pumps [e.g., the ATP-binding cassette (ABC) transporters].15

The predominant CFTR mutation is a 3-bp deletion thateliminates the phenylalanine of CFTR at position 508, theso-called DF508 mutation.16 This deletion has been detectedin 66% of more than 20,000 CF patient chromosomes analyzed worldwide,2,31,35 but its prevalence varies consider-ably from population to population (Table 38.1). Ingeneral, DF508 is more prevalent in northern Europeanthan southern European or in Middle Eastern popula-tions.31 More than 1000 other mutations of the CF genehave been reported but all at a relatively low frequency. The occurrence of known mutations accounts for only 90%of all CF gene abnormalities.31 CFTR mutations includeother deletions, missense mutations, nonsense mutations,frameshift mutations, and introduction of new splice sites.31

Correlations between genotype and phenotype are begin-ning to emerge. For example, homozygosity for the DF508mutation almost always confers exocrine pancreatic insuffi-ciency.32 A severe phenotype, including meconium ileus andliver disease, is strongly associated with the presence of two“severe” (i.e., pancreatic exocrine insufficiency) alleles.36–38

A region on human chromosome 19q13 has recently beenidentified as a modifier locus for meconium ileus.39 Con-flicting data exist on whether DF508 homozygosity versusother severe alleles is associated with a more severe form of

Table 38.1 Frequency of the DF508 Mutation

Cystic Fibrosis Population Chromosomes (%)

North American Caucasians 76

North American Hispanics 46

United Kingdom 74

Spain 49

Italy 43

Ashkenazi Jews 30

From Leinna WK, Feldman GL, Kerem B, et al: Mutation analysis forheterozygote detection and the prenatal diagnosis of cystic fibrosis. N Engl J Med 322:291–296, 1990.

38.qxd 1/25/05 2:47 PM Page 1218

Page 3: Cystic Fibrosis

chronic lung disease.33 On the other hand, “mild” muta-tions with some residual CFTR function and preservationof pancreatic exocrine function have been identified that areassociated with normal concentrations of sweat chloride,exocrine pancreatic sufficiency, or both.40,41 Moreover, vari-able phenotypes with “mono-organ” disease are also emerg-ing; for example, idiopathic pancreatitis and congenitalbilateral absence of the vas deferens are associated withmutations in the CFTR gene.42–44 In summary, it is nowclear that CF is indeed a syndrome caused by many combi-nations of mutations at a single gene locus, each of whichmay confer a slightly different phenotype.45 The ultimatephenotype of each person with CF is undoubtedly also influ-enced by genetic background (i.e., “modifier genes”46) aswell as postnatal environmental factors.

Deoxyribonucleic acid (DNA) analysis can now be usedto confirm the diagnosis, make prenatal diagnoses, andscreen for carrier status in selected cases. For example,probes for 28 of the most common CF mutations in NorthCarolina provided definitive diagnostic information in fewerthan 90% of persons with CF. Screening for CF carriersusing the DF508 probe identifies only 50% to 60% ofcouples at risk of having a CF child.47

The high frequency of CF gene mutations in many pop-ulations has been ascribed to an unknown heterozygoteadvantage. Some evidence suggests a reproductive advan-tage for the carrier state.48,49 Others have postulated thatreduced capacity to generate a secretory diarrhea inresponse to cholera infection because of diminished intes-tinal chloride transport may have provided a historical survival advantage to heterozygotes.50–52

PATHOLOGY

Soon after the original description of the CF syndrome,Farber pointed out the prominent accumulation of mucusin the respiratory and gastrointestinal tracts.5 Subsequently,mucus stasis has been described in numerous sites, includ-ing the conducting airways of the lung,53 paranasal sinuses,mucus-secreting salivary glands, apocrine sweat glands,small intestine, appendix, pancreas, biliary system, uterinecervix, and wolffian duct structures. However, the eccrinesweat gland, which figures prominently in the pathophysi-ology of CF, is morphologically normal at all ages. Patho-logic changes in the lung, which is the primary site of organdysfunction, also reflect chronic infection.

LUNG PATHOLOGY

It is clear that disease of the conducting airways in CF isacquired postnatally. The airways of children with CF whohave died within the first days of life display only subtleabnormalities. The earliest macroscopic pathologic lesion is reported to be mucus obstruction of bronchioles.53

However, the numbers and distribution of mucus-produc-ing goblet cells and the numbers and size of submucosalglands appear to be within normal ranges at birth. A carefulmorphometric analysis of CF airways early in life demon-strated dilation of submucosal gland acinar and ductallumens before reaction to chronic infection would beexpected.54 This finding suggests that either hypersecretionor, more likely, failure to clear secretions at an early ageaccounts for mucus accumulation in bronchial regions,

121938 • Cystic Fibrosis

R domain

Outside

NH2NBD1

DF508 Mutation

Chargedside chains

Protein kinase C

Protein kinase A

NBD2 CO2H

N-linkedcarbohydrate

Calculated net charge on the CFTR isindicated by color intensity. The darkestpurple is +12 and the darkest gray is –6.

NBD = Nucleotide Binding Domain

ATP bindingdomains

0 +6 +12–6

Figure 38.1 Proposed structure for the cystic fibrosis transmembrane regulator (CFTR) protein. Two repeat segments each consist ofsix transmembrane spans followed by a nucleotide-binding fold (NBF). The segments are joined by a highly charged region thatcontains multiple phosphorylation sites, the R domain. Much of the CFTR is intracytoplasmic. Glycosylation occurs on an extracellularloop of the second motif.

38.qxd 1/25/05 2:47 PM Page 1219

Page 4: Cystic Fibrosis

1220 Section J • OBSTRUCTIVE DISEASES

whereas the mucus retention in bronchioles presumablyreflects the failure to clear mucus secreted by surface secre-tory (goblet) cells. Failure to clear secretions from theairway lumens likely initiates infection.

With the progression of lung disease, evidence for bron-chiolitis and bronchitis becomes more prominent, the submucosal glands hypertrophy, and goblet cells not onlybecome more numerous but also propagate into the bron-chioles. Small airways may be completely obstructed bysecretions (Fig. 38.2).55 Bronchiolectasis and thenbronchiectasis are consequences of persistent obstruc-tion–infection cycles. Bronchiectasis had been thought tomanifest in the second decade of life but now is beingdetected earlier in life with increased use of computedtomography (CT) scans. Pneumonia, when present, gener-ally assumes a peribronchial pattern.

Detailed pathologic descriptions of lung disease are basedon examination of lungs at autopsy or lung transplant, andthey reflect advanced changes.56–59 Bronchiectatic cystsoccupy as much as 50% of the cross-sectional area of thelate-stage CF lung.60 In general, bronchiectasis is moresevere in upper lobes than in lower lobes. In addition todilation of the small airways, bronchioles may be stenoticor even obliterated. The extent of obliterative bronchiolitisappears to be directly correlated with age at death.57 Autop-sied lungs also show extensive overinflation of air spaces.Small amounts of destructive emphysema are seen in manypatients, especially those who have lived for two to threedecades.57 Absence of more extensive alveolar wall destruc-tion can be explained by the confinement of chronic infec-tion to conducting airways. Several patterns of interstitialpneumonia have also been described in autopsied lungs,including usual interstitial pneumonitis, interstitial pneu-monitis with organizing pneumonia, and diffuse alveolardamage.61 Fibrosis is extensive in peribronchiolar and peri-bronchial regions and may contribute to the restrictive lungfunction pattern that is superimposed on obstruction inend-stage lung disease.62 Subpleural cysts often occur on themediastinal surfaces of the upper lobes and are thought tobe related to the frequent occurrence of pneumothorax in

patients with advanced lung disease.63 The bronchial arteries become large and tortuous,64 contributing to apropensity for hemoptysis in ectatic airways. The pulmonaryarteries display varying degrees of change reflecting pul-monary hypertension.

OTHER RESPIRATORY TRACT PATHOLOGY

Hypertrophy and hyperplasia of secretory elements, mucusaccumulation, and chronic inflammatory changes are alsofeatures of the paranasal sinuses and the nasal passages. Acommon feature of nasal pathology is inflammatory edemaof the mucosa with subsequent pedunculation and forma-tion of polyps.65

NONRESPIRATORY PATHOLOGIC FEATURES

Most of the nonpulmonary pathology in CF occurs in thegastrointestinal tract and related organs. Striking changesare seen in the exocrine pancreas.66 Obstruction of ducts byinspissated secretions is an early feature, followed by dila-tion of secretory ducts and acini and flattening of the epithe-lium. Loss of acinar cells is widespread, and areas ofdestruction are replaced by fibrous tissue and fat. Intra-luminal calcifications may occur and may be recognizedroentgenographically. Small cysts are common and gener-ally represent dilated ducts. Inflammatory changes are notprominent. The islets of Langerhans are spared until laterperiods of life. Changes in the islets include disruption byfibrous tissue bands that may provide a barrier betweenhormone-secreting cells and the vascular spaces.67 Patho-logic changes in the pancreas are used occasionally to makea postmortem diagnosis in atypical or missed cases of CF.

The pancreas is abnormal in almost all patients with CFand is virtually destroyed in approximately 90% of CFpatients studied at autopsy. Liver changes are not as fre-quent or consistent.68 In 25% or more of all autopsies,islands of relatively normal parenchymal cells are divided byfibrotic bands, creating a distinctive multilobular appear-ance. Microscopically, this focal biliary cirrhosis is

A B

Figure 38.2 A, Hypertrophied submucosal gland in the trachea of an 18-year-old woman with cystic fibrosis is shown. Mucus-containing acini are distended. The gland occupies almost the entire thickness of the tracheal wall. B, Large and small bronchioles inthe lungs of a 21-year-old man with cystic fibrosis. These airways are completely obstructed with secretions and display chronicinflammation of the walls and surrounding tissues. Peribronchiolar fibrosis also can be demonstrated with appropriate stains. Airspace enlargement is prominent (right), but more normal-appearing peripheral lung architecture is present (left). (A, B: Hematoxylinand eosin stain; ¥42.)

38.qxd 1/25/05 2:47 PM Page 1220

Page 5: Cystic Fibrosis

characterized by inspissation of secretions within the bileductules, bile duct proliferation, inflammatory reaction, anda paucity of evidence for bile stasis. Approximately 30% ofpatients also display fatty infiltration of the liver, whichcannot always be ascribed to malnutrition. In approximately25% of cases, the gallbladder is hypoplastic and contains agelatinous mucus-like material. Cholelithiasis is an increas-ingly frequent observation.

Changes in the intestinal tract itself are less prominent.69

Brunner’s glands of the duodenum are hypertrophied, withdilated ducts and acinar lumens filled with mucus. There islittle if any primary change of the small intestinal tractmucosa. The appendix frequently displays goblet cell hyper-plasia of the epithelium and accumulation of secretionswithin crypts and in the lumen, changes that may be diag-nostic of CF. A number of investigators have claimed theability to diagnose CF by rectal biopsy based on goblet cell hyperplasia and accumulation of mucus in the crypts.However, subsequent studies have demonstrated that thesefindings are not consistent in the rectal mucosa of patientswith CF.70

Reproductive abnormalities are frequent and characteris-tic.71 In a large majority of male patients with CF, the vasdeferens, the tail and body of the epididymis, and seminalvesicles are obliterated by fibrous tissue or are completelyabsent. Women with CF have mucus-distended uterine cer-vical glands and plugging of the cervical canal with tena-cious mucus secretions. Endocervicitis is a frequent finding.

Non-mucus-secreting glands, such as the sweat glandsand the parotid salivary glands, have normal gross and his-tologic appearances. Mucus-secreting salivary glands areusually somewhat enlarged, contain prominent mucus-secreting cells, and display a varied amount of mucus plug-ging in duct lumens.

PATHOPHYSIOLOGY

CYSTIC FIBROSIS TRANSMEMBRANE REGULATORPROTEIN: STRUCTURE, METABOLISM, AND FUNCTION

The CFTR is a single peptide chain that contains 1480amino acids. The calculated molecular weight is 170 kD, butCFTR migrates in its mature form as an approximately 180-kD glycoprotein due to variable N-glycosylation.31,72 Figure38.1 demonstrates the domain structure of CFTR, with tworepeat motifs, each consisting of six transmembrane span-ning segments followed by a consensus nucleotide-bindingfold (ATP-binding region). These two segments are linkedby a highly charged region, the R (regulatory) domain,which contains multiple phosphorylation sites.15 The N andC termini contain important motifs that mediate protein–protein interactions that position CFTR within specializedcellular domains.73 The CFTR structure closely resemblesthat of a number of other solute transport proteins and placesthe CF gene product in the ABC superfamily of proteins.74

CFTR mRNA expression is largely restricted to epithelialcells and is found at moderate levels in human pancreas, sali-vary glands, sweat glands, intestine, and the reproductivetract.15,72 In the lung, CFTR mRNA is expressed at lowlevels in the surface epithelium of the airways and perhapsat higher levels within certain regions of submucosal

glands.75 These sites of CFTR mRNA expression are con-sistent with locations of tissue dysfunction in patients withCF. Biochemical and immunohistochemical studies describean apical membrane location for CFTR in most epithelia,although CFTR may also be inserted into the basolateralmembrane of sweat ductal epithelia.76

Most studies indicate that a central feature of the molecu-lar pathophysiology of CF is that mutated CFTR (e.g.,DF508) is not normally folded and processed; hence it doesnot reach the Golgi or apical plasma membranes (Fig.38.3).24,77 The abnormal CFTR DF508 trafficking is temperature-sensitive, and some degree of normal process-ing and delivery of CFTR DF508 to the plasma membranecan be detected when temperatures are lowered.78 Most(>80%) of the other identified CFTR mutations also producea protein that is not normally processed.79 Some studies sug-gested that abnormal folding of DF508 may not occur in allaffected tissues in vivo, including the airways and colonicepithelia.80,81 However, more recent studies indicate that theprocessing defect is likely a feature of these epithelia in vivo.82

Abnormal processing is not characteristic of all CFTRmutations.79,83 Mutant CFTR may reach the plasma mem-brane but be refractory to regulatory influences or exhibitconduction abnormalities as a consequence of several typesof CFTR gene mutations.84

The CFTR protein appears to have multiple functions(Fig. 38.3). It serves as a Cl- channel,20,21 modulates theactivity of other plasma membrane channels,22,23 and perhapsregulates exocytotic and endocytotic events.85,86 AlthoughCFTR resembles known solute pumps, as yet CFTR has notbeen shown to function as a pump. Its chloride channelactivity (open probability, or Po) is regulated through cyclicadenosine monophosphate (cAMP)-dependent kinase,

122138 • Cystic Fibrosis

Lumen

Blood

Golgi

RER

Nuclei

cAMP

Otherchannel(s)

Cl-CFTR +/–

+

Normal cell

CFTR

Otherchannel(s)

Cystic fibrosis cell

Figure 38.3 Schema of biogenesis of CFTR in a normal cell (L)and a DF508 CF cell (R). In the normal cell, the mature CFTRpolypeptide has been synthesized in the rough endoplasmicreticulum (RER), glycosylated in the Golgi, and localized to theapical membrane to function as a cAMP-regulated Cl- channeland regulator of other channels, e.g., as an inhibitor of theepithelial Na+ channel (ENaC). In the CF cell, the DF508 CFTRpolypeptide has misfolded and been degraded by intracellularproteolytic airways. Hence, it is not positioned in the apicalmembrane to act as a cAMP-regulated Cl- channel or to inhibitENaC.

38.qxd 1/25/05 2:47 PM Page 1221

Page 6: Cystic Fibrosis

1222 Section J • OBSTRUCTIVE DISEASES

C-kinase, and tyrosine kinase pathways.87–89 Phosphorylationof R-domain serine residues and ATP binding and hydroly-sis by the nucleotide binding folds are necessary, independ-ent steps in the activation of chloride conductance.90,91

ABNORMAL AIRWAY MUCOSAL SALT AND WATER TRANSPORT

Cystic fibrosis is a disease that affects the epithelia liningvarious organs of the body. Because the native functions ofaffected epithelia in these organs vary widely—some are salt-absorbing but not volume-absorbing (sweat duct), some are volume-absorbing (airways), and others are volume-secreting (pancreas)—it is not surprising that the physio-logic consequences of mutant CFTR in these epithelia differwidely.

In the lung, airway but not alveolar epithelia are primarilyaffected. It is not yet clear whether the predominant patho-physiology of CF in the airway occurs in the superficial

epithelium or the glandular elements in the airway wall. Thebest studied defects in CF airway epithelia are the Na+ andCl- transport abnormalities identified in the superficialepithelia covering nasal and tracheobronchial surfaces.9,92

The likelihood that the superficial epithelium may be themore important affected epithelium in the lung is suggestedby data indicating that the earliest functional manifestationin the CF lung is airflow obstruction in the bronchioles, aregion that contains no glands.93 For discussion of the speci-ficity of tests of function in the small airways, see Chapter 24.

A classic premise of CF research has been that abnor-malities of Cl- and likely Na+ transport lead to abnormalregulation of airway surface liquid (ASL) volume, that is,depletion of both salt and water (volume) on the CF airwaysurface (Fig. 38.4). As a result of ASL volume reduction,mucus clearance mechanisms are impaired, which causesairflow obstruction and ultimately bacterial infection. Asecond hypothesis linking abnormal Na+ and Cl- trans-port to CF pathogenesis focused on differences in salt

7 µm

7 µm

Basal state

Cystic fibrosis

Basal state

MCT

NaCl + waterabsorption

NaCl + waterhyperabsorption

Cystic fibrosis

MCT

Figure 38.4 Schema describing the mucociliary transport (MCT) apparatus in normal and cystic fibrosis airway epithelia. In normalairways (upper left panel), the volume and composition of airway surface liquids (stars between cilia) are adjusted so the extendedcilium can transmit energy to the mucus layer that rests on the surface of the periciliary liquid layer. The height (volume) and thecomposition of this liquid layer are regulated by the superficial epithelial cells. The low-power electron micrograph (upper rightpanel) shows the 7 mm deep periciliary liquid layer on the surface of a cultured normal airway epithelium that exhibits mucustransport.98 In cystic fibrosis airways (lower left panel), hyperabsorption of volume in response to excessive Na+ transport depletesthe height (volume) of the airway surface periciliary layer. It is postulated that this depletion of liquid contributes to slower airwayclearance in cystic fibrosis by impacting mucus on cilial shafts and by altering cough clearance properties of mucus. The low powerelectron micrograph (lower right panel) shows the depleted periciliary layer on the surface of a CF airway epithelial culture that haslost the ability to perform mucus transport.98 Other composition abnormalities of the periciliary secretory layer also may be importantbut are as yet unidentified.

38.qxd 1/25/05 2:47 PM Page 1222

Page 7: Cystic Fibrosis

concentration and activity of antimicrobial factors (e.g.,defensins) in CF versus normal ASL.94,95 This hypothesissuggests that airway epithelia regulate the ionic concentra-tion, not volume, of ASL, and NaCl concentrations are pre-dicted to be low (<50 mmol/L) in the airway surface liquidsof normal individuals and “high” in CF (>100 mmol/L).Defensins are active in low-salt ASL, whereas they are inac-tive in high-salt ASL. The result of inactive defensins in CFwould be chronic infection. Table 38.2 describes the ASLcomposition in normal subjects and CF subjects prior toinfection.143,398 These data do not support a difference inASL NaCl composition as central to the initiation of CFlung disease. Furthermore, defensins are found in very smallquantities in both normal and CF ASL.96

As discussed in Chapter 12, the role of active ion systemsin the pulmonary epithelium can be reviewed on two levels:(1) the requirement of pulmonary epithelium to coordinateintrapulmonary surface liquid volume during “axial” (i.e.,surface) flow between different pulmonary regions, such asalveolar and airway surfaces; and (2) the requirement toregulate the surface-liquid volume in local microenviron-ments to mediate efficient mucus transport. Recent studieshave increased the level of understanding of how airwayepithelia normally regulate ASL volume under a variety ofconditions and how CF airway epithelia fail to do so. Forexample, confocal microscopy studies of well differentiatedhuman bronchial epithelial cultures that exhibit rotationalmucus transport have revealed that the mucus and peri-ciliary liquid (PCL) layers are both transported axially,imposing a requirement for volume absorption as ASL istransported cephalad in vivo.97 Further studies with thissystem revealed that net volume hyperabsorption is a featureof CF airway epithelia and abolishes mucus transport, bothby depletion of the 7 mm PCL layer and concentration ofmucins in the mucus layer.98 The depletion of PCL is a“severe” lesion, as it is predicted to hinder both cilial-dependent and cough-dependent mucus clearance. Consis-tent with this prediction, the evidence that mucociliary andparticularly cough clearances are depressed in CF patientsin vivo is mounting.99,100

ACTIVE ION TRANSPORT PROPERTIES OF AIRWAY EPITHELIA

Normal airway epithelia exhibit the capacity to activelyabsorb Na+ and actively secrete Cl- ions. Each of these

processes is electrogenic, and they are coordinately regu-lated.101 The relative physiologic role of each process variesdepending on the volume of liquid on airway surfaces.

A parameter that describes the overall airway transportactivity of airway epithelia is the transepithelial electricpotential difference (PD). The transepithelial PD can beanalyzed simply as the product of the active ion transportrate [short-circuit current (Isc)] and the ionic resistance ofthe epithelium (Rt). A diagnostic hallmark of CF airwayepithelia is a raised PD. A series of in vivo ion substitutionstudies,9 complemented by in vitro isotopic flux studies ofexcised tissues,9,92,102 revealed that the raised PD in CFairway epithelia reflects contributions of both increased iontransport (Na+ absorption) and increased epithelial resist-ance (due to decreased cellular Cl- permeability). Indirectevidence from studies measuring the effects of amiloride, aNa+ channel blocker, in vivo indicates that the elevated Na+

absorptive rate rather than the defective Cl- conductancedominates the resting electric abnormalities of the superfi-cial epithelium of CF airways.

Abnormalities of Superficial Epithelial Cl- Transport

In vivo PD,9 Ussing flux chamber,102 and microelectrodestudies103–107 all agree that CF airway epithelial cells exhibitan abnormally low apical membrane conductive Cl- perme-ability. Studies utilizing double-barreled Cl--selectivemicroelectrodes indicate that the basolateral membrane Cl-

conductance and Na+/K+/2Cl- cotransport activities of CFcells are not different from those of normal cells.105,106,108

The abnormality in the resting apical membrane Cl- con-ductance of airway epithelia reflects the absence of tonicCFTR function at this site. Like other affected epithelia, CFairway epithelia do not respond with Cl- secretion to agentsthat raise cellular cAMP (e.g., beta-agonists), whereasnormal airway epithelia do, if the apical membrane Na+ per-meability is blocked.88,92,109–112 This abnormal response inCF airway epithelia does not reflect dysfunction of adeny-lyl cyclase or cAMP-dependent protein kinase activities.Rather, CF airway epithelial cells fail to secrete Cl- due to the absence of CFTR in the apical membrane or to intrinsic defects in mutant CFTR.

Unlike normal airway epithelia, CF airway epithelia alsodo not respond with Cl- secretion to activators of proteinkinase C (PKC).88,111,113,114 Studies with the expressed CFTRprotein have indicated that the CFTR Cl- channel is directly

122338 • Cystic Fibrosis

Table 38.2 Composition of Airway Surface Liquid in Normal Subjects and Uninfected Cystic Fibrosis Patients

Region/Phenotype Study No. Na+ (mmol/L) Cl- (mmol/L) K+ (mmol/L)

Normal nose Knowles143 8 109 ± 5 126 ± 5 29 ± 3Hull398 10 106 ± 4 115 ± 4 ND

CF nose Knowles143 8 110 ± 6 132 ± 5 28 ± 3Hull398 10 116 ± 7 125 ± 4 ND

Normal lower airway Knowles143 11 91 ± 7 81 ± 6 17 ± 3Hull398 7 85 ± 10 108 ± 5 ND

CF lower airway Knowles143 8 87 ± 4 77 ± 6 21 ± 2Hull398 5 78 ± 16 77 ± 7 ND

38.qxd 1/25/05 2:47 PM Page 1223

Page 8: Cystic Fibrosis

1224 Section J • OBSTRUCTIVE DISEASES

regulated by PKC phosphorylation.115 Thus, the failure ofCF airway epithelia to respond to PKC also reflects absenceor dysfunction of the mutated CFTR Cl- channel itself. Anarea of intense interest at present is how CFTR is func-tionally and physically associated with regulatory kinasesand phosphatases in the apical domains of epithelialcells.116,117

Recent studies have placed in perspective the function ofdefective CFTR-mediated Cl- transport in airway epithelia.In normal airway epithelia, the maintenance of a normalASL volume depends on both the regulation of Na+ trans-port rates (see later) and the ability to secrete Cl- (Fig.38.5). Specifically, as ASL volume diminishes to about

7 mm, Na+ absorption is inhibited and CFTR-mediated Cl- secretion is initiated. In CF, the absence of CFTR-dependent Cl- secretion contributes to the failure to main-tain an adequate ASL volume on airway surfaces.

The CF airway epithelial cells retain their normal capac-ity to secrete Cl- in response to elevations in intracellularCa2+.118,119 Data derived from studies of cultured monolay-ers impaled with double-barreled Cl- selective microelec-trodes indicate that elevation of intracellular Ca2+ withcalcium ionphores (e.g., ionomycin) initiate Cl- secretion in both CF and normal cells. The Cl- channel activated byraised Ca2+ must be molecularly distinct from CFTR, as evi-denced by retention of this activity in gene-targeted (“CF”)

A

High/NormalASL Volume

Na+ Cl-

Na+ Na+ 2Cl-K+

K+

H2OAbsorption

Secretion

~

ENaC CaCCCFTR

Va = ~-30 mVDFaCl- = 0 mV

Low ASL Volume

Na+Cl-

Na+ Na+ 2Cl-K+

K+

H2O

~

ENaC CaCCCFTR

Va = -35 mVDFaCl- = -5 mV

B

High/NormalASL Volume

Na+ Cl-

Na+ Na+ 2Cl-K+

K+

H2O

Absorption

~

ENaC CaCC

Cl-CFTR

Low

Na+

K+K+ K+

~K+

Na+ Cl-

Na+ Na+ 2Cl-K+

K+

H2O

~

ENaC CaCC

Cl-CFTR

Na+

K+

~K+

Figure 38.5 A, Normal human airway epithelia can switch between absorptive and secretory ion transport modes. Left. When excessairway surface liquid is present, epithelial Na+ channel (ENaC)-mediated Na+ absorption is dominant. Cl- is projected to be absorbedpassively via the paracellular path, reflecting the fact that there is no electrochemical driving force (DFa

Cl-) favoring Cl- exit from thecell. In contrast, both the negative apical membrane potential (Va) and low intracellular Na+ activity (-20 mM) favor Na+ entry intothe cell. Water is absorbed in response to the osmotic gradients generated by combined NaCl absorption. Right. When the ASL volumeis low, ENaC is inhibited, the apical membrane potential becomes more negative, and a driving force for Cl- secretion is generated.NaCl and water are thus secreted. B, CF airway epithelia exhibit accelerated ASL volume absorption under “high/normal” ASL volumeconditions and cannot switch to a secretory mode during low ASL volume conditions. Left. Routes of raised Na+, Cl-, and H2Oabsorption and cellular mechanisms for raised Na+ transport in CF airway epithelia under high/normal ASL volume conditions. Right.The absence of CFTR from the apical membrane eliminates regulation of the Na+ channel (ENaC) and limits capacity for Cl- secretion.

38.qxd 1/25/05 2:47 PM Page 1224

Page 9: Cystic Fibrosis

mice.120 This Ca2+-activated Cl- channel (CaCC), however,has still not been adequately described biophysically withsingle-channel (patch electrode) techniques. It has also not been resolved whether CaCC responds to raised Ca2+

itself or requires participation of a kinase (e.g., a Ca2+

calmodulin-dependent kinase).118,119,121,122

The observation that Ca2+-regulated Cl- secretion can betriggered in CF airway epithelia has therapeutic implications.In a search for compounds that interact with receptors onthe lumen of airway epithelial cells that regulate intracellularCa2+ levels, it was discovered that the triphosphatenucleotides [ATP and uridine triphosphate (UTP)] werehighly effective in initiating Cl- secretion in CF and normalairway epithelial cells.123–126 Triphosphate nucleotidesincrease intracellular Ca2+ by interacting with purinoceptors(P2Y2) that, via activation of phospholipase C, increase inos-itol triphosphate levels. In vivo studies found that ATP andUTP were effective Cl- secretagogues in the nasal mucosa ofCF subjects.125 Indeed, an intriguing finding was that thetriphosphate nucleotides were more effective in initiating Cl-

secretion in CF than in normal subjects. These observationshave led to the clinical testing of analogues of UTP that byvirtue of being resistant to hydrolysis in CF airways may havea long duration of action.127,128

Abnormalities of Superficial Epithelial Na+ Transport

Abnormalities in Na+ transport in CF appear to be confinedto epithelia that are involved in volume absorption. Thus,data have been reported that accelerated Na+ absorption isa feature of the proximal tubule of the kidney,129,130 thejejunum and the ileum of the small intestine,131–133 and theairways of CF patients.92 Interestingly, the mechanisms ofNa+ hyperabsorption may differ in the three affected epithe-lia. Na+-coupled solute movements may be the majoraffected pathways in the intestine, whereas defective regu-lation of the epithelial Na+ channel (ENaC) is the majorabnormality in the lung.

Studies of freshly excised airway specimens establishedthat the raised PD that characterizes CF airway epitheliareflects in part an accelerated basal rate of active Na+ absorp-tion.103 These studies also identified a defect in acute regu-lation of Na+ transport rates in CF.103 Beta-agonists andother reagents that raise intracellular cAMP paradoxicallyraise the already accelerated basal rate of Na+ transport inCF tissues but do not affect Na+ transport rates in normaltissues.103

Studies with microelectrodes in freshly excised tissuesindicated that the principal abnormality in the Na+ trans-port path in CF airway epithelia resided in the apical mem-brane Na+ conductance.103 Studies of cultured airwayepithelia with double-barreled Na+ selective microelectrodesshowed that the apical membrane Na+ permeability in CFwas twofold to threefold higher than in normal cells.134,135

Thus, these studies placed a second ion permeability abnor-mality in the CF apical membrane and strongly suggestedthat the Na+ transport abnormality was not secondary to the changes in the electrochemical driving forces for Na+

entry consequent to the absence of apical membrane Cl-

permeability.The effects of the CF defect on the regulation of Na+

transport rates at the single-channel level are not under-

stood. The available evidence indicates that the fraction oftime the Na+ channel is capable of conducting ions (Po) isapproximately threefold greater in CF than in normalcells.136–138 This finding suggests that the defect responsiblefor accelerated Na+ transport in airway epithelia reflects anabsence of the tonic inhibition of ENaC provided by wild-type CFTR.

A key issue with respect to the Na+ transport defect relatesto its role in the pathogenesis of airway infection. As dis-cussed above, recent data indicate that both regulation ofNa+ transport rates as a function of the available volume onan airway surface and the capacity to secrete Cl- are requiredfor ASL volume homeostasis (Fig. 38.5). Furthermore,these studies have shown that CF airways fail to normallyregulate Na+ absorption, and this defect contributes toabnormally low ASL volumes on CF airway surfaces. Forexample, in vitro studies of cultured CF bronchial epitheliahave shown that in CF Na+ hyperabsorption persists despitelow ASL volume, and that this defect contributes to PCLdepletion and mucus stasis (Fig. 38.6). Further evidence forthe role of increased Na+ absorption in the pathogenesis ofCF has emerged from in vivo studies of mice with airway-specific overexpression of ENaC. These mice exhibitedincreased Na+ absorption and a lung phenotype that resem-bles CF (e.g., mucus obstruction, neutrophilic inflamma-tion, and defects in bacterial clearance).139 The recentreports that nucleotides also slow Na+ transport in CFairways140,141 suggest that data from therapeutic trials withthese agents may be informative with respect to the role ofNa+ absorption in CF lung disease.

Ion Transport Defects of Glandular Epithelium

CFTR may be expressed at high levels in certain regions ofproximal airway glands. In situ hybridization studies indi-cate that lung CFTR is expressed at the highest level insome serous cells contained in submucosal gland acini anda minor cell type in the ductal epithelium, the so-calledoncocyte.142

In vivo studies of nasal gland secretion have suggested asimilar capacity for gland acini to secrete liquid volume inCF and normal subjects in vivo.143 Furthermore, thesestudies raised the possibility that submucosal gland ductsmodify the gland secretion so it is hypotonic (i.e., has areduced NaCl concentration). Unlike the sweat duct, butsimilar to the parotid duct, the functions of the ductalepithelium in nasal glands of CF patients appear similar tothose in normal subjects.

In vitro studies indicate that cultured CF bronchial glandacinar cells, unlike the superficial airway epithelium of CFsubjects, do not respond to raised intracellular Ca2+ with Cl-

secretion, whereas normal glandular cells do. Thus, thebronchial glands could have a Cl- transport phenotype thatis more similar to gastrointestinal epithelia than the super-ficial airway epithelium.133,144,145 Studies of bronchial glandshave more recently been performed on freshly excisedpreparations from lung transplant procedures. CF airwaygland acini, like superficial airway epithelia, do not secreteCl- in response to beta-agonists, whereas glands fromnormal subjects exhibit moderate responses.144,145 Interest-ingly, both normal and CF glands secrete Cl- in response to cholinergic agents, suggesting that Ca2+-regulated Cl-

122538 • Cystic Fibrosis

38.qxd 1/25/05 2:47 PM Page 1225

Page 10: Cystic Fibrosis

1226 Section J • OBSTRUCTIVE DISEASES

secretion is preserved in freshly excised gland preparations.The predicted functional correlate of these data is that CFglands fail to secrete sufficient volume to normally hydratemucins secreted by glands or, possibly, hydrate the airwaysurface. The relative role of the gland defect in CF ASLvolume depletion is presently unknown, but at a minimumthis deficiency would serve to exacerbate the ASL volumedefect generated by the CF superficial epithelium.

MUCIN MACROMOLECULE SECRETION IN THE CYSTICFIBROSIS AIRWAY

Following Farber’s report5 of mucus obstruction in manyexocrine glands, a search was initiated for abnormal macro-molecules that might change the physical properties of CFexocrine secretions. Particular emphasis was placed onanalysis of mucin glycoproteins, the high-molecular-weightglycoconjugates that are the determinants of the viscoelas-tic properties of mucus.146 One of the difficulties in com-paring these substances sampled from CF and controlairways is that they are susceptible to proteolytic andperhaps other degradative enzymes to which they areexposed in vivo. In addition, these glycoproteins displayextensive microheterogeneity of carbohydrate constituentsand, therefore, are difficult to compare from subject tosubject as well as patient group to control group.

Overall, purified mucins from CF patients and controlsubjects show considerable similarity. Early suggestions thatthe fucose content of CF glycoproteins was elevated and thesialic acid content decreased appears to reflect the effects ofchronic infection and inflammation and is not a CF-specificdefect.147 In addition, several investigators have reportedthat CF mucus glycoproteins from both respiratory and gas-trointestinal sources contained increased numbers of sul-fated sugars.148–151 Goblet cells in the bronchial epitheliumof patients with CF have an elevated sulfur content,152

which, if present as sulfate, could reflect increased sulfationof secretory glycoconjugates. However, again, most evi-dence suggests that increased sulfation likely is a reflectionof chronic infection rather than a consequence of CFTRmutations.147

There is also no convincing evidence that CF epithelialcells hypersecrete mucin macromolecules before the onset ofairway injury.153 However, hypersecretion of mucus doesoccur in response to proteases, oxidative metabolites, and other products of chronic infection and inflammation.This induced hypersecretion, coupled with goblet cell

A

pO2

B

pO2

C

pO2

Figure 38.6 A, Failure of mucus transport coupled withpersistent mucin secretion over time produces adherent mucusmasses/plugs. The raised epithelial QO2 required to fuelaccelerated CF Na+ absorption generates steep oxygen gradients(red Æ blue stippling in bar) in thickened mucus masses. B, Pseudomonas aeruginosa deposited on mucus masses growsand adapts to hypoxic niches within these masses asmacrocolonies (biofilms). C, The thickened mucus, coupled withmacrocolony growth, resists secondary defenses, includingneutrophils, setting the stage for chronic infection. Thepresence of increased macrocolony density and, to a lesserextent, neutrophils renders these mucopurulent masses franklyanaerobic (blue bar).

38.qxd 1/25/05 2:47 PM Page 1226

Page 11: Cystic Fibrosis

hyperplasia, likely significantly exacerbates the mucus accu-mulation and obstruction that is initiated by ASL volumedepletion.154

In the future, an important area of research will focus on the behavior of mucins in reduced (“low”) ASL volumeenvironments. Of particular interest are studies on thenature of the adhesive interactions that may occur when a“thickened” (concentrated) mucus layer comes into contactwith cell surface (tethered) mucins in the absence of PCL(Fig. 38.2). Elucidation of the mechanism of this interac-tion may lead to therapies to reverse this adhesion and allowclearance of adherent mucus plaques.

Analyses of other macromolecular secretory productsfrom patients with CF have failed to uncover substantialabnormalities. These macromolecules have included alpha-amylase, ribonuclease, and lysozyme from salivary secretionsand pancreatic enzymes, including chymotrypsin, trypsin,carboxypeptidase, amylase, and lipase.

PATHOPHYSIOLOGY OF INFECTION

Failure of the airways to clear mucus normally is likely theprimary pathophysiologic event in CF (Fig. 38.6). Adher-ent mucus plaques and plugs are the sites of the chronicbacterial infection of CF airways.155 These bacteria areusually found in macrocolonies that appear to represent thebiofilm type of growth characteristic of CF infections. Ingeneral, biofilms represent a type of bacterial growth inwhich bacteria form and become enmeshed in an extensiveextracellular matrix. Bacterial cellular physiology maychange in this environment, and the matrix may provide abarrier to antimicrobial agents and inflammatory cells,thereby promoting bacterial persistence. Generation ofthese immobile mucus surfaces thus provides the nidus forthe chronic respiratory tract infection that is the major con-tributor to the loss of lung function and eventual demise ofpatients with CF.

The chronic infection of the CF lung usually is localizedto endobronchial regions, and the bacterial species mostcommonly cultured are Staphylococcus aureus andPseudomonas aeruginosa. Once established, infection of theCF lung is rarely eradicated. Bacterial infection extendingbeyond the airways, however, is distinctly uncommon.Therefore, local airway defense mechanisms, rather thangeneralized host or pulmonary defense mechanisms, appearto be impaired in CF.

As reviewed above, the old concept that a generalizedfailure of mucus clearance from airways is the principal airwaydefense mechanism degraded in CF has been supplementedwith new clinical and laboratory data. For example, studies of airway mucus transport velocity had found that clearanceof inhaled radiotracers from the central airways is usuallyreduced.100 More recent studies of mucus clearance from themore peripheral airways, the site of early disease in CF, havefound that clearance is routinely reduced in this region.156

Virtually all studies agree that ciliary morphology and beatfrequency in CF airway epithelia are normal.157,158 In con-trast, data from model systems indicate that the volume ofliquid on CF airway surfaces is severely reduced.98 Thus, it islikely that ASL volume depletion, by both removing the PCLand concentrating the mucus layer, leads to failure of bothmucociliary and cough clearance. The resulting mucus stasis,

adhesion, and obstruction likely are responsible for thesevere, unrelenting lung infections (Fig. 38.6).

Staphylococcus aureus and P. aeruginosa are infrequent res-piratory tract pathogens of children and adults, except inCF. The role played by these organisms in endobronchialinfection has prompted suggestions that surface propertiesof CF airway epithelia are altered in a fashion that promotestheir adherence. S. aureus does not preferentially adhere toCF airway epithelial cells,159 and studies of respiratory cellinteractions with P. aeruginosa also have not demonstratedconsistent differences between CF and normal cells.160,161 Inan opposite formulation, recent studies have suggested thatthe wild-type CFTR expressed on normal epithelial cellsbinds Pseudomonas and mediates bacterial internalization,which ultimately kills the microorganism. CF cells report-edly fail to perform this function.162 Although intriguing,there is little pathologic evidence for P. aeruginosa inter-nalization in normal well differentiated airway epithelialcells, and this hypothesis does not account for infection inCF patients who express mutant CFTR in the apical mem-brane (e.g., G551D CFTR).

As noted above, there is increasing evidence that CFairway infection reflects an infection of the mucus layer,forming biofilms of bacteria that are resistant to the host’sdefense.155 No differences in bacterial binding to CF versusnormal mucins have been detected for Staphylococcus orPseudomonas.159 Rather, it is likely that it is increased bac-terial mucus adherence due to nonspecific increases in“tackiness” of the mucins concentrated by ASL volumedepletion that accounts for the presence of these bacteria inmucus. In addition, there may be a unique interactionbetween P. aeruginosa and concentrated mucus that pro-motes biofilm-type growth.163

An important new concept is that the mucus adherent toairway surfaces may be hypoxic and even frankly anaerobic(Fig. 38.6).164 Prior to infection, the increased oxygen con-sumption by CF airway epithelia (required to fuel increasedNa+ absorption), coupled with increased diffusion distancesfor oxygen through mucus plaques, can create relativelyhypoxic zones near the epithelial surface. As it has recentlybeen shown that P. aeruginosa can adapt to and grow in ahypoxic environment, this environment may exert a subtlepressure that favors P. aeruginosa acquisition in CF. Perhapsmore importantly, as bacterial densities in CF mucus reach106 CFU/mL or more, the bacteria consume the remain-ing oxygen in the environment. Indeed, direct measure-ments of PO2 in vivo with oxygen electrodes have recordedPO2 of less than 2 mm Hg in infected CF mucus.164

This observation has two important implications for CFtherapy. First, P. aeruginosa sensitivities to antimicrobialschange in oxygen-depleted versus oxygen-rich environ-ments; for example, macrolides become more effective inoxygen-depleted environments. Thus, clinical testing ofPseudomonas isolates for antimicrobial sensitivity mayrequire studies under anaerobic conditions. Second, P.aeruginosa physiology differs under anaerobic and aerobicconditions.165 Identification of essential genes for anaerobicgrowth may represent novel therapeutic targets.

Once bacterial infection of mucus has occurred, bacteriarelease products that diffuse to the underlying epithelial cellsand stimulate up-regulation of mucin production166,167 andcytokine responses,168 and they mediate cell damage. The

122738 • Cystic Fibrosis

38.qxd 1/25/05 2:47 PM Page 1227

Page 12: Cystic Fibrosis

1228 Section J • OBSTRUCTIVE DISEASES

infection may be worsened after cell damage by bacteriabinding to the injured cells (but not the uninjured cells).169

Pulmonary immunology in CF has been investigatedextensively. There is little reason to believe that a primaryimmunologic deficiency state exists in CF. Although CFsubjects tend to have low levels of serum immunoglobulinG (IgG) in the first decade of life, these levels increase dramatically as chronic infection is established.170 Secretoryantibody levels appear to be normal or enhanced.171 Anti-body responses specific for infecting organisms in the res-piratory tract are brisk.172,173 The number of B lymphocytesand plasma cells that differentiate into immunoglobulin-secreting cells are not depressed.174 T-lymphocyte numbersare adequate, and these lymphocytes proliferate in responseto nonspecific mitogens.175 With advancing severity of pul-monary disease, lymphocytes of CF patients may proliferateless briskly in response to P. aeruginosa and other gram-negative organisms.176 This acquired dysfunction can bereversed in some patients by intensive antibiotic treatment,but this may contribute to subsequent resistance of end-stage lung disease to antimicrobial therapy.

Polymorphonuclear leukocytes and macrophages areplentiful in CF airways. CF pulmonary macrophages func-tion normally when removed from their usual environ-ment.177 Polymorphonuclear cells also function normallyoutside the host environment. These observations suggestthat potential phagocytic defects in vivo could reflect infec-tion-induced degradation of opsonins (e.g., IgG and com-plement) and possibly defective leukocyte migrationthrough thickened, adhesive mucus.178

Much recent work has focused on the magnitude of theinflammatory response to airways infection in CF and theregulatory systems that modulate this response.179 Studiesfrom lung lavage fluid from CF neonates indicate that themagnitude of the neutrophil response normalized to bacte-rial counts in lavage fluid is increased in CF compared tothat of non-CF infants with lung infections.180 Otherworkers have suggested that the cytokine interleukin-10(IL-10), which normally serves to dampen epithelial cell IL-6 and IL-8 responses to luminal bacterial infection, is notproduced at normal levels in CF airway epithelia.181,182 It isimportant to learn whether the increased neutrophil influxreflects persistent failure to kill bacteria growing in biofilmswithin thickened mucus plaques, failure of an epithelial regulatory mechanism for neutrophil influx, epithelial com-pensation to a chronic stimulus, or a combination thereof.

The mechanisms by which progressive obstructive lungdisease and destruction of airway walls occur in CF havereceived attention recently. The role of proteinases in thisprocess in particular has been intensively studied.183 Leuko-cytes and bacteria produce large quantities of elastase thatexceed the neutralization capacity of airway antiproteinases.Both leukocyte and bacterial elastases generate C5a non-immunologically in CF secretions.184 High levels of thischemotactic factor stimulate further polymorphonuclearleukocyte recruitment, increasing the burden of free elas-tase in the airway lumen. Ultimately, proteolytic injury toairway walls contributes to their destruction with the devel-opment of widespread bronchiolectasis and bronchiecta-sis.185 The fact that destructive emphysema is not aprominent feature of CF lungs, especially early in the course of lung disease, can be attributed to confinement of

infection to endobronchial spaces in mild to moderate lungdisease. Other mechanisms of conducting airway tissueinjury undoubtedly exist but have not yet been described indetail.

CLINICAL MANIFESTATIONS

Cystic fibrosis presents in many ways and mimics a numberof other clinical entities. Usual presentations include earlyonset of respiratory tract symptoms, particularly persistentcough and recurrent or refractory lung infiltrates. Usualgastrointestinal presentations include meconium ileus inapproximately 15% of patients and failure to thrive withsteatorrhea. A surprising number of people with CF escapedetection in the first decade or two of life, often becausesymptoms are unusual, subtle, or even absent. Patients with“mild” mutations may present in childhood or later in life.35

unusual presentations are compiled in Table 38.3, and presentations that may be expected at adolescence or earlyadulthood are indicated. Recognition of the protean mani-festations of CF and a high index of suspicion are requiredto detect all cases, either in childhood or later in life.

LOWER RESPIRATORY TRACT DISEASE

The earliest manifestation of CF lung disease is generallycough. At first it is intermittent, coinciding with episodes ofacute respiratory tract infection and persisting longer thanexpected. With time, the cough becomes a daily event. It isoften worse at night and especially on arising in the morning.

Table 38.3 Unusual Presentations of Cystic Fibrosis

RespiratoryBronchiolitis/asthmaPseudomonas aeruginosa or Staphylococcus aureus colonization

of the respiratory tract*Staphylococcal pneumoniaNasal polyposis*Nontuberculous mycobacterial infection*

GastrointestinalMeconium plug syndromeRectal prolapseRecurrent abdominal pain and/or right lower quadrant mass*Hypoproteinemic edemaProlonged neonatal jaundiceBiliary cirrhosis with portal hypertension*Vitamin deficiency states (A, D, E, K)Acrodermatitis enteropathica-like eruption with fatty acid and

zinc deficiencyRecurrent pancreatitis*

GenitourinaryMale infertility*Female infertility*

OtherHypochloremic, hyponatremic alkalosis

Mother of a child with cystic fibrosis*

* Presentations that may occur in adolescents or adults with cysticfibrosis.

38.qxd 1/25/05 2:47 PM Page 1228

Page 13: Cystic Fibrosis

With progression of lung disease, the cough becomes pro-ductive and then paroxysmal, with associated gagging andemesis. Sputum is usually tenacious, purulent, and oftengreen, reflecting P. aeruginosa infection. Hyperinflation ofthe lungs is noted early in the progression of lung disease. Infact, it is unusual to see CF respiratory tract disease withoutat least a moderate increase in lung volume. Asthmatic orbronchiolitic-type wheezing is most common during thefirst 2 years of life but may be encountered at any age.Wheezing may occur with or without evidence of atopy,which is probably not more frequent in patients with CF.186

Lung sounds are often unremarkable for extended periods oftime, sometimes for years. Not infrequently, diminution inthe intensity of breath sounds may be the only abnormalitynoted, correlating with the extent of hyperinflation. Adven-titious lung sounds are usually heard first over the upperlobes, and they often are localized to the right side.

Cystic fibrosis patients may have only mild bronchiticsymptoms for long periods of time, in some cases for adecade or two, but eventually these periods of stability arepunctuated with exacerbations of symptoms, includingincreased intensity of cough, tachypnea, shortness of breath,decreased activity and appetite, and weight loss. It has beenassumed that these exacerbations are triggered by acute res-piratory infections, perhaps of viral or mycoplasmal origin,although studies differ concerning evidence for these infec-tions during periods of increased lung symptoms.187–192

Environmental factors, including cigarette smoke,193 alsomay evoke more respiratory symptoms. Intense antibiotictherapy and assistance with clearance of mucus are usuallyrequired to reduce lung symptoms and improve lung func-tion. Exacerbations characteristically occur with increasingfrequency. Frank limitation of activity or disability is associ-ated with end-stage lung disease and heralds a sequence ofterminal events, including substantial hypoxemia, pul-monary hypertension, cor pulmonale, and death.

Microbiology

The airways of patients with CF are colonized early withbacteria; and once established, infection is rarely if ever

eradicated. S. aureus and Haemophilus influenzae are oftenthe first organisms detected.194 P. aeruginosa characteristi-cally is cultured from respiratory secretions months to yearslater, although this organism is present at diagnosis withincreasing frequency. Age at acquisition of P. aeruginosa,shown in Figure 38.7, is dependent in part on genotype,with earlier acquisition occurring in persons homozygousfor DF508.33 The possibility of nosocomial acquisition of P.aeruginosa has been raised.195 With progression of lungdisease, P. aeruginosa is often the only organism recoveredfrom sputum, and it may be present in several types ofcolony, usually with different antibiotic sensitivity patterns.Typically, one of these types is mucoid due to elaborationof large amounts of alginate. These mucoid organisms arefound in microcolonies of pseudomonads embedded andgrowing in biofilms of alginate.196 Mucoid properties arethought to inhibit opsonic and nonopsonic phagocytosis,enhance adherence properties, and decrease sensitivity toantibiotics and toxic, reactive intermediates produced byleukocytes.155,197–199 The recovery of P. aeruginosa, particu-larly the mucoid form, from the lower respiratory tract of achild or young adult with chronic lung symptoms is virtu-ally diagnostic of CF, although it may also occur in patientswith primary ciliary dyskinesia.200

Recently, other species have been recovered from CFlungs with increasing frequency, particularly Burkholderiacepacia. This organism has become prevalent in a few CFcare centers and is particularly difficult to treat because it isusually resistant to most antimicrobial drugs. Evidence forpatient-to-patient spread is strong and has led to recom-mendations for stringent infection control measures in CFcare settings.201,202 Colonization has been linked to the rapiddemise of a small percentage of patients203 with what iscalled the “cepacia syndrome.” Molecular analyses have dis-tinguished genomovars of B. cepacia, some of which com-prise distinct species.204–207 Genomovars II (Burkholderiamultivorans) and III (Burkholderia cenocepacia) have beenassociated with the cepacia syndrome, and genomovar IIIincludes the highly transmissible strain that may be linkedto expression of the cable pilus.206–208 Occasionally, othergram-negative rods are present in sputum, including

122938 • Cystic Fibrosis

Per

cent

80

70

60

50

40

30

20

10

00-1 2-5 6-10 11-15 16-20 21-25 26-30 31-35 36+

OverallPrevalence

61.9%58.2%

FemalesMales

Age (years)

Figure 38.7 Prevalence of Pseudomonas aeruginosainfection by age and sex in cystic fibrosis patientsenrolled in the U.S. Cystic Fibrosis Foundation Registry,1991, based on an analysis of 16,417 patients.(Courtesy of Dr. S. FitzSimmons.)

38.qxd 1/25/05 2:47 PM Page 1229

Page 14: Cystic Fibrosis

1230 Section J • OBSTRUCTIVE DISEASES

mucoid Escherichia coli, Stenotrophomonas maltophilia,Alcaligenes xylosoxidans, Klebsiella, and Proteus. Anaerobeshave been recovered from CF lung tissue; they may beundetected pathogens and are found in large abscess cavities on rare occasions.209

Sputum bacteriology correlates reasonably well with spec-imens obtained directly from the lower respiratory tract.Oropharyngeal swab cultures that yield S. aureus or P.aeruginosa are modestly predictive of results from bron-choscopic specimens, but negative pharyngeal cultures donot rule out the presence of these organisms in lowerairways.210 Quantitative bacteriology may be particularlyuseful for determining the relative contributions of organ-isms isolated.

A large number of sputum specimens contain yeast andAspergillus fumigatus. Neither organism has been consid-ered a serious pathogen, although the latter causes symp-toms of allergic bronchopulmonary aspergillosis in a smallpercentage of patients.211,212 Colonization of lungs withrapidly growing mycobacteria has been documented withincreasing frequency. Up to 20% of adult patients in someclinics are colonized by nontuberculous mycobacteria, occa-sionally prompting treatment with agents directed againstthese organisms.213–217 Mycobacterium tuberculosis infectionhas been seen in only very sporadic cases.

Radiology

The earliest radiographic change is usually hyperinflation ofthe lungs, reflecting obstruction of small airways. Thedegree of hyperinflation generally increases with age. Asbronchitis progresses, peribronchial cuffing becomesincreasingly prominent. Mucus impaction in airways may be seen as branching finger-like shadows. Evidence ofbronchiectasis, such as enlarged ring shadows and cysts, iscommon by 5 to 10 years of age. Frequently, peripheralrounded densities are noted during acute exacerbations;they may disappear with treatment, leaving residual cysts.Subpleural blebs often become evident during the seconddecade of life and are most prominent along the mediasti-nal border. For reasons that remain unexplained, the rightupper lobe usually displays the earliest and most severechanges. With advancing disease, the pulmonary artery seg-ments become more prominent. A relatively small, verticalcardiac shadow is typical, but the heart enlarges with onsetof cor pulmonale. Hilar adenopathy is rarely prominent.Lobar or segmental atelectasis is uncommon but is mostoften seen in infants or small children. Chest roentgeno-graphic scoring systems have been introduced and may beuseful for clinical studies and to chart the course of lunginjury.218

Roentgenographic improvement with intensive treatmentis not readily appreciated because of the fixed nature of theairway changes. The most striking improvement often isreduced inflation of the lungs, a change that tends to makethe fixed lung markings more prominent.

Computed tomography of the chest has not been usedroutinely for monitoring the course of CF lung changes.The cost is high, and resolution for young patients was initially poor. With the advent of rapid scanning, however,mild bronchiectasis and mucoid impaction can be observedat all ages.219,220 CT scanning detects these lesions, even

when the chest roentgenogram is nearly normal. The sever-ity of bronchiectasis can be graded in older patients.221 Theindications for CT scanning in place of routine chestroentgenography are not established. One indication maybe the evaluation of a patient considered for lobectomy.Magnetic resonance imaging (MRI) also defines peri-bronchial thickening, mucoid impaction, and bronchiecta-sis but has no well documented advantage over CT scans in these areas of imaging.222 MRI may be superior for dis-tinguishing hilar adenopathy and prominent pulmonaryvessels.223

Lung Function

Newborns with CF are thought to have normal lung func-tion. However, within weeks to months, many infants withCF show evidence of increased airway resistance, gas trap-ping, and diminished flow rates.224 When children reach anage that makes cooperation possible, more complete testingfirst demonstrates airway obstruction, which presumablyoccurs in small airways, as indicated by reduced maximummidexpiratory flow rates, reduced flows at low lungvolumes, and elevation of the residual volume to total lungcapacity ratio (RV/TLC).225–227 Other sensitive indicators oflung pathology include an increased alveolar-arterial oxygengradient, frequency dependence of dynamic compliance,reduced response of flows to a helium-oxygen mixture, elevated slope of phase III of the single-breath nitrogenwashout, and an elevated physiologic dead space.224 Teststhat are used most often to follow the course of pulmonaryfunction include spirometry, lung volume measurements,and measures of oxygenation. In general, patients progressfrom initial reductions in maximum midexpiratory flowrates to reductions in FEV1/FVC and then to diminishedvital capacity and total lung volumes. This progression fromperipheral airway obstruction to more generalized obstruc-tion and then to acquisition of a restrictive component isillustrated in Table 38.4. These tests are described and evaluated in Chapter 24.

By the time a diagnosis is made, many children with CFshow mild decrements of arterial PO2. Oxygenation declinesslowly throughout life. As a rule, patients who maintain satisfactory oxygenation do well, independent of the extentof the obstructive impairment. When arterial PO2 values dipbelow 55 mm Hg on a sustained basis, symptomatic pul-monary hypertension should be expected.228,229 Exaggeratednocturnal desaturation experienced by patients with CF may be a contributing factor to pulmonary hypertension.230

Hypoxemia generally is not accompanied by polycythemia,at least in part due to an expanded plasma volume and, insome persons, to suppressed erythropoiesis secondary tochronic infection. Elevation of arterial PCO2 generallyoccurs with FEV1 volumes less than 30% of those predictedand constitutes an end-stage event for most patients. Lessthan 50% of patients with FEV1 less than 30% of predicted,arterial PCO2 greater than 50 mm Hg, or arterial PO2 lessthan 55 mm live for 24 additional months.231

Airway hyperreactivity is a frequent feature of CF lungdisease and can be demonstrated by exercise testing, hista-mine challenge, or response to bronchodilators. Up to 68%of the CF population demonstrate decreased flows after histamine, and flows improve in as many as 40% with

38.qxd 1/25/05 2:47 PM Page 1230

Page 15: Cystic Fibrosis

aerosolized bronchodilators.232 In contrast to cross-sectional studies, repeated tests every 1 to 3 months for ayear have demonstrated bronchodilator responsiveness atleast once in 95% of subjects.232 Responsiveness is unrelatedto the severity of pulmonary disease or indices of atopy butseems to be more prevalent during winter than duringsummer months. Responsiveness diminishes with exacerba-tions of lung disease but returns as lung function improvesover 2 weeks of intensive in-hospital therapy. Several inves-tigators have noted that occasional patients with CF have aparadoxical response to bronchodilators, perhaps due tofurther loss of tone in bronchiectatic airways. The patho-genesis of bronchial hyperreactivity in CF is unclear. Airwayhyperreactivity is discussed in Chapter 37.

Exercise tolerance is related to the severity of the airwayobstruction.233,234 Most patients with CF maintain adequateoxygen saturation, even with maximal exercise. However, inone study, 12 of 29 patients with an FEV1 less than 50% offorced vital capacity (FVC) had arterial oxygen saturationbelow 90% at peak exercise, compared with no drop forpatients with higher FEV1 values.235 Persons with CF havehigher than expected ventilatory muscle endurance, and thisendurance can be further improved with inspiratory muscletraining. However, improved inspiratory muscle strengthand endurance do not augment exercise performance.236

Exercise does improve cardiorespiratory fitness but does notimprove standard pulmonary function test results. Variedweight training, however, in one study increased weight and

muscle size and, in addition, reduced the RV/TLC by12%.237 Another study demonstrated that adults with CFand advanced lung disease improve with exercise, but thosewith only moderate lung disease have decreased end-expiratory lung volumes after graded exercise on a cycleergometer.238 Maximum oxygen consumption during exer-cise appears to be a better predictor of survival than routinepulmonary function testing.239 The physiology of exerciseand exercise testing are discussed in Chapter 25.

Resting energy expenditure is variably elevated in patientswith CF. Although some have claimed that there are genet-ically determined increases in energy expenditure (e.g.,increased ATP hydrolysis) related to certain CFTR muta-tions,240 others have found increased work of breathing tobe a more plausible explanation.241

UPPER RESPIRATORY TRACT DISEASE

Virtually all patients with CF have roentgenographic opaci-fication of the paranasal sinuses242 and intermittent symp-toms of increased upper airway secretions and moderateairflow obstruction.243 Nasal polyps occur in 15% to 20% ofpatients and are most common toward the end of the firstdecade and during the second decade of life.65 Manifesta-tions include severe or complete nasal airflow obstruction,rhinorrhea, and occasionally widening of the bridge of thenose. Despite the presence of roentgenographic abnormal-ities, acute or chronic symptoms of sinusitis occur in fewerthan 10% of children244 and in about 24% of adults.245,246

COMPLICATIONS OF RESPIRATORY TRACT DISEASE

Lobar and segmental atelectasis occurs in about 5% ofpatients.247 This complication is most prevalent in the first 5years of life and thereafter has a diminishing frequency. Theright lung is the site of atelectasis in the majority of patients.Many episodes occur in conjunction with an exacerbation ofclinical symptoms, but silent atelectasis is common. Occa-sionally, volume loss is associated with allergic aspergillosisand endobronchial mucus plugs. However, in mostinstances, a discrete plug is not evident on bronchoscopy.

Pneumothorax is a more frequent complication and iscaused by rupture of subpleural blebs. The overall incidenceis about 1% per year and increases with age and diseaseseverity, so less than 20% of CF adults experience at leastone pneumothorax.248 Pneumothorax occurs equally inboth sexes and is more frequent in the right chest. A smallasymptomatic pneumothorax may be discovered at the timeof routine chest roentgenographic examination. More com-monly, patients develop acute onset of shortness of breath,chest pain, and/or hemoptysis. The incidence of tensionpneumothorax is probably higher in patients with CF thanin those with less severe or no lung disease; and under thesecircumstances, the accumulation of pleural air may becomea life-threatening event. Simultaneous bilateral pneumoth-oraces have been described and constitute an urgent situa-tion. Recurrent pneumothoraces are very common.

Hemoptysis is a common event in older CF patients. It cor-relates with clinical and radiologic evidence of bronchiecta-sis and is usually due to airway inflammation rather thanvigorous activity or trauma to the chest. Blood streaking inthe sputum (“minor hemoptysis”) is common, and massive

123138 • Cystic Fibrosis

Table 38.4 Representative Pulmonary Function Test Resultsfrom Three Young Adult Males with Mild, Moderate, andSevere Lung Disease*

Patient with Patient withPatient with Moderate SevereMild Lung Lung Lung

Test Disease Disease Disease

FVC 98 72 48

FEV1 92 46 34

FEV1/FVC (0.81) (0.70) (0.64)

MMEF 83 15 6

Vmax50 91 19 11

Vmax25 52 10 5

FRC 162 112 75

RV 189 200 120

TLC 131 105 62

RV/TLC (0.29) (0.45) (0.50)

PaO2 (room air) [87] [74] [48]

* Values are percent predicted, except those in parentheses, which aresimple ratios, and those in brackets, which are millimeters of mercury.Patient with mild disease coughs several times a day; the cough isoccasionally productive. He shows no restriction of activity. Patientwith moderate disease coughs frequently, expectorates moderatelylarge amounts of mucus, but is able to jog 3 miles daily and is a full-time student in a professional school. Patient with severe disease haschronic right heart failure but is able to work daily as a hair stylist.

38.qxd 1/25/05 2:47 PM Page 1231

Page 16: Cystic Fibrosis

1232 Section J • OBSTRUCTIVE DISEASES

hemoptysis (>240 mL of blood in 24 hours) occurs inapproximately 5% of patients.249 There is a strong correlationbetween the occurrence of both small- and large-volumehemoptysis and the exacerbation of lung infection. Patientswith relatively large-volume hemoptysis may be able to local-ize the site of bleeding by describing a bubbling or gurglingsensation in one area of the chest. Any lobe may be involved,and localization is difficult in most patients. Even emergentbronchoscopy often does not detect the location of blood inthe airways. In the past the immediate mortality with massivehemoptysis may have been as high as 10%, but with propertreatment most patients with massive hemoptysis have aprognosis that is not much different from that for theremainder of the CF population.250

About 1% to 10% of patients develop allergic bron-chopulmonary aspergillosis (ABPA) at some time in theirlives.251 In one study, more than 50% of patients with CFhad precipitating antibodies to Aspergillus fumigatus intheir serum, and this organism can be recovered from thesputum with a similar frequency.252 New lung infiltrates,increased cough, respiratory distress, wheezing, and theexpectoration of rusty brown plugs of sputum containingmany eosinophils suggest this diagnosis. Atelectasis due toplugging of a lobar or segmental bronchus with hyphae-laden mucus may occur. Chronic airway colonization withA. fumigatus (or other species) is common, and diagnosisof ABPA requires new clinical findings plus skin hypersen-sitivity and elevated levels of IgG and IgE antibodies againstA. fumigatus or other fungi.211,212

Pleural effusions and empyema are uncommon in patientswith CF, but pleuritic symptoms and signs may accompanyexacerbations of lung disease. Staphylococcal andpseudomonal empyemas have been described, but respira-tory tract infections usually spare the pleural space.253

Digital clubbing occurs in virtually all patients with CF andis usually present early in the course of symptomatic lungdisease. The cause of clubbing is unknown, but its severitygenerally correlates with the severity of lung disease.254

Hypertrophic pulmonary osteoarthropathy occurs in as manyas 15% of older adolescents and adults.175,255 If roentgeno-graphic evidence for periostitis is used as the definition, theincidence is 8%. The most common sites are the distal aspectsof the tibia, fibula, radius, and ulna. Signs and symptomsinclude pain, bone tenderness, swelling, and warmth overthe involved areas. Effusions in nearby joints may occur.There is often discomfort with ambulation. Hypertrophicpulmonary osteoarthropathy frequently becomes moresymptomatic with pulmonary disease exacerbations andtends to subside when control of pulmonary symptoms isachieved. There are rare instances of cutaneous vasculitis due to CF, which causes a self-limited, painless, palpablepurpura, typically involving the lower extremities.255

Respiratory failure leads to death in more than 90% ofCF patients. Hypoxemia often develops during exertion256

or sleep257 and progresses over years. Hypercapnia reflectssevere airway obstruction and increases with disease sever-ity and acute exacerbations. Hepatic congestion and periph-eral edema caused by pulmonary hypertension and corpulmonale may develop in the last year of life,258 particu-larly if hypoxemia is not adequately treated. Occasionally,infections such as respiratory syncytial virus or influenzacause acute respiratory failure that is fully reversible.259

GASTROINTESTINAL MANIFESTATIONS

Meconium ileus occurs in at least 15% of newborns with CFand is virtually diagnostic for CF.69 These infants fail to passmeconium in the first day or two of life, develop abdomi-nal distention, and proceed to bilious emesis. Occasionally,perforation occurs, and peritonitis accompanied by shockintervenes. Flat and upright abdominal films reveal multi-ple dilated loops of intestine and air-fluid levels. The lowerabdomen often takes on a granular appearance, represent-ing accumulated meconium containing small air bubbles. Abarium enema demonstrates a small colon; and if contrastmaterial can be refluxed into the ileum, the point of ilealobstruction can be identified. In utero, ileal perforationresults in peritoneal and scrotal calcification. Occasionally,obstruction occurs lower in the intestinal tract and causesonly delayed passage of meconium. This condition has beentermed the meconium plug syndrome and is much less spe-cific for CF.260 Meconium ileus is also a prominent featureof transgenic mice with a disrupted CFTR gene.27

Beyond the newborn period, intestinal obstruction mayoccur for a variety of reasons. Perhaps the most common(occurring in 20% of patients) is called the meconium ileus equivalent or the distal intestinal obstruction syn-drome.36,69,261 As with meconium ileus of the newborn,obstruction occurs in the terminal ileum and is usually asso-ciated with voluminous, sticky, incompletely digested intes-tinal contents. In some instances, a partial obstructionoccurs, accompanied by intermittent abdominal pain. Com-plete obstruction is associated with failure to pass stools,abdominal distention, and vomiting. A mobile right lowerquadrant mass may be palpable. Obstruction beyond theimmediate newborn period is also characteristic of the CFmouse model. Other causes of abdominal pain associatedwith obstruction include intussusception, intestinal adhe-sions from previous abdominal surgery (a particularproblem in patients with CF), low-grade appendicitis (par-tially suppressed by antibiotic therapy) and periappendicealabscess.262 Nonfilling of the appendix with contrast enemais frequent in patients with CF, even in the absence ofappendicitis.263 Duodenal irritation, caused by failure tobuffer gastric acid, may underlie radiographic changes inthis area and recurrent epigastric pain.264

Rectal prolapse occurs in nearly 20% of children but is aninfrequent event for adults with CF.265 In fact, CF is said tobe the most common cause of rectal prolapse in the UnitedStates. Factors contributing to rectal prolapse include thepresence of bulky, sticky stools that adhere to rectal mucosa,loss of the perirectal fat that normally supports the rectum,and increased frequency of high intra-abdominal pressuredue to paroxysmal coughing. Pneumatosis coli has beenreported in association with rectal prolapse in an 18-year-old with CF.

PANCREATIC DISEASE

Pancreatic exocrine insufficiency is present from birth in thelarge majority of patients with CF.266 It has been estimatedthat only 5% to 10% of patients have significant pancreaticenzyme release. Exocrine pancreatic sufficiency has beenassociated with several mutations, including 3789 + 10 KbC to T, R117H, R334W, R347H, R347P, A455E, P574H,

38.qxd 1/25/05 2:47 PM Page 1232

Page 17: Cystic Fibrosis

and Y563N. Enzyme deficiency results in fat and proteinmaldigestion, producing a distended abdomen and fre-quent, bulky, greasy, foul-smelling stools. Uncorrectedmaldigestion results in failure to gain weight and ultimatelyfailure of linear growth. However, poor growth also may beassociated with increased expenditure of energy to accom-plish the work of breathing, a point that is often overlookedin the assessment of patients who are short or excessivelythin.267 Fat loss in stools may be as high as 50% to 70% oftotal intake. Nitrogen malabsorption is roughly comparableor perhaps somewhat less severe. In general, carbohydratesare adequately absorbed in CF. Deficient absorption of fat-soluble vitamins occasionally produces symptoms. Sympto-matic vitamin A deficiency, initially a prominent part of the CF syndrome, is rare and occurs only in patients whodo not take supplementary vitamins or pancreatic enzymes.Increased intracranial pressure, xerophthalmia, and nightblindness may result. Rickets due to vitamin D deficiency israrely seen. However, bone demineralization is common in CF patients.268–271 Vitamin E deficiency is common inunsupplemented patients but only rarely causes symptomsor signs, including increased red blood cell destruction andneuroaxonal dystrophy. Vitamin K-dependent coagulationfactors may also be deficient, resulting in a hemorrhagicdiathesis. Although severe hemorrhagic problems occurmostly in young children, associated symptoms such ashemoptysis are occasionally seen in older patients.

It is now recognized that symptoms of pancreatitis areencountered in less than 1% of identified adolescent or adultCF patients and are limited to those who have retainedsome exocrine pancreatic function.272 However, recurrentpancreatitis has recently been reported to be associated withmutations in CFTR and is becoming more frequently rec-ognized as a presenting symptom in adults with CF.43,273,274

Pancreatic calcifications are occasionally seen roentgeno-graphically but do not seem to correlate with symptomaticpancreatitis.

HEPATOBILIARY DISEASE

Focal biliary cirrhosis is characteristic of CF but producessymptoms in less than 5% of CF patients and causes deathin about 2%.37,275 Hepatic abnormalities can present ashepatosplenomegaly or as persistent elevation of hepaticenzymes (particularly alkaline phosphatase). Rarely, patientspresent with esophageal varices and hemorrhage due toportal hypertension. Fatty liver is more common and mayimprove with adequate nutrition. Gallbladder disease iscommon in adults with CF. Altogether, 10% to 30% ofpatients have dysfunctional gallbladders276 or gallstones.277

Common bile duct stenosis secondary to CF is rare.278

GENITOURINARY TRACT ABNORMALITIES

More than 99% of male patients with CF have obstructiveazoospermia due to obstruction of the vas deferens.44 Rel-atively high incidences of inguinal hernia and hydrocele alsohave been noted. Failure of reproductive function does notbecome an issue until well into the second decade of life.Semen analysis may be required to identify the 1% of maleCF patients who are fertile. The volume of ejaculate isusually one third to one half of normal. There usually is

complete absence of spermatozoa, and a number of chem-ical abnormalities of semen that reflect absence of secretionsfrom the seminal vesicles can be demonstrated.279

Female infertility in CF may be as high as 20%.279 Somewomen with CF are anovulatory because of chronic lungdisease and malnutrition. Another obstacle to conceptionmay be the presence of thick, tenacious cervical mucus,which is difficult to dislodge from the os. This mucus isdehydrated and has abnormal electrolyte concentrations,preventing the usual ferning at midcycle and perhapsimpeding normal sperm migration.280 Endocervicitis iscommonly noted.

More than 600 pregnancies in CF females have beenreported. A longitudinal study of 325 pregnant women withCF demonstrated 258 live births (79%) and 67 therapeuticabortions.281 Compared to 1142 age- and severity-matchedcontrols, pregnancy in a woman with CF did not have anindependent negative effect on pulmonary status or mor-tality over 2 years.282 However, it is essential that CF femalesconsider their own health and expected life span in thecontext of family planning. Women with CF can breast-feedsuccessfully.283

SWEAT GLAND DYSFUNCTION

Sweat chloride is elevated in most CF patients due to abnor-mal NaCl reabsorption in the sweat ducts.284 Excessive loss ofsalt in the sweat predisposes young children to depletionepisodes, especially at times when there is additional salt lossdue to vomiting or diarrhea. These children present withlethargy, anorexia, and hypochloremic alkalosis, a presenta-tion that is more common in warm arid zones. Suchhypochloremic alkalosis is rare in older children and adults.285

DIAGNOSIS

The diagnosis of CF is based on carefully defined clinical cri-teria and laboratory evidence of CFTR dysfunction, includ-ing abnormal sweat Cl- levels.284,286 Accepted diagnosticcriteria are listed in Table 38.5. Any one of these clinical fea-tures, if accompanied by a sweat chloride level greater than60 mEq/L, is sufficient to make the diagnosis. Characteris-tic nasal epithelial bioelectric abnormalities can also serve as laboratory evidence of CFTR dysfunction and be used to diagnose CF when phenotypic clinical features arepresent.284,287 Many diagnoses are verified by identifying amutation in CFTR on both alleles. At this time, however, asubstantial number of patients have unidentified mutations.A few persons with sweat chloride values persistently in thediagnostic range but without clinical features of CF or afamily history have been identified. So long as they remainsymptom-free and mutations of both CFTR genes are notidentified, the diagnosis of CF cannot be made. Other clin-ical entities may be accompanied by elevated sweat chlorideconcentrations (Table 38.6). None of these disorders iseasily confused with CF. It is now recognized that a smallpercentage of persons with CF have sweat values in an inter-mediate (40–60 mEq/L) or even normal range.35,288,289

For the 1% to 2% of patients with compatible clinical fea-tures of CF but normal sweat chloride levels, severalapproaches to the diagnosis are available. Clinical criteria for

123338 • Cystic Fibrosis

38.qxd 1/25/05 2:47 PM Page 1233

Page 18: Cystic Fibrosis

1234 Section J • OBSTRUCTIVE DISEASES

diagnosing CF in the presence of a normal sweat chloridevalue were proposed prior to modern diagnostic options.288

The discovery that bioelectric potential differences acrossrespiratory epithelia are abnormal in patients with CF9,102,287

offers a quantitative diagnostic test that has been useful forevaluating atypical patients. Also, DNA analysis techniquesare now available for detecting CF gene mutations, partic-ularly in clinical variants. For example, a single mutation of

the CFTR gene, 3789 + 10 Kb C to T, is associated withmany of the suspected cases of CF whose sweat chloridevalues are in the normal range.35,40

Although DNA analysis is becoming a standard of care inCF, the sweat test remains the diagnostic standard. Onlyone method is recognized by the Cystic Fibrosis Founda-tion as adequate for a definitive diagnosis of CF. This testinvolves collection of sweat by pilocarpine iontophoresiscoupled with chemical determination of the chloride con-centration.290,291 The procedure must be carried out in ameticulous fashion to avoid errors that frequently con-tribute to misleading values. As many as 40% of patientsreferred to CF centers have been inaccurately diagnosedelsewhere because of false-positive or false-negative sweattest results. In addition to frequent laboratory errors, faultyvalues can be obtained if the sweat rate is not sufficientlyhigh. At least 50 mg of sweat must be collected in a 30-minute period. Potential pitfalls in interpretation includethe presence of hypoproteinemic edema, failure to considerage-related effects, and concurrent administration of corti-costeroids. Although using a chloride threshold of 60 mEq/Lin sweat appears to distinguish nearly all adults with CFfrom those with other lung conditions,292 normal values ofsweat chloride do increase with age, and some investigatorsinsist on documentation of sweat chloride levels in excess of70 mEq/L for the diagnosis of CF in adults.

Newborn screening for CF is carried out on all infants inseveral states. The test used is an assay of immunoreactivetrypsin in dried blood spots collected on filter paper in thefirst several days of life. Experience thus far suggests thatmany, but not all, newborns with CF may be identified withthis test.293 The test has an acceptable rate of false-positiveresults. Screening for gene mutations is currently precludedby the large number of mutations. If screening were basedon DF508 alone, fewer than half of all newborns with CFwould be identified. If parents were to be screened by DNAanalysis, only 70% of carriers or 50% of carrier couples wouldbe detected. At present, routine newborn screening cannotbe strongly recommended, largely because there is not yeta specific highly efficacious therapy for this condition thatdemands early intervention. However, studies of newbornswith CF discovered by screening have detected nutritionaldeficits, which suggests that early identification and treat-ment may be useful for protecting against loss of lung func-tion.294–297a In addition, pharmacologic and gene transfertherapies, currently at an experimental stage, are theoreti-cally most effective early in life; if their efficacy is docu-mented, newborn screening can play an important role.

Antenatal diagnosis can now be carried out, especiallywhen the genotype of parents is established. A recentNational Institutes of Health Consensus Statement recom-mended that genetic testing for CF be offered to couplescurrently planning a pregnancy.298

TREATMENT

The primary objectives of CF treatment are to control infec-tion, promote mucus clearance, and improve nutrition.299

In addition, experience has repeatedly demonstrated thatattention to preventive aspects of lung care and psychoso-cial factors are important. The efficacy of a number of

Table 38.5 Criteria for the Diagnosis of Cystic Fibrosis

Phenotypic Clinical FeaturesChronic Sinopulmonary DiseaseChronic cough and sputum productionPersistent infection with characteristic pathogens

(Staphylococcus aureus and Pseudomonas aeruginosa)Airflow obstructionChronic chest radiographic abnormalitiesSinus disease; nasal polypsGastrointestinal and Nutritional AbnormalitiesPancreatic exocrine insufficiency; recurrent pancreatitisMeconium ileus; distal intestinal obstruction syndromeObstructive Azoospermia in Males

Laboratory Evidence of CFTR DysfunctionElevated sweat Cl-

Mutation in the CF gene on both allelesCharacteristic bioelectric abnormalities (potential difference)

in nasal epithelium in vivo

CFTR, cystic fibrosis transmembrane conductance regulator.Adapted from Rosenstein BJ, Cutting GR: The diagnosis of cysticfibrosis: A consensus statement; Cystic Fibrosis Foundation ConsensusPanel. J Pediatr 132:589–595, 1998.

Table 38.6 Conditions Other Than Cystic Fibrosis Associatedwith Elevated Sweat Chloride Levels

Adrenal insufficiency

Anorexia nervosa

Atopic dermatitis

Pseudohypoaldosteronism

Hypothyroidism

Hypoparathyroidism

Nephrogenic diabetes insipidus

Ectodermal dysplasia

Glycogen storage disease (type I)

Mucopolysaccharidosis

Fucosidosis

Malnutrition

Mauriac’s syndrome

Familial cholestasis syndrome

Prostaglandin E1 administration

Hypogammaglobulinemia

38.qxd 1/25/05 2:47 PM Page 1234

Page 19: Cystic Fibrosis

currently used and new approaches is being tested in con-trolled multicenter clinical trials.

AMBULATORY CARE

At diagnosis, most patients are introduced to a care programthat includes postural drainage with chest percussion, admin-istration of antibiotics as indicated, and a nutritional regimenincluding pancreatic enzymes and fat-soluble vitamins.Other therapeutic modalities are prescribed as indicated.

Airway Clearance

The use of postural drainage with chest percussion to clearmucus is based on the concept that cough clears mucusfrom large airways, but chest vibrations are necessary tomove secretions from the small airways, where expiratoryflow rates are low. The most compelling argument for theuse of this therapeutic modality comes from a study of olderchildren with mild to moderate airflow limitation.300 Whenpatients were receiving chest physical therapy on a regularbasis, the only immediate effect documented was an increasein peak expiratory flow rate 30 minutes after therapy.However, after 3 weeks without chest physical therapy, bothFVC and flow rates were significantly reduced. The diminu-tion in lung function could be reversed with renewal ofregular chest physical therapy. A recent meta-analysis of 35studies concluded that standard chest physical therapyincreases sputum production and improves expiratoryairflow (FEV1).301 Most care centers prescribe this therapyone to four times a day, depending on the severity of illness.A variety of mechanical percussors are available to assist,especially for patients who perform their own therapy.Several studies in the literature suggest that repeated forcedexpiratory maneuvers,302 use of positive expiratory pres-sure,303 and vigorous exercise304–306 may substitute for chestphysical therapy. The advisability of chest physical therapyfor patients who have minimal cough and no apparentsputum production is open to question. Study results arenot available to answer this question. Theoretically, chestphysical therapy might prevent or delay inspissation ofmucus in small airways, which is the earliest lesion. In addi-tion, caregivers have found that many patients who do notreceive chest physical therapy on a regular basis are unlikelyto initiate this therapy when pulmonary exacerbations arise.

Antibiotics

Lung infection is the major source of morbidity and mor-tality in CF. Therefore, antibiotic therapy is the mainstay of therapy designed to control progression of disease. Ingeneral, antibiotic therapy should be predicated on the pres-ence of symptoms and guided by the identification of organ-isms from the lower respiratory tract. There is evidence thatearly, vigorous use of antibiotics produces better results thandelaying the administration of antibiotics until symptomsare well developed or advanced.307 Similarly, P. aeruginosainfection has been treated by regularly scheduled courses ofintravenous antibiotics.308 Data justifying these approachesare lacking.

Another principle of antimicrobial therapy in CF is thatdosages need to be higher than for non-CF-related chest

infections. Both total body clearance and volume of distri-bution are considerably greater for CF patients than forother patients.309 In addition, large doses are needed toachieve therapeutic levels in the infected and mucus- or pus-filled endobronchial space. Sputum levels achieved arehighly varied but usually are less than half of the levels meas-ured in serum.310 Experience also has led many caregivers ofCF patients to use longer than usual courses of antibiotics,continuing for at least 2 weeks and frequently 3 to 4 weeks.

The choice of antibiotics is optimally based on the resultsof sputum culture and sensitivity testing. For patients withnonproductive cough, specimens are best obtained by har-vesting secretions with a throat swab placed just above theglottis during repetitive coughing. Nasopharyngeal culturesare not useful as a guide for selecting antibiotics. If S. aureusis present or expected, the choice may include a semisyntheticpenicillin, a combination of ampicillin and clavulanic acid, acephalosporin, or clindamycin. Suggested drugs and dosesare given in Table 38.7. Haemophilus infections are besttreated with ampicillin, trimethoprim-sulfamethoxazole,cefuroxime, or cefaclor. In the majority of cases, these antibi-otics eradicate S. aureus and H. influenzae from the airwayson a temporary basis. In older children and adults, tetracy-clines may provide useful empirical therapy. Ciprofloxacinappears to control symptoms and reduces numbers ofPseudomonas organisms in CF airways, but its usefulness islimited by rapid emergence of resistant organisms.311

Because of limited options for treatment of Pseudomonaswith oral antibiotics, parenteral preparations of aminogly-cosides or other antibiotics have been delivered to the lowerrespiratory tract by aerosol.312,313 Recent studies show clearshort-term benefit of aerosolized tobramycin (300 mg in anebulizer) taken twice daily.314 This treatment, taken inalternate months for three cycles, improved lung function,decreased the bacterial burden, and decreased the relativerisk for hospitalization. The rate of acquired tobramycinresistance was about 7% over 24 weeks in the treated group.

Azithromycin

Long-term macrolide antibiotics effectively treat diffusepanbronchiolitis and have been tested in CF. Randomized,controlled clinical trials in CF children in the UnitedKingdom,315 in adults who were chronically infected with P.aeruginosa in Australia,316 and in chronically P. aeruginosa-infected children and adults in the United States317 demon-strated clinical improvement (better pulmonary functionand fewer pulmonary exacerbations) with azithromycin. Asputum acid-fast bacillus (AFB) smear and culture shouldbe obtained prior to initiating chronic macrolide therapybecause of the risk that undiagnosed nontuberculousmycobacterial infection could develop macrolide resistance.For patients on chronic macrolide therapy, an AFB smearand culture should be obtained every 6 months.

Aerosolized Recombinant Human DNase

The lysis of viscous DNA with the recombinant enzymeDNase offers benefit to some patients with purulent airwaysecretions.318 When taken once daily, aerosolized DNasereduced the relative risk of respiratory exacerbations by 28%and improved the FEV1 about 6% above baseline over 6

123538 • Cystic Fibrosis

38.qxd 1/25/05 2:47 PM Page 1235

Page 20: Cystic Fibrosis

1236 Section J • OBSTRUCTIVE DISEASES

months. Not all patients had objective improvement in lungfunction, but symptomatic benefit occurred in many of thetreated patients without improved lung function. The long-term effect of DNase on lung function is currently beingevaluated in an open-label multinational study.

Other Aerosol Therapy

Other solutions given by aerosol have the objective of pro-viding a volume of liquid to hydrate inspissated mucus secre-tions. One frequently used solution is normal saline, but theefficacy of this approach has not been substantiated. Studiesfrom Australia suggest that aerosolized hypertonic saline(3–12%) may offer short-term benefit with acute improve-ment in lung function and mucociliary clearance.319,320,320a

Bronchodilators

As detailed in the clinical manifestations section, manypatients demonstrate bronchial lability, prompting frequentuse of bronchodilators,224 particularly beta-adrenoceptoragonists. Although their immediate effectiveness can be documented in the pulmonary function laboratory, overall

improvement or long-term benefit has not been established.Indications for the use of bronchodilators include trouble-some wheezing and a documented improvement in pul-monary function (e.g., at least 15% improvement of FEV1).Beta-agonists can be nebulized, administered by metered-dose inhalers, or given orally. Caution should be exercised con-cerning long-term therapy with these agents because animalstudies show that administration of large amounts of beta-adrenoceptor agonists cause submucosal gland hypertrophyand presumably a hypersecretory state.321 Theophylline prepa-rations also are effective in selected cases. However, CFpatients seem to be less tolerant of theophylline because of fre-quent gastrointestinal irritation.322 Ipratropium bromide is atleast as effective as beta-agonists in CF,323–325 although distalintestinal obstruction syndrome has been reported coincidentwith use of this agent.326 Cromolyn sodium has also been used;a single study failed to demonstrate efficacy.327

Corticosteroids

An initial double-blind controlled study of alternate-dayoral corticosteroid administration V. placebo demonstratedbetter maintenance of pulmonary function and fewer exac-

Table 38.7 Antimicrobial Agents Used to Treat CF Lung Infection

Dose

Route and PediatricOrganism Agent (mg/kg/day) Adult (g/day) Doses/day

OralS. aureus Dicloxacillin 50–100 2–4 4

Cefaclor 40 0.75–1.5 3Clindamycin 15–25 0.6–1.2 4Erythromycin 50 2 4Amoxicillin/clavulanate 40 1.5 3Azithromycin — 0.5 day 1, then 1

0.25 daily for 2–5 daysH. influenzae Amoxicillin 40–60 1.5 3

Trimethoprim-sulfamethoxazole 20* 0.32–0.64* 2–4P. aeruginosa Ciprofloxacin — 1.5–2.25 3

Ofloxacin — 0.8–1.2 2–3Empirical Tetracycline 50–100 2 3–4

IntravenousS. aureus Oxacillin 150–200 † 4

Vancomycin 40 2 2–4P. aeruginosa Gentamicin or 8–20 † 2–3

Tobramycin 8–20 † 2–3Amikacin 15–30 † 4–6Carbenicillin or 250–450 † 4–6Ticarcillin 250–450 † 4–6Piperacillin, mezlocillin, or 45–90 4 3–4

azlocillin ticarcillin/clavulanateimipenem/cilastatin

AerosolP. aeruginosa and Ceftazidime 150 4–6 3

B. cepaciaP. aeruginosa Gentamicin 40–600‡ 80–600‡ 2–4

Tobramycin 40–600‡ 80–600‡ 2–4

* Quantity of trimethoprim.† Usually dosed by milligrams per kilogram per day as with children.‡ Total dose in milligrams.

38.qxd 1/25/05 2:47 PM Page 1236

Page 21: Cystic Fibrosis

erbations of lung disease requiring hospitalization over a 4-year period.328 However, a subsequent large multicenterstudy of corticosteroids administered in a dose of 1.0 or 2.0 mg/kg every other day failed to confirm efficacy andwas attended by an unacceptable rate of side effects, includ-ing growth suppression and hyperglycemia. Widespread useof corticosteroids cannot be advocated. They may, however,be used for specific indications such as allergic bronchopul-monary aspergillosis.

Anti-inflammatory Therapy With Ibuprofen

A recent study of high-dose ibuprofen over 4 years indicatesthat young CF patients (<13 years) with mild lung diseasehave remarkable slowing of the decline of lung function(eightfold) compared to placebo control subjects.329 Noeffect was documented in patients older than 13 years, evenamong patients with mild lung disease. There were few sideeffects, but most of the patients were taking antacids or H2-receptor blockers.

Other Respiratory Therapies

Mucolytics, expectorants, and cough suppressants havebeen used for relief of chest symptoms. In general, coughis an important mucus clearance mechanism for patientswith CF and should not be suppressed. Expectorants, whichassist in the elimination of airway secretions during cough,probably do not achieve that objective. Rather than beinghelpful, long-term administration of iodides to patients withCF has been associated with a high incidence of goiter andhypothyroidism.330 Mucolytics such as N-acetylcysteine areinjurious to respiratory epithelium and promote bronchitiswhen used regularly. Therefore, aerosol inhalation of thissubstance should be used selectively and only for shortperiods. Another experimental approach, delivery of elastaseinhibitors to airways by aerosol, is the subject of several clinical trials. Experimental studies implicate a cascadeinvolving epidermal growth factor receptor (EGFR) activa-tion in mucin production330a by a wide variety of stimuliincluding products of Pseudomonas bacteria,330b and selec-tive EGFR inhibitors may provide useful therapy for mucushyper secretion in CF.

Other experimental drugs include Na+ conductance inhi-bitors331,332 and triphosphate nucleotides.125,127,156 Both act toincrease salt and water availability at the epithelial surface ofthe airway and may improve mucus clearance properties.

Exercise is generally considered beneficial for patients withCF and should be encouraged for all but those with the mostsevere lung disease and hypoxemia.333 A 12-week exerciseprogram consisting of three 1-hour sessions a week, duringwhich jogging was used to produce a heart rate averaging70% to 85% of peak heart rate, has been shown to increaseexercise tolerance and cardiorespiratory fitness, probably by increasing respiratory muscle endurance.237 However,this program did not improve pulmonary function. Mostcaregivers of patients with CF believe that regular, vigorousexercise promotes a positive self-concept and increases theperception of wellness. In addition to cardiopulmonary con-ditioning exercises, isometric exercises such as weight liftingmay build abdominal and upper body muscle strength andin this way promote deep breathing and effective coughing.

Gene Transfer Technology

It is technically possible using genetically engineered viralvectors to transfer the wild-type CFTR gene to respiratoryepithelial cells in vitro, where it can be expressed for at leastdays to weeks.334 Systems for efficient targeted delivery andoptimal integration in vivo are being studied and contributeto the hope that genetic reconstitution will be a feasibleapproach to lung therapy in the future.

Nutrition

Approximately 90% of patients with CF require mealtimepancreatic enzymes, packaged as granules that are coatedwith acid-resistant material to promote delivery to the smallintestine. Capsules containing 4000 to 24,000 units of lipase are available. The number of capsules taken should beadjusted based on weight gain, the presence or absence ofabdominal cramping, and the character of the stools. Dosageshould be limited to current guidelines (about 2500 lipaseunits/kg/meal) because a large total daily dose of enzymeshas been associated with fibrosing colonopathy.335–338 Vita-mins A and D are generally supplied by a daily multiple-vitamin preparation. Also, 100 to 200 units of vitamin E dailyare recommended because serum levels are generally low inunsupplemented patients. Vitamin K is given sporadically totreat bleeding complications or to correct prolonged pro-thrombin times. Other vitamins and trace minerals may bedeficient and require supplementation on a selective basis.267

Many patients with CF have a higher than normal caloricneed because of the increased work of breathing. In general,patients should be encouraged to eat a balanced diet,including at least a moderate amount of fat. When theanorexia of chronic infection supervenes, there is failure togain weight or even weight loss. Further encouragement toeat high-calorie foods may be helpful. Supplementationwith elemental dietary preparations by mouth is unlikely tobe sustained over an extended period of time. Some CF carecenters administer supplementary elemental feedings noc-turnally by nasogastric tube or percutaneous duodenos-tomy.267,339 Although short-term benefits such as increasedweight gain can be achieved, long-term beneficial effects onpulmonary function have not been established.

Patients with CF who retain exocrine pancreatic functionand maintain good nutrition experience a slower rate ofdecline of pulmonary function than those with pancreaticdysfunction.340 This observation has been used as a ration-ale for emphasizing nutritional interventions. Although thisapproach may have merit, it is generally more efficacious tomaintain good nutrition by preventing progression of lungdisease than to maintain good lung function by emphasiz-ing nutritional therapy.

Psychosocial Factors

As with any chronic disease, compliance with therapy andthe ability to function fully are highly dependent on thepatient’s attitude. Therefore, approaches that promote apositive self-concept, foster the ability of patients to takecontrol of their medical management, and enable them toparticipate fully in life events are likely to promote well-being and perhaps longevity. Medical care that providescontinuity and fosters trust may pay large dividends.

123738 • Cystic Fibrosis

38.qxd 1/25/05 2:47 PM Page 1237

Page 22: Cystic Fibrosis

1238 Section J • OBSTRUCTIVE DISEASES

Personnel who specifically provide psychosocial support areimportant contributors to CF care teams.341

Immunization for Lung Pathogens

Rubeola, pertussis, and influenza infections are particularlyinjurious to CF lungs and may trigger a downward spiral oflung function. Adequate immunization early in life for per-tussis and measles is mandatory. In addition, patients of allages should be adequately immunized for influenza virusinfection on a yearly basis. The early use of amantadine foracute respiratory illnesses during epidemics of influenza Ainfections may further prevent adverse consequences of thisinfection. Other routine vaccines should be administered as for the general population. There is no evidence thatadministration of pneumococcal vaccine is useful.

HOSPITAL THERAPY

Indications for hospitalization and intensive pulmonarytherapy include increased cough or wheezing, respiratorydistress with decreased activity tolerance, weight loss, a sus-tained downward trend in pulmonary function, increasinghypoxemia, or one of the major pulmonary complicationsof CF. Although all the modalities of therapy can be inten-sified in the hospital, the major advantage of hospitalizationis the ability to administer intravenous antimicrobial agentsthat control Pseudomonas infection and intensify physicalmaneuvers to clear airway secretions.

Antibiotics for intravenous administration should beselected on the basis of respiratory tract cultures and sus-ceptibility studies. Two-drug treatment of Pseudomonasinfection is the rule. A third antibiotic may be added as necessary for control of S. aureus or other organisms. Drugsand dosages currently used for intravenous therapy are listedin Table 38.7. A clinical response is often not seen for 4 to7 days after initiation of therapy. In general, a 2-week courseprovides good improvement in pulmonary function342,343

and more sustained benefit than shorter courses. Withrefractory infection, treatment for 3 weeks or more is notunusual. Some advocate intensive therapy until pulmonaryfunction has returned to a previous baseline or untilimprovement has reached a plateau. Prolonged courses ofantibiotic therapy should include periodic monitoring ofrespiratory tract organisms and their antibiotic sensitivities,as shifts in either are common.

Aminoglycosides have been the mainstay of anti-Pseudomonas therapy for many years. A major advantage isthe ability to monitor and adjust blood levels. For gentamicinor tobramycin, two to three times a day dosage and achieve-ment of peaks in the range of 10 to 12 mg/mL and troughsless than 2 mg/mL seem to be optimal. Patients should bemonitored during aminoglycoside therapy for nephrotoxic-ity and ototoxicity. An aminoglycoside is usually paired withone of the penicillin derivatives or with ceftazidime.

Patients with CF, particularly those who are in school orholding full-time jobs, may opt to administer intravenousantibiotics at home for all or a portion of the treatmentcourse. This seems to work well in selected cases and maybe as effective as in-hospital treatment.344,345 For patientsrequiring frequent courses or long-term home antibiotictherapy, central intravenous catheters have been surgicallyplaced and used for prolonged periods.

One study suggested that intravenous infusion of immu-noglobulin (500 mg/kg) during a course of intravenousantibiotics enhances the pulmonary function response.346

Further studies are needed to confirm these observations.Interest in bronchial lavage as a therapeutic modality has

been tempered by the realization that long-term benefits areunlikely to accrue from this approach and that acute dete-rioration of lung function is a possible outcome.347 In fact,there is no evidence that bronchoscopy and lavage, whetherit be whole lung, lobar, or segmental, is superior to inten-sive antibiotic therapy and chest percussion.

Lung Complications

Hypercapnic and hypoxemic respiratory failure in CF areprimarily due to progressive obstructive airway disease withalveolar hypoventilation and ventilation/perfusion mis-matching. Treatment of airway infection and inflammationwith antibiotics as well as the airway clearance measuressummarized above is essential. Low-flow oxygen is effectiveat relieving nocturnal, exertional, and resting hypoxemiaand does not usually cause significant hypercapnia.348 Theuse of supplemental oxygen in children has been discour-aged349 because nocturnal oxygen did not improve survivalor prevent the development of cor pulmonale in one con-trolled trial.350 A larger study demonstrated that the devel-opment of pulmonary hypertension in adults with CF isstrongly correlated with hypoxemia and is associated withincreased mortality.351 Therefore, supplemental oxygen inaccordance with the guidelines established for chronicobstructive pulmonary disease352 is recommended.353

Diuretics, inotropic agents, and theophylline produce fewbenefits and are rarely used. Cor pulmonale is a late com-plication of airway obstruction, and no treatment optionsbeyond those for the primary disease processes are effective.

Ventilatory assistance is effective in CF patients with acuterespiratory failure caused by reversible insults,259 but it pro-duces few long-term benefits in patients with respiratoryfailure due to irreversible lung damage caused by CF bronch-iectasis.354 Assisted ventilation using nasal or face masks355 orendotracheal tubes356,357 can effectively treat patients awaitinglung transplantation. The airway disease generally progresses,however, and long-term ventilatory support is rarely feasible.

Atelectasis is best treated by vigorous standard therapy,including airway clearance and antibiotics. Corticosteroidsmay be helpful in the presence of reactive airways or severeairway inflammation. There is no evidence that bron-choscopy and lavage are effective in expanding collapsedsegments of lobes.247 ABPA responds to standard doses ofsystemic corticosteroids.358 Inhaled steroids359 and the oralantifungal agent itraconazole360 may prove to be useful.

Pneumothorax can be observed, allowing spontaneousresolution, if it is small and minimally symptomatic. A largepneumothorax (>20% of the hemithorax volume, compro-mising ventilation or causing hypotension) requires tubedrainage.229 Recurrences are very common and may requirethoracoscopic talc poudrage or surgical pleural abrasion.253

Hemoptysis requires treatment of airway infection andsupplemental vitamin K if the prothrombin time is pro-longed due to inadequate absorption. Massive hemoptysismay resolve with such conservative therapy and modestcough suppression for 1 to 2 days,249 but bronchial artery

38.qxd 1/25/05 2:47 PM Page 1238

Page 23: Cystic Fibrosis

embolization250,361 provides more definitive control, whichusually persists for more than 6 months.

Gastrointestinal Complications

Meconium ileus can be relieved in a number of cases withenemas using meglumine diatrizoate (Gastrografin) orother contrast materials, which are refluxed into the termi-nal ileum under fluoroscopy. If this fails, or if there is evidence of perforation, surgical intervention is required.Distal intestinal obstruction syndrome also can be relievedwith contrast enemas that reach the terminal ileum.However, this approach has been largely supplanted byintestinal “flushes” with a balanced salt solution; 1 to 2 litersrapidly instilled into the stomach is usually effective. Occa-sionally, surgical intervention is necessary. Rectal prolapseusually can be reduced voluntarily by older patients usingabdominal, perineal, and gluteal muscles, but in small chil-dren rectal prolapse must be reduced manually by continu-ous gentle pressure with the patient in the knee-chestposition265; sedation may be helpful. Adequate pancreaticenzyme therapy, decreased fat in the diet, and control ofpulmonary infection usually prevent recurrences. A fewpatients may require surgical stabilization of the rectum.

Cirrhosis is generally focal and usually does not require spe-cific therapy. Localization of CFTR to bile ductule cells sug-gests that abnormal bile secretion may be germane to CF liverdisease. Ursodeoxycholic acid (URSO) is effective treatmentfor primary biliary cirrhosis.362 Early trials of URSO in CFpatients showed improved liver function test results, but thelong-term effect on liver function is not known.363,364 Thethresholds for initiating therapy and clinical outcomes areunder investigation. Bleeding esophageal varices that com-plicate cirrhosis often can be managed with banding or scle-rotherapy. In the past, significant bleeding has been treatedsuccessfully with portosystemic shunting.365 Splenorenal anas-tomoses have been most effective, and hepatic encephalopa-thy has not been a problem. The transjugular intrahepaticportosystemic shunt procedure366,367 can relieve portal hyper-tension and reduce bleeding episodes. Liver failure and ascitesare treated as in other patients. Liver transplantation has beensuccessfully performed, with the 2-year survival exceeding50%.368,369 Pancreatitis, when it occurs in adolescents oryoung adults with CF, is treated with standard measures.

Hyperglycemia as a complication of CF can occur at anyage, but it is generally a problem of the second and thirddecades of life.268 Ketoacidosis is rarely encountered. Whenblood glucose levels are only intermittently elevated andglycosuria is not present, no treatment is necessary. Withthe advent of sustained glycosuria, insulin treatment shouldbe instituted. Oral hypoglycemic agents, considered inef-fective in the past, may be helpful in selected patients.370

The development of significant hyperglycemia may notchange longevity.371,372 However, vascular disease affectingthe retina and kidneys has been documented in CF patientswho have had prolonged hyperglycemia.373,374 Therefore,careful control of blood sugar levels is desirable.

Surgical Therapy

Nasal polypectomy to relieve obstruction is the mostcommon surgical procedure in CF, and most patients get

symptomatic improvement.375 Recurrence of polyps iscommon,376 but the incidence of polyposis usually wanesafter the second decade. Gallstones are common, and symp-tomatic disease may require elective cholecystectomy in asmany as 5% of CF adults.377 Lobectomy is occasionally indi-cated for massive hemoptysis that is refractory to bronchialartery embolization. Partial lung resection has been advo-cated for apparently localized disease and recurrent severeexacerbations.378 However, the generalized lung diseasecontinues to progress; the limited probability of long-termbenefits dictates caution in patient selection.

Transplantation

See Chapter 89 for a full discussion of lung transplantation.Lung transplantation has become an accepted treatment

for respiratory failure secondary to CF.379 Heart-lung trans-plant has been largely replaced by sequential double-lungtransplant because of limited organ availability. Patientsshould be referred when their prognosis is about equal tothe waiting time for donor lungs, currently about 2 yearsafter acceptance as a lung transplant candidate. More than1600 lung transplants have been performed for CF aroundthe world. The transplanted lungs remain free of CF but aresubject to secondary infection, acute rejection, and chronicrejection (bronchiolitis obliterans syndrome). The 5-yearsurvival is 48%—as good as that of lung transplant re-cipients with other causes of lung disease. Living lobartransplantation is an effective alternative to conventionalcadaveric lung transplants.380 The lobe donors must havesufficiently large lungs that their lower lobe fills the recipi-ent’s hemithorax. Survival appears to be similar to that fol-lowing conventional lung transplantation.

COURSE OF THE DISEASE AND PROGNOSIS

Cystic fibrosis runs a highly varied course, ranging fromdeath due to complications of meconium ileus in the firstdays of life or death from severe respiratory tract problemswithin the first months of life to essentially asymptomaticexistence for 10 to 20 years381 and protracted survival. A fewpatients with CF do live into the sixth and seventh decadesof life.261 More than 7500 (36%) of the patients in the U.S.Cystic Fibrosis Registry are 18 years of age or older.382 Thechanging age distribution is demonstrated in Figure 38.8.

At most care centers, patients with CF are monitored bygeneral clinical assessment, which often involves the use ofa scoring system,383,384 periodic monitoring of respiratorytract pathogens, periodic chest roentgenograms, serial pul-monary function testing, and ongoing nutritional assess-ment. Initial intensive treatment is usually followed byimprovement in pulmonary function tests as well as sub-stantial improvement of weight-to-height ratios and accel-erated linear growth in children. After initial improvementor stabilization, there is often an extended period of stablelung function, which may last 5, 10, or even 15 years. Pro-gressive dysfunction inevitably supervenes. Longitudinalpatterns of pulmonary function in older individuals with CFare highly variable.385,386

The most recent statistics from the Cystic Fibrosis Foun-dation indicate that 50% of patients can now be expected

123938 • Cystic Fibrosis

38.qxd 1/25/05 2:47 PM Page 1239

Page 24: Cystic Fibrosis

1240 Section J • OBSTRUCTIVE DISEASES

to survive beyond age 30 years. These data were generatedfrom all patients under care in Cystic Fibrosis Foundation-sponsored centers in 1997 (Fig. 38.9). Analyses of survivalcurves over the years have suggested that longevity hasincreased steadily to the present time. Some of the improve-ment in survival may have resulted from the diagnosis ofmilder forms of the disease. However, there is considerableopinion that improved symptomatic therapy has also con-tributed to better survival.

Multiple factors determine the prognosis for individualswith CF.387 Data from all U.S. CF centers suggest that sur-vival is better for patients living in northern climates thanfor those in the South. On average, male patients live 3 yearslonger than do female patients. Blacks who survive the firstseveral years of life may have a better prognosis than whites.Age at diagnosis has not emerged as a clear determinant ofsurvival. However, a study of sibling pairs suggests that theyounger sibling who is diagnosed before 1 year of age,before the onset of symptoms, usually has better pulmonaryfunction at 7 years of age than does the older sibling diag-nosed at an older age because of symptoms.388 Outcomeanalyses of patients who have been identified at birth byscreening procedures and those who have been diagnosedafter onset of symptoms should provide more definitive evi-dence for the efficacy of early diagnosis and treatment.

Several studies suggest that the clinical features presentwhen the patient is first seen make a difference. Patients whoinitially have only steatorrhea and failure to thrive generallyimprove remarkably after institution of therapy and do wellfor extended periods of time. On the other hand, patientswho have respiratory tract symptoms when first seen usuallycontinue to have respiratory tract problems and have a lessfavorable prognosis.389 When the entire CF population isconsidered, those patients who retain pancreatic exocrinefunction display a slower progression of pulmonarydisease.32,390 Meconium ileus does not influence longevity ifthe patient survives the first months of life. Initial claimsthat CF patients with allergies have improved survivalcannot be substantiated. There are numerous indicators ofpoor prognosis once airway disease has been established,

Per

cent

50

40

30

20

10

01969 1978 1992 1997 2002

8.0

18.0

32.8

36.1

40.2

Year Year

n = 624 2350 5244 6432 9288

Figure 38.8 Percent of cystic fibrosis patientsaged 18 or older for 1969 to 2002. Data are fromthe Patient Registry, Cystic Fibrosis Foundation,Bethesda, MD.

100

80

60

40

20

0

100

80

60

40

20

0

10 20 30 40

Median survival

30.6 Years

Per

cent

sur

viva

l

Age (y)

A

B 10 20 30 40

Age (y)

Per

cent

sur

viva

l

Median survival28.2 Years

Figure 38.9 Survival curve for male (A) and female (B) cysticfibrosis patients followed in all U.S. centers in 1991. This curvedemonstrates that the median survival age, or age to which50% of the patient population may be expected to survive,exceeds 30 years for males and 28 years for females. Aggregatemedian survival is 29.4 years. Data are from the PatientRegistry, Cystic Fibrosis Foundation, Bethesda, MD. (Courtesy ofDr. S. Fitzsimmons.)

38.qxd 1/25/05 2:47 PM Page 1240

Page 25: Cystic Fibrosis

including colonization with P. aeruginosa or B. cepacia andoccurrence of such pulmonary complications as pneumoth-orax and massive hemoptysis.391 The onset of insulin-dependent hyperglycemia has been considered a riskfactor,372 but some evidence suggests that survival rates ofthese patients parallel those of patients who have no overtproblem of glucose metabolism.392

Little is known about the influence of medical care onlongevity. Several studies suggest that patients followed atestablished care centers have a greater median survival thanthose who receive care in nonspecialized settings.393 In addi-tion, evidence suggests that center to center variation in sur-vival of patients is directly related to the intensity of carerendered in each center, including frequency of monitoring,antibiotic use, and hospitalization for treatment of pul-monary exacerbations.307

Finally, psychosocial factors undoubtedly play a promi-nent role in outcome, although this is difficult to docu-ment.394–396 The role of a supportive family is crucial. Suicideis uncommon, but it is widely recognized that a substantialnumber of adolescent and adult patients do not comply fully with therapy regimens because of denial, unresolveddependence/independence issues, and depression. Clearly,attitude and the ability to cope with a fatal illness duringmaturation and early adulthood do influence outcome.

Even though faced with a life-threatening illness and bur-dened with staggering demands on time, energy, and finan-cial resources to comply with therapy, most adolescents andyoung adults cope admirably and are able to achieve a sat-isfactory quality of life. Successful relationships,397 advancededucation, and a full-time occupation are frequentlyachieved (Table 38.8), indicating that independent lifestylesand full participation in life events are attainable goals formany patients.

SUMMARY

Cystic fibrosis is a prevalent autosomal-recessive syndromethat causes life-threatening chronic lung disease of child-hood and adulthood. It is caused by one or two of more

than 1000 identified mutations occurring in a gene on chro-mosome 7 that codes for an integral membrane protein ofepithelial cells (CFTR). This protein is a chloride channeland also regulates other ion channels in epithelial cells. Dis-turbed function results in alterations of electrolyte content,intraluminal volume, and diminished clearance of exocrinesecretions. Lung pathophysiology reflects airway obstruc-tion by accumulated mucus and secondary bacterial infection.

Diagnosis requires the identification of typical pulmonaryor pancreatic manifestations or a family history of CF com-bined with documentation of elevated chloride concentra-tions in sweat. Genotype analyses confirm the diagnosis andhave predictive implications for the clinical course.

Treatment currently focuses on control of mucus reten-tion and chronic infection in the lungs, replacement of pan-creatic enzymes, and nutritional therapy. Close monitoring,vigorous attempts to reverse deterioration of lung functionand nutritional deficits, and psychosocial support offer additional benefits.

The outcome is highly variable, but longevity hasincreased steadily and median survival now exceeds 31years. Quality of life for patients with CF has also improved.New approaches to therapy, including lung transplantation,pharmacologic interventions targeted to epithelial cellpathophysiology, and gene transfer to airway epitheliumoffer hope for further substantial therapeutic advances.

REFERENCES

1. Davis PB, Drumm M, Konstan MW: Cystic fibrosis. Am JRespir Crit Care Med 154:1229–1256, 1996.

2. Zielenski J, Tsui LC: Cystic fibrosis: Genotypic andphenotypic variations. Annu Rev Genet 29:777–807, 1995.

3. Taussig LM. Cystic Fibrosis. New York: Thieme-Stratton,1984.

4. Andersen DH: Cystic fibrosis of the pancreas and itsrelation to celiac disease: A clinical and pathologic study.Am J Dis Child 56:344–399, 1938.

5. Farber S: Some organic digestive disturbances in early life. J Mich Med Sci 44:587–594, 1945.

6. Andersen DH, Hodges RG: Celiac syndrome. V. Geneticsof cystic fibrosis of the pancreas with a consideration ofetiology. Am J Dis Child 72:62–80, 1946.

7. Di Sant’Agnese PA, Darling RC, Perera GA, Shea E:Abnormal electrolyte composition of sweat in cystic fibrosisof the pancreas. Pediatrics 12:549–563, 1953.

8. Gibson LE, Cooke RE: A test for concentration ofelectrolytes in sweat in cystic fibrosis of the pancreasutilizing pilocarpine by iontophoresis. Pediatrics23:545–549, 1959.

9. Knowles M, Gatzy J, Boucher R: Increased bioelectricpotential difference across respiratory epithelia in cysticfibrosis. N Engl J Med 305:1489–1495, 1981.

10. Quinton PM, Bijman J: Higher bioelectric potentials dueto decreased chloride absorption in the sweat glands ofpatients with cystic fibrosis. N Engl J Med308:1185–1189, 1983.

11. Tsui LC, Buchwald M, Barker D, et al: Cystic fibrosis locusdefined by a genetically linked polymorphic DNA marker.Science 230:1054–1057, 1985.

12. Zengerling S, Tsui LC, Grzeschik KH, et al: Mapping ofDNA markers linked to the cystic fibrosis locus on the longarm of chromosome 7. Am J Hum Genet 40:228–236,1987.

124138 • Cystic Fibrosis

Table 38.8 Educational and Employment Records of Adultswith Cystic Fibrosis

Parameter %

Highest Educational Degree (n = 686)Associate 10Bachelor’s 14Master’s 5Doctorate 2Other 4No advanced degree or no response 65

Employment (n = 679)Full time 37Part time 21Unemployed, seeking work 12Unemployed, not seeking work 30

Data are from a national survey by the Cystic Fibrosis Foundation.Adapted from Lewiston NJ: Psychosocial impact of cystic fibrosis.Semin Respir Med 6:321–332, 1985.

38.qxd 1/25/05 2:47 PM Page 1241

Page 26: Cystic Fibrosis

1242 Section J • OBSTRUCTIVE DISEASES

13. White R, Woodward S, Leppert M, et al: A closely linkedgenetic marker for cystic fibrosis. Nature 318:382–384,1985.

14. Wainwright BJ, Scambler PJ, Schmidtke J, et al:Localization of cystic fibrosis locus to human chromosome7cen-q22. Nature 318:384–385, 1985.

15. Riordan JR, Rommens JM, Kerem B-T, et al: Identificationof the cystic fibrosis gene: Cloning and characterization ofcomplementary DNA. Science 245:1066–1073, 1989.

16. Kerem B, Rommens JM, Buchanan JA, et al: Identificationof the cystic fibrosis gene: Genetic analysis. Science245:1073–1080, 1989.

17. Drumm ML, Pope HA, Cliff WH, et al: Correction of thecystic fibrosis defect in vitro by retrovirus-mediated genetransfer. Cell 62:1227–1233, 1990.

18. Rich DP, Anderson MP, Gregory RJ, et al: Expression ofcystic fibrosis transmembrane conductance regulatorcorrects defective chloride channel regulation in cysticfibrosis airway epithelial cells. Nature 347:358–363, 1990.

19. Anderson MP, Rich DP, Gregory RJ, et al: Generation ofcAMP-activated chloride currents by expression of CFTR.Science 251:679–682, 1991.

20. Anderson MP, Gregory RJ, Thompson S, et al:Demonstration that CFTR is a chloride channel byalteration of its anion selectivity. Science 253:202–205,1991.

21. Bear CE, Li C, Kartner N, et al: Purification and functionalreconstitution of the cystic fibrosis transmembraneconductance regulator (CFTR). Cell 68:809–818, 1992.

22. Egan M, Flotte T, Afione S, et al: Defective regulation ofoutwardly rectifying Cl- channels by protein kinase Acorrected by insertion of CFTR. Nature 358:581–584, 1992.

23. Stutts MJ, Canessa CM, Olsen JC, et al: CFTR as a cAMP-dependent regulator of sodium channels. Science269:847–850, 1995.

24. Cheng SH, Gregory RJ, Marshall J, et al: Defectiveintracellular transport and processing of CFTR is themolecular basis of most cystic fibrosis. Cell 63:827–834,1990.

25. Jensen TJ, Loo MA, Pind S, et al: Multiple proteolyticsystems, including the proteasome, contribute to CFTRprocessing. Cell 83:129–135, 1995.

26. Gunderson KL, Kopito RR: Conformational states ofCFTR associated with channel gating: The role of ATPbinding and hydrolysis. Cell 82:231–239, 1995.

27. Snouwaert JN, Brigman KK, Latour AM, et al: An animalmodel for cystic fibrosis made by gene targeting. Science257:1083–1088, 1992.

28. Clarke LL, Grubb BR, Gabriel SE, et al: Defectiveepithelial chloride transport in a gene targeted mousemodel of cystic fibrosis. Science 257:1125–1128, 1992.

29. Dorin JR, Dickinson P, Alton EWFW, et al: Cystic fibrosisin the mouse by targeted insertional mutagenesis. Nature359:211–215, 1992.

30. Grubb BR, Boucher RC: Pathophysiology of gene-targetedmouse models for cystic fibrosis. Physiol Rev79:S193–S214, 1999.

31. Tsui L-C, Buchwald M: Biochemical and moleculargenetics of cystic fibrosis. Adv Hum Genet 20:153–266,1991.

32. Kerem E, Corey M, Kerem B-S, et al: The relationbetween genotype and phenotype in cystic fibrosis: Analysisof the most common mutation (DF508). N Engl J Med323:1517–1522, 1990.

33. Liechti-Gallati S, Bonsall I, Malik N, et al:Genotype/phenotype association in cystic fibrosis: Analysesof the delta F508, R553X, and 3905insT mutations.Pediatr Res 32:175–178, 1992.

34. Cystic Fibrosis Foundation. Patient Registry Annual DataReport 2002. Bethesda, MD: Cystic Fibrosis Foundation,2003.

35. Knowles MR, Friedman KJ, Silverman L: Genetics,diagnosis, and clinical phenotype. In Cystic Fibrosis inAdults. Philadelphia: Lippincott-Raven, 1999, pp 27–42.

36. Gaskin KJ: Intestines. In Cystic Fibrosis in Adults.Philadelphia: Lippincott-Raven, 1999, pp 325–342.

37. Colombo C, Crosignani A, Melzi ML, et al: Hepatobiliarysystem. In Cystic Fibrosis in Adults. Philadelphia:Lippincott-Raven, 1999, pp 309–324.

38. Duthie A, Doherty DG, Williams C, et al: Genotypeanalysis for delta F508, G551D and R553X mutations inchildren and young adults with cystic fibrosis with andwithout chronic liver disease. Hepatology 15:660–664,1992.

39. Zielenski J, Corey M, Rozmahel R, et al: Detection of acystic fibrosis modifier locus for meconium ileus on humanchromosome 19q13. Nat Genet 22:128–129, 1999.

40. Highsmith WE, Burch LH, Zhou Z, et al: A novelmutation in the cystic fibrosis gene in patients withpulmonary disease but normal sweat chlorideconcentrations. N Engl J Med 331:974–980, 1994.

41. Strong TV, Smit LS, Turpin SV, et al: Cystic fibrosis genemutation in two sisters with mild disease and normal sweatelectrolyte levels. N Engl J Med 325:1630–1634, 1991.

42. Noone PG, Knowles MR: ‘CFTR-opathies’: Diseasephenotypes associated with cystic fibrosis transmembraneregulator gene mutations. Respir Res 2:328–332, 2001.

43. Cohn JA, Friedman KJ, Noone PG, et al: Relation betweenmutations of the cystic fibrosis gene and idiopathicpancreatitis. N Engl J Med 339:653–658, 1998.

44. Wilschanski M, Corey M, Durie P, et al: Diversity ofreproductive tract abnormalities in men with cystic fibrosis.JAMA 276:607–608, 1996.

45. Knowles MR, Durie PR: What is cystic fibrosis? N Engl JMed 347:439–442, 2002.

46. Merlo CA, Boyle MP: Modifier genes in cystic fibrosis lungdisease. J Lab Clin Med 141:237–241, 2003.

47. Wilfond BS, Fost N: The cystic fibrosis gene: Medical andsocial implications for heterozygote detection. JAMA263:2777–2783, 1990.

48. Danks DM, Allan J, Anderson CM: A genetic study offibrocystic disease of the pancreas. Ann Hum Genet28:323–356, 1965.

49. Ten Kate LP: A method for analysing fertility ofheterozygotes for autosomal recessive disorders, withspecial reference to cystic fibrosis, Tay-Sachs disease andphenylketonuria. Ann Hum Genet 40:287–297, 1977.

50. Romeo G, Devoto M, Galietta LJV: Why is the cysticfibrosis gene so frequent? Hum Genet 84:1–5, 1989.

51. Boat TF, Dearborn DG: Etiology and pathogenesis. InCystic Fibrosis. New York: Thieme-Stratton, 1984, pp25–84.

52. Gabriel SE, Brigman KN, Koller BH, et al: Cystic fibrosisheterozygote resistance to cholera toxin in the cysticfibrosis mouse model. Science 266:107–109, 1994.

53. Zuelzer WW, Newton WA Jr: The pathogenesis offibrocystic disease of the pancreas: A study of 36 cases withspecial reference to the pulmonary lesions. Pediatrics4:53–69, 1949.

54. Sturgess J, Imrie J: Quantitative evaluation of thedevelopment of tracheal submucosal glands in infants withcystic fibrosis and control infants. Am J Pathol106:303–311, 1982.

55. Baker D, Kupke KG, Ingram P, et al: Microprobe analysisin human pathology. Scan Electron Microsc Pt 2:659–680,1985.

38.qxd 1/25/05 2:47 PM Page 1242

Page 27: Cystic Fibrosis

56. Tomashefski JF Jr, Bruce M, Goldberg HI, Dearborn DG:Regional distribution of macroscopic lung disease in cysticfibrosis. Am Rev Respir Dis 133:535–540, 1986.

57. Sobonya RE, Taussig LM: Quantitative aspects of lungpathology in cystic fibrosis. Am Rev Respir Dis134:290–295, 1986.

58. Ogrinc G, Kampalath B, Tomashefski JF Jr: Destructionand loss of bronchial cartilage in cystic fibrosis. HumPathol 29:65–73, 1998.

59. Hamutcu R, Rowland JM, Horn MV, et al: Clinicalfindings and lung pathology in children with cystic fibrosis.Am J Respir Crit Care Med 165:1172–1175, 2002.

60. Tomashefski JF Jr, Dahms BB, Abramowsky CR: Thepathology of cystic fibrosis. In Cystic Fibrosis, vol 1. NewYork: Marcel Dekker, 1993, pp 435–489.

61. Tomashefski JF Jr, Konstan MW, Bruce M: The pathologyof interstitial pneumonia in cystic fibrosis. Am Rev RespirDis 133:A365, 1986.

62. Tomashefski JF Jr, Bruce M, Stern RC, et al: Pulmonaryair cysts in cystic fibrosis: Relation of pathologic features toradiologic findings and history of pneumothorax. HumPathol 16:253–261, 1985.

63. Boat TF, di Sant’Agnese PA, Warwick WJ, HandwergerSA: Pneumothorax in cystic fibrosis. JAMA209:1498–1504, 1969.

64. Mack JF, Moss AF, Harper WW, O’Loughlin BJ: Thebronchial arteries in cystic fibrosis. JAMA 209:1498–1504,1965.

65. Stern RC, Boat TF, Wood RE, et al: Treatment andprognosis of nasal polyps in cystic fibrosis. Am J Dis Child136:1067–1070, 1982.

66. Di Sant’Agnese PA, Hubbard VS: The pancreas. In CysticFibrosis. New York: Thieme-Stratton, 1984, pp 230–295.

67. Handwerger S, Roth J, Gorden P, et al: Glucoseintolerance in cystic fibrosis. N Engl J Med 281:451–461,1969.

68. Di Sant’Agnese PA, Hubbard VS: The hepatobiliarysystem. In Cystic Fibrosis. New York: Thieme-Stratton,1984, pp 296–322.

69. Di Sant’Agnese PA, Hubbard VS: The gastrointestinaltract. In Cystic Fibrosis. New York: Thieme-Stratton, 1984,pp 212–229.

70. Neutra MR, Trier JS: Rectal mucosa in cystic fibrosis:Morphological features before and after short term organculture. Gastroenterology 75:701–710, 1978.

71. Brugman SM, Taussig LM: The reproductive system. InCystic Fibrosis. New York: Thieme-Stratton, 1984, pp323–337.

72. Gregory RJ, Cheng SH, Rich DP, et al: Expression andcharacterization of the cystic fibrosis transmembraneconductance regulator. Nature 347:382–386, 1990.

73. Short DB, Trotter KW, Reczek D, et al: An apical PDZprotein anchors the cystic fibrosis transmembraneconductance regulator to the cytoskeleton. J Biol Chem273:19797–19801, 1998.

74. Hyde SC, Emsley P, Hartshorn MJ, et al: Structural modelof ATP-binding proteins associated with cystic fibrosis,multidrug resistance and bacterial transport. Nature346:362–365, 1990.

75. Engelhardt JF, Yankaskas JR, Ernst SA, et al: Submucosalglands are the predominant site of CFTR expression inhuman bronchus. Nat Genet 2:240–247, 1992.

76. Reddy MM, Quinton PM: Intracellular potentials ofmicroperfused human sweat duct cells. Pfluegers Arch410:471–475, 1987.

77. Riordan JR: Cystic fibrosis as a disease of misprocessing ofthe cystic fibrosis transmembrane conductance regulatorglycoprotein. Am J Hum Genet 64:1499–1504, 1999.

78. Denning GM, Anderson MP, Amara JF, et al: Processing ofmutant cystic fibrosis transmembrane conductanceregulator is temperature-sensitive. Nature 358:761–764,1992.

79. Gregory RJ, Rich DP, Cheng SH, et al: Maturation andfunction of cystic fibrosis transmembrane conductanceregulator variants bearing mutations in putative nucleotide-binding domains 1 and 2. Mol Cell Biol 11:3886–3893,1991.

80. Kalin N, Claabeta A, Sommer M, et al: DeltaF508 CFTRprotein expression in tissues from patients with cysticfibrosis. J Clin Invest 103:1379–1389, 1999.

81. Bronsveld I, Mekus F, Bijman J, et al: Residual chloridesecretion in intestinal tissue of deltaF508 homozygoustwins and siblings with cystic fibrosis: The European CFTwin and Sibling Study Consortium. Gastroenterology119:32–40, 2000.

82. Mall M, Kreda SM, Mengos A, et al: The DeltaF508mutation results in loss of CFTR function and matureprotein in native human colon. Gastroenterology126:32–41, 2004.

83. Anderson MP, Welsh MJ: Regulation by ATP and ADP ofCFTR chloride channels that contain mutant nucleotide-binding domains. Science 257:1701–1704, 1992.

84. Drumm M. What happens to Delta F508 in vivo? J ClinInvest 103:1369–1370, 1999.

85. Bradbury NA, Jilling T, Berta G, et al: Regulation ofplasma membrane recycling by CFTR. Science256:530–532, 1992.

86. Bradbury NA, Cohn JA, Venglarik CJ, Bridges RJ:Biochemical and biophysical identification of cystic fibrosistransmembrane conductance regulator chloride channels ascomponents of endocytic clathrin-coated vesicles. J BiolChem 269:8296–8302, 1994.

87. Berger HA, Anderson MP, Gregory RJ, et al: Identificationand regulation of the cystic fibrosis transmembraneconductance regulator-generated chloride channel. J ClinInvest 88:1422–1431, 1991.

88. Hwang T-C, Lu L, Zeitlin PL, et al: Cl- channels in CF:Lack of activation by protein kinase C and cAMP-dependent protein kinase. Science 244:1351–1356, 1989.

89. Fischer H, Machen TE: CFTR displays voltage dependenceand two gating modes during stimulation. J Gen Physiol104:541–566, 1994.

90. Anderson MP, Berger HA, Rich DP, et al: Nucleosidetriphosphates are required to open the CFTR chloridechannel. Cell 67:775–784, 1991.

91. Cheng SH, Rich DP, Marshall J, et al: Phosphorylation ofthe R domain by cAMP-dependent protein kinase regulatesthe CFTR chloride channel. Cell 66:1027–1036, 1991.

92. Knowles MR, Stutts MJ, Spock A, et al: Abnormal ionpermeation through cystic fibrosis respiratory epithelium.Science 221:1067–1070, 1983.

93. Davis PB: Pathophysiology of the lung disease in cysticfibrosis. In Cystic Fibrosis (Lung Biology in Health andDisease), vol 64. New York: Marcel Dekker, 1993, pp193–218.

94. Smith JJ, Travis SM, Greenberg EP, Welsh MJ: Cysticfibrosis airway epithelia fail to kill bacteria because ofabnormal airway surface fluid. Cell 85:229–236, 1996.

95. Zabner J, Smith JJ, Karp PH, et al: Loss of CFTR chloridechannels alters salt absorption by cystic fibrosis airwayepithelia in vitro. Mol Cell 2:397–403, 1998.

96. Ganz T: Antimicrobial polypeptides in host defense of therespiratory tract. J Clin Invest 109:693–697, 2002.

97. Matsui H, Randell SH, Peretti SW, et al: Coordinatedclearance of periciliary liquid and mucus from airwaysurfaces. J Clin Invest 102:1125–1131, 1998.

124338 • Cystic Fibrosis

38.qxd 1/25/05 2:47 PM Page 1243

Page 28: Cystic Fibrosis

1244 Section J • OBSTRUCTIVE DISEASES

98. Matsui H, Grubb BR, Tarran R, et al: Evidence forpericiliary liquid layer depletion, not abnormal ioncomposition, in the pathogenesis of cystic fibrosis airwaysdisease. Cell 95:1005–1015, 1998.

99. Kohler D, App E, Schmitz-Schumann M, et al: Inhalationof amiloride improves the mucociliary and the coughclearance in patients with cystic fibroses. Eur J Respir Dis69(Suppl 146):319–326, 1986.

100. Regnis JA, Robinson M, Bailey DL, et al: Mucociliaryclearance in patients with cystic fibrosis and in normalsubjects. Am J Respir Crit Care Med 150:66–71, 1994.

101. Tarran R, Grubb BR, Gatzy JT, et al: The relative roles ofpassive surface forces and active ion transport in themodulation of airway surface liquid volume andcomposition. J Gen Physiol 118:223–236, 2001.

102. Knowles M, Gatzy J, Boucher R: Relative ion permeabilityof normal and cystic fibrosis nasal epithelium. J Clin Invest71:1410–1417, 1983.

103. Boucher RC, Stutts MJ, Knowles MR, et al: Na+ transportin cystic fibrosis respiratory epithelia: Abnormal basal rateand response to adenylate cyclase activation. J Clin Invest78:1245–1252, 1986.

104. Cotton CU, Stutts MJ, Knowles MR, et al: Abnormalapical cell membrane in cystic fibrosis respiratoryepithelium: An in vitro electrophysiologic analysis. J ClinInvest 79:80–85, 1987.

105. Widdicombe JH, Welsh MJ, Finkbeiner WE: Cystic fibrosisdecreases the apical membrane chloride permeability ofmonolayers cultured from cells of tracheal epithelium. ProcNatl Acad Sci USA 82:6167–6171, 1985.

106. Willumsen NJ, Boucher RC: Shunt resistance and ionpermeabilities in normal and cystic fibrosis airwayepithelium. Am J Physiol 256:C1054–C1063, 1989.

107. Willumsen NJ, Davis CW, Boucher RC: Intracellular Cl-

activity and cellular Cl- pathways in cultured human airwayepithelium. Am J Physiol 256:C1033–C1044, 1989.

108. Willumsen NJ, Davis CW, Boucher RC: Cellular Cl-

transport in cultured cystic fibrosis airway epithelium. Am JPhysiol 256:C1045–C1053, 1989.

109. Frizzell RA, Rechkemmer G, Shoemaker RL: Alteredregulation of airway epithelial cell chloride channels incystic fibrosis. Science 233:558–560, 1986.

110. Welsh MJ: Abnormal regulation of ion channels in cysticfibrosis epithelia. FASEB J 4:2718–2725, 1990.

111. Welsh MJ, Liedtke CM: Chloride and potassium channelsin cystic fibrosis airway epithelia. Nature 322:467–470,1986.

112. Boucher RC, Cheng EHC, Paradiso AM, et al: Chloridesecretory response of cystic fibrosis human airway epithelia:Preservation of calcium but not protein kinase C- and A-dependent mechanisms. J Clin Invest 84:1424–1431,1989.

113. Schoumacher RA, Shoemaker RL, Halm DR, et al:Phosphorylation fails to activate chloride channels fromcystic fibrosis airway cells. Nature 330:752–754, 1987.

114. Li M, McCann JD, Anderson MP, et al: Regulation ofchloride channels by protein kinase C in normal and cysticfibrosis airway epithelia. Science 244:1353–1356, 1989.

115. Tabcharani JA, Chang XB, Riordan JR, Hanrahan JW:Phosphorylation-regulated Cl- channel in CHO cells stablyexpressing the cystic fibrosis gene. Nature 352:628–631,1991.

116. Luo J, Pato MD, Riordan JR, Hanrahan JW: Differentialregulation of single CFTR channels by PP2C, PP2A, andother phosphatases. Am J Physiol 274:C1397–C1410,1998.

117. Berger HA, Travis SM, Welsh MJ: Regulation of the cysticfibrosis transmembrane conductance regulator Cl- channel

by specific protein kinases and protein phosphatases. J BiolChem 268:2037–2047, 1993.

118. Willumsen NJ, Boucher RC: Activation of an apical Cl-

conductance by Ca2+ ionophores in cystic fibrosis airwayepithelia. Am J Physiol 256:C226–C233, 1989.

119. Wagner JA, McDonald TV, Nghiem PT, et al: Antisenseoligodeoxynucleotides to the cystic fibrosis transmembraneconductance regulator inhibit cAMP-activated but notcalcium-activated chloride currents. Proc Natl Acad SciUSA 89:6785–6789, 1992.

120. Clarke LL, Grubb BR, Yankaskas JR, et al: Relationship ofa non-CFTR mediated chloride conductance to organ-leveldisease in cftr(-/-) mice. Proc Natl Acad Sci USA91:479–483, 1994.

121. Anderson MP, Welsh MJ: Calcium and cAMP activatedifferent chloride channels in the apical membrane ofnormal and cystic fibrosis epithelia. Proc Natl Acad SciUSA 88:6003–6007, 1991.

122. Worrell RT, Frizzell RA: CaMKII mediates stimulation ofchloride conductance by calcium in T84 cells. Am J Physiol260:C877–C882, 1991.

123. Brown HA, Lazarowski ER, Boucher RC, Harden TK:Evidence that UTP and ATP regulate phospholipase Cthrough a common extracellular 5¢-nucleotide receptor inhuman airway epithelial cells. Mol Pharmacol 40:648–655,1991.

124. Mason SJ, Paradiso AM, Boucher RC: Regulation oftransepithelial ion transport and intracellular calcium byextracellular adenosine triphosphate in human normal andcystic fibrosis airway epithelium. Br J Pharmacol103:1649–1656, 1991.

125. Knowles MR, Clarke LL, Boucher RC: Activation byextracellular nucleotides of chloride secretion in the airwayepithelia of patients with cystic fibrosis. N Engl J Med325:533–538, 1991.

126. Clarke LL, Boucher RC: Chloride secretory response toextracellular ATP in normal and cystic fibrosis nasalepithelia. Am J Physiol 263:C348–C356, 1992.

127. Noone PG, Hamblett N, Accurso F, et al: Safety ofaerosolized INS 365 in patients with mild to moderatecystic fibrosis: Results of a phase I multi-center study.Pediatr Pulmonol 32:122–128, 2001.

128. Yerxa BR, Sabater JR, Davis CW, et al: Pharmacology ofINS37217 [P1-(uridine 5¢)-P4-(2¢deoxycytidine 5¢)tetraphosphate, tetrasodium salt], a next generation P2Y2

receptor agonist for the treatment of cystic fibrosis. JPharmacol Exp Ther 302:871–880, 2002.

129. Robson AM, Tateishi S, Ingelfinger JR, et al: Renalfunction in cystic fibrosis. J Pediatr 79:42–50, 1971.

130. Stenvinkel P, Hjelte L, Alvan G, et al: Decreased renalclearance of sodium in cystic fibrosis. Acta Paediatr Scand80:194–198, 1991.

131. O’Loughlin EV, Hunt DM, Gaskin KJ, et al: Abnormalepithelial transport in cystic fibrosis jejunum. Am J Physiol260:G758–G763, 1991.

132. Baxter P, Goldhill J, Hardcastle J, et al: Enhancedintestinal glucose and alanine transport in cystic fibrosis.Gut 31:817–820, 1990.

133. Berschneider HM, Knowles MR, Azizkhan RG, et al:Altered intestinal chloride transport in cystic fibrosis.FASEB J 2:2625–2629, 1988.

134. Willumsen NJ, Boucher RC: Sodium transport andintracellular sodium activity in cultured human nasalepithelium. Am J Physiol 261:C319–C331, 1991.

135. Willumsen NJ, Boucher RC: Transcellular sodiumtransport in cultured cystic fibrosis human nasal epithelium.Am J Physiol 261:C332–C341, 1991.

38.qxd 1/25/05 2:47 PM Page 1244

Page 29: Cystic Fibrosis

136. Chinet T, Fullton J, Yankaskas J, et al: Characterization ofsodium channels in the apical membrane of nasal epithelialcells. Am Rev Respir Dis 141:A164, 1990.

137. Chinet T, Fullton J, Boucher R, Stutts J: Sodium channelsin the apical membrane of normal and CF nasal epithelialcells. Pediatr Pulmonol (Suppl 5):209, 1990

138. Chinet T, Fullton J, Yankaskas J, et al: Characteristics ofcation channels in cell-attached patches of normal and CFnasal epithelial cells. Am Rev Respir Dis 143:A148, 1991.

139. Mall M, Grubb BR, Harkema JR, et al: Increased airwayepithelial Na+ absorption produces cystic fibrosis-like lungdisease in mice. Nat Med 10:487–493, 2004.

140. Devor DC, Pilewski JM: UTP inhibits Na+ absorption inwild-type and DeltaF508 CFTR-expressing humanbronchial epithelia. Am J Physiol 276:C827–C837, 1999.

141. Mall M, Wissner A, Gonska T, et al: Inhibition ofamiloride-sensitive epithelial Na(+) absorption byextracellular nucleotides in human normal and cysticfibrosis airways. Am J Respir Cell Mol Biol 23:755–761,2000.

142. Engelhardt JF, Stratford-Perricaudet LD, Yang Y, et al:Direct transfer of recombinant genes into human bronchialepithelia of xenografts with E1 deleted adenoviruses. NatGenet 4:27–34, 1993.

143. Knowles MR, Robinson JM, Wood RE, et al: Ioncomposition of airway surface liquid of patients with cysticfibrosis as compared to normal and disease-controlsubjects. J Clin Invest 100:2588–2595, 1997.

144. Yamaya M, Finkbeiner WE, Widdicombe JH: Ion transportby cultures of human tracheobronchial submucosal glands.Am J Physiol 261:L485–L490, 1991.

145. Yamaya M, Finkbeiner WE, Widdicombe JH: Altered iontransport by tracheal glands in cystic fibrosis. Am J Physiol261:L491–L494.

146. Boat TF, Cheng PW: Biochemistry of airway mucussecretions. Fed Proc 39:3067–3074, 1980.

147. Davril M, Degroote S, Humbert P, et al: The sialylation ofbronchial mucins secreted by patients suffering from cysticfibrosis or from chronic bronchitis is related to the severityof airway infection. Glycobiology 9:311–321, 1999.

148. Wesley A, Forstner J, Qureshi R, et al: Human intestinalmucin in cystic fibrosis. Pediatr Res 17:65–69, 1983.

149. Cheng P-W, Boat TF, Cranfill K, et al: Increased sulfationof glycoconjugates by cultured nasal epithelial cells frompatients with cystic fibrosis. J Clin Invest 84:68–72, 1989.

150. Frates RC Jr, Kaizu TT, Last JA: Mucus glycoproteinssecreted by respiratory epithelial tissue from cystic fibrosispatients. Pediatr Res 17:30–34, 1983.

151. Zhang Y, Jiang Q, Dudus L, et al: Vector-specificcomplementation profiles of two independent primarydefects in cystic fibrosis airways. Hum Gene Ther9:635–648, 1998.

152. Roomans GM, von Euler AM, Muller RM, Gilljam H: X-ray microanalysis of goblet cells in bronchial epitheliumof patients with cystic fibrosis. J Submicrosc Cytol18:613–615, 1986.

153. Holmen JM, Karlsson NG, Abdullah LH, et al: Mucinsand their O-Glycans from human bronchial epithelial cellcultures. Am J Physiol Lung Cell Mol Physiol287:L824–L834, 2004.

154. Leigh MW, Boat TF: Airway secretions. In BasicMechanisms of Pediatric Respiratory Disease: Cellular and Integrative. Philadelphia: B.C. Decker, 1991, pp 328–345.

155. Lam J, Chan R, Lam K, Costerton JW: Production ofmucoid microcolonies by Pseudomonas aeruginosa withininfected lungs in cystic fibrosis. Infect Immun 28:546–556,1980.

156. Bennett WD, Olivier KN, Zeman KL, et al: Effect ofuridine 5’-triphosphate plus amiloride on mucociliaryclearance in adult cystic fibrosis. Am J Respir Crit CareMed 153:1796–1801, 1996.

157. Katz SM, Holsclaw DS Jr: Ultrastructural features ofrespiratory cilia in cystic fibrosis. Am J Clin Pathol73:682–685, 1980.

158. Rutland J, Cole PJ: Nasal mucociliary clearance and ciliarybeat frequency in cystic fibrosis compared with sinusitis andbronchiectasis. Thorax 36:654–658, 1981.

159. Ulrich M, Herbert S, Berger J, et al: Localization ofStaphylococcus aureus in infected airways of patients withcystic fibrosis and in a cell culture model of S. aureusadherence. Am J Respir Cell Mol Biol 19:83–91, 1998.

160. Saiman L, Cacalano G, Gruenert D, Prince A: Comparisonof adherence of Pseudomonas aeruginosa to respiratoryepithelial cells from cystic fibrosis patients and healthysubjects. Infect Immun 60:2808–2814, 1992.

161. Plotkowski MC, Chevillard M, Pierrot D, et al: Epithelialrespiratory cells from cystic fibrosis patients do not possessspecific Pseudomonas aeruginosa-adhesive properties. J Med Microbiol 36:104–111, 1992.

162. Pier GB, Grout M, Zaidi TS, et al: Role of mutant CFTRin hypersusceptibility of cystic fibrosis patients to lunginfections. Science 271:64–67, 1996.

163. Puchelle E, de Bentzmann S, Zahm JM: Physical andfunctional properties of airway secretions in cystic fibrosis:Therapeutic approaches. Respiration 62(Suppl 1):2–12,1995.

164. Worlitzsch D, Tarran R, Ulrich M, et al: Effects of reducedmucus oxygen concentration in airway Pseudomonasinfections of cystic fibrosis patients. J Clin Invest109:317–325, 2002.

165. Yoon SS, Hennigan RF, Hilliard GM, et al: Pseudomonasaeruginosa anaerobic respiration in biofilms: Relationshipsto cystic fibrosis pathogenesis. Dev Cell 3:593–603, 2002.

166. Li JD, Dohrman AF, Gallup M, et al: Transcriptionalactivation of mucin by Pseudomonas aeruginosalipopolysaccharide in the pathogenesis of cystic fibrosislung disease. Proc Natl Acad Sci USA 94:967–972, 1997.

167. Dohrman A, Miyata S, Gallup M, et al: Mucin gene (MUC2 and MUC 5AC) upregulation by gram-positive and gramnegative bacteria. Biochim Biophys Acta 1406:251–259,1998.

168. DiMango E, Zar HJ, Bryan R, Prince A: DiversePseudomonas aeruginosa gene products stimulaterespiratory epithelial cells to produce interleukin-8. J ClinInvest 96:2204–2210, 1995.

169. Plotkowski MC, Zahm JM, Tournier JM, Puchelle E:Pseudomonas aeruginosa adhesion to normal and injuredrespiratory mucosa. Mem Inst Oswaldo Cruz 87(SupplV):61–68, 1992.

170. Matthews WJ Jr, Williams M, Oliphint B, et al:Hypogammaglobulinemia in patients with cystic fibrosis. N Engl J Med 302:245–249, 1980.

171. Gugler E, Pallavicini JC, Swedlow H, et al: Immunologicalstudies of submaxillary saliva from patients with cystic fibrosisand from normal children. J Pediatr 73:548–559, 1968.

172. Doring G, Obernesser HJ, Botzenhart K, et al: Proteasesof Pseudomonas aeruginosa in patients with cystic fibrosis.J Infect Dis 147:744–750, 1983.

173. Baltimore RS, Fick RB Jr, Fino L: Antibody to multiplemucoid strains of Pseudomonas aeruginosa in patients withcystic fibrosis, measured by an enzyme-linkedimmunosorbent assay. Pediatr Res 20:1085–1090, 1986.

174. Sorensen RU, Ruuskanen O, Miller K, Stern RC: B-lymphocyte function in cystic fibrosis. Eur J Respir Dis64:524–533, 1983.

124538 • Cystic Fibrosis

38.qxd 1/25/05 2:47 PM Page 1245

Page 30: Cystic Fibrosis

1246 Section J • OBSTRUCTIVE DISEASES

175. Sorensen RU, Stern RC, Polmar SH: Cellular immunity tobacteria: Impairment of in vitro lymphocyte responses toPseudomonas aeruginosa in cystic fibrosis patients. InfectImmun 18:735–740, 1977.

176. Sorensen RU, Stern RC, Chase P, Polmar SH: Defectivecellular immunity to gram-negative bacteria in cysticfibrosis patients. Infect Immun 23:398–402, 1979.

177. Thomassen MJ, Demko CA, Wood RE, et al:Ultrastructure and function of alveolar macrophages fromcystic fibrosis patients. Pediatr Res 14:715–721, 1980.

178. Matsui H: Behavior of neutrophils and Pseudomonas inthick airway mucus. Pediatr Pulmonol (Suppl25):154–155, 2003.

179. Konstan MW, Berger M: Current understanding of theinflammatory process in cystic fibrosis: Onset and etiology.Pediatr Pulmonol 24:137–142, 1997.

180. Noah TL, Black HR, Cheng P-W, et al: Nasal andbronchoalveolar lavage fluid cytokines in early cysticfibrosis. J Infect Dis 175:638–647, 1997.

181. Bonfield TL, Konstan MW, Burfeind P, et al: Normalbronchial epithelial cells constitutively produce the anti-inflammatory cytokine interleukin-10, which isdownregulated in cystic fibrosis. Am J Respir Cell Mol Biol13:257–261, 1995.

182. Bonfield TL, Panuska JR, Konstan MW, et al:Inflammatory cytokines in cystic fibrosis lungs. Am J RespirCrit Care Med 152:2111–2118, 1995.

183. Birrer P, McElvaney NG, Rudeberg A, et al: Protease-antiprotease imbalance in the lungs of children with cysticfibrosis. Am J Respir Crit Care Med 150:207–213, 1994.

184. Fick RB Jr, Robbins RA, Squier SU, et al: Complementactivation in cystic fibrosis respiratory fluids: In vivo and invitro generation of C5a and chemotactic activity. PediatrRes 20:1258–1268, 1986.

185. Bruce MC, Poncz L, Klinger JD, et al: Biochemical andpathologic evidence for proteolytic destruction of lungconnective tissue in cystic fibrosis. Am Rev Respir Dis132:529–535, 1985.

186. Davis PB: Clinical pathophysiology and manifestations oflung disease. In Cystic Fibrosis in Adults. Philadelphia:Lippincott-Raven, 1999, pp 45–67.

187. Wang EEL, Prober CG, Manson B, et al: Association ofrespiratory viral infections with pulmonary deterioration inpatients with cystic fibrosis. N Engl J Med311:1653–1658, 1984.

188. Petersen NT, Hoiby N, Mordhorst CH, et al: Respiratoryinfections in cystic fibrosis patients caused by virus,Chlamydia and Mycoplasma-possible synergism withPseudomonas aeruginosa. Acta Paediatr Scand70:623–628, 1981.

189. Conway SP, Simmonds EJ, Littlewood JM: Acute severedeterioration in cystic fibrosis associated with influenza Avirus infection. Thorax 47:112–114, 1992.

190. Winnie GB, Cowan RG: Association of Epstein-Barr virusinfection and pulmonary exacerbations in patients withcystic fibrosis. Pediatr Infect Dis 11:722–726, 1992.

191. Abman SH, Ogle JW, Butler-Simon N, et al: Role ofrespiratory syncytial virus in early hospitalizations forrespiratory distress of young infants with cystic fibrosis. JPediatr 113:826–830, 1988.

192. Ramsey BW, Gore EJ, Smith AL, et al: The effect ofrespiratory viral infections on patients with cystic fibrosis.Am J Dis Child 143:662–668, 1989.

193. Campbell PW III, Parker RA, Roberts BT, et al:Association of poor clinical status and heavy exposure totobacco smoke in patients with cystic fibrosis who arehomozygous for the F508 deletion. J Pediatr120:261–264, 1992.

194. Gilligan PH: Microbiology of cystic fibrosis lung disease.In Cystic Fibrosis in Adults. Philadelphia: Lippincott-Raven, 1999, pp 93–114.

195. Tummler B, Koopmann U, Grothues D, et al: Nosocomialacquisition of Pseudomonas aeruginosa by cystic fibrosispatients. J Clin Microbiol 29:1265–1267, 1991.

196. Costerton JW, Stewart PS, Greenberg EP: Bacterialbiofilms: A common cause of persistent infections. Science284:1318–1322, 1999.

197. May TB, Shinabarger D, Maharaj R, et al: Alginatesynthesis by Pseudomonas aeruginosa: A key pathogenicfactor in chronic pulmonary infections of cystic fibrosispatients. Clin Microbiol Rev 4:191–206, 1991.

198. Blackwood LL, Pennington JE: Influence of mucoidcoating on clearance of Pseudomonas aeruginosa fromlungs. Infect Immun 32:443–448, 1981.

199. May TB, Chakrabarty AM. Pseudomonas aeruginosa:Genes and enzymes of alginate synthesis. Trends Microbiol2:151–157, 1994.

200. Noone PG, Leigh MW, Sannuti A, et al: Primary ciliarydyskinesia: Diagnostic and phenotypic features. Am JRespir Crit Care Med 169:459–467, 2004.

201. LiPuma JJ, Dasen SE, Nielson DW, et al: Person-to-persontransmission of Pseudomonas cepacia between patients withcystic fibrosis. Lancet 336:1094–1096, 1990.

202. Saiman L, Siegel J: Infection control recommendations forpatients with cystic fibrosis: Microbiology, importantpathogens, and infection control practices to preventpatient-to-patient transmission. Infect Control HospEpidemiol 24:S6–52, 2003.

203. Isles A, MacLusky I, Corey M, et al: Pseudomonas cepaciainfection in cystic fibrosis: An emerging problem. J Pediatr104:206–210, 1984.

204. Fisher MC, LiPuma JJ, Dasen SE, et al: Source ofPseudomonas cepacia: Ribotyping of isolates from patientsand from the environment. J Pediatr 123:745–747, 1993.

205. Steinbach S, Sun L, Jiang RZ, et al: Transmissibility ofPseudomonas cepacia in clinic patients and lung-transplantrecipients with cystic fibrosis. N Engl J Med 331:981–987,1994.

206. Sun L, Jiang RZ, Steinbach S, et al: The emergence of ahighly transmissible lineage of cbl+ Pseudomonas(Burkholderia) cepacia causing CF centre epidemics inNorth America and Britain. Nat Med 1:661–666, 1995.

207. Vermis K, Coenye T, Mahenthiralingam E, et al:Evaluation of species-specific recA-based PCR tests forgenomovar level identification within the Burkholderiacepacia complex. J Med Microbiol 51:937–940, 2002.

208. Goldstein R, Sun L, Jiang RZ, et al: Structurally variantclasses of pilus appendage fibers coexpressed fromBurkholderia (Pseudomonas) cepacia. J Bacteriol177:1039–1052, 1995.

209. Lester LA, Egge A, Hubbard VS, di Sant’Agnese PA:Aspiration and lung abscess in cystic fibrosis. Am RevRespir Dis 127:786–787, 1983.

210. Ramsey BW, Wentz KR, Smith AL, et al: Predictive valueof oropharyngeal cultures for identifying lower airwaybacteria in cystic fibrosis patients. Am Rev Respir Dis144:331–337, 1991.

211. Hutcheson PS, Rejent AJ, Slavin RG: Variability inparameters of allergic bronchopulmonary aspergillosis inpatients with cystic fibrosis. J Allergy Clin Immunol88:390–394, 1991.

212. Stevens DA, Moss RB, Kurup VP, et al: Allergicbronchopulmonary aspergillosis in cystic fibrosis—state ofthe art: Cystic Fibrosis Foundation Consensus Conference.Clin Infect Dis 37(Suppl 3):S225–S264, 2003.

38.qxd 1/25/05 2:47 PM Page 1246

Page 31: Cystic Fibrosis

213. Kilby JM, Gilligan PH, Yankaskas JR, et al:Nontuberculous mycobacteria in adult patients with cysticfibrosis. Chest 102:70–75, 1992.

214. Aitken ML, Burke W, McDonald G, et al: Nontuberculousmycobacterial disease in adult cystic fibrosis patients. Chest103:1096–1099, 1993.

215. Olivier KN, Weber DJ, Wallace RJ Jr, et al: Nontuberculousmycobacteria in cystic fibrosis. I. Multicenter prevalencestudy of a potential pathogen in a susceptible population.Am J Respir Crit Care Med 167:835–840, 2003.

216. Olivier KN, Weber DJ, Lee JH, et al: Nontuberculousmycobacteria. II. Nested cohort study of impact on cysticfibrosis lung disease. Am J Respir Crit Care Med167:835–840, 2003.

217. Olivier KN, Yankaskas JR, Knowles MR: Nontuberculousmycobacterial pulmonary disease in cystic fibrosis. SeminRespir Infect 11:272–284, 1996.

218. Brasfield D, Hicks G, Soong S, Tiller R: The chestroentgenogram in cystic fibrosis: A new scoring system.Pediatrics 63:24–29, 1979.

219. Nathanson I, Conboy K, Murphy S, et al: Ultrafastcomputerized tomography of the chest in cystic fibrosis: Anew scoring system. Pediatr Pulmonol 11:81–86, 1991.

220. Robinson TE, Leung AN, Northway WH, et al:Composite spirometric-computed tomography outcomemeasure in early cystic fibrosis lung disease. Am J RespirCrit Care Med 168:588–593, 2003.

221. Bhalla M, Turcios N, Aponte V, et al: Cystic fibrosis:Scoring system with thin-section CT. Radiology179:783–788, 1991.

222. Kinsella D, Hamilton A, Goddard P, et al: The role ofmagnetic resonance imaging in cystic fibrosis. Clin Radiol44:23–26, 1991.

223. Fiel SB, Friedman AC, Caroline DF, et al: Magneticresonance imaging in young adults with cystic fibrosis.Chest 91:181–184, 1987.

224. Taussig LM, Landau LI, Marks MI: Respiratory system. In Cystic Fibrosis. New York: Thieme-Stratton, 1984, pp 115–174.

225. Levison H, Godfrey S: Pulmonary aspects of cystic fibrosis.In Cystic Fibrosis: Projections into the Future. New York:Stratton Intercontinental, 1976, pp 3–24.

226. Hanrahan JP, Brown RW, Carey VJ, et al: Passiverespiratory mechanics in healthy infants: Effects of growth,gender, and smoking. Am J Respir Crit Care Med154:670–680, 1996.

227. Feher A, Castile R, Kisling J, et al: Flow limitation innormal infants: A new method for forced expiratorymaneuvers from raised lung volumes. J Appl Physiol80:2019–2025, 1996.

228. Siassi B, Moss AJ, Dooley RR: Clinical recognition of corpulmonale in cystic fibrosis. J Pediatr 78:794–805, 1971.

229. Yankaskas JR, Egan TM, Mauro MA: Major complications.In Cystic Fibrosis in Adults. Philadelphia: Lippincott-Raven, 1999, pp 175–193.

230. Francis PW, Muller NL, Gurwitz D, et al: Hemoglobindesaturation: Its occurrence during sleep in patients withcystic fibrosis. Am J Dis Child 134:734–740, 1980.

231. Boat T: Comparison of lung function and survival patternsbetween cystic fibrosis and emphysema or chronicbronchitis patients. In Perspectives in Cystic Fibrosis.Mississauga, Ontario: Imperial Press, 1980, pp 236–245.

232. Hordvik NL, Koenig P, Morris D, et al: A longitudinalstudy of bronchodilator responsiveness in cystic fibrosis.Am Rev Respir Dis 131:889–893, 1985.

233. Godfrey S, Mearns M: Pulmonary function and response toexercise in cystic fibrosis. Arch Dis Child 46:144–152,1971.

234. Lands LC, Coates AL: Cardiopulmonary and skeletalmuscle function and their effects on exercise limitation. InCystic Fibrosis in Adults. Philadelphia: Lippincott-Raven,1999, pp 365–382.

235. Henke KG, Orenstein DM: Oxygen saturation duringexercise in cystic fibrosis. Am Rev Respir Dis 129:708–711,1984.

236. Asher MI, Pardy RL, Coates AL, et al: The effects ofinspiratory muscle training in patients with cystic fibrosis.Am Rev Respir Dis 126:855–859, 1982.

237. Orenstein DM, Franklin BA, Doershuk CF, et al: Exerciseconditioning and cardiopulmonary fitness in cystic fibrosis.Chest 80:392–398, 1981.

238. Regnis JA, Alison JA, Henke KG, et al: Changes in end-expiratory lung volume during exercise in cystic fibrosisrelate to severity of lung disease. Am Rev Respir Dis144:507–512, 1991.

239. Nixon PA, Orenstein DM, Kelsey SF, Doershuk CF: Theprognostic value of exercise testing in patients with cysticfibrosis. N Engl J Med 327:1785–1788, 1992.

240. O’Rawe A, McIntosh I, Dodge JA, et al: Increased energyexpenditure in cystic fibrosis is associated with specificmutations. Clin Sci 82:71–76, 1992.

241. Davies PSW, Bronstein MN: Energy expenditures of infantswith cystic fibrosis. Pediatr Pulmonol (Suppl 8):175–176,1992.

242. Gharib R, Allen RP, Joos HA, Bravo LR: Paranasal sinusesin cystic fibrosis: Incidence of roentgen abnormalities. AmJ Dis Child 108:499–502, 1964.

243. Stern RC, Jones K. Nasal and sinus disease. In CysticFibrosis in Adults. Philadelphia: Lippincott-Raven, 1999,pp 221-231.

244. Cepero R, Smith RJH, Catlin FI, et al: Cystic fibrosis: Anotolaryngologic perspective. Otolaryngol Head Neck Surg97:356–360, 1987.

245. Shwachman H, Kowalski M, Shaw K-T: Cystic fibrosis: Anew outlook; seventy patients above 25 years of age.Medicine (Baltimore) 56:129–149, 1977.

246. Jaffe BF, Strome M, Khaw K-T, Shwachman H: Nasalpolypectomy and sinus surgery for cystic fibrosis: A 10 yearreview. Otolaryngol Clin North Am 10:81–90, 1977.

247. Stern RC, Boat TF, Orenstein DM, et al: Treatment andprognosis of lobar and segmental atelectasis in cysticfibrosis. Am Rev Respir Dis 118:821–826, 1978.

248. Spector ML, Stern RC: Pneumothorax in cystic fibrosis: A 26-year experience. Ann Thorac Surg 47:204–207,1989.

249. Stern RC, Wood RE, Boat TF, et al: Treatment andprognosis of massive hemoptysis in cystic fibrosis. Am RevRespir Dis 117:825–828, 1978.

250. Brinson GM, Noone PG, Mauro MA, et al: Bronchialartery embolization for the treatment of hemoptysis inpatients with cystic fibrosis. Am J Respir Crit Care Med157:1951–1958, 1998.

251. Becker JW, Burke W, McDonald G, et al: Prevalence ofallergic bronchopulmonary aspergillosis and atopy in adultpatients with cystic fibrosis. Chest 109:1536–1540, 1996.

252. Schonheyder H, Jensen T, Hoiby N, et al: Frequency ofAspergillus fumigatus isolates and antibodies to Aspergillusantigens in cystic fibrosis. Acta Pathol Microbiol Scand93:105–112, 1985.

253. Egan TM: Thoracic surgery for patients with cystic fibrosis.In Treatment of the Hospitalized Patient with CysticFibrosis. New York: Marcel Dekker, 1998, pp 231–247.

254. Lemen RJ, Gates AJ, Mathe AA, et al: Relationshipsamong digital clubbing, disease severity, and serumprostaglandins F2alpha and E concentrations in cystic fibrosispatients. Am Rev Respir Dis 117:639–646, 1978.

124738 • Cystic Fibrosis

38.qxd 1/25/05 2:47 PM Page 1247

Page 32: Cystic Fibrosis

1248 Section J • OBSTRUCTIVE DISEASES

255. Noone PG, Bresnihan B: Rheumatic disease in cysticfibrosis. In Cystic Fibrosis in Adults. Philadelphia:Lippincott-Raven, 1999, pp 439–447.

256. Nixon PA, Orenstein DM, Curtis SE, Ross EA: Oxygensupplementation during exercise in cystic fibrosis. Am RevRespir Dis 142:807–811, 1990.

257. Tepper RS, Skatrud JB, Dempsey JA: Ventilation andoxygenation changes during sleep in cystic fibrosis. Chest84:388–393, 1983.

258. Stern RC, Borkat G, Hirschfeld SS, et al: Heart failure incystic fibrosis: Treatment and prognosis of cor pulmonalewith failure of the right side of the heart. Am J Dis Child134:267–272, 1980.

259. Garland JS, Chan YM, Kelly KJ, Rice TB: Outcome ofinfants with cystic fibrosis requiring mechanical ventilationfor respiratory failure. Chest 96:136–138, 1989.

260. Rosenstein BJ, Langbaum TS: Incidence of meconiumabnormalities in newborn infants with cystic fibrosis. Am JDis Child 134:72–73, 1980.

261. Di Sant’Agnese PA, Davis PB: Cystic fibrosis in adults: 75cases and a review of 232 cases in the literature. Am J Med66:121–132, 1979.

262. McCarthy VP, Mischler EH, Hubbard VS, et al:Appendiceal abscess in cystic fibrosis: A diagnosticchallenge. Gastroenterology 86:564–568, 1984.

263. Fletcher BD, Abramowsky CR: Contrast enemas in cysticfibrosis: Implications of appendiceal nonfilling. AJR Am JRoentgenol 137:323–326, 1981.

264. Taussig LM, Saldino RM, di Sant’Agnese PA: Radiographicabnormalities of the duodenum and small bowel in cysticfibrosis of the pancreas (mucoviscidosis). Radiology106:369–376, 1973.

265. Stern RC, Izant RJ Jr, Boat TF, et al: Treatment andprognosis of rectal prolapse in cystic fibrosis.Gastroenterology 82:707–710, 1982.

266. Durie PR, Forstner GG: The exocrine pancreas. In CysticFibrosis in Adults. Philadelphia: Lippincott-Raven, 1999,pp 261–287.

267. Kalnins D, Stewart C, Tullis E, Pencharz PB: Nutrition. InCystic Fibrosis in Adults. Philadelphia: Lippincott-Raven,1999, pp 289–307.

268. Robbins MK, Ontjes DA: Endocrine and renal disorders incystic fibrosis. In Cystic Fibrosis in Adults. Philadelphia:Lippincott-Raven, 1999, pp 383–418.

269. Henderson RC, Madsen CD: Bone density in children and adolescents with cystic fibrosis. J Pediatr 128:28–34,1996.

270. Salamoni F, Roulet M, Gudinchet F, et al: Bone mineralcontent in cystic fibrosis patients: Correlation with fat-freemass. Arch Dis Child 74:314–318, 1996.

271. Aris RM, Neuringer IP, Weiner MA, et al: Severeosteoporosis before and after lung transplantation. Chest109:1176–1183, 1996.

272. Shwachman H, Lebenthal E, Khaw KT: Recurrent acute pancreatitis in patients with cystic fibrosis withnormal pancreatic enzymes. Pediatrics 55:86–95, 1975.

273. Sharer N, Schwarz M, Malone G, et al: Mutations of thecystic fibrosis gene in patients with chronic pancreatitis. N Engl J Med 339:645–652, 1998.

274. Noone PG, Zhou Z, Silverman LM, et al: Cystic fibrosisgene mutations and pancreatitis risk: Relation to epithelialion transport and trypsin inhibitor gene mutations.Gastroenterology 121:1310–1319, 2001.

275. Sokol RJ, Durie PR: Recommendations for management of liver and biliary tract disease in cystic fibrosis; CysticFibrosis Foundation Hepatobiliary Disease ConsensusGroup. J Pediatr Gastroenterol Nutr 28:S1–S13, 1999.

276. Jebbink MC, Heijerman HG, Masclee AA, Lamers CB:Gallbladder disease in cystic fibrosis. Neth J Med41:123–126, 1992.

277. Nagel RA, Westaby D, Javaid A, et al: Liver disease andbile duct abnormalities in adults with cystic fibrosis. Lancet2:1422–1425, 1989.

278. O’Brien S, Keogan M, Casey M, et al: Biliarycomplications of cystic fibrosis. Gut 33:387–391, 1992.

279. Flume PA, Yankaskas JR. Reproductive issues. In CysticFibrosis in Adults. Philadelphia: Lippincott-Raven, 1999,pp 449–464.

280. Kopito LE, Kosasky HJ, Shwachman H: Water andelectrolytes in human cervical mucus from patients withcystic fibrosis. Fertil Steril 24:512–516, 1973.

281. FitzSimmons SC, Fitzpatrick S, Thompson B, et al: Alongitudinal study of the effects of pregnancy on 325women with cystic fibrosis. Pediatr Pulmonol (Suppl13):99–101, 1996.

282. Goss CH, Rubenfeld GD, Otto K, Aitken ML: The effectof pregnancy on survival in women with cystic fibrosis.Chest 124:1460–1468, 2003.

283. Michel SH, Mueller DH: Impact of lactation on womenwith cystic fibrosis and their infants: A review of five cases.J Am Diet Assoc 94:159–165, 1994.

284. Rosenstein BJ, Cutting GR: The diagnosis of cysticfibrosis: A consensus statement; Cystic Fibrosis FoundationConsensus Panel. J Pediatr 132:589–595, 1998.

285. Nussbaum E, Boat TF, Wood RE, Doershuk CF: Cysticfibrosis with acute hypoelectrolytemia and metabolicalkalosis in infancy. Am J Dis Child 133:965–966, 1979.

286. Stern RC: The diagnosis of cystic fibrosis. N Engl J Med336:487–491, 1997.

287. Knowles MR, Paradiso AM, Boucher RC: In vivo nasalpotential difference: Techniques and protocols for assessingefficacy of gene transfer in cystic fibrosis. Hum Gene Ther6:447–457, 1995.

287a. Standaert TA, Boitano L, Emerson J, et al: Standardizedprocedure for measurement of nasal potential difference: anoutcome measure in multicenter cystic fibrosis clinicaltrials. Pediatr Pulmonol 37:385–392, 2004.

288. Stern RC, Boat TF, Doershuk CF: Obstructiveazoospermia as a diagnostic criterion for the cystic fibrosissyndrome. Lancet 1:1401–1404, 1982.

289. Stern RC, Boat TF, Abramowsky CR, et al: Intermediate-range sweat chloride concentration and Pseudomonasbronchitis: A cystic fibrosis variant with preservation ofexocrine pancreatic function. JAMA 239:2676–2680,1978.

290. Sweat Testing: Sample Collection and QuantitativeAnalysis; Approved Guideline. Document C34-A. Wayne,PA: National Committee for Clinical Laboratory Standards,1994, No. 14.

291. LeGrys VA: Sweat testing for the diagnosis of cysticfibrosis: Practical considerations. J Pediatr 129:892–897,1996.

292. Davis PB, Del Rio S, Muntz JA, Dieckman L: Sweatchloride concentration in adults with pulmonary diseases.Am Rev Respir Dis 128:34–37, 1983.

293. Rock MJ, Mischler EH, Farrell PM, et al: Newbornscreening for cystic fibrosis is complicated by age-relateddecline in immunoreactive trypsinogen levels. Pediatrics85:1001–1007, 1990.

294. Reardon MC, Hammond KB, Accurso FJ, et al:Nutritional deficits exist before 2 months of age in someinfants with cystic fibrosis identified by screening test. J Pediatr 105:271–274, 1984.

295. Ramsey BW, Farrell PM, Pencharz P: Nutritionalassessment and management in cystic fibrosis: A consensus

38.qxd 1/25/05 2:47 PM Page 1248

Page 33: Cystic Fibrosis

report; The Consensus Committee. Am J Clin Nutr55:108–116, 1992.

296. Farrell PM, Kosorok MR, Laxova A, et al: Nutritionalbenefits of neonatal screening for cystic fibrosis; WisconsinCystic Fibrosis Neonatal Screening Study Group. N Engl JMed 337:963–969, 1997.

297. Waters DL, Wilcken B, Irwig L, et al: Clinical outcomes ofnewborn screening for cystic fibrosis. Arch Dis Child FetalNeonatal Ed 80:F1–F7, 1999.

297a. Grosse SD, Boyle CA, Botkin JR, et al and CDC:Newborn screening for cystic fibrosis: evaluation of benefitsand risks and recommendations for state newbornscreening programs. MMWR Recomm Rep 53(RR-13):1–36, 2004.

298. Genetic Testing for Cystic Fibrosis, April 14-16. NIHConsensus Statement Online 1997;15:1–20, 1997.

299. Yankaskas JR, Marshall BC, Sufian B, et al: Cystic fibrosisadult care: Consensus conference report. Chest125:S1–S39, 2004.

300. Desmond KJ, Schwenk WF, Thomas E, et al: Immediateand long-term effects of chest physiotherapy in patientswith cystic fibrosis. J Pediatr 103:538–542, 1983.

301. Thomas J, Cook DJ, Brooks D: Chest physical therapymanagement of patients with cystic fibrosis: A meta-analysis. Am J Respir Crit Care Med 151:846–850, 1995.

302. Mortensen J, Falk M, Groth S, Jensen C: The effects ofpostural drainage and positive expiratory pressurephysiotherapy on tracheobronchial clearance in cysticfibrosis. Chest 100:1350–1357, 1991.

303. Oberwaldner B, Evans JC, Zach MS: Forced expirationsagainst a variable resistance: A new chest physiotherapymethod in cystic fibrosis. Pediatr Pulmonol 2:358–367,1986.

304. Blomquist M, Freyschuss U, Wiman L-G, Strandvik B:Physical activity and self treatment in cystic fibrosis. ArchDis Child 61:362–367, 1986.

305. Schneiderman-Walker J, Pollock SL, Corey M, et al: Arandomized controlled trial of a 3-year home exerciseprogram in cystic fibrosis. J Pediatr 136:304–310, 2000.

306. Baldwin DR, Hill AL, Peckham DG, Knox AJ: Effect ofaddition of exercise to chest physiotherapy on sputumexpectoration and lung function in adults with cysticfibrosis. Respir Med 88:49–53, 1994.

307. Wood RE: Determinants of survival in cystic fibrosis. CysticFibrosis Club Abstr 26:69, 1985.

308. Pedersen SS, Jensen T, Hoiby N, et al: Management ofPseudomonas aeruginosa lung infection in Danish cysticfibrosis patients. Acta Paediatr Scand 76:955–961, 1987.

309. Smith A, Cohen M, Ramsey B: Pharmacotherapy. In CysticFibrosis in Adults. Philadelphia: Lippincott-Raven, 1999,pp 345–364.

310. Mendelman PM, Smith AL, Levy J, et al: Aminoglycosidepenetration, inactivation, and efficacy in cystic fibrosissputum. Am Rev Respir Dis 132:761–765, 1985.

311. LeBel M, Bergeron MG, Vallee F, et al: Pharmacokineticsand pharmacodynamics of ciprofloxacin in cystic fibrosispatients. Antimicrob Agents Chemother 30:260–266,1986.

312. Hodson ME, Penketh ARL, Batten JC: Aerosolcarbenicillin and gentamicin treatment of Pseudomonasaeruginosa infection in patients with cystic fibrosis. Lancet2:1137–1139, 1981.

313. Wall MA, Terry AB, Eisenberg J, et al: Inhaled antibioticsin cystic fibrosis. Lancet 1:1325, 1983.

314. Ramsey BW, Pepe MS, Quan JM, et al: Intermittentadministration of inhaled tobramycin in patients with cysticfibrosis; The Cystic Fibrosis Inhaled Tobramycin StudyGroup. N Engl J Med 340:23–30, 1999.

315. Equi A, Balfour-Lynn IM, Bush A, Rosenthal M: Longterm azithromycin in children with cystic fibrosis: Arandomised, placebo-controlled crossover trial. Lancet360:978–984, 2002.

316. Wolter J, Seeney S, Bell S, et al: Effect of long termtreatment with azithromycin on disease parameters in cysticfibrosis: A randomised trial. Thorax 57:212–216, 2002.

317. Saiman L, Marshall BC, Mayer-Hamblett N, et al:Azithromycin in patients with cystic fibrosis chronicallyinfected with Pseudomonas aeruginosa: A randomizedcontrolled trial. JAMA 290:1749–1756, 2003.

318. Fuchs HJ, Borowitz DS, Christiansen DH, et al: Effect ofaerosolized recombinant human DNase on exacerbations ofrespiratory symptoms and on pulmonary function inpatients with cystic fibrosis; The Pulmozyme Study Group.N Engl J Med 331:637–642, 1994.

319. Eng PA, Morton J, Douglass JA, et al: Short-term efficacyof ultrasonically nebulized hypertonic saline in cysticfibrosis. Pediatr Pulmonol 21:77–83, 1996.

320. Robinson M, Regnis JA, Bailey DL, et al: Effect ofhypertonic saline, amiloride, and cough on mucociliaryclearance in patients with cystic fibrosis. Am J Respir CritCare Med 153:1503–1509, 1996.

320a. Bye P, Elkins M, Robinson M, et al: Long-term inhalationof hypertonic saline in patients with cystic fibrosis—Arandomized controlled trial. Pediatr Pulmonol (Suppl27):329, 2004.

321. Sturgess J, Reid L: The effect of isoprenaline andpilocarpine on (a) bronchial mucus-secreting tissue and (b)pancreas, salivary glands, heart, thymus, liver and spleen.Br J Exp Pathol 54:388–403, 1973.

322. Shapiro GG, Bamman J, Kanerek P, Bierman CW: Theparadoxical effect of adrenergic and methylxanthine drugsin cystic fibrosis. Pediatrics 58:740–743, 1976.

323. Avital A, Sanchez I, Chernick V: Efficacy of salbutamoland ipratropium bromide in decreasing bronchialhyperreactivity in children with cystic fibrosis. PediatrPulmonol 13:34–37, 1992.

324. Sanchez I, Holbrow J, Chernick V: Acute bronchodilatorresponse to a combination of beta-adrenergic andanticholinergic agents in patients with cystic fibrosis. J Pediatr 120:486–488, 1992.

325. Weintraub SJ, Eschenbacher WL: The inhaledbronchodilators ipratropium bromide and metaproterenolin adults with CF. Chest 95:861–864, 1989.

326. Mulherin D, FitzGerald MX: Meconium ileus equivalent inassociation with nebulized ipratropium bromide in cysticfibrosis. Lancet 1:552, 1990.

327. Sivan Y, Arce P, Eigen H, et al: A double-blind,randomized study of sodium cromoglycate versus placeboin patients with cystic fibrosis and bronchial hyperreactivity.J Allergy Clin Immunol 85:649–654, 1990.

328. Auerbach HS, Williams M, Kirkpatrick JA, Colten HR:Alternate-day prednisone reduces morbidity and improvespulmonary function in cystic fibrosis. Lancet 2:686–688,1985.

329. Konstan MW, Byard PJ, Hoppel CL, Davis PB: Effect ofhigh-dose ibuprofen in patients with cystic fibrosis. N EnglJ Med 332:848–854, 1995.

330. Dolan TF Jr, Gibson LE: Complications of iodide therapyin patients with cystic fibrosis. J Pediatr 79:684–687, 1971.

330a. Takeyama K, Dabbagh K, Lee HM, et al: Epidermalgrowth factor system regulates mucin production inairways. Proc Natl Acad Sci U S A 96:3081–3086, 1999.

330b. Kohri K, Ueki IF, Shim JJ, et al: Pseudomonas aeruginosainduces MUC5AC production via epidermal growth factorreceptor. Eur Respir J 20:1263–1270, 2002.

124938 • Cystic Fibrosis

38.qxd 1/25/05 2:47 PM Page 1249

Page 34: Cystic Fibrosis

1250 Section J • OBSTRUCTIVE DISEASES

331. Knowles MR, Church NL, Waltner WE, et al: A pilot studyof aerosolized amiloride for the treatment of lung diseasein cystic fibrosis. N Engl J Med 322:1189–1194, 1990.

332. Hofmann T, Stutts MJ, Ziersch A, et al: Effects of topicallydelivered benzamil and amiloride on nasal potentialdifference in cystic fibrosis. Am J Respir Crit Care Med157:1844–1849, 1998.

333. Sims DE, Westfall JA, Kiorpes AL, Horne MM:Preservation of tracheal mucus by nonaqueous fixative.Biotech Histochem 66:173–180, 1991.

334. Johnson LG, Knowles MR: New therapeutic strategies forcystic fibrosis lung disease. In Cystic Fibrosis in Adults.Philadelphia: Lippincott-Raven, 1999, pp 233–258.

335. Smyth RL, van Velzen D, Smyth AR, et al: Strictures ofascending colon in cystic fibrosis and high strengthpancreatic enzymes. Lancet 343:85–86, 1994.

336. Borowitz DS, Grand RJ, Durie PR: Use of pancreaticenzyme supplements for patients with cystic fibrosis in thecontext of fibrosing colonopathy; Consensus Committee. J Pediatr 127:681–684, 1995.

337. Smyth RL, Ashby D, O’Hea U, et al: Fibrosingcolonopathy in cystic fibrosis: Results of a case-controlstudy. Lancet 346:1247–1251, 1995.

338. FitzSimmons SC, Burkhart GA, Borowitz D, et al: High-dose pancreatic-enzyme supplements and fibrosingcolonopathy in children with cystic fibrosis. N Engl J Med336:1283–1289, 1997.

339. Bertrand JM, Morin CL, Lasalle R, et al: Short-termclinical, nutritional, and functional effects of continuouselemental enteral alimentation in children with cysticfibrosis. J Pediatr 104:41–47, 1984.

340. Gaskin K, Gurevit D, Durie P, et al: Improved respiratoryprognosis in patients with cystic fibrosis with normal fatabsorption. J Pediatr 100:857–862, 1982.

341. Orenstein DM. Cystic Fibrosis: A Guide for Patient andFamily. Philadelphia: Lippincott-Raven, 2004, p 3.

342. Redding GJ, Restuccia R, Cotton EK, Brooks JG: Serialchanges in pulmonary functions in children hospitalizedwith cystic fibrosis. Am Rev Respir Dis 126:31–36, 1982.

343. Regelmann WE, Elliott GR, Warwick WJ, Clawson CC:Reduction of sputum Pseudomonas aeruginosa density byantibiotics improves lung function in cystic fibrosis morethan do bronchodilators and chest physiotherapy alone.Am Rev Respir Dis 141:914–921, 1990.

344. Donati MA, Guenette G, Auerbach H: Prospectivecontrolled study of home and hospital therapy of cysticfibrosis pulmonary disease. J Pediatr 111:28–33, 1987.

345. Pond MN, Newport M, Joanes D, Conway SP: Homeversus hospital intravenous antibiotic therapy in thetreatment of young adults with cystic fibrosis. Eur Respir J7:1640–1644, 1994.

346. Winnie GB, Cowan RG, Wade NA: Intravenous immuneglobulin treatment of pulmonary exacerbations in cysticfibrosis. J Pediatr 114:309–314, 1989.

347. Braunstein MS, Fleet WF: Failure of bronchopulmonarylavage in cystic fibrosis. Chest 66:96–99, 1974.

348. Spier S, Rivlin J, Hughes D, Levison H: The effect ofoxygen on sleep, blood gases, and ventilation in cysticfibrosis. Am Rev Respir Dis 129:712–718, 1984.

349. Coates AL: Oxygen therapy, exercise, and cystic fibrosis.Chest 101:2–4, 1992.

350. Zinman R, Corey M, Coates AL, et al: Nocturnal homeoxygen in the treatment of hypoxemic cystic fibrosispatients. J Pediatr 114:368–377, 1989.

351. Fraser KL, Tullis DE, Sasson Z, et al: Pulmonaryhypertension and cardiac function in adult cystic fibrosis:Role of hypoxemia. Chest 115:1321–1328, 1999.

352. Continuous or nocturnal oxygen therapy in hypoxemicchronic obstructive lung disease: A clinical trial; NocturnalOxygen Therapy Trial Group. Ann Intern Med93:391–398, 1980.

353. Schidlow DV, Taussig LM, Knowles MR: Cystic FibrosisFoundation consensus conference report on pulmonarycomplications of cystic fibrosis. Pediatr Pulmonol15:187–198, 1993.

354. Davis PB, di Sant’Agnese PA: Assisted ventilation forpatients with cystic fibrosis. JAMA 239:1851–1854, 1978.

355. Hodson ME, Madden BP, Steven MH, et al: Non-invasivemechanical ventilation for cystic fibrosis patients: Apotential bridge to transplantation. Eur Respir J4:524–527, 1991.

356. Flume PA, Egan TM, Westerman JH, et al: Lungtransplantation for mechanically ventilated patients. J HeartLung Transplant 13:15–21, 1994.

357. Sood N, Paradowski LJ, Yankaskas JR: Outcomes ofintensive care unit care in adults with cystic fibrosis. Am JRespir Crit Care Med 163:335–338, 2001.

358. Knutsen AP, Slavin RG: Allergic bronchopulmonarymycosis complicating cystic fibrosis. Semin Respir Infect7:179–192, 1992.

359. Imbeault B, Cormier Y: Usefulness of inhaled high-dosecorticosteroids in allergic bronchopulmonary aspergillosis.Chest 103:1614–1617, 1993.

360. Mannes GP, van der Heide S, van Aalderen WM, GerritsenJ: Itraconazole and allergic bronchopulmonary aspergillosisin twin brothers with cystic fibrosis. Lancet 341:492, 1993.

361. Fellows KE, Khaw KT, Schuster S, Shwachman H:Bronchial artery embolization in cystic fibrosis: Techniqueand long-term results. J Pediatr 95:959–963, 1979.

362. Hofmann AF, Popper H: Ursodeoxycholic acid for primarybiliary cirrhosis. Lancet 2:398–399, 1987.

363. Galabert C, Montet JC, Lengrand D, et al: Effects ofursodeoxycholic acid on liver function in patients withcystic fibrosis and chronic cholestasis. J Pediatr121:138–141, 1992.

364. Colombo C, Battezzati PM, Podda M, et al:Ursodeoxycholic acid for liver disease associated with cysticfibrosis: A double-blind multicenter trial; The ItalianGroup for the Study of Ursodeoxycholic Acid in CysticFibrosis. Hepatology 23:1484–1490, 1996.

365. Stern RC, Stevens DP, Boat TF, et al: Symptomatic hepaticdisease in cystic fibrosis: Incidence, course, and outcome ofportal systemic shunting. Gastroenterology 70:645–649,1976.

366. Rossle M, Haag K, Ochs A, et al: The transjugularintrahepatic portosystemic stent-shunt procedure forvariceal bleeding. N Engl J Med 330:165–171, 1994.

367. Berger KJ, Schreiber RA, Tchervenkov J, et al:Decompression of portal hypertension in a child with cysticfibrosis after transjugular intrahepatic portosystemic shuntplacement. J Pediatr Gastroenterol Nutr 19:322–325,1994.

368. Mieles LA, Orenstein D, Teperman L, et al: Livertransplantation in cystic fibrosis. Lancet 1:1073, 1989.

369. Noble-Jamieson G, Barnes N, Jamieson N, et al: Livertransplantation for hepatic cirrhosis in cystic fibrosis. J RSoc Med 89(Suppl 27):31–37, 1996.

370. Rosenbloom AL: Diabetes mellitus in cystic fibrosis.Pediatr Pulmonol (Suppl 6):209–210, 1991.

371. Rodman HM, Doershuk CF, Roland JM: The interactionof 2 diseases: Diabetes mellitus and cystic fibrosis.Medicine (Baltimore) 65:389–397, 1986.

372. Finkelstein SM, Wielinski CL, Elliot GR, et al: Diabetesmellitus associated with cystic fibrosis. J Pediatr112:373–377, 1988.

38.qxd 1/25/05 2:47 PM Page 1250

Page 35: Cystic Fibrosis

373. Lass JH, Spurney RV, Dutt RM, et al: A morphologic andfluorophotometric analysis of the corneal endothelium intype I diabetes mellitus and cystic fibrosis. Am JOphthalmol 100:783–788, 1985.

374. Allen JL: Progressive nephropathy in a patient with cysticfibrosis and diabetes. N Engl J Med 315:764, 1986.

375. Duplechain JK, White JA, Miller RH: Pediatric sinusitis:The role of endoscopic sinus surgery in cystic fibrosis andother forms of sinonasal disease. Arch Otolaryngol HeadNeck Surg 117:422–426, 1991.

376. Cuyler JP: Follow-up of endoscopic sinus surgery onchildren with cystic fibrosis. Arch Otolaryngol Head NeckSurg 118:505–506, 1992.

377. Stern RC, Rothstein FC, Doershuk CF: Treatment and prognosis of symptomatic gallbladder disease in patients withcystic fibrosis. J Pediatr Gastroenterol Nutr 5:35–40, 1986.

378. Smith MB, Hardin WD Jr, Dressel DA, et al: Predictingoutcome following pulmonary resection in cystic fibrosispatients. J Pediatr Surg 26:655–659, 1991.

379. Yankaskas JR, Mallory GB Jr: Lung transplantation incystic fibrosis: Consensus conference statement. Chest113:217–226, 1998.

380. Barr ML, Schenkel FA, Cohen RG, et al: Recipient anddonor outcomes in living related and unrelated lobartransplantation. Transplant Proc 30:2261–2263, 1998.

381. Stern RC, Boat TF, Doershuk CF, et al: Cystic fibrosisdiagnosed after age 13: Twenty-five teenage and adultpatients including three asymptomatic men. Ann InternMed 87:188–191, 1977.

382. FitzSimmons SC: Cystic Fibrosis Foundation PatientRegistry; 1997 Annual Data Report. Bethesda, MD: CysticFibrosis Foundation, 1998.

383. Shwachman H, Kulczycki LL: Long-term study of onehundred five patients with cystic fibrosis: Study made overa five- to fourteen-year period. J Dis Child 96:6–15, 1958.

384. Taussig LM, Kattwinkel J, Friedewald WT, di Sant’AgnesePA: A new prognostic score and clinical evaluation systemfor cystic fibrosis. J Pediatr 82:380–390, 1973.

385. Fink RJ, Doershuk CF, Tucker AS, et al: Pulmonaryfunction and morbidity in 40 adult patients with cysticfibrosis. Chest 74:643–647, 1978.

386. Corey M, Levison H, Crozier D: Five- to seven-year courseof pulmonary function in cystic fibrosis. Am Rev Respir Dis114:1085–1092, 1976.

387. Wood RE: Prognosis. In Cystic Fibrosis. New York:Thieme-Stratton, 1984, pp 434–460.

388. Orenstein DM, Boat TF, Stern RC, et al: The effect ofearly diagnosis and treatment in cystic fibrosis: A seven-yearstudy of 16 sibling pairs. Am J Dis Child 131:973–975,1977.

389. Katz JN, Horwitz RI, Dolan TF, Shapiro ED: Clinicalfeatures as predictors of functional status in children withcystic fibrosis. J Pediatr 108:352–358, 1986.

390. Hamosh A, Corey M: Correlation between genotype andphenotype in patients with cystic fibrosis; The CysticFibrosis Genotype-Phenotype Consortium. N Engl J Med329:1308–1313, 1993.

391. Knoke JD, Stern RC, Doershuk CF, et al: Cystic fibrosis:The prognosis for five-year survival. Pediatr Res12:676–679, 1978.

392. Reisman J, Corey M, Canny G, Levison H: Diabetesmellitus in patients with cystic fibrosis: Effect on survival.Pediatrics 86:374–377, 1990.

393. Hill DJ, Martin AJ, Davidson GP, Smith GS: Survival ofcystic fibrosis patients in South Australia: Evidence thatcystic fibrosis centre care leads to better survival. Med JAust 143:230–232, 1985.

394. Denning CR, Gluckson MM: Psychosocial aspects of cysticfibrosis. In Cystic Fibrosis. New York: Thieme-Stratton,1984, pp 461–492.

395. Lewiston NJ: Psychosocial impact of cystic fibrosis. SeminRespir Med 6:321–332, 1985.

396. Yankaskas JR, Fernald GW: Adult social issues. In CysticFibrosis in Adults. Philadelphia: Lippincott-Raven, 1999,pp 465–476.

397. Levine SB, Stern RC: Sexual function in cystic fibrosis:Relationship to overall health status and pulmonary disease severity in 30 married patients. Chest 81:422–428,1982.

398. Hull J, Skinner W, Robertson C, Phelan P: Elementalcontent of airway surface liquid from infants with cysticfibrosis. Am J Respir Crit Care Med 157:10–14, 1998.

125138 • Cystic Fibrosis

38.qxd 1/25/05 2:47 PM Page 1251